Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 92
Filtrar
1.
J Neurosci ; 37(11): 3072-3084, 2017 03 15.
Artigo em Inglês | MEDLINE | ID: mdl-28188219

RESUMO

Neurogenesis is essential to brain development and plays a central role in the response to brain injury. Stroke and head trauma stimulate proliferation of endogenous neural stem cells (NSCs); however, the survival of young neurons is sharply reduced by postinjury inflammation. Cellular mitochondria are critical to successful neurogenesis and are a major target of inflammatory injury. Mitochondrial protection was shown to improve survival of young neurons. This study tested whether reducing cellular microRNA-210 (miR-210) would enhance mitochondrial function and improve survival of young murine neurons under inflammatory conditions. Several studies have demonstrated the potential of miR-210 inhibition to enhance and protect mitochondrial function through upregulation of mitochondrial proteins. Here, miR-210 inhibition significantly increased neuronal survival and protected the activity of mitochondrial enzymes cytochrome c oxidase and aconitase in differentiating NSC cultures exposed to inflammatory mediators. Unexpectedly, we found that reducing miR-210 significantly attenuated NSC proliferation upon induction of differentiation. Further investigation revealed that increased mitochondrial function suppressed the shift to primarily glycolytic metabolism and reduced mitochondrial length characteristic of dividing cells. Activation of AMP-regulated protein kinase-retinoblastoma signaling is important in NSC proliferation and the reduction of this activation observed by miR-210 inhibition is one mechanism contributing to the reduced proliferation. Postinjury neurogenesis occurs as a burst of proliferation that peaks in days, followed by migration and differentiation over weeks. Our studies suggest that mitochondrial protective miR-210 inhibition should be delayed until after the initial burst of proliferation, but could be beneficial during the prolonged differentiation stage.SIGNIFICANCE STATEMENT Increasing the success of endogenous neurogenesis after brain injury holds therapeutic promise. Postinjury inflammation markedly reduces newborn neuron survival. This study found that enhancement of mitochondrial function by reducing microRNA-210 (miR-210) levels could improve survival of young neurons under inflammatory conditions. miR-210 inhibition protected the activity of mitochondrial enzymes cytochrome c oxidase and aconitase. Conversely, we observed decreased precursor cell proliferation likely due to suppression of the AMP-regulated protein kinase-retinoblastoma axis with miR-210 inhibition. Therefore, mitochondrial protection is a double-edged sword: early inhibition reduces proliferation, but inhibition later significantly increases neuroblast survival. This explains in part the contradictory published reports of the effects of miR-210 on neurogenesis.


Assuntos
Proliferação de Células , Sobrevivência Celular/imunologia , Encefalite/imunologia , MicroRNAs/imunologia , Mitocôndrias/imunologia , Neurogênese/imunologia , Neurônios/imunologia , Animais , Citocinas/imunologia , Encefalite/patologia , Feminino , Inflamação/imunologia , Inflamação/patologia , Masculino , Camundongos , Mitocôndrias/patologia , Neurônios/patologia
2.
Stroke ; 49(6): 1488-1495, 2018 06.
Artigo em Inglês | MEDLINE | ID: mdl-29748423

RESUMO

BACKGROUND AND PURPOSE: Inflammatory cells play a significant role in secondary injury after ischemic stroke. Recent studies have suggested that a lack of autophagy in myeloid cells causes augmented proinflammatory cytokine release and prolonged inflammation after tissue injury. In this study, we investigated the roles of myeloid cell autophagy in ischemic brain injury. METHODS: Focal cerebral ischemia was induced via transient middle cerebral artery occlusion in mice with autophagy-deficient myeloid lineage cells (Atg5flox/flox LysMCre+) and in their littermate controls (Atg5flox/flox). Infarct volume, neurological function, inflammatory cell infiltration, and proinflammatory cytokine expression levels were evaluated. RESULTS: Mice lacking autophagy in myeloid lineage cells had a lower survival rate for 14 days than control mice (20% versus 70%; P<0.05). Although there was no difference in infarct volume at 12 hours between the 2 groups, mice lacking autophagy in myeloid lineage cells had larger infarct volumes at later time points (3 and 7 days after reperfusion) with worse neurological deficit scores and lower grip test scores. There were a higher number of ionized calcium binding adaptor molecule 1-positive cells and cells expressing M1 marker CD16/32 in mice lacking autophagy in myeloid cells at the later time points. Moreover, these mice had higher expression levels of proinflammatory cytokines at later time points; however, there was no difference in ionized calcium binding adaptor molecule 1-positive cells or mRNA levels of proinflammatory cytokines at the earlier time point (12 hours after reperfusion). CONCLUSIONS: These data suggest that the lack of myeloid cell autophagy aggravates secondary injury by augmenting and prolonging inflammation after ischemic stroke without affecting the initial injury.


Assuntos
Autofagia/fisiologia , Lesões Encefálicas/metabolismo , Linhagem da Célula/fisiologia , Células Mieloides/metabolismo , Acidente Vascular Cerebral/metabolismo , Animais , Encéfalo/metabolismo , Isquemia Encefálica/metabolismo , Citocinas/metabolismo , Modelos Animais de Doenças , Infarto da Artéria Cerebral Média/metabolismo , Inflamação/metabolismo , Camundongos Transgênicos
3.
Mol Cell Neurosci ; 82: 118-125, 2017 07.
Artigo em Inglês | MEDLINE | ID: mdl-28522364

RESUMO

Whether the effect of miR-181a is sexually dimorphic in stroke is unknown. Prior work showed protection of male mice with miR-181a inhibition. Estrogen receptor-α (ERα) is an identified target of miR181 in endometrium. Therefore we investigated the separate and joint effects of miR-181a inhibition and 17ß-estradiol (E2) replacement after ovariectomy. Adult female mice were ovariectomized and implanted with an E2- or vehicle-containing capsule for 14d prior to 1h middle cerebral artery occlusion (MCAO). Each group received either miR-181a antagomir or mismatch control by intracerebroventricular injection 24h before MCAO. After MCAO neurologic deficit and infarct volume were assessed. Primary male and female astrocyte cultures were subjected to glucose deprivation with miR-181a inhibitor or transfection control, and E2 or vehicle control, with/without ESRα knockdown with small interfering RNA. Cell death was assessed by propidium iodide staining, and lactate dehydrogenase assay. A miR-181a/ERα target site blocker (TSB), with/without miR-181a mimic, was used to confirm targeting of ERα by miR-181a in astrocytes. Individually, miR-181a inhibition or E2 decreased infarct volume and improved neurologic score in female mice, and protected male and female astrocyte cultures. Combined miR-181a inhibition plus E2 afforded greater protection of female mice and female astrocyte cultures, but not in male astrocyte cultures. MiR-181a inhibition only increased ERα levels in vivo and in female cultures, while ERα knockdown with siRNA increased cell death in both sexes. Treatment with ERα TSB was strongly protective in both sexes. In conclusion, the results of the present study suggest miR-181a inhibition enhances E2-mediated stroke protection in females in part by augmenting ERα production, a mechanism detected in female mice and female astrocytes. Sex differences were observed with combined miR-181a inhibition/E2 treatment, and miR-181a targeting of ERα.


Assuntos
Astrócitos/metabolismo , Isquemia Encefálica/genética , Receptor alfa de Estrogênio/genética , Ataque Isquêmico Transitório/metabolismo , MicroRNAs/genética , Animais , Astrócitos/efeitos dos fármacos , Isquemia Encefálica/metabolismo , Modelos Animais de Doenças , Feminino , Ataque Isquêmico Transitório/genética , Masculino , Camundongos Endogâmicos C57BL , Neurônios/metabolismo , Fármacos Neuroprotetores/farmacologia , Traumatismo por Reperfusão/genética , Traumatismo por Reperfusão/metabolismo , Fatores Sexuais
4.
Stroke ; 46(8): 2271-6, 2015 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-26130091

RESUMO

BACKGROUND AND PURPOSE: Interleukin (IL)-4 protects from middle cerebral artery occlusion in male mice. Females generally show less injury in response to the same ischemic challenge, but the underlying mechanisms are not fully understood. We tested the importance of IL-4 in female protection using IL-4 knockout (KO) mice. METHODS: IL-4 KO and wild-type (WT) mice of both sexes were subjected to middle cerebral artery occlusion. Infarct volume was assessed by triphenyltetrazolium chloride staining and neurobehavioral outcome by neuroscore. T cell proliferation was assessed after Concanavalin A exposure. Ischemic brain immune cell populations were analyzed by fluorescence-activated cell sorting and immunostaining. RESULTS: Infarction in WT females during estrus and proestrus phases was significantly smaller than in males; neurological score was better. Infarction volume was larger and neurological score worse in IL-4 KO compared with WT in both sexes, with no sex difference. Proliferation of T cells was inhibited in WT females with higher proliferation and no sex difference in IL-4 KO. Macrophage numbers and total T cells in the ischemic hemisphere were lower in WT females, and monocytes increased markedly in IL-4 KOs with no sex difference. The reduced macrophage infiltration in WT-females was predominantly M2. Loss of IL-4 increased CD68+ and iNOS+ cells and reduced YM1+ and Arg1+ cells in both sexes. CONCLUSIONS: IL-4 is required for female neuroprotection during the estrus phase of the estrus cycle. Protected WT females show a predominance of M2-activated microglia/macrophages and reduced inflammatory infiltration. Increasing macrophage M2 polarization, with or without added inhibition of infiltration, may be a new approach to stroke treatment.


Assuntos
Isquemia Encefálica/metabolismo , Isquemia Encefálica/prevenção & controle , Interleucina-4/deficiência , Caracteres Sexuais , Animais , Isquemia Encefálica/patologia , Feminino , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Knockout , Distribuição Aleatória
5.
Stroke ; 46(2): 551-6, 2015 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-25604249

RESUMO

BACKGROUND AND PURPOSE: MicroRNA (miR)-200c increases rapidly in the brain after transient cerebral ischemia but its role in poststroke brain injury is unclear. Reelin, a regulator of neuronal migration and synaptogenesis, is a predicted target of miR-200c. We hypothesized that miR-200c contributes to injury from transient cerebral ischemia by targeting reelin. METHODS: Brain infarct volume, neurological score and levels of miR-200c, reelin mRNA, and reelin protein were assessed in mice subjected to 1 hour of middle cerebral artery occlusion with or without intracerebroventricular infusion of miR-200c antagomir, mimic, or mismatch control. Direct targeting of reelin by miR-200c was assessed in vitro by dual luciferase assay and immunoblot. RESULTS: Pretreatment with miR-200c antagomir decreased post-middle cerebral artery occlusion brain levels of miR-200c, resulting in a significant reduction in infarct volume and neurological deficit. Changes in brain levels of miR-200c inversely correlated with reelin protein expression. Direct targeting of the Reln 3' untranslated region by miR-200c was verified with dual luciferase assay. Inhibition of miR-200c resulted in an increase in cell survival subsequent to in vitro oxidative injury. This effect was blocked by knockdown of reelin mRNA, whereas application of reelin protein afforded protection. CONCLUSIONS: These findings suggest that the poststroke increase in miR-200c contributes to brain cell death by inhibiting reelin expression, and that reducing poststroke miR-200c is a potential target to mitigate stroke-induced brain injury.


Assuntos
Isquemia Encefálica/metabolismo , Moléculas de Adesão Celular Neuronais/biossíntese , Proteínas da Matriz Extracelular/biossíntese , MicroRNAs/administração & dosagem , MicroRNAs/biossíntese , Proteínas do Tecido Nervoso/biossíntese , Serina Endopeptidases/biossíntese , Animais , Isquemia Encefálica/patologia , Moléculas de Adesão Celular Neuronais/antagonistas & inibidores , Células Cultivadas , Proteínas da Matriz Extracelular/antagonistas & inibidores , Marcação de Genes , Injeções Intraventriculares , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Proteínas do Tecido Nervoso/antagonistas & inibidores , Proteína Reelina
6.
J Neurosci Res ; 93(11): 1703-12, 2015 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-26147710

RESUMO

Recent studies have demonstrated that neural stem cell (NSC) culture at physiologically normoxic conditions (2-5% O2) is advantageous in terms of neuronal differentiation and survival. Neuronal differentiation is accompanied by a remarkable shift to mitochondrial oxidative metabolism compared with preferentially glycolytic metabolism of proliferating cells. However, metabolic changes induced by growth in a normoxic (5%) O2 culture environment in NSCs have been minimally explored. This study demonstrates that culturing under 5% O2 conditions results in higher levels of mitochondrial oxidative metabolism, decreased glycolysis, and reduced levels of reactive oxygen species in NSC cultures. Inflammation is one of the major environmental factors limiting postinjury NSC neuronal differentiation and survival. Our results show that NSCs differentiated under 5% O2 conditions possess better resistance to in vitro inflammatory injury compared with those exposed to 20% O2. The present work demonstrates that lower, more physiologically normal O2 levels support metabolic changes induced during NSC neuronal differentiation and provide increased resistance to inflammatory injury, thus highlighting O2 tension as an important determinant of cell fate and survival in various stem cell therapies.


Assuntos
Técnicas de Cultura de Células/métodos , Diferenciação Celular/fisiologia , Células-Tronco Neurais/citologia , Células-Tronco Neurais/metabolismo , Oxigênio/farmacologia , Animais , Apoptose/fisiologia , Diferenciação Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Células Cultivadas , Glicólise/efeitos dos fármacos , Glicólise/fisiologia , Imuno-Histoquímica , Marcação In Situ das Extremidades Cortadas , Inflamação/metabolismo , Camundongos
7.
Neurochem Res ; 40(2): 301-7, 2015 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-24993363

RESUMO

Astrocytes are critical regulators of neuronal function and an effective target for stroke therapy in animal models. Identifying individual targets with the potential for simultaneous activation of multiple downstream pathways that regulate astrocyte homeostasis may be a necessary element for successful clinical translation. Mitochondria and microRNAs each represent individual targets with multi-modal therapeutic potential. Mitochondria regulate metabolism and apoptosis, while microRNAs have the capacity to bind and inhibit numerous mRNAs. By combining strategies targeted at maintaining astrocyte function during and following cerebral ischemia, a synergistic therapeutic effect may be achieved.


Assuntos
Astrócitos/fisiologia , MicroRNAs/fisiologia , Mitocôndrias/fisiologia , Acidente Vascular Cerebral/terapia , Animais , Modelos Animais de Doenças , Humanos , Acidente Vascular Cerebral/patologia
8.
Anesthesiology ; 123(4): 810-9, 2015 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-26270940

RESUMO

BACKGROUND: Isoflurane induces cell death in neurons undergoing synaptogenesis via increased production of pro-brain-derived neurotrophic factor (proBDNF) and activation of postsynaptic p75 neurotrophin receptor (p75). Astrocytes express p75, but their role in neuronal p75-mediated cell death remains unclear. The authors investigated whether astrocytes have the capacity to buffer increases in proBDNF and protect against isoflurane/p75 neurotoxicity. METHODS: Cell death was assessed in day in vitro (DIV) 7 mouse primary neuronal cultures alone or in co-culture with age-matched or DIV 21 astrocytes with propidium iodide 24 h after 1 h exposure to 2% isoflurane or recombinant proBDNF. Astrocyte-targeted knockdown of p75 in co-culture was achieved with small-interfering RNA and astrocyte-specific transfection reagent and verified with immunofluorescence microscopy. proBDNF levels were assessed by enzyme-linked immunosorbent assay. Each experiment used six to eight replicate cultures/condition and was repeated at least three times. RESULTS: Exposure to isoflurane significantly (P < 0.05) increased neuronal cell death in primary neuronal cultures (1.5 ± 0.7 fold, mean ± SD) but not in co-culture with DIV 7 (1.0 ± 0.5 fold) or DIV 21 astrocytes (1.2 ± 1.2 fold). Exogenous proBDNF dose dependently induced neuronal cell death in both primary neuronal and co-cultures, an effect enhanced by astrocyte p75 inhibition. Astrocyte-targeted p75 knockdown in co-cultures increased media proBDNF (1.2 ± 0.1 fold) and augmented isoflurane-induced neuronal cell death (3.8 ± 3.1 fold). CONCLUSIONS: The presence of astrocytes provides protection to growing neurons by buffering increased levels of proBDNF induced by isoflurane. These findings may hold clinical significance for the neonatal and injured brain where increased levels of proBDNF impair neurogenesis.


Assuntos
Astrócitos/efeitos dos fármacos , Astrócitos/metabolismo , Fator Neurotrófico Derivado do Encéfalo/biossíntese , Isoflurano/toxicidade , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Precursores de Proteínas/biossíntese , Animais , Astrócitos/patologia , Fator Neurotrófico Derivado do Encéfalo/antagonistas & inibidores , Células Cultivadas , Técnicas de Cocultura , Camundongos , Neurônios/patologia , Precursores de Proteínas/antagonistas & inibidores
9.
PLoS Comput Biol ; 10(2): e1003471, 2014 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-24516376

RESUMO

Overexpression of the inducible heat shock protein 70, Hsp72, has broadly cytoprotective effects and improves outcome following stroke. A full understanding of how Hsp72 protects cells against injury is elusive, though several distinct mechanisms are implicated. One mechanism is its anti-inflammatory effects. We study the effects of Hsp72 overexpression on activation of the transcription factor NF-κB in microglia combining experimentation and mathematical modeling, using TNFα to stimulate a microglial cell line stably overexpressing Hsp72. We find that Hsp72 overexpression reduces the amount of NF-κB DNA binding activity, activity of the upstream kinase IKK, and amount of IκBα inhibitor phosphorylated following TNFα application. Simulations evaluating several proposed mechanisms suggest that inhibition of IKK activation is an essential component of its regulatory activities. Unexpectedly we find that Hsp72 overexpression reduces the initial amount of the RelA/p65 NF-κB subunit in cells, contributing to the attenuated response. Neither mechanism in isolation, however, is sufficient to attenuate the response, providing evidence that Hsp72 relies upon multiple mechanisms to attenuate NF-κB activation. An additional observation from our study is that the induced expression of IκBα is altered significantly in Hsp72 expressing cells. While the mechanism responsible for this observation is not known, it points to yet another means by which Hsp72 may alter the NF-κB response. This study illustrates the multi-faceted nature of Hsp72 regulation of NF-κB activation in microglia and offers further clues to a novel mechanism by which Hsp72 may protect cells against injury.


Assuntos
Proteínas de Choque Térmico HSP72/metabolismo , Microglia/metabolismo , NF-kappa B/metabolismo , Animais , Linhagem Celular , Biologia Computacional , DNA/metabolismo , Proteínas de Choque Térmico HSP72/genética , Quinase I-kappa B/antagonistas & inibidores , Quinase I-kappa B/metabolismo , Proteínas I-kappa B/genética , Proteínas I-kappa B/metabolismo , Conceitos Matemáticos , Camundongos , Modelos Neurológicos , Inibidor de NF-kappaB alfa , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Transdução de Sinais , Fator de Transcrição RelA/metabolismo , Fator de Necrose Tumoral alfa/metabolismo , Regulação para Cima
10.
J Neurosci ; 32(18): 6391-410, 2012 May 02.
Artigo em Inglês | MEDLINE | ID: mdl-22553043

RESUMO

Reactive astrogliosis is characterized by a profound change in astrocyte phenotype in response to all CNS injuries and diseases. To better understand the reactive astrocyte state, we used Affymetrix GeneChip arrays to profile gene expression in populations of reactive astrocytes isolated at various time points after induction using two mouse injury models, ischemic stroke and neuroinflammation. We find reactive gliosis consists of a rapid, but quickly attenuated, induction of gene expression after insult and identify induced Lcn2 and Serpina3n as strong markers of reactive astrocytes. Strikingly, reactive astrocyte phenotype strongly depended on the type of inducing injury. Although there is a core set of genes that is upregulated in reactive astrocytes from both injury models, at least 50% of the altered gene expression is specific to a given injury type. Reactive astrocytes in ischemia exhibited a molecular phenotype that suggests that they may be beneficial or protective, whereas reactive astrocytes induced by LPS exhibited a phenotype that suggests that they may be detrimental. These findings demonstrate that, despite well established commonalities, astrocyte reactive gliosis is a highly heterogeneous state in which astrocyte activities are altered to respond to the specific injury. This raises the question of how many subtypes of reactive astrocytes exist. Our findings provide transcriptome databases for two subtypes of reactive astrocytes that will be highly useful in generating new and testable hypotheses of their function, as well as for providing new markers to detect different types of reactive astrocytes in human neurological diseases.


Assuntos
Astrócitos/metabolismo , Lesões Encefálicas/metabolismo , Encéfalo/metabolismo , Gliose/genética , Proteínas do Tecido Nervoso/metabolismo , Proteoma/metabolismo , Animais , Modelos Animais de Doenças , Perfilação da Expressão Gênica/métodos , Genoma/genética , Camundongos , Proteínas do Tecido Nervoso/genética , Proteoma/genética
11.
Glia ; 61(11): 1784-94, 2013 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-24038396

RESUMO

Following transient forebrain ischemia, astrocytes play a key role in determining whether or not neurons in the hippocampal CA1 sector go on to die in a delayed fashion. MicroRNAs (miRNAs) are a novel class of RNAs that control gene expression at the post-transcriptional level and the miR-29 family is highly expressed in astrocytes. In this study we assessed levels of miR-29 in hippocampus following forebrain ischemia and found that after transient forebrain ischemia and short periods of reperfusion, miR-29a significantly increased in the resistant dentate gyrus, but decreased in the vulnerable CA1 region of the hippocampus. We demonstrate that miR-29a targets BH3-only proapoptotic BCL2 family member PUMA by luciferase reporter assay and by Western blot. Comparing primary neuron and astrocyte cultures, and postnatal brain, we verified the strongly astrocytic expression of miR-29a. We further found that miR-29a mimic protects and miR-29a inhibitor aggravates cell injury and mitochondrial function after ischemia-like stresses in vitro. Lastly, by overexpressing and reducing miR-29a we demonstrate the protective effect of miR-29a on CA1 delayed neuronal death after forebrain ischemia. Our data suggest that by targeting a pro-apoptotic BCL2 family member, increasing levels of miR-29a might emerge as a strategy for protection against ischemia-reperfusion injury.


Assuntos
Proteínas Reguladoras de Apoptose/metabolismo , Astrócitos/metabolismo , Ataque Isquêmico Transitório/metabolismo , MicroRNAs/metabolismo , Neurônios/metabolismo , Prosencéfalo/metabolismo , Animais , Proteínas Reguladoras de Apoptose/genética , Morte Celular/fisiologia , Mitocôndrias/genética , Mitocôndrias/metabolismo , Ratos , Traumatismo por Reperfusão/metabolismo
12.
Stroke ; 44(3): 764-70, 2013 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-23349191

RESUMO

BACKGROUND AND PURPOSE: Brain ischemia causes immediate and delayed cell death that is exacerbated by inflammation. Recent studies show that hypocretin-1/orexin-A (Hcrt-1) reduces ischemic brain injury, and Hcrt-positive neurons modulate infection-induced inflammation. Here, we tested the hypothesis that Hcrt plays a protective role against ischemia by modulating inflammation. METHODS: Orexin/ataxin-3 (AT) mice, a transgenic strain in which Hcrt-producing neurons degenerate in early adulthood, and wild-type mice were subjected to transient middle cerebral artery occlusion (MCAO). Infarct volume, neurological score, and spontaneous home cage activity were assessed. Inflammation was measured using immunohistochemistry, ELISA, and assessment of cytokine mRNA levels. RESULTS: Infarct volumes 24 and 48 hours after MCAO were significantly larger, neurological score was worse, and spontaneous activity decreased in AT compared with wild-type mice. Macrophage/microglial infiltration and myeloperoxidase-positive cells were higher in AT compared with wild-type mice. Pre-MCAO intracerebroventricular injection of Hcrt-1 significantly reduced infarct volume and macrophage/microglial infiltration in both genotypes and improved neurological score in AT mice. Post-MCAO treatment decreased infarct size in both wild-type and AT mice, but had no effect on neurological score in either genotype. Microglia express the Hcrt-1 receptor after MCAO. Tumor necrosis factor-α production by lipopolysaccharide-stimulated microglial BV2 cells was significantly reduced by Hcrt-1 pretreatment. Sham AT mice exhibit increased brain tumor necrosis factor-α and interleukin-6 mRNA, suggesting chronic inflammation. CONCLUSIONS: Loss of Hcrt neurons in AT mice resulted in worsened stroke outcomes, which were reversed by administration of exogenous Hcrt-1. The mechanism underlying Hcrt-mediated neuroprotection includes attenuation of inflammatory responses after ischemic insult.


Assuntos
Isquemia Encefálica/prevenção & controle , Isquemia Encefálica/fisiopatologia , Encefalite/prevenção & controle , Encefalite/fisiopatologia , Peptídeos e Proteínas de Sinalização Intracelular/fisiologia , Peptídeos e Proteínas de Sinalização Intracelular/uso terapêutico , Neuropeptídeos/fisiologia , Neuropeptídeos/uso terapêutico , Animais , Isquemia Encefálica/patologia , Movimento Celular , Encefalite/patologia , Infarto da Artéria Cerebral Média/complicações , Injeções Intraventriculares , Interleucina-6/metabolismo , Peptídeos e Proteínas de Sinalização Intracelular/genética , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Microglia/metabolismo , Microglia/patologia , Modelos Animais , Neuropeptídeos/genética , Receptores de Orexina , Orexinas , RNA Mensageiro/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Receptores de Neuropeptídeos/metabolismo , Fatores de Tempo , Fator de Necrose Tumoral alfa/metabolismo
13.
J Neurosci Res ; 91(6): 799-807, 2013 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-23426889

RESUMO

Activation of protein kinase C∊ (PKC∊) confers protection against neuronal ischemia/reperfusion. Activation of PKC∊ leads to its translocation to multiple intracellular sites, so a mitochondria-selective PKC∊ activator was used to test the importance of mitochondrial activation to the neuroprotective effect of PKC∊. PKC∊ can regulate key cytoprotective mitochondrial functions, including electron transport chain activity, reactive oxygen species (ROS) generation, mitochondrial permeability transition, and detoxification of reactive aldehydes. We tested the ability of mitochondria-selective activation of PKC∊ to protect primary brain cell cultures or mice subjected to ischemic stroke. Pretreatment with either general PKC∊ activator peptide, TAT-Ψ∊RACK, or mitochondrial-selective PKC∊ activator, TAT-Ψ∊HSP90, reduced cell death induced by simulated ischemia/reperfusion in neurons, astrocytes, and mixed neuronal cultures. The protective effects of both TAT-Ψ∊RACK and TAT-Ψ∊HSP90 were blocked by the PKC∊ antagonist ∊V1-2 , indicating that protection requires PKC∊ interaction with its anchoring protein, TAT-∊RACK. Further supporting a mitochondrial mechanism for PKC∊, neuroprotection by TAT-Ψ∊HSP90 was associated with a marked delay in mitochondrial membrane depolarization and significantly attenuated ROS generation during ischemia. Importantly, TAT-Ψ∊HSP90 reduced infarct size and reduced neurological deficit in C57/BL6 mice subjected to middle cerebral artery occlusion and 24 hr of reperfusion. Thus selective activation of mitochondrial PKC∊ preserves mitochondrial function in vitro and improves outcome in vivo, suggesting potential therapeutic value clinically when brain ischemia is anticipated, including neurosurgery and cardiac surgery.


Assuntos
Isquemia Encefálica/enzimologia , Mitocôndrias/enzimologia , Proteína Quinase C-épsilon/metabolismo , Acidente Vascular Cerebral/enzimologia , Animais , Western Blotting , Isquemia Encefálica/patologia , Circulação Cerebrovascular/fisiologia , Ativação Enzimática/fisiologia , Potencial da Membrana Mitocondrial , Camundongos , Espécies Reativas de Oxigênio/metabolismo , Traumatismo por Reperfusão/enzimologia , Traumatismo por Reperfusão/patologia , Acidente Vascular Cerebral/patologia
14.
Neurobiol Dis ; 45(1): 555-63, 2012 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-21983159

RESUMO

MicroRNAs (miRNA) are short (~22nt) single stranded RNAs that downregulate gene expression. Although recent studies indicate extensive miRNA changes in response to ischemic brain injury, there is currently little information on the roles of specific miRNAs in this setting. Heat shock proteins (HSP) of the HSP70 family have been extensively studied for their multiple roles in cellular protection, but there is little information on their regulation by miRNAs. We used bioinformatics to identify miR-181 as a possible regulator of several HSP70 family members. We validated GRP78/BIP as a target by dual luciferase assay. In response to stroke in the mouse we find that miR-181 increases in the core, where cells die, but decreases in the penumbra, where cells survive. Increased levels of miR-181a are associated with decreased GRP78 protein levels, but increased levels of mRNA, implicating translational arrest. We manipulated levels of miR-181a using plasmid overexpression of pri-miR-181ab or mimic to increase, and antagomir or inhibitor to reduce levels. Increased miR-181a exacerbated injury both in vitro and in the mouse stroke model. Conversely, reduced levels were associated with reduced injury and increased GRP78 protein levels. Studies in C6 cells show that if GRP78 levels are maintained miR-181a no longer exerts a toxic effect. These data demonstrate that miR-181 levels change in response to stroke and inversely correlate with levels of GRP78. Importantly, reducing or blocking miR-181a protects the brain from stroke.


Assuntos
Isquemia Encefálica/genética , Encéfalo/metabolismo , Proteínas de Choque Térmico/genética , MicroRNAs/genética , Acidente Vascular Cerebral/genética , Animais , Isquemia Encefálica/metabolismo , Chaperona BiP do Retículo Endoplasmático , Expressão Gênica , Proteínas de Choque Térmico/metabolismo , Camundongos , MicroRNAs/metabolismo , Neurônios/metabolismo , Acidente Vascular Cerebral/metabolismo
15.
Ann Neurol ; 70(4): 606-15, 2011 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-22028221

RESUMO

OBJECTIVE: Reperfusion after stroke leads to infiltration of inflammatory cells into the ischemic brain. Nicotinamide adenine dinucleotide phosphate oxidase (NOX2) is a major enzyme system that generates superoxide in immune cells. We studied the effect of NOX2 derived from the immune cells in the brain and in blood cells in experimental stroke. METHODS: To establish whether NOX2 plays a role in brain ischemia, strokes were created in mice, then mice were treated with the NOX2 inhibitor apocynin or vehicle and compared to mice deficient in NOX2's gp91 subunit and their wild-type littermates. To determine whether NOX2 in circulating cells versus brain resident cells contribute to ischemic injury, bone marrow chimeras were generated by transplanting bone marrow from wild-type or NOX2-deficient mice into NOX2 or wild-type hosts, respectively. RESULTS: Apocynin and NOX2 deletion both significantly reduced infarct size, blood-brain barrier disruption, and hemorrhagic transformation of the infarcts, compared to untreated wild-type controls. This was associated with decreased matrix metalloproteinase 9 expression and reduced loss of tight junction proteins. NOX2-deficient mice receiving wild-type marrow had better outcomes compared to the wild-type mice receiving wild-type marrow. Interestingly, wild-type mice receiving NOX2-deficient marrow had even smaller infarct sizes and less hemorrhage than NOX2-deficient mice receiving wild-type marrow. INTERPRETATION: This indicates that NOX2, whether present in circulating cells or brain resident cells, contributes to ischemic brain injury and hemorrhage. However, NOX2 from the circulating cells contributed more to the exacerbation of stroke than that from brain resident cells. These data suggest the importance of targeting the peripheral immune system for treatment of stroke.


Assuntos
Isquemia Encefálica/tratamento farmacológico , Encéfalo/efeitos dos fármacos , Glicoproteínas de Membrana/deficiência , NADPH Oxidases/deficiência , Acidente Vascular Cerebral/tratamento farmacológico , Acetofenonas/administração & dosagem , Acetofenonas/farmacologia , Animais , Células da Medula Óssea/enzimologia , Encéfalo/patologia , Isquemia Encefálica/patologia , Inibidores Enzimáticos/administração & dosagem , Inibidores Enzimáticos/farmacologia , Masculino , Glicoproteínas de Membrana/antagonistas & inibidores , Glicoproteínas de Membrana/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , NADPH Oxidase 2 , NADPH Oxidases/antagonistas & inibidores , NADPH Oxidases/genética , Acidente Vascular Cerebral/patologia , Resultado do Tratamento
16.
J Neurosci ; 30(37): 12242-51, 2010 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-20844120

RESUMO

The impairment of hippocampal neurogenesis has been linked to the pathogenesis of neurological disorders from chronic neurodegenerative disease to the progressive cognitive impairment of children who receive brain irradiation. Numerous studies provide evidence that inflammation downregulates neurogenesis, with multiple factors contributing to this impairment. Although mitochondria are one of the primary targets of inflammatory injury, the role of mitochondrial function in the modulation of neurogenesis remains relatively unstudied. In this study, we used neurosphere-derived cells to show that immature doublecortin (Dcx)-positive neurons are uniquely sensitive to mitochondrial inhibition, demonstrating rapid loss of mitochondrial potential and cell viability compared with glial cells and more mature neurons. Mitochondrial inhibition for 24 h produced no significant changes in astrocyte or oligodendrocyte viability, but reduced viability of mature neurons by 30%, and reduced survival of Dcx(+) cells by 60%. We demonstrate that protection of mitochondrial function with mitochondrial metabolites or the mitochondrial chaperone mtHsp75/mortalin partially reverses the inflammation-associated impairment of neurogenesis in vitro and in irradiated mice in vivo. Our findings highlight mitochondrial mechanisms involved in neurogenesis and indicate mitochondria as a potential target for protective strategies to prevent the impairment of neurogenesis by inflammation.


Assuntos
Metabolismo Energético/fisiologia , Mediadores da Inflamação/fisiologia , Mitocôndrias/patologia , Inibição Neural/fisiologia , Neurogênese/fisiologia , Animais , Diferenciação Celular/genética , Linhagem Celular , Células Cultivadas , Proteínas do Domínio Duplacortina , Proteína Duplacortina , Metabolismo Energético/genética , Proteínas de Choque Térmico HSP70/biossíntese , Proteínas de Choque Térmico HSP70/genética , Proteínas de Choque Térmico HSP70/fisiologia , Mediadores da Inflamação/antagonistas & inibidores , Mediadores da Inflamação/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Proteínas Associadas aos Microtúbulos/antagonistas & inibidores , Proteínas Associadas aos Microtúbulos/biossíntese , Mitocôndrias/genética , Mitocôndrias/fisiologia , Inibição Neural/genética , Neurogênese/genética , Neurônios/metabolismo , Neurônios/patologia , Neuropeptídeos/antagonistas & inibidores , Neuropeptídeos/biossíntese , Tiamina/administração & dosagem
17.
BMC Bioinformatics ; 12: 276, 2011 Jul 05.
Artigo em Inglês | MEDLINE | ID: mdl-21729324

RESUMO

BACKGROUND: Activation of the NF-κB transcription factor and its associated gene expression in microglia is a key component in the response to brain injury. Its activation is dynamic and is part of a network of biochemical species with multiple feedback regulatory mechanisms. Mathematical modeling, which has been instrumental for understanding the NF-κB response in other cell types, offers a valuable tool to investigate the regulation of NF-κB activation in microglia at a systems level. RESULTS: We quantify the dynamic response of NF-κB activation and activation of the upstream kinase IKK using ELISA measurements of a microglial cell line following treatment with the pro-inflammatory cytokine TNFα. A new mathematical model is developed based on these data sets using a modular procedure that exploits the feedback structure of the network. We show that the new model requires previously unmodeled dynamics involved in the stimulus-induced degradation of the inhibitor IκBα in order to properly describe microglial NF-κB activation in a statistically consistent manner. This suggests a more prominent role for the ubiquitin-proteasome system in regulating the activation of NF-κB to inflammatory stimuli. We also find that the introduction of nonlinearities in the kinetics of IKK activation and inactivation is essential for proper characterization of transient IKK activity and corresponds to known biological mechanisms. Numerical analyses of the model highlight key regulators of the microglial NF-κB response, as well as those governing IKK activation. Results illustrate the dynamic regulatory mechanisms and the robust yet fragile nature of the negative feedback regulated network. CONCLUSIONS: We have developed a new mathematical model that incorporates previously unmodeled dynamics to characterize the dynamic response of the NF-κB signaling network in microglia. This model is the first of its kind for microglia and provides a tool for the quantitative, systems level study the dynamic cellular response to inflammatory stimuli.


Assuntos
Isquemia Encefálica/metabolismo , Microglia/metabolismo , NF-kappa B/metabolismo , Transdução de Sinais , Animais , Isquemia Encefálica/patologia , Linhagem Celular , Citocinas/metabolismo , Regulação da Expressão Gênica , Quinase I-kappa B/metabolismo , Inflamação/metabolismo , Camundongos , NF-kappa B/antagonistas & inibidores , Fator de Transcrição RelA/metabolismo , Fator de Necrose Tumoral alfa/metabolismo
18.
Stroke ; 42(7): 2026-32, 2011 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-21597016

RESUMO

BACKGROUND AND PURPOSE: Stroke causes brain injury with activation of an inflammatory response that can contribute to injury. We tested the hypothesis that the anti-inflammatory cytokine interleukin-4 (IL-4) reduces injury after stroke using IL-4 knockout (KO) adult male mice. METHODS: IL-4 KO and wild-type mice were subjected to transient middle cerebral artery occlusion. Outcome was assessed by triphenyltetrazolium chloride staining for infarct volume, neuroscore and spontaneous activity for behavioral outcome, and immunostaining and stereological counting for cellular response. RESULTS: Infarction volume at 24 hours was significantly larger in IL-4 KO mice, neurological score was significantly worse, and spontaneous activity was reduced compared with wild-type mice. Increased macrophage/microglial infiltration, increased numbers of myeloperoxidase-positive cells, and increased Th1/Th2 ratio were observed in the infarct core in IL-4 KO mice. Reduced astrocyte activation was observed in the cortical penumbra in IL-4 KO mice. Recombinant IL-4 administered intracerebroventricularly before middle cerebral artery occlusion significantly reduced infarct volume, improved neurological score, reduced macrophages/microglia, and lowered the Th1/Th2 ratio in IL-4 KO mice, but not in wild-type. CONCLUSIONS: Loss of IL-4 signaling in KO mice was associated with worse outcome, and this was reversed by giving exogenous IL-4. Worsened outcome was associated with increased inflammation in the core, which was reversed in IL-4 KO but not significantly changed in wild-type mice by exogenous IL-4. This is consistent with IL-4 signaling leading to reduced inflammation in the core and a possible beneficial role for activated astrocytes in the penumbra.


Assuntos
Lesões Encefálicas/etiologia , Lesões Encefálicas/genética , Interleucina-4/genética , Ataque Isquêmico Transitório/complicações , Animais , Astrócitos/metabolismo , Modelos Animais de Doenças , Inflamação , Ataque Isquêmico Transitório/genética , Macrófagos/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Knockout , Proteínas Recombinantes/uso terapêutico , Transdução de Sinais , Resultado do Tratamento
19.
J Neurosci Res ; 89(12): 1989-96, 2011 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-21910136

RESUMO

The process of neurogenesis continues throughout life, with thousands of new neurons generated every day in the mammalian brain. Impairment of hippocampal neurogenesis has been suggested to be involved in neurodegenerative conditions, including the cognitive decline associated with aging, Alzheimer's disease, Parkinson's disease, and ionizing radiation. These neurodegenerative conditions are all characterized by proinflammatory changes and increased numbers of activated microglia. Activated microglia produce a variety of proinflammatory factors, including interleukin-6, tumor necrosis factor-α, reactive oxygen species, and nitric oxide, all of which are antineurogenic. These same factors have also been shown to suppress mitochondrial function, but the role of mitochondria in neurogenesis remains barely investigated. This brief review summarizes the findings of several studies that support a role for mitochondrial impairment as part of the mechanism of the reduction of neurogenesis associated with inflammation.


Assuntos
Células-Tronco Adultas/metabolismo , Inflamação/fisiopatologia , Mitocôndrias/metabolismo , Mitocôndrias/patologia , Células-Tronco Neurais/metabolismo , Neurogênese/fisiologia , Células-Tronco Adultas/patologia , Animais , Humanos , Degeneração Neural/fisiopatologia , Células-Tronco Neurais/patologia
20.
Anesthesiology ; 114(4): 891-900, 2011 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-21317632

RESUMO

BACKGROUND: Problems with learning and memory are common after surgery in the elderly and are associated with high morbidity. Heat shock protein 72 (Hsp72) confers neuroprotection against acute neurologic injury. We hypothesized that overexpression of Hsp72 would prevent the development of postoperative memory loss. METHODS: C57BL/6 wild-type and Hsp72 overexpressing transgenic mice were randomly allocated to the following: control, isoflurane anesthesia alone, or tibial fracture during isoflurane anesthesia. Animals were trained 24 h before surgery using a fear conditioning protocol and assessed in their training environment and in a novel context on posttreatment days 1, 3, and 7. Microglial activation was assessed by immunostaining. RESULTS: Adult male C57BL/6 wild-type mice exhibited reduced memory evidenced by a decreased percentage freezing time on days 1 and 3 after anesthesia alone (58.8 ± 5, 46.5 ± 5 mean ± SEM) and after surgery (53.4 ± 6, 44.1 ± 7), compared with controls (78.8 ± 5, 63.4 ± 6; P < 0.05 and P < 0.001, respectively). Hsp72 mice showed no difference by treatment on any day. Similarly, nonhippocampal-dependent memory was significantly impaired on days 1 and 3 after surgery and day 3 after anesthesia. The genotype effect was significant on days 1 and 7. CD68-immunopositive activated microglia in the hippocampus varied modestly with subregion and time; on day 7, there was a significant treatment effect with no genotype effect, with more activated microglia after surgery in all regions. CONCLUSION: Hsp72 overexpression is associated with prevention of postoperative hippocampal-dependent and -independent memory deficit induced by anesthesia and/or surgery. Memory deficit is not correlated with numbers of activated hippocampal microglia.


Assuntos
Anestesia Geral/efeitos adversos , Proteínas de Choque Térmico HSP72/metabolismo , Transtornos da Memória/induzido quimicamente , Transtornos da Memória/prevenção & controle , Ortopedia , Animais , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Período Pós-Operatório , Distribuição Aleatória , Fatores de Tempo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA