Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 65
Filtrar
Mais filtros

Base de dados
Tipo de documento
País de afiliação
Intervalo de ano de publicação
1.
Int J Mol Sci ; 21(19)2020 Sep 25.
Artigo em Inglês | MEDLINE | ID: mdl-32992769

RESUMO

Stimulator of interferon genes (STING)-mediated type-I interferon signaling is a well characterized instigator of the innate immune response following bacterial or viral infections in the periphery. Emerging evidence has recently linked STING to various neuropathological conditions, however, both protective and deleterious effects of the pathway have been reported. Elevated oxidative stress, such as neuroinflammation, is a feature of a number of neuropathologies, therefore, this study investigated the role of the STING pathway in cell death induced by elevated oxidative stress. Here, we report that the H2O2-induced activation of the STING pathway is protective against cell death in wildtype (WT) MEFSV40 cells as compared to STING-/- MEF SV40 cells. This protective effect of STING can be attributed, in part, to an increase in autophagy flux with an increased LC3II/I ratio identified in H2O2-treated WT cells as compared to STING-/- cells. STING-/- cells also exhibited impaired autophagic flux as indicated by p62, LC3-II and LAMP2 accumulation following H2O2 treatment, suggestive of an impairment at the autophagosome-lysosomal fusion step. This indicates a previously unrecognized role for STING in maintaining efficient autophagy flux and protecting against H2O2-induced cell death. This finding supports a multifaceted role for the STING pathway in the underlying cellular mechanisms contributing to the pathogenesis of neurological disorders.


Assuntos
Autofagia , Morte Celular , Proteínas de Membrana/fisiologia , Animais , Linhagem Celular , Técnicas de Inativação de Genes , Peróxido de Hidrogênio/toxicidade , Camundongos , Estresse Oxidativo , Espécies Reativas de Oxigênio/metabolismo
2.
Brain Behav Immun ; 79: 75-90, 2019 07.
Artigo em Inglês | MEDLINE | ID: mdl-31029798

RESUMO

Autism Spectrum Disorder (ASD) is a neurodevelopmental disorder characterised by deficits in social communication and restricted or repetitive behaviours. The clinical presentation of ASD is highly variable and diagnosis is based on the presence of impaired social communication and repetitive and/or restricted behaviours. Although the precise pathophysiologies underlying ASD are unclear, growing evidence supports a role for dysregulated neuroinflammation. The potential involvement of microglia and astrocytes reactive to inflammatory stimuli in ASD has generated much interest due to their varied roles including in mounting an immune response and regulating synaptic function. Increased numbers of reactive microglial and astrocytes in both ASD postmortem tissue and animal models have been reported. Whether dysregulation of glial subtypes exacerbates alterations in neural connectivity in the brain of autistic patients is not well explored. A role for the gut-brain axis involving microbial-immune-neuronal cross talk is also a growing area of neuroinflammation research. Greater understanding of these interactions under patho/physiological conditions and the identification of consistent immune profile abnormalities can potentially lead to more reliable diagnostic measures and treatments in ASD.


Assuntos
Transtorno do Espectro Autista/imunologia , Inflamação/imunologia , Neuroimunomodulação/imunologia , Animais , Astrócitos/imunologia , Astrócitos/fisiologia , Transtorno do Espectro Autista/fisiopatologia , Encéfalo/imunologia , Encéfalo/fisiopatologia , Modelos Animais de Doenças , Microbioma Gastrointestinal/imunologia , Humanos , Inflamação/metabolismo , Microglia/imunologia , Microglia/fisiologia , Neuroimunomodulação/fisiologia , Neurônios/imunologia , Neurônios/metabolismo
3.
J Neuroinflammation ; 15(1): 323, 2018 Nov 21.
Artigo em Inglês | MEDLINE | ID: mdl-30463579

RESUMO

BACKGROUND: Traumatic brain injury (TBI) represents a major cause of disability and death worldwide with sustained neuroinflammation and autophagy dysfunction contributing to the cellular damage. Stimulator of interferon genes (STING)-induced type-I interferon (IFN) signalling is known to be essential in mounting the innate immune response against infections and cell injury in the periphery, but its role in the CNS remains unclear. We previously identified the type-I IFN pathway as a key mediator of neuroinflammation and neuronal cell death in TBI. However, the modulation of the type-I IFN and neuroinflammatory responses by STING and its contribution to autophagy and neuronal cell death after TBI has not been explored. METHODS: C57BL/6J wild-type (WT) and STING-/- mice (8-10-week-old males) were subjected to controlled cortical impact (CCI) surgery and brains analysed by QPCR, Western blot and immunohistochemical analyses at 2 h or 24 h. STING expression was also analysed by QPCR in post-mortem human brain samples. RESULTS: A significant upregulation in STING expression was identified in late trauma human brain samples that was confirmed in wild-type mice at 2 h and 24 h after CCI. This correlated with an elevated pro-inflammatory cytokine profile with increased TNF-α, IL-6, IL-1ß and type-I IFN (IFN-α and IFN-ß) levels. This expression was suppressed in the STING-/- mice with a smaller lesion volume in the knockout animals at 24 h post CCI. Wild-type mice also displayed increased levels of autophagy markers, LC3-II, p62 and LAMP2 after TBI; however, STING-/- mice showed reduced LAMP2 expression suggesting a role for STING in driving dysfunctional autophagy after TBI. CONCLUSION: Our data implicates a detrimental role for STING in mediating the TBI-induced neuroinflammatory response and autophagy dysfunction, potentially identifying a new therapeutic target for reducing cellular damage in TBI.


Assuntos
Lesões Encefálicas Traumáticas/complicações , Encéfalo/metabolismo , Encefalite/etiologia , Encefalite/metabolismo , Regulação da Expressão Gênica/genética , Proteínas de Membrana/metabolismo , Animais , Autofagia/genética , Encéfalo/patologia , Lesões Encefálicas Traumáticas/patologia , Proteínas de Ligação ao Cálcio/metabolismo , Citocinas/genética , Citocinas/metabolismo , Modelos Animais de Doenças , Lateralidade Funcional , Proteína Glial Fibrilar Ácida/metabolismo , Fator 3-alfa Nuclear de Hepatócito/metabolismo , Humanos , Proteína 2 de Membrana Associada ao Lisossomo/genética , Proteína 2 de Membrana Associada ao Lisossomo/metabolismo , Masculino , Proteínas de Membrana/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Proteínas dos Microfilamentos/metabolismo , Proteínas do Tecido Nervoso/metabolismo
4.
J Neural Transm (Vienna) ; 125(5): 797-807, 2018 05.
Artigo em Inglês | MEDLINE | ID: mdl-28676934

RESUMO

Past research in Alzheimer's disease (AD) has largely been driven by the amyloid hypothesis; the accompanying neuroinflammation seen in AD has been assumed to be consequential and not disease modifying or causative. However, recent data from both clinical and preclinical studies have established that the immune-driven neuroinflammation contributes to AD pathology. Key evidence for the involvement of neuroinflammation in AD includes enhanced microglial and astroglial activation in the brains of AD patients, increased pro-inflammatory cytokine burden in AD brains, and epidemiological evidence that chronic non-steroidal anti-inflammatory drug use prior to disease onset leads to a lower incidence of AD. Identifying critical mediators controlling this neuroinflammation will prove beneficial in developing anti-inflammatory therapies for the treatment of AD. The type-I interferons (IFNs) are pleiotropic cytokines that control pro-inflammatory cytokine secretion and are master regulators of the innate immune response that impact on disorders of the central nervous system. This review provides evidence that the type-I IFNs play a critical role in the exacerbation of neuroinflammation and actively contribute to the progression of AD.


Assuntos
Doença de Alzheimer/imunologia , Inflamação/imunologia , Interferon Tipo I/imunologia , Degeneração Neural/imunologia , Doença de Alzheimer/patologia , Animais , Encéfalo/imunologia , Encéfalo/patologia , Humanos , Inflamação/patologia , Degeneração Neural/patologia
5.
J Neurochem ; 141(1): 75-85, 2017 04.
Artigo em Inglês | MEDLINE | ID: mdl-28029694

RESUMO

Evidence from post-mortem human brains, animal studies and cell culture models has implicated neuroinflammation in the aetiology of chronic neuropathologies including Alzheimer's and Parkinson's diseases. Although the neuroinflammatory response is considered detrimental in contributing to these pathologies, the underlying mechanisms are still not well understood. The type-I interferons (IFNs) have been well characterised in the periphery and are known to initiate/modulate the immune response. Recently, they have been implicated in ageing and we have also demonstrated increased type-I IFN expression in post-mortem human Alzheimer's and Parkinson's disease brains. We hypothesise that the type-I IFNs are key drivers of the damaging, self-perpetuating pro-inflammatory response that contributes to these chronic neuropathologies. In support of this, we have recently confirmed in models of Alzheimer's and Parkinson's disease that mice lacking the type-I IFN receptor (IFNAR1), display an attenuated neuroinflammatory response with subsequent neuroprotection. To further investigate type-I IFN-mediated neuroinflammation and the specific CNS cell types involved, this study treated primary cultured wild-type and IFNAR1-/- neurons or mixed glia with the mitochondrial complex I inhibitor, rotenone. Wild-type neurons and glia treated with 3 nM and 25 nM rotenone, respectively, exhibited a pro-inflammatory response, including increased type-I IFN expression that was attenuated in cells lacking IFNAR1. Reduced type-I IFN signalling in IFNAR1-/- neurons also conferred protection against caspase-3-mediated rotenone-induced cell death. Further, this reduced pro-inflammatory response in the IFNAR1-/- glia subsequently diminished their neurotoxic effects to wild-type neurons. In support of this, we confirmed that therapeutically targeting the type-I IFN glial response to rotenone through a specific IFNAR1 blocking monoclonal antibody was neuroprotective. Our data has confirmed that both neurons and glia contribute to the pro-inflammatory response induced by rotenone with attenuation of this response beneficial in reducing neuronal cell death. Read the Editorial Comment for this article on page 9.


Assuntos
Imunidade Inata/fisiologia , Mediadores da Inflamação/metabolismo , Interferon Tipo I/fisiologia , Síndromes Neurotóxicas/metabolismo , Rotenona/toxicidade , Animais , Anticorpos Monoclonais/administração & dosagem , Sobrevivência Celular/efeitos dos fármacos , Sobrevivência Celular/fisiologia , Células Cultivadas , Técnicas de Cocultura , Relação Dose-Resposta a Droga , Feminino , Imunidade Inata/efeitos dos fármacos , Inflamação/tratamento farmacológico , Inflamação/metabolismo , Inflamação/patologia , Mediadores da Inflamação/antagonistas & inibidores , Interferon Tipo I/antagonistas & inibidores , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Síndromes Neurotóxicas/tratamento farmacológico , Síndromes Neurotóxicas/patologia , Gravidez
6.
Glia ; 64(9): 1590-604, 2016 09.
Artigo em Inglês | MEDLINE | ID: mdl-27404846

RESUMO

Type-1 interferons (IFNs) are pleiotropic cytokines with a critical role in the initiation and regulation of the pro-inflammatory response. However, the contribution of the type-1 IFNs to CNS disorders, specifically chronic neuropathologies such as Parkinson's disease is still unknown. Here, we report increased type-1 IFN signaling in both post mortem human Parkinson's disease samples and in the 1-methyl-4-phenyl-1, 2, 3, 6-tetrahydropyridine (MPTP) mouse model. In response to MPTP, mice lacking the type-1 IFN receptor (IFNAR1(-/-) ) displayed decreased type-1 IFN signaling, an attenuated pro-inflammatory response and reduced loss of dopaminergic neurons. The neuroprotective potential of targeting the type-1 IFN pathway was confirmed by reduced neuroinflammation and DA cell death in mice treated with a blocking monoclonal IFNAR1 (MAR-1) antibody. The MPTP/MAR-1 treated mice also displayed increased striatal dopamine levels and improved behavioural outcomes compared to their MPTP/IgG controls. These data, implicate for the first time, a deleterious role for the type-1 IFNs as key modulators of the early neuroinflammatory response and therefore the neuronal cell death in Parkinson's disease. GLIA 2016;64:1590-1604.


Assuntos
Neurônios Dopaminérgicos/metabolismo , Interferon Tipo I/genética , Doença de Parkinson/genética , Animais , Morte Celular/genética , Citocinas/metabolismo , Modelos Animais de Doenças , Dopamina/metabolismo , Inflamação/genética , Interferon Tipo I/metabolismo , Camundongos Endogâmicos C57BL , Camundongos Knockout , Microglia/metabolismo , Doença de Parkinson/patologia , Substância Negra/patologia
7.
J Neurochem ; 136(3): 457-74, 2016 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-26509334

RESUMO

Alzheimer's disease (AD) is a progressive neurodegenerative disease and the most common cause of dementia. Deposition of amyloid-ß (Aß) remains a hallmark feature of the disease, yet the precise mechanism(s) by which this peptide induces neurotoxicity remain unknown. Neuroinflammation has long been implicated in AD pathology, yet its contribution to disease progression is still not understood. Recent evidence suggests that various Aß complexes interact with microglial and astrocytic expressed pattern recognition receptors that initiate innate immunity. This process involves secretion of pro-inflammatory cytokines, chemokines and generation of reactive oxygen species that, in excess, drive a dysregulated immune response that contributes to neurodegeneration. The mechanisms by which a neuroinflammatory response can influence Aß production, aggregation and eventual clearance are now becoming key areas where future therapeutic intervention may slow progression of AD. This review will focus on evidence supporting the combined neuroinflammatory-amyloid hypothesis for pathogenesis of AD, describing the key cell types, pathways and mediators involved. Alzheimer's disease (AD) is a progressive neurodegenerative disorder and the leading cause of dementia worldwide. Deposition of intracellular plaques containing amyloid-beta (Aß) is a hallmark proteinopathy of the disease yet the precise mechanisms by which this peptide induces neurotoxicity remains unknown. A neuroinflammatory response involving polarized microglial activity, enhanced astrocyte reactivity and elevated pro-inflammatory cytokine and chemokine load has long been implicated in AD and proposed to facilitate neurodegeneration. In this issue we discuss key receptor systems of innate immunity that detect Aß, drive pro-inflammatory cytokine and chemokine production and influence Aß aggregation and clearance. Evidence summarized in this review supports the combined neuroinflammatory-amyloid hypothesis for pathogenesis of AD and highlights the potential of immunomodulatory agents as potential future therapies for AD patients.


Assuntos
Doença de Alzheimer , Proteínas Amiloidogênicas/metabolismo , Citocinas/metabolismo , Encefalite/complicações , Imunidade Inata , Doença de Alzheimer/complicações , Doença de Alzheimer/metabolismo , Doença de Alzheimer/patologia , Animais , Humanos , Neuroglia/patologia
8.
Clin Sci (Lond) ; 130(13): 1039-50, 2016 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-27215677

RESUMO

Chronic obstructive pulmonary disease (COPD) is characterized by progressive airflow limitation and loss of lung function, and is currently the third largest cause of death in the world. It is now well established that cardiovascular-related comorbidities such as stroke contribute to morbidity and mortality in COPD. The mechanisms linking COPD and stroke remain to be fully defined but are likely to be interconnected. The association between COPD and stroke may be largely dependent on shared risk factors such as aging and smoking, or the association of COPD with traditional stroke risk factors. In addition, we propose that COPD-related systemic inflammation and oxidative stress may play important roles by promoting cerebral vascular dysfunction and platelet hyperactivity. In this review, we briefly discuss the pathogenesis of COPD, acute exacerbations of COPD (AECOPD) and cardiovascular comorbidities associated with COPD, in particular stroke. We also highlight and discuss the potential mechanisms underpinning the link between COPD and stroke, with a particular focus on the roles of systemic inflammation and oxidative stress.


Assuntos
Inflamação/complicações , Estresse Oxidativo/fisiologia , Doença Pulmonar Obstrutiva Crônica/complicações , Doença Pulmonar Obstrutiva Crônica/metabolismo , Fumar/metabolismo , Acidente Vascular Cerebral/metabolismo , Animais , Humanos , Inflamação/fisiopatologia , Doença Pulmonar Obstrutiva Crônica/fisiopatologia , Fatores de Risco , Fumar/fisiopatologia , Acidente Vascular Cerebral/complicações , Acidente Vascular Cerebral/fisiopatologia
9.
J Neuroinflammation ; 11: 37, 2014 Feb 27.
Artigo em Inglês | MEDLINE | ID: mdl-24576351

RESUMO

BACKGROUND: Lysophosphatidic acid (LPA) is a bioactive phospholipid with a potentially causative role in neurotrauma. Blocking LPA signaling with the LPA-directed monoclonal antibody B3/Lpathomab is neuroprotective in the mouse spinal cord following injury. FINDINGS: Here we investigated the use of this agent in treatment of secondary brain damage consequent to traumatic brain injury (TBI). LPA was elevated in cerebrospinal fluid (CSF) of patients with TBI compared to controls. LPA levels were also elevated in a mouse controlled cortical impact (CCI) model of TBI and B3 significantly reduced lesion volume by both histological and MRI assessments. Diminished tissue damage coincided with lower brain IL-6 levels and improvement in functional outcomes. CONCLUSIONS: This study presents a novel therapeutic approach for the treatment of TBI by blocking extracellular LPA signaling to minimize secondary brain damage and neurological dysfunction.


Assuntos
Lesões Encefálicas/tratamento farmacológico , Lesões Encefálicas/imunologia , Imunoglobulina G/uso terapêutico , Fatores Imunológicos/uso terapêutico , Lisofosfolipídeos/imunologia , Adulto , Idoso de 80 Anos ou mais , Animais , Lesões Encefálicas/líquido cefalorraquidiano , Citocinas/metabolismo , Modelos Animais de Doenças , Feminino , Escala de Coma de Glasgow , Humanos , Lisofosfolipídeos/líquido cefalorraquidiano , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Pessoa de Meia-Idade , Método Simples-Cego , Adulto Jovem
10.
Br J Pharmacol ; 2024 May 06.
Artigo em Inglês | MEDLINE | ID: mdl-38710660

RESUMO

BACKGROUND AND PURPOSE: Traumatic brain injury (TBI) remains a major public health concern worldwide with unmet effective treatment. Stimulator of interferon genes (STING) and its downstream type-I interferon (IFN) signalling are now appreciated to be involved in TBI pathogenesis. Compelling evidence have shown that STING and type-I IFNs are key in mediating the detrimental neuroinflammatory response after TBI. Therefore, pharmacological inhibition of STING presents a viable therapeutic opportunity in combating the detrimental neuroinflammatory response after TBI. EXPERIMENTAL APPROACH: This study investigated the neuroprotective effects of the small-molecule STING inhibitor n-(4-iodophenyl)-5-nitrofuran-2-carboxamide (C-176) in the controlled cortical impact mouse model of TBI in 10- to 12-week-old male mice. Thirty minutes post-controlled cortical impact surgery, a single 750-nmol dose of C-176 or saline (vehicle) was administered intravenously. Analysis was conducted 2 h and 24 h post-TBI. KEY RESULTS: Mice administered C-176 had significantly smaller cortical lesion area when compared to vehicle-treated mice 24 h post-TBI. Quantitative temporal gait analysis conducted using DigiGait™ showed C-176 administration attenuated TBI-induced impairments in gait symmetry, stride frequency and forelimb stance width. C-176-treated mice displayed a significant reduction in striatal gene expression of pro-inflammatory cytokines Tnf-α, Il-1ß and Cxcl10 compared to their vehicle-treated counterparts 2 h post-TBI. CONCLUSION AND IMPLICATIONS: This study demonstrates the neuroprotective activity of C-176 in ameliorating acute neuroinflammation and preventing white matter neurodegeneration post-TBI. This study highlights the therapeutic potential of small-molecule inhibitors targeting STING for the treatment of trauma-induced inflammation and neuroprotective potential.

11.
Am J Respir Cell Mol Biol ; 48(1): 17-26, 2013 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-23002098

RESUMO

Oxidative stress caused by excessive reactive oxygen species production is implicated in influenza A virus-induced lung disease. Glutathione peroxidase (GPx)-1 is an antioxidant enzyme that may protect lungs from such damage. The objective of this study was to determine if GPx-1 protects the lung against influenza A virus-induced lung inflammation in vivo. Male wild-type (WT) or GPx-1(-/-) mice were inoculated with HKx31 (H3N2, 1 × 10(4) plaque-forming units), and bronchoalveolar lavage fluid (BALF)/lung compartments were analyzed on Days 3 and 7 after infection for inflammatory marker expression, histology, and viral titer. WT mice infected with HKx31 had significantly more BALF total cells, macrophages, neutrophils, and lymphocytes at Days 3 and 7 compared with naive WT animals (n = 5-8; P < 0.05). However, infected GPx-1(-/-) mice had significantly more BALF inflammation, which included more total cells, macrophages, and neutrophils, compared with WT mice, and this was abolished by treatment with the GPx mimetic ebselen. BALF inflammation persisted in GPx-1(-/-) mice on Day 10 after infection, and GPx-1(-/-) mice had significantly more influenza-specific CD8(+) T cells in spleen compared with WT mice (n = 3-4; P < 0.05). Infected GPx-1(-/-) mice had greater peribronchial and parenchymal inflammation than WT mice, and viral titer was significantly reduced in GPx-1(-/-) mice at Day 3 (n = 5; P < 0.05). Gene expression analysis revealed that infected GPx-1(-/-) mice had higher whole lung TNF-α, macrophage inflammatory protein (MIP)-1α, MIP-2, KC, and matrix metalloproteinase (MMP)-12 mRNA compared with infected WT mice. GPx-1(-/-) mice had more MIP-2 protein in BALF at Day 3 and more active MMP-9 protease in BALF at Days 3 and 7 than WT mice. These data indicate that GPx-1 reduces influenza A virus-induced lung inflammation.


Assuntos
Glutationa Peroxidase/fisiologia , Vírus da Influenza A Subtipo H3N2 , Infecções por Orthomyxoviridae/enzimologia , Pneumonia/enzimologia , Pneumonia/prevenção & controle , Imunidade Adaptativa , Animais , Azóis/farmacologia , Líquido da Lavagem Broncoalveolar/citologia , Linfócitos T CD8-Positivos/imunologia , Quimiocinas/genética , Citocinas/genética , Glutationa Peroxidase/deficiência , Glutationa Peroxidase/genética , Vírus da Influenza A Subtipo H3N2/imunologia , Isoindóis , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Compostos Organosselênicos/farmacologia , Infecções por Orthomyxoviridae/etiologia , Infecções por Orthomyxoviridae/patologia , Peptídeo Hidrolases/genética , Pneumonia/etiologia , Pneumonia/patologia , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Espécies Reativas de Oxigênio , Carga Viral , Glutationa Peroxidase GPX1
12.
Microcirculation ; 18(1): 12-23, 2011 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-21166922

RESUMO

OBJECTIVE: The aim of this study was to assess the ability of Gpx1 to regulate leukocyte-endothelial cell interactions in the cerebral microcirculation under inflammatory conditions associated with oxidative stress. METHODS: To induce cerebral inflammation, wild-type and Gpx1(-/-) mice underwent systemic treatment with TNF or transient focal cerebral ischemia via MCAO. Leukocyte rolling and adhesion in cerebral postcapillary venules were assessed by intravital microscopy. RESULTS: Absence of Gpx1(-/-) resulted in increased cerebral oxidant production in response to TNF. Under these conditions, leukocyte rolling in cerebral venules was significantly elevated in Gpx1(-/-) mice, whereas leukocyte adhesion was lower than that in wild-type mice. Despite this, expression of key adhesion molecules did not differ between the strains. Following MCAO, Gpx1(-/-) mice displayed significant reductions in rolling and adhesion associated with severe blood flow restriction. In contrast, following treatment with the anti-oxidant ebselen to equalize postischemic cerebral blood flow in wild-type and Gpx1(-/-) mice, absence of Gpx1 was associated with significant elevations in leukocyte interactions. CONCLUSIONS: These data show that under some inflammatory conditions, Gpx1 regulates leukocyte-endothelial cell interactions in the cerebral microvasculature, but that this is affected by the nature of the inflammatory insult.


Assuntos
Encéfalo/irrigação sanguínea , Comunicação Celular , Células Endoteliais/enzimologia , Glutationa Peroxidase/metabolismo , Migração e Rolagem de Leucócitos , Leucócitos/enzimologia , Vasculite do Sistema Nervoso Central/enzimologia , Animais , Encéfalo/enzimologia , Encéfalo/patologia , Circulação Cerebrovascular , Células Endoteliais/patologia , Glutationa Peroxidase/genética , Inflamação/enzimologia , Inflamação/genética , Inflamação/patologia , Leucócitos/patologia , Camundongos , Camundongos Knockout , Microcirculação , Vasculite do Sistema Nervoso Central/genética , Vasculite do Sistema Nervoso Central/patologia , Glutationa Peroxidase GPX1
13.
Blood ; 114(9): 1937-46, 2009 Aug 27.
Artigo em Inglês | MEDLINE | ID: mdl-19584397

RESUMO

Tissue-type plasminogen activator (tPA) is an extracellular protease that converts plasminogen into plasmin. For tPA to generate plasmin under biologic conditions, a cofactor must first bring tPA and plasminogen into physical proximity. Fibrin provides this cofactor for tPA-mediated plasmin generation in blood. Despite being naturally devoid of fibrin(ogen), tPA-mediated plasmin formation also occurs in the brain. The fibrin-like cofactor(s) that facilitates plasmin formation in the injured brain has remained unknown. Here we show that protein aggregates formed during neuronal injury provide a macromolecular, nonfibrin cofactor that promotes tPA-mediated plasmin formation and subsequent cell breakdown. The binding of plasminogen and tPA to these protein aggregates occurs via distinct mechanisms. Importantly, nonneuronal cell types also exhibit this cofactor effect upon injury, indicating a general phenomenon. This novel cofactor identified in nonviable cells has ramifications for ischemic stroke where tPA is used clinically and where plasmin activity within the injured brain is unwanted. A means of selectively inhibiting the binding of tPA to nonviable cells while preserving its association with fibrin may be of benefit for the treatment of ischemic stroke.


Assuntos
Fibrina/química , Fibrinolisina/química , Ativador de Plasminogênio Tecidual/metabolismo , Animais , Linhagem Celular , Células Cultivadas , Fibrinolisina/metabolismo , Isquemia/patologia , Substâncias Macromoleculares , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Modelos Biológicos , Neurônios/metabolismo , Ratos , Acidente Vascular Cerebral/patologia
14.
Cell Microbiol ; 12(3): 372-85, 2010 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-19888989

RESUMO

Gram-negative bacterial peptidoglycan is specifically recognized by the host intracellular sensor NOD1, resulting in the generation of innate immune responses. Although epithelial cells are normally refractory to external stimulation with peptidoglycan, these cells have been shown to respond in a NOD1-dependent manner to Gram-negative pathogens that can either invade or secrete factors into host cells. In the present work, we report that Gram-negative bacteria can deliver peptidoglycan to cytosolic NOD1 in host cells via a novel mechanism involving outer membrane vesicles (OMVs). We purified OMVs from the Gram-negative mucosal pathogens: Helicobacter pylori, Pseudomonas aeruginosa and Neisseria gonorrhoea and demonstrated that these peptidoglycan containing OMVs upregulated NF-kappaB and NOD1-dependent responses in vitro. These OMVs entered epithelial cells through lipid rafts thereby inducing NOD1-dependent responses in vitro. Moreover, OMVs delivered intragastrically to mice-induced innate and adaptive immune responses via a NOD1-dependent but TLR-independent mechanism. Collectively, our findings identify OMVs as a generalized mechanism whereby Gram-negative bacteria deliver peptidoglycan to cytosolic NOD1. We propose that OMVs released by bacteria in vivo may promote inflammation and pathology in infected hosts.


Assuntos
Células Epiteliais/imunologia , Bactérias Gram-Negativas/imunologia , NF-kappa B/imunologia , Proteína Adaptadora de Sinalização NOD1/imunologia , Peptidoglicano/metabolismo , Vesículas Secretórias/metabolismo , Animais , Feminino , Células HeLa , Helicobacter pylori/imunologia , Humanos , Masculino , Camundongos , Neisseria/imunologia , Peptidoglicano/imunologia , Pseudomonas aeruginosa/imunologia , Vesículas Secretórias/imunologia
15.
Bioorg Med Chem Lett ; 21(17): 5102-6, 2011 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-21470863

RESUMO

3',4'-Dihydroxyflavonol (DiOHF) is a cardioprotective flavonol that reduces injury associated with myocardial ischaemia and reperfusion. We hypothesized that the efficacy of DiOHF could be enhanced through its targeting to hypoxic regions of partial reperfusion. Copper(I)-catalyzed ligation of an azide-modified DiOHF analogue to 2-propargyl-nitroimidazole afforded a DiOHF-nitroimidazole conjugate (DiOHF-NIm). When incubated with Con8 cells under normoxic conditions DiOHF-NIm could be detected in both the culture supernatant and cell lysate, whereas under hypoxic conditions it was present in substantially reduced amounts consistent with its selective metabolism under hypoxia. DiOHF-NIm possessed antioxidant activity comparable to DiOHF through scavenging of superoxide produced by NADPH/NADPH oxidase, but had significantly attenuated vasorelaxant activity. DiOHF-NIm treatment significantly reduced lactate dehydrogenase release following ischaemia/reperfusion in hindlimbs of anaesthetized rats (p <0.05), to a level similar to DiOHF treatment but also at earlier time points. DiOHF-NIm significantly reduced levels of myeloperoxidase (p <0.05), a biomarker of neutrophil accumulation, whereas the reduction afforded by DiOHF was not significant. DiOHF-NIm therefore represents a promising potential therapeutic for ischaemia/reperfusion injury.


Assuntos
Cardiotônicos/síntese química , Flavonóis/síntese química , Traumatismo por Reperfusão/prevenção & controle , Animais , Cardiotônicos/farmacologia , Cromatografia Líquida de Alta Pressão , Flavonóis/farmacologia , Ratos
16.
Front Neurosci ; 15: 621501, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33633536

RESUMO

Neuroinflammation driven by type-I interferons in the CNS is well established to exacerbate the progression of many CNS pathologies both acute and chronic. The role of adaptor protein Stimulator of Interferon Genes (STING) is increasingly appreciated to instigate type-I IFN-mediated neuroinflammation. As an upstream regulator of type-I IFNs, STING modulation presents a novel therapeutic opportunity to mediate inflammation in the CNS. This review will detail the current knowledge of protective and detrimental STING activity in acute and chronic CNS pathologies and the current therapeutic avenues being explored.

17.
ACS Chem Neurosci ; 12(22): 4224-4235, 2021 11 17.
Artigo em Inglês | MEDLINE | ID: mdl-34634903

RESUMO

Parkinson's disease (PD) is a progressive neurological disorder, in which dopaminergic midbrain neurons degenerate, leading to dopamine depletion that is associated with neuronal death. In this Review, we initially describe the pathogenesis of PD and established therapies that unfortunately only delay progression of the disease. With a rapidly escalating incidence in PD, there is an urgent need to develop new therapies that not only halt progression but even reverse degeneration. Biomaterials are playing critical roles in these new therapies which include controlled and site-specific delivery of neurotrophins, increased engraftment of implanted neural stem cells, and redirection of endogenous stem cell populations away from their niche to encourage reparative mechanisms. This Review will therefore cover important design features of biomaterials used in regenerative medicine and tissue engineering strategies targeted at PD.


Assuntos
Células-Tronco Neurais , Doença de Parkinson , Materiais Biocompatíveis , Dopamina , Neurônios Dopaminérgicos , Humanos , Doença de Parkinson/terapia
18.
Am J Physiol Lung Cell Mol Physiol ; 299(3): L425-33, 2010 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-20511341

RESUMO

Reactive oxygen species (ROS) produced from cigarette smoke cause oxidative lung damage including protein denaturation, lipid peroxidation, and DNA damage. Glutathione peroxidase-1 (gpx-1) is a detoxifying enzyme that may protect lungs from such damage. The aim of this study was to determine whether gpx-1 protects the lung against oxidative stress-induced lung inflammation in vivo. Male wild-type (WT) or gpx-1(-/-) mice were exposed to cigarette smoke generated from nine cigarettes per day for 4 days to induce oxidative stress and lung inflammation. The effect of the gpx mimetic ebselen on cigarette smoke-induced lung inflammation was evaluated when given prophylactically and therapeutically, i.e., during established inflammation. Mice were killed, and the lungs were lavaged with PBS and then harvested for genomic and proteomic analysis. Gpx-1(-/-) mice exposed to cigarette smoke had enhanced BALF neutrophils, macrophages, proteolytic burden, whole lung IL-17A, and MIP1alpha mRNA compared with WT mice. The gpx mimetic ebselen (10 and 100 microM) inhibited cigarette smoke extract-induced oxidation of MH-S cells in vitro and inhibited cigarette smoke-induced increases in BALF macrophages, neutrophils, proteolytic burden, and macrophage and neutrophil chemotactic factor gene expression when administered prophylactically. In addition, ebselen inhibited established BALF inflammation when administered therapeutically. These data show that gpx-1 protects against cigarette smoke-induced lung inflammation, and agents that mimic the actions of gpx-1 may have therapeutic utility in inflammatory lung diseases where cigarette smoke plays a role.


Assuntos
Glutationa Peroxidase/metabolismo , Pneumonia/etiologia , Pneumonia/prevenção & controle , Fumar , Animais , Antioxidantes/farmacologia , Azóis/farmacologia , Líquido da Lavagem Broncoalveolar/química , Líquido da Lavagem Broncoalveolar/citologia , Contagem de Células , Fatores Quimiotáticos/metabolismo , Expressão Gênica/efeitos dos fármacos , Glutationa Peroxidase/deficiência , Peptídeos e Proteínas de Sinalização Intercelular/sangue , Isoindóis , Macrófagos/metabolismo , Macrófagos/patologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Neutrófilos/metabolismo , Neutrófilos/patologia , Compostos Organosselênicos/farmacologia , Oxirredução/efeitos dos fármacos , Peptídeo Hidrolases/metabolismo , Pneumonia/patologia , Proteínas/metabolismo , Glutationa Peroxidase GPX1
19.
J Neurotrauma ; 37(5): 706-723, 2020 03 01.
Artigo em Inglês | MEDLINE | ID: mdl-32027210

RESUMO

Traumatic brain injury (TBI) is a major health problem causing disability and death worldwide. There is no effective treatment, due in part to the complexity of the injury pathology and factors affecting its outcome. The extent of brain injury depends on the type of insult, age, sex, lifestyle, genetic risk factors, socioeconomic status, other co-injuries, and underlying health problems. This review discusses the genes that have been directly tested in TBI models, and whether their effects are known to be sex-dependent. Sex differences can affect the incidence, symptom onset, pathology, and clinical outcomes following injury. Adult males are more susceptible at the acute phase and females show greater injury in the chronic phase. TBI is not restricted to a single sex; despite variations in the degree of symptom onset and severity, it is important to consider both female and male animals in TBI pre-clinical research studies.


Assuntos
Lesões Encefálicas Traumáticas/genética , Caracteres Sexuais , Animais , Feminino , Masculino , Modelos Animais , Fatores Sexuais , Fatores Socioeconômicos
20.
Sci Rep ; 10(1): 3153, 2020 02 21.
Artigo em Inglês | MEDLINE | ID: mdl-32081950

RESUMO

Neuroinflammation and accompanying microglial dysfunction are now appreciated to be involved in Alzheimer's disease (AD) pathogenesis. Critical to the process of neuroinflammation are the type-I interferon (IFN) family of cytokines. Efforts to phenotypically characterize microglia within AD identify distinct populations associated with type-I IFN signalling, yet how this affects underlying microglial function is yet to be fully elucidated. Here we demonstrate that Aß1-42 exposure increases bioactive levels of type-I IFN produced by primary microglia alongside increased expression of type-I IFN related genes. Primary microglia isolated from brains of APPswePS1ΔE9 mice with ablated type-I IFN signalling show an increased phagocytic ability to uptake FITC-Aß1-42. Correlative assessment of plaque sizes in aged APPswePS1ΔE9 mice with abrogated type-I IFN signalling show unchanged deposition levels. Microglia from these mice did however show alterations in morphology. This data further highlights the role of type-I IFN signalling within microglia and identifies a role in phagocytosis. As such, targeting both microglial and global type-I IFN signalling presents as a novel therapeutic strategy for AD management.


Assuntos
Peptídeos beta-Amiloides/metabolismo , Encéfalo/metabolismo , Interferon Tipo I/metabolismo , Microglia/metabolismo , Fragmentos de Peptídeos/metabolismo , Animais , Receptor 1 de Quimiocina CX3C/metabolismo , Citocinas/metabolismo , Genótipo , Imunidade Inata , Camundongos , Fagocitose , RNA/metabolismo , Transdução de Sinais
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA