Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Mol Ther ; 32(3): 783-799, 2024 Mar 06.
Artigo em Inglês | MEDLINE | ID: mdl-38196192

RESUMO

We recently described a novel ribosome-based regulatory mechanism/checkpoint that controls innate immune gene translation and microglial activation in non-sterile inflammation orchestrated by RNA binding protein SRSF3. Here we describe a role of SRSF3 in the regulation of microglia/macrophage activation phenotypes after experimental stroke. Using a model-system for analysis of the dynamic translational state of microglial ribosomes we show that 24 h after stroke highly upregulated immune mRNAs are not translated resulting in a marked dissociation of mRNA and protein networks in activated microglia/macrophages. Next, microglial activation after stroke was characterized by a robust increase in pSRSF3/SRSF3 expression levels. Targeted knockdown of SRSF3 using intranasal delivery of siRNA 24 h after stroke caused a marked knockdown of endogenous protein. Further analyses revealed that treatment with SRSF3-siRNA alleviated translational arrest of selected genes and induced a transient but significant increase in innate immune signaling and IBA1+ immunoreactivity peaking 5 days after initial injury. Importantly, delayed SRSF3-mediated increase in immune signaling markedly reduced the size of ischemic lesion measured 7 days after stroke. Together, our findings suggest that targeting SRSF3 and immune mRNA translation may open new avenues for molecular/therapeutic reprogramming of innate immune response after ischemic injury.


Assuntos
Isquemia Encefálica , Acidente Vascular Cerebral , Humanos , Microglia/metabolismo , Isquemia Encefálica/genética , Isquemia Encefálica/terapia , Macrófagos/metabolismo , Acidente Vascular Cerebral/patologia , Biossíntese de Proteínas , RNA Mensageiro/metabolismo , RNA Interferente Pequeno/metabolismo , Fatores de Processamento de Serina-Arginina/genética , Fatores de Processamento de Serina-Arginina/metabolismo
2.
Mol Neurodegener ; 18(1): 95, 2023 Dec 16.
Artigo em Inglês | MEDLINE | ID: mdl-38104136

RESUMO

BACKGROUND: Microglia, the brain-resident macrophages perform immune surveillance and engage with pathological processes resulting in phenotype changes necessary for maintaining homeostasis. In preceding studies, we showed that antibiotic-induced perturbations of the gut microbiome of APPPS1-21 mice resulted in significant attenuation in Aß amyloidosis and altered microglial phenotypes that are specific to male mice. The molecular events underlying microglial phenotypic transitions remain unclear. Here, by generating 'APPPS1-21-CD11br' reporter mice, we investigated the translational state of microglial/macrophage ribosomes during their phenotypic transition and in a sex-specific manner. METHODS: Six groups of mice that included WT-CD11br, antibiotic (ABX) or vehicle-treated APPPS1-21-CD11br males and females were sacrificed at 7-weeks of age (n = 15/group) and used for immunoprecipitation of microglial/macrophage polysomes from cortical homogenates using anti-FLAG antibody. Liquid chromatography coupled to tandem mass spectrometry and label-free quantification was used to identify newly synthesized peptides isolated from polysomes. RESULTS: We show that ABX-treatment leads to decreased Aß levels in male APPPS1-21-CD11br mice with no significant changes in females. We identified microglial/macrophage polypeptides involved in mitochondrial dysfunction and altered calcium signaling that are associated with Aß-induced oxidative stress. Notably, female mice also showed downregulation of newly-synthesized ribosomal proteins. Furthermore, male mice showed an increase in newly-synthesized polypeptides involved in FcγR-mediated phagocytosis, while females showed an increase in newly-synthesized polypeptides responsible for actin organization associated with microglial activation. Next, we show that ABX-treatment resulted in substantial remodeling of the epigenetic landscape, leading to a metabolic shift that accommodates the increased bioenergetic and biosynthetic demands associated with microglial polarization in a sex-specific manner. While microglia in ABX-treated male mice exhibited a metabolic shift towards a neuroprotective phenotype that promotes Aß clearance, microglia in ABX-treated female mice exhibited loss of energy homeostasis due to persistent mitochondrial dysfunction and impaired lysosomal clearance that was associated with inflammatory phenotypes. CONCLUSIONS: Our studies provide the first snapshot of the translational state of microglial/macrophage cells in a mouse model of Aß amyloidosis that was subject to ABX treatment. ABX-mediated changes resulted in metabolic reprogramming of microglial phenotypes to modulate immune responses and amyloid clearance in a sex-specific manner. This microglial plasticity to support neuro-energetic homeostasis for its function based on sex paves the path for therapeutic modulation of immunometabolism for neurodegeneration.


Assuntos
Doença de Alzheimer , Amiloidose , Microbiota , Doenças Mitocondriais , Camundongos , Animais , Masculino , Feminino , Doença de Alzheimer/metabolismo , Microglia/metabolismo , Camundongos Transgênicos , Antibacterianos/metabolismo , Antibacterianos/farmacologia , Amiloidose/metabolismo , Macrófagos/metabolismo , Peptídeos/metabolismo , Doenças Mitocondriais/metabolismo , Doenças Mitocondriais/patologia , Epigênese Genética , Peptídeos beta-Amiloides/metabolismo , Modelos Animais de Doenças
3.
Int J Mol Sci ; 24(6)2023 Mar 07.
Artigo em Inglês | MEDLINE | ID: mdl-36982140

RESUMO

Amyotrophic lateral sclerosis (ALS) is a clinically highly heterogeneous disease with a survival rate ranging from months to decades. Evidence suggests that a systemic deregulation of immune response may play a role and affect disease progression. Here, we measured 62 different immune/metabolic mediators in plasma of sporadic ALS (sALS) patients. We show that, at the protein level, the majority of immune mediators including a metabolic sensor, leptin, were significantly decreased in the plasma of sALS patients and in two animal models of the disease. Next, we found that a subset of patients with rapidly progressing ALS develop a distinct plasma assess immune-metabolic molecular signature characterized by a differential increase in soluble tumor necrosis factor receptor II (sTNF-RII) and chemokine (C-C motif) ligand 16 (CCL16) and further decrease in the levels of leptin, mostly dysregulated in male patients. Consistent with in vivo findings, exposure of human adipocytes to sALS plasma and/or sTNF-RII alone, induced a significant deregulation in leptin production/homeostasis and was associated with a robust increase in AMP-activated protein kinase (AMPK) phosphorylation. Conversely, treatment with an AMPK inhibitor restored leptin production in human adipocytes. Together, this study provides evidence of a distinct plasma immune profile in sALS which affects adipocyte function and leptin signaling. Furthermore, our results suggest that targeting the sTNF-RII/AMPK/leptin pathway in adipocytes may help restore assess immune-metabolic homeostasis in ALS.


Assuntos
Esclerose Lateral Amiotrófica , Leptina , Animais , Humanos , Masculino , Proteínas Quinases Ativadas por AMP , Receptores do Fator de Necrose Tumoral , Homeostase
4.
Brain Commun ; 2(2): fcaa124, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-33134918

RESUMO

Despite wide genetic, environmental and clinical heterogeneity in amyotrophic lateral sclerosis, a rapidly fatal neurodegenerative disease targeting motoneurons, neuroinflammation is a common finding. It is marked by local glial activation, T cell infiltration and systemic immune system activation. The immune system has a prominent role in the pathogenesis of various chronic diseases, hence some of them, including some types of cancer, are successfully targeted by immunotherapeutic approaches. However, various anti-inflammatory or immunosuppressive therapies in amyotrophic lateral sclerosis have failed. This prompted increased scrutiny over the immune-mediated processes underlying amyotrophic lateral sclerosis. Perhaps the biggest conundrum is that amyotrophic lateral sclerosis pathogenesis exhibits features of three otherwise distinct immune dysfunctions-excessive inflammation, autoimmunity and inefficient immune responses. Epidemiological and genome-wide association studies show only minimal overlap between amyotrophic lateral sclerosis and autoimmune diseases, so excessive inflammation is usually thought to be secondary to protein aggregation, mitochondrial damage or other stresses. In contrast, several recently characterized amyotrophic lateral sclerosis-linked mutations, including those in TBK1, OPTN, CYLD and C9orf72, could lead to inefficient immune responses and/or damage pile-up, suggesting that an innate immunodeficiency may also be a trigger and/or modifier of this disease. In such cases, non-selective immunosuppression would further restrict neuroprotective immune responses. Here we discuss multiple layers of immune-mediated neuroprotection and neurotoxicity in amyotrophic lateral sclerosis. Particular focus is placed on individual patient mutations that directly or indirectly affect the immune system, and the mechanisms by which these mutations influence disease progression. The topic of immunity in amyotrophic lateral sclerosis is timely and relevant, because it is one of the few common and potentially malleable denominators in this heterogenous disease. Importantly, amyotrophic lateral sclerosis progression has recently been intricately linked to patient T cell and monocyte profiles, as well as polymorphisms in cytokine and chemokine receptors. For this reason, precise patient stratification based on immunophenotyping will be crucial for efficient therapies.

5.
J Clin Invest ; 129(4): 1581-1595, 2019 02 25.
Artigo em Inglês | MEDLINE | ID: mdl-30667370

RESUMO

The cytoplasmic aggregation of TAR DNA-binding protein-43 (TDP-43) is a hallmark of degenerating neurons in amyotrophic lateral sclerosis (ALS) and subsets of frontotemporal dementia (FTD). In order to reduce TDP-43 pathology, we generated single-chain (scFv) antibodies against the RNA recognition motif 1 (RRM1) of TDP-43, which is involved in abnormal protein self-aggregation and interaction with p65 NF-κB. Virus-mediated delivery into the nervous system of a scFv antibody, named VH7Vk9, reduced microgliosis in a mouse model of acute neuroinflammation and mitigated cognitive impairment, motor defects, TDP-43 proteinopathy, and neuroinflammation in transgenic mice expressing ALS-linked TDP-43 mutations. These results suggest that antibodies targeting the RRM1 domain of TDP-43 might provide new therapeutic avenues for the treatment of ALS and FTD.


Assuntos
Esclerose Lateral Amiotrófica/terapia , Proteínas de Ligação a DNA , Dependovirus , Demência Frontotemporal/terapia , Anticorpos de Cadeia Única , Transdução Genética , Motivos de Aminoácidos , Esclerose Lateral Amiotrófica/genética , Esclerose Lateral Amiotrófica/metabolismo , Esclerose Lateral Amiotrófica/patologia , Animais , Linhagem Celular , Proteínas de Ligação a DNA/antagonistas & inibidores , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/metabolismo , Modelos Animais de Doenças , Demência Frontotemporal/genética , Demência Frontotemporal/metabolismo , Demência Frontotemporal/patologia , Camundongos , Camundongos Transgênicos , Mutação , Anticorpos de Cadeia Única/biossíntese , Anticorpos de Cadeia Única/genética
6.
Neurobiol Aging ; 40: 50-60, 2016 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-26973103

RESUMO

Estrogens are known to exert neuroprotective and immuneomodulatory effects after stroke. However, at present, little is known about the role of estrogens and its receptors in postischemic inflammation after menopause. Here, we provide important in vivo evidence of a distinct shift in microglial phenotypes in the model of postmenopause brain. Using a model-system for live imaging of microglial activation in the context of chronic estrogen- and ERα-deficiency associated with aging, we observed a marked deregulation of the TLR2 signals and/or microglial activation in ovariectomized and/or ERα knockout mice. Further analysis revealed a 5.7-fold increase in IL-6, a 4.7-fold increase in phospho-Stat3 levels suggesting an overactivation of JAK/STAT3 pathway and significantly larger infarction in ERα knockouts chronically deprived of estrogen. Taken together, our results suggest that in the experimental model of menopause and/or aging, ERα mediates innate immune responses and/or microglial activation, and ischemia-induced production of IL-6. Based on our results, we propose that the loss of functional ERα may lead to deregulation of postischemic inflammatory responses and increased vulnerability to ischemic injury in aging female brains.


Assuntos
Envelhecimento , Isquemia Encefálica/patologia , Encéfalo/patologia , Receptor alfa de Estrogênio/deficiência , Receptor alfa de Estrogênio/fisiologia , Menopausa , Envelhecimento/metabolismo , Envelhecimento/patologia , Animais , Encéfalo/metabolismo , Isquemia Encefálica/metabolismo , Estrogênios/deficiência , Estrogênios/fisiologia , Feminino , Imunidade Inata , Interleucina-6/metabolismo , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Microglia/patologia , Modelos Animais , Fator de Transcrição STAT3/metabolismo
7.
Mol Ther ; 22(3): 498-510, 2014 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-24394188

RESUMO

There is emerging evidence that the misfolding of superoxide dismutase 1 (SOD1) may represent a common pathogenic event in both familial and sporadic amyotrophic lateral sclerosis (ALS). To reduce the burden of misfolded SOD1 species in the nervous system, we have tested a novel therapeutic approach based on adeno-associated virus (AAV)-mediated tonic expression of a DNA construct encoding a secretable single-chain fragment variable (scFv) antibody composed of the variable heavy and light chain regions of a monoclonal antibody (D3H5) binding specifically to misfolded SOD1. A single intrathecal injection of the AAV encoding the single-chain antibody in SOD1(G93A) mice at 45 days of age resulted in sustained expression of single-chain antibodies in the spinal cord, and it delayed disease onset and extension of life span by up to 28%, in direct correlation with scFv titers in the spinal cord. The treatment caused attenuation of neuronal stress signals and reduction in levels of misfolded SOD1 in the spinal cord of SOD1(G93A) mice. From these results, we propose that an immunotherapy based on intrathecal inoculation of AAV encoding a secretable scFv against misfolded SOD1 should be considered as potential treatment for ALS, especially for individuals carrying SOD1 mutations.


Assuntos
Esclerose Lateral Amiotrófica/terapia , Dependovirus/genética , Anticorpos de Cadeia Única/imunologia , Medula Espinal/imunologia , Superóxido Dismutase/imunologia , Esclerose Lateral Amiotrófica/imunologia , Animais , Modelos Animais de Doenças , Progressão da Doença , Terapia Genética , Vetores Genéticos/administração & dosagem , Gliose/patologia , Gliose/terapia , Células HEK293 , Humanos , Imunoterapia , Injeções Espinhais , Camundongos , Dobramento de Proteína , Proteínas Recombinantes/genética , Proteínas Recombinantes/imunologia , Anticorpos de Cadeia Única/farmacologia , Superóxido Dismutase/metabolismo , Superóxido Dismutase-1
8.
PLoS One ; 8(1): e55039, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23383048

RESUMO

Stroke induces the recruitment of neuronal precursors from the subventricular zone (SVZ) into the ischemic striatum. In injured areas, de-routed neuroblasts use blood vessels as a physical scaffold to their migration, in a process that resembles the constitutive migration seen in the rostral migratory stream (RMS). The molecular mechanism underlying injury-induced vasculature-mediated migration of neuroblasts in the post-stroke striatum remains, however, elusive. Using adult mice we now demonstrate that endothelial cells in the ischemic striatum produce brain-derived neurotrophic factor (BDNF), a neurotrophin that promotes the vasculature-mediated migration of neuronal precursors in the RMS, and that recruited neuroblasts maintain expression of p75NTR, a low-affinity receptor for BDNF. Reactive astrocytes, which are widespread throughout the damaged area, ensheath blood vessels and express TrkB, a high-affinity receptor for BDNF. Despite the absence of BDNF mRNA, we observed strong BDNF immunolabeling in astrocytes, suggesting that these glial cells trap extracellular BDNF. Importantly, this pattern of expression is reminiscent of the adult RMS, where TrkB-expressing astrocytes bind and sequester vasculature-derived BDNF, leading to the entry of migrating cells into the stationary phase. Real-time imaging of cell migration in acute brain slices revealed a direct role for BDNF in promoting the migration of neuroblasts to ischemic areas. We also demonstrated that cells migrating in the ischemic striatum display higher exploratory behavior and longer stationary periods than cells migrating in the RMS. Our findings suggest that the mechanisms involved in the injury-induced vasculature-mediated migration of neuroblasts recapitulate, at least partially, those observed during constitutive migration in the RMS.


Assuntos
Isquemia Encefálica/patologia , Isquemia Encefálica/fisiopatologia , Fator Neurotrófico Derivado do Encéfalo/metabolismo , Movimento Celular , Neostriado/irrigação sanguínea , Neostriado/patologia , Neurônios/patologia , Animais , Astrócitos/metabolismo , Astrócitos/patologia , Isquemia Encefálica/genética , Isquemia Encefálica/metabolismo , Fator Neurotrófico Derivado do Encéfalo/genética , Regulação da Expressão Gênica , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Neovascularização Fisiológica , Células-Tronco Neurais/patologia , Neurônios/metabolismo , Receptor trkB/genética , Receptores de Fator de Crescimento Neural/genética
9.
J Neurosci ; 32(30): 10383-95, 2012 Jul 25.
Artigo em Inglês | MEDLINE | ID: mdl-22836271

RESUMO

Growing evidence suggests that galectin-3 is involved in fine tuning of the inflammatory responses at the periphery, however, its role in injured brain is far less clear. Our previous work demonstrated upregulation and coexpression of galectin-3 and IGF-1 in a subset of activated/proliferating microglial cells after stroke. Here, we tested the hypothesis that galectin-3 plays a pivotal role in mediating injury-induced microglial activation and proliferation. By using a galectin-3 knock-out mouse (Gal-3KO), we demonstrated that targeted disruption of the galectin-3 gene significantly alters microglia activation and induces ∼4-fold decrease in microglia proliferation. Defective microglia activation/proliferation was further associated with significant increase in the size of ischemic lesion, ∼2-fold increase in the number of apoptotic neurons, and a marked deregulation of the IGF-1 levels. Next, our results revealed that contrary to WT cells, the Gal3-KO microglia failed to proliferate in response to IGF-1. Moreover, the IGF-1-mediated mitogenic microglia response was reduced by N-glycosylation inhibitor tunicamycine while coimmunoprecipitation experiments revealed galectin-3 binding to IGF-receptor 1 (R1), thus suggesting that interaction of galectin-3 with the N-linked glycans of receptors for growth factors is involved in IGF-R1 signaling. While the canonical IGF-1 signaling pathways were not affected, we observed an overexpression of IL-6 and SOCS3, suggesting an overactivation of JAK/STAT3, a shared signaling pathway for IGF-1/IL-6. Together, our findings suggest that galectin-3 is required for resident microglia activation and proliferation in response to ischemic injury.


Assuntos
Isquemia Encefálica/metabolismo , Encéfalo/metabolismo , Proliferação de Células/efeitos dos fármacos , Galectina 3/metabolismo , Microglia/metabolismo , Animais , Encéfalo/efeitos dos fármacos , Encéfalo/patologia , Isquemia Encefálica/genética , Isquemia Encefálica/patologia , Células Cultivadas , Galectina 3/genética , Fator de Crescimento Insulin-Like I/farmacologia , Interleucina-6/metabolismo , Macrófagos/efeitos dos fármacos , Macrófagos/metabolismo , Macrófagos/patologia , Camundongos , Camundongos Knockout , Microglia/efeitos dos fármacos , Microglia/patologia , Fator de Transcrição STAT3/metabolismo , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/fisiologia , Regulação para Cima/efeitos dos fármacos , Regulação para Cima/fisiologia
10.
J Exp Med ; 208(12): 2429-47, 2011 Nov 21.
Artigo em Inglês | MEDLINE | ID: mdl-22084410

RESUMO

TDP-43 (TAR DNA-binding protein 43) inclusions are a hallmark of amyotrophic lateral sclerosis (ALS). In this study, we report that TDP-43 and nuclear factor κB (NF-κB) p65 messenger RNA and protein expression is higher in spinal cords in ALS patients than healthy individuals. TDP-43 interacts with and colocalizes with p65 in glial and neuronal cells from ALS patients and mice expressing wild-type and mutant TDP-43 transgenes but not in cells from healthy individuals or nontransgenic mice. TDP-43 acted as a co-activator of p65, and glial cells expressing higher amounts of TDP-43 produced more proinflammatory cytokines and neurotoxic mediators after stimulation with lipopolysaccharide or reactive oxygen species. TDP-43 overexpression in neurons also increased their vulnerability to toxic mediators. Treatment of TDP-43 mice with Withaferin A, an inhibitor of NF-κB activity, reduced denervation in the neuromuscular junction and ALS disease symptoms. We propose that TDP-43 deregulation contributes to ALS pathogenesis in part by enhancing NF-κB activation and that NF-κB may constitute a therapeutic target for the disease.


Assuntos
Esclerose Lateral Amiotrófica/metabolismo , Proteínas de Ligação a DNA/metabolismo , Junção Neuromuscular/metabolismo , Transdução de Sinais/fisiologia , Fator de Transcrição RelA/metabolismo , Análise de Variância , Animais , Western Blotting , Primers do DNA/genética , Proteínas de Ligação a DNA/genética , Ensaio de Desvio de Mobilidade Eletroforética , Ensaio de Imunoadsorção Enzimática , Humanos , Imunoprecipitação , Espectrometria de Massas , Camundongos , Camundongos Transgênicos , Microscopia de Fluorescência , Neuroglia/metabolismo , Junção Neuromuscular/efeitos dos fármacos , Neurônios/metabolismo , Interferência de RNA , Reação em Cadeia da Polimerase em Tempo Real , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Medula Espinal/citologia , Medula Espinal/patologia , Fator de Transcrição RelA/antagonistas & inibidores , Transgenes/genética , Vitanolídeos/farmacologia
11.
J Neuroinflammation ; 8: 137, 2011 Oct 11.
Artigo em Inglês | MEDLINE | ID: mdl-21989292

RESUMO

BACKGROUND: Mounting evidence supports a significant role of inflammation in Parkinson's disease (PD) pathophysiology, with several inflammatory pathways being suggested as playing a role in the dopaminergic degeneration seen in humans and animal models of the disease. These include tumor necrosis factor, prostaglandins and oxidative-related stress components. However, the role of innate immunity has not been established in PD. METHODS: Based on the fact that the myeloid differentiation primary response gene (88) (MyD88) is the most common adaptor protein implicated in toll-like receptor (TLR) signaling, critical in the innate immune response, we undertook a study to investigate the potential contribution of this specific pathway to MPTP-induced brain dopaminergic degeneration using MyD88 knock out mice (MyD88-/-), following our observations that the MyD88-dependent pathway was critical for MPTP dopaminergic toxicity in the enteric nervous system. Post-mortem analyses assessing nigrostriatal dopaminergic degeneration and inflammation were performed using HPLC, western blots, autoradiography and immunofluorescence. RESULTS: Our results demonstrate that MyD88-/- mice are as vulnerable to MPTP-induced dopamine and DOPAC striatal depletion as wild type mice. Furthermore, MyD88-/- mice show similar striatal dopamine transporter and tyrosine hydroxylase loss, as well as dopaminergic cell loss in the substantia nigra pars compacta in response to MPTP. To evaluate the extent of the inflammatory response created by the MPTP regimen utilized, we further performed bioluminescence imaging using TLR2-luc/gfp transgenic mice and microglial density analysis, which revealed a modest brain microglial response following MPTP. This was accompanied by a significant astrocytic reaction in the striatum, which was of similar magnitude both in wild type and MyD88-/- mice. CONCLUSIONS: Our results suggest that subacute MPTP-induced dopaminergic degeneration observed in the central nervous system is MyD88-independent, in contrast to our recent observations that this pathway, in the same cohort of animals, is critical in the loss of dopaminergic neurons in the enteric nervous system.


Assuntos
1-Metil-4-Fenil-1,2,3,6-Tetra-Hidropiridina/farmacologia , Corpo Estriado/patologia , Neurônios Dopaminérgicos , Intoxicação por MPTP/patologia , Fator 88 de Diferenciação Mieloide/metabolismo , Neurotoxinas/farmacologia , Substância Negra/patologia , Ácido 3,4-Di-Hidroxifenilacético/metabolismo , Animais , Astrócitos/efeitos dos fármacos , Astrócitos/metabolismo , Astrócitos/patologia , Autorradiografia , Corpo Estriado/citologia , Corpo Estriado/metabolismo , Dopamina/metabolismo , Neurônios Dopaminérgicos/efeitos dos fármacos , Neurônios Dopaminérgicos/metabolismo , Neurônios Dopaminérgicos/patologia , Proteína Glial Fibrilar Ácida/metabolismo , Humanos , Intoxicação por MPTP/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Fator 88 de Diferenciação Mieloide/genética , Transdução de Sinais/fisiologia , Substância Negra/citologia , Substância Negra/metabolismo , Receptores Toll-Like/genética , Receptores Toll-Like/metabolismo
12.
PLoS One ; 5(10): e13742, 2010 Oct 29.
Artigo em Inglês | MEDLINE | ID: mdl-21060793

RESUMO

BACKGROUND: Toll-like receptors (TLRs) play a crucial role in the activation of innate immunity in response to many viruses. We previously reported the implication of TLR2 in the recognition of Epstein-Barr virus (EBV) by human monocytes. Because murine gammaherpesvirus-68 (MHV-68) is a useful model to study human gammaherpesvirus pathogenesis in vivo, we evaluated the importance of mouse TLR2 in the recognition of MHV-68. METHODOLOGY/PRINCIPAL FINDINGS: In studies using transfected HEK293 cells, MHV-68 lead to the activation of NF-κB reporter through TLR2. In addition, production of interleukin-6 (IL-6) and interferon-α (IFN-α) upon MHV-68 stimulation was reduced in murine embryonic fibroblasts (MEFs) derived from TLR2-/- and MyD88-/- mice as compared to their wild type (WT) counterpart. In transgenic mice expressing a luciferase reporter gene under the control of the mTLR2 promoter, MHV-68 challenge activated TLR2 transcription. Increased expression levels of TLR2 on blood granulocytes (CD115(-)Gr1(+)) and inflammatory monocytes (CD115(+)Gr1(+)), which mobilized to the lungs upon infection with MHV-68, was also confirmed by flow cytometry. Finally, TLR2 or MyD88 deficiency was associated with decreased IL-6 and type 1 IFN production as well as increased viral burden during short-term challenges with MHV-68. CONCLUSIONS/SIGNIFICANCE: TLR2 contributes to the production of inflammatory cytokines and type 1 IFN as well as to the control of viral burden during infection with MHV-68. Taken together, our results suggest that the TLR2 pathway has a relevant role in the recognition of this virus and in the subsequent activation of the innate immune response.


Assuntos
Gammaherpesvirinae/fisiologia , Infecções por Herpesviridae/fisiopatologia , Receptor 2 Toll-Like/fisiologia , Animais , Linhagem Celular , Células Cultivadas , Gammaherpesvirinae/isolamento & purificação , Genes Reporter , Infecções por Herpesviridae/virologia , Humanos , Interferon gama/biossíntese , Interleucina-6/biossíntese , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Fator 88 de Diferenciação Mieloide/metabolismo , NF-kappa B/metabolismo , Receptor 2 Toll-Like/genética
13.
Stroke ; 39(3): 935-42, 2008 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-18258827

RESUMO

BACKGROUND AND PURPOSE: We sought to develop a model system for live analysis of brain inflammatory response in ischemic injury. METHODS: Using a reporter mouse-expressing luciferase gene under transcriptional control of the murine glial fibrillary acidic protein (GFAP) promoter (GFAP-luc mice) and biophotonic/bioluminescent imaging as tools, we developed a model system for in vivo analysis of astrocyte activation/response in cerebral ischemia. RESULTS: Analysis of photon emissions from the brains of living animals revealed marked sex differences in astrocyte response to ischemic injury. The increase in GFAP signals was significantly higher in female mice in the metestrus/diestrus period compared with male transgenic mice (1.71 x 10(7)+/-0.19 x 10(7) vs 0.92 x 10(7)+/-0.15 x 10(7), P<0.001). Similar results were obtained by quantitative immunohistochemistry (males vs females: 13.4+/-0.5 vs 16.96+/-0.64, P<0.0001). However, astrocyte activation/GFAP signals showed cyclic, estrus-dependent variations in response to ischemic injury. Physiologically higher levels of estrogen and application of pharmacologic doses of estrogen during replacement therapy attenuated GFAP upregulation after stroke. Interestingly, contrary to a positive correlation between the intensities of GFAP signals and infarct size in male mice, no such correlation was observed in any of the experimental groups of female GFAP-luc mice. CONCLUSIONS: Our results suggest that GFAP upregulation in ischemic injury may have different functional significance in female and male experimental animals and may not directly reflect the extent of ischemia-induced neuronal damage in female GFAP-luc mice. Using a novel live imaging approach, we demonstrated that the early-phase brain inflammatory response to ischemia may be associated with sex-specific biomarkers of brain damage.


Assuntos
Astrócitos/patologia , Isquemia Encefálica/metabolismo , Isquemia Encefálica/patologia , Encefalite/diagnóstico , Estrogênios/metabolismo , Proteína Glial Fibrilar Ácida/metabolismo , Fatores Sexuais , Animais , Isquemia Encefálica/complicações , Infarto Cerebral/etiologia , Infarto Cerebral/patologia , Diestro , Encefalite/etiologia , Encefalite/metabolismo , Estrogênios/farmacologia , Feminino , Proteína Glial Fibrilar Ácida/genética , Imuno-Histoquímica , Luminescência , Masculino , Metestro , Camundongos , Camundongos Transgênicos , Regulação para Cima/efeitos dos fármacos
14.
Nano Lett ; 7(8): 2513-20, 2007 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-17638392

RESUMO

Astrocytes are the principle macroglial brain cells. They are activated by different stressors and brain injuries. Quantum dots (QDs) can cause oxidative stress. This study shows a real-time imaging of primary cortical cultures and assessment of QD-induced activation of astrocytes in the brains of transgenic mice with the luciferase gene driven by the murine astrocyte promoter. This approach may be widely applicable for assessing the astroglia/tissue response to nanoparticles in live animals.


Assuntos
Astrócitos/citologia , Astrócitos/efeitos dos fármacos , Microscopia de Fluorescência/métodos , Nanopartículas/administração & dosagem , Pontos Quânticos , Animais , Sobrevivência Celular/efeitos dos fármacos , Sistemas Computacionais , Células PC12 , Ratos
15.
Exp Neurol ; 204(1): 433-42, 2007 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-17234187

RESUMO

Considering that several pathways leading to cell death are activated in cerebral ischemia, we tested in mouse models of transient and permanent ischemia a drug cocktail aiming at distinct pharmacological targets during the evolution of ischemic injury. It consists of minocycline--an antibiotic with anti-inflammatory properties, riluzole--a glutamate antagonist, and nimodipine--a blocker of voltage-gated calcium channels. Administered 2 h after transient or permanent MCAO, it significantly decreased the size of infarction, by approximately 65% after transient and approximately 35% after permanent ischemia and markedly improve clinical recovery of mice. In both experimental models a three-drug cocktail achieved significantly more efficient neuroprotection than any of the components tested alone. However, some interesting observation emerged from the single-drug studies. Treatment with minocycline alone was efficient in both experimental models while treatment with glutamate antagonist riluzole conferred neuroprotection only after transient MCAO. Immunohistochemical analysis following three-drug treatment revealed reduced microglia/macrophages and caspase-3 activation as well as preserved GFAP immunoreactivity following transient ischemia. No detectable differences in the levels of Mac-2, GFAP and caspase-3 immunoreactivities were observed 72 h after permanent MCAO. These marked differences in the brain tissue responses to ischemic injury and to treatments suggest that different pathological mechanisms may be operating in transient and permanent ischemia. However, the three-drug cocktail exerted significant neuroprotection in both experimental models thus demonstrating that simultaneous targeting of several pathophysiological pathways involved in the evolution of ischemic injury may represent a rational therapeutic strategy for stroke.


Assuntos
Isquemia Encefálica/patologia , Ataque Isquêmico Transitório/patologia , Minociclina/farmacologia , Fármacos Neuroprotetores/farmacologia , Nimodipina/farmacologia , Riluzol/farmacologia , Animais , Encéfalo/enzimologia , Encéfalo/patologia , Caspase 3/metabolismo , Combinação de Medicamentos , Ativação Enzimática , Proteína Glial Fibrilar Ácida/metabolismo , Imuno-Histoquímica , Ataque Isquêmico Transitório/metabolismo , Macrófagos/patologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Microglia/patologia , Infarto do Miocárdio/patologia , Fatores de Tempo
16.
Biochim Biophys Acta ; 1762(11-12): 1013-24, 2006.
Artigo em Inglês | MEDLINE | ID: mdl-16675207

RESUMO

The discovery of missense mutations in the gene coding for the Cu/Zn superoxide dismutase 1 (SOD1) in subsets of familial cases was rapidly followed by the generation of transgenic mice expressing various forms of SOD1 mutants. The mice overexpressing high levels of mutant SOD1 mRNAs do develop motor neuron disease but unraveling the mechanisms of pathogenesis has been very challenging. Studies with mouse lines suggest that the toxicity of mutant SOD1 is unrelated to copper-mediated catalysis but rather to propensity of a subfraction of mutant SOD1 proteins to form misfolded protein species and aggregates. However, the mechanism of toxicity of SOD1 mutants remains to be elucidated. Involvement of cytoskeletal components in ALS pathogenesis is supported by several mouse models of motor neuron disease with neurofilament abnormalities and with genetic defects in microtubule-based transport. Here, we describe how transgenic mouse models have been used for understanding pathogenic pathways of motor neuron disease and for pre-clinical drug testing.


Assuntos
Esclerose Lateral Amiotrófica/genética , Esclerose Lateral Amiotrófica/terapia , Modelos Neurológicos , Neurônios Motores/patologia , Superóxido Dismutase/genética , Animais , Fatores de Troca do Nucleotídeo Guanina/genética , Humanos , Filamentos Intermediários/genética , Filamentos Intermediários/metabolismo , Filamentos Intermediários/patologia , Camundongos , Camundongos Transgênicos , Microtúbulos/fisiologia , Neurônios Motores/metabolismo , Mutação , Superóxido Dismutase/metabolismo , Superóxido Dismutase-1 , Fator A de Crescimento do Endotélio Vascular/genética
17.
J Neurosci ; 23(6): 2131-40, 2003 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-12657672

RESUMO

There is growing evidence for involvement of members of the cyclin-dependent kinase (Cdk) family in neurodegenerative disorders and in apoptotic death of neurons subjected to various insults. After our recent report that a deregulation of Cdk5 activity by p25 may contribute to pathogenesis of amyotrophic lateral sclerosis (ALS), we further examined the possible involvement of other Cdks in mice expressing a mutant form of superoxide dismutase (SOD1(G37R)) linked to ALS. No substantial changes in Cdk2 or Cdk6 distribution and kinase activities were detected in spinal motor neurons from SOD1(G37R) mice when compared with normal mice. Of particular interest was the upregulation and mislocalization of Cdk4, a regulator of the G1-S checkpoint of the cell cycle, in motor neurons of SOD1(G37R) mice. The increase of Cdk4 activity in SOD1(G37R) mice was associated with an increase in nuclear Cdk4, cyclin D1, its coactivator, and with the abnormal phosphorylation of the retinoblastoma (Rb) protein at Cdk phosphorylation sites. Pharmacological treatment of SOD1(G37R) mice with minocycline, a compound that attenuates microgliosis and slows down disease, lessened the dysregulation of Cdk5/Cdk4 and the phosphorylation of Rb. Interestingly, phospho-Rb was immunoprecipitated with anti-Cdk4 but not with anti-Cdk5 antibodies, suggesting a key role for Cdk4 in the phosphorylation of Rb. Remarkably, the overexpression of a transgene coding for human neurofilament H, a phosphorylation sink for deregulated Cdk5 activity by p25, resulted in a reduction in levels of nuclear Cdk4 and Rb phosphorylation. These results indicate that a cell cycle signaling at the neuronal G1-S checkpoint subsequent to Cdk5 deregulation may constitute a critical step of the neuronal death pathway in ALS caused by mutant SOD1.


Assuntos
Esclerose Lateral Amiotrófica/genética , Esclerose Lateral Amiotrófica/metabolismo , Proteínas de Ciclo Celular/metabolismo , Neurônios Motores/metabolismo , Proteínas Proto-Oncogênicas , Superóxido Dismutase/biossíntese , Substituição de Aminoácidos , Esclerose Lateral Amiotrófica/patologia , Animais , Antibacterianos/farmacologia , Morte Celular , Núcleo Celular/metabolismo , Ciclina D1/metabolismo , Quinase 4 Dependente de Ciclina , Quinase 5 Dependente de Ciclina , Quinases Ciclina-Dependentes/metabolismo , Progressão da Doença , Fase G1/fisiologia , Humanos , Camundongos , Camundongos Mutantes Neurológicos , Camundongos Transgênicos , Minociclina/farmacologia , Neurônios Motores/efeitos dos fármacos , Neurônios Motores/patologia , Proteínas de Neurofilamentos/biossíntese , Proteínas de Neurofilamentos/genética , Fosforilação/efeitos dos fármacos , Proteína do Retinoblastoma/metabolismo , Fase S/fisiologia , Transdução de Sinais , Superóxido Dismutase/genética , Transgenes , Regulação para Cima
18.
Brain Res ; 946(2): 153-61, 2002 Aug 16.
Artigo em Inglês | MEDLINE | ID: mdl-12137917

RESUMO

Peripherin is a type III intermediate filament predominantly expressed in neurons having direct axonal projections toward peripheral structures. Here, we report that brain injuries can trigger expression of peripherin and the formation of peripherin accumulations in neurons that are normally silent for this gene. Stab lesions made with nitrocellulose implants induced within 4 days the formation of peripherin accumulations, devoid of neurofilament proteins, in thalamic neurites at the site of the lesion. The local administration of interleukin-6 or leukemia inhibitory factor at the site of the stab lesion extended the expression pattern of peripherin to other neuronal subsets in areas of the cortex and/or of the hippocampus adjacent to injury. We also show that transient focal ischemia in mice, a model of stroke, can trigger within 72 h the formation of neuronal peripherin accumulations in neurons of the cortex, thalamus and hippocampus. This new type of potentially noxious intermediate filament protein accumulations, composed of peripherin, may be of relevance to many brain degenerative disorders with occurrence of proinflammatory cytokines.


Assuntos
Química Encefálica/fisiologia , Lesões Encefálicas/metabolismo , Isquemia Encefálica/metabolismo , Proteínas de Filamentos Intermediários/biossíntese , Glicoproteínas de Membrana , Proteínas do Tecido Nervoso/biossíntese , Neurônios/metabolismo , Animais , Western Blotting , Encéfalo/citologia , Lesões Encefálicas/patologia , Isquemia Encefálica/patologia , Técnica Indireta de Fluorescência para Anticorpo , Inibidores do Crescimento/farmacologia , Imuno-Histoquímica , Interleucina-6/farmacologia , Fator Inibidor de Leucemia , Linfocinas/farmacologia , Camundongos , Camundongos Endogâmicos C57BL , Proteínas de Neurofilamentos/metabolismo , Periferinas , Tálamo/citologia , Tálamo/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA