Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 20
Filtrar
1.
Stroke ; 49(3): 630-637, 2018 03.
Artigo em Inglês | MEDLINE | ID: mdl-29371434

RESUMO

BACKGROUND AND PURPOSE: Oxidative stress is an early response to cerebral ischemia and is likely to play an important role in the pathogenesis of cerebral ischemic injury. We sought to evaluate whether hyperacute plasma concentrations of biomarkers of oxidative stress, inflammation, and tissue damage predict infarct growth (IG). METHODS: We prospectively measured plasma F2-isoprostane (F2-isoP), urinary 8-oxo-7,8-dihydro-2'-deoxyguoanosine, plasma oxygen radical absorbance capacity assay, high sensitivity C reactive protein, and matrix metalloproteinase 2 and 9 in consecutive patients with acute ischemic stroke presenting within 9 hours of symptom onset. Patients with baseline diffusion-weighted magnetic resonance imaging and follow-up diffusion-weighted imaging or computed tomographic scan were included to evaluate the final infarct volume. Baseline diffusion-weighted imaging volume and final infarct volume were analyzed using semiautomated volumetric method. IG volume was defined as the difference between final infarct volume and baseline diffusion-weighted imaging volume. RESULTS: A total of 220 acute ischemic stroke subjects were included in the final analysis. One hundred seventy of these had IG. Baseline F2-isoP significantly correlated with IG volume (Spearman ρ=0.20; P=0.005) and final infarct volume (Spearman ρ=0.19; P=0.009). In a multivariate binary logistic regression model, baseline F2-isoP emerged as an independent predictor of the occurrence of IG (odds ratio, 2.57; 95% confidence interval, 1.37-4.83; P=0.007). In a multivariate linear regression model, baseline F2-isoP was independently associated with IG volume (B, 0.38; 95% confidence interval, 0.04-0.72; P=0.03). CONCLUSIONS: Elevated hyperacute plasma F2-isoP concentrations independently predict the occurrence of IG and IG volume in patients with acute ischemic stroke. If validated in future studies, measuring plasma F2-isoP might be helpful in the acute setting to stratify patients with acute ischemic stroke for relative severity of ischemic injury and expected progression.


Assuntos
Infarto Encefálico/sangue , Lesões Encefálicas/sangue , Proteína C-Reativa/metabolismo , F2-Isoprostanos/sangue , Estresse Oxidativo , Idoso , Idoso de 80 Anos ou mais , Biomarcadores/sangue , Infarto Encefálico/patologia , Lesões Encefálicas/patologia , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Valor Preditivo dos Testes , Estudos Prospectivos
2.
Cell Mol Life Sci ; 71(6): 1055-66, 2014 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-24132511

RESUMO

It is now relatively well accepted that the cerebrovascular system does not merely provide inert pipes for blood delivery to the brain. Cerebral endothelial cells may compose an embedded bunker of trophic factors that contribute to brain homeostasis and function. Recent findings suggest that soluble factors from cerebral endothelial cells nourish neighboring cells, such as neurons and astrocytes. Although data are strongest in supporting mechanisms of endothelial-neuron and/or endothelial-astrocyte trophic coupling, it is likely that similar interactions also exist between cerebral endothelial cells and oligodendrocyte lineage cells. In this mini-review, we summarize current advances in the field of endothelial-oligodendrocyte trophic coupling. These endothelial-oligodendrocyte interactions may comprise the oligovascular niche to maintain their cellular functions and sustain ongoing angiogenesis/oligodendrogenesis. Importantly, it should be noted that the cell-cell interactions are not static-the trophic coupling is disturbed under acute phase after brain injury, but would be recovered in the chronic phase to promote brain remodeling and repair. Oligodendrocyte lineage cells play critical roles in white matter function, and under pathological conditions, oligodendrocyte dysfunction lead to white matter damage. Therefore, a deeper understanding of the mechanisms of endothelial-oligodendrocyte trophic coupling may lead to new therapeutic approaches for white matter-related diseases, such as stroke or vascular dementia.


Assuntos
Comunicação Celular/fisiologia , Células Endoteliais/metabolismo , Endotélio Vascular/metabolismo , Oligodendroglia/metabolismo , Adrenomedulina/metabolismo , Animais , Encéfalo/irrigação sanguínea , Fator Neurotrófico Derivado do Encéfalo/metabolismo , Endotélio Vascular/citologia , Fator 2 de Crescimento de Fibroblastos/metabolismo , Humanos , Metaloproteinases da Matriz/metabolismo , Neovascularização Fisiológica , Neurônios/metabolismo , Oligodendroglia/citologia , Ratos , Transdução de Sinais , Fator de Crescimento Transformador beta/metabolismo , Fator A de Crescimento do Endotélio Vascular/metabolismo
3.
Proc Natl Acad Sci U S A ; 109(19): 7505-10, 2012 May 08.
Artigo em Inglês | MEDLINE | ID: mdl-22529378

RESUMO

Crosstalk between the brain and systemic responses in blood is increasingly suspected of playing critical roles in stroke. However, how this communication takes place remains to be fully understood. Here, we show that reactive astrocytes can release a damage-associated molecular-pattern molecule called high-mobility-group-box-1 (HMGB1) that promotes endothelial progenitor cell (EPC)-mediated neurovascular remodeling during stroke recovery. Conditioned media from reactive astrocytes increase EPC proliferation in vitro. siRNA suppression of HMGB1 in astrocytes or blockade of the HMGB1 receptor for advanced glycation endproducts in EPCs prevents this effect. In a mouse model of focal cerebral ischemia, reactive astrocytes in the peri-infarct cortex up-regulate HMGB1 at 14 d poststroke, along with an accumulation of endogenous EPCs. In vivo siRNA suppression of HMGB1 blocks this EPC response, reduces peri-infact angiogenesis, and worsens neurological deficits. Taken together, these molecular and in vivo findings support a previously undescribed mechanism of crosstalk between reactive astrocytes and EPCs wherein HMGB1 promotes neurovascular remodeling and functional recovery after stroke and brain injury.


Assuntos
Astrócitos/metabolismo , Células Endoteliais/metabolismo , Proteína HMGB1/metabolismo , Células-Tronco/metabolismo , Animais , Animais Recém-Nascidos , Astrócitos/citologia , Astrócitos/efeitos dos fármacos , Western Blotting , Isquemia Encefálica/genética , Isquemia Encefálica/metabolismo , Isquemia Encefálica/fisiopatologia , Proliferação de Células/efeitos dos fármacos , Células Cultivadas , Infarto Cerebral/genética , Infarto Cerebral/metabolismo , Infarto Cerebral/fisiopatologia , Meios de Cultivo Condicionados/farmacologia , Modelos Animais de Doenças , Células Endoteliais/citologia , Células Endoteliais/efeitos dos fármacos , Proteína HMGB1/genética , Proteína HMGB1/farmacologia , Humanos , Imuno-Histoquímica , Interleucina-1beta/farmacologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Interferência de RNA , Ratos , Ratos Sprague-Dawley , Recuperação de Função Fisiológica , Células-Tronco/citologia , Células-Tronco/efeitos dos fármacos , Acidente Vascular Cerebral/genética , Acidente Vascular Cerebral/metabolismo , Acidente Vascular Cerebral/fisiopatologia , Fatores de Tempo
4.
Stroke ; 44(12): 3516-21, 2013 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-24072001

RESUMO

BACKGROUND AND PURPOSE: White matter injury caused by cerebral hypoperfusion may contribute to the pathophysiology of vascular dementia and stroke, but the underlying mechanisms remain to be fully defined. Here, we test the hypothesis that oxidative stress interferes with endogenous white matter repair by disrupting renewal processes mediated by oligodendrocyte precursor cells (OPCs). METHODS: In vitro, primary rat OPCs were exposed to sublethal CoCl2 for 7 days to induce prolonged chemical hypoxic stress. Then, OPC proliferation/differentiation was assessed. In vivo, prolonged cerebral hypoperfusion was induced by bilateral common carotid artery stenosis in mice. Then, reactive oxygen species production, myelin density, oligodendrocyte versus OPC counts, and cognitive function were evaluated. To block oxidative stress, OPCs and mice were treated with the radical scavenger edaravone. RESULTS: Prolonged chemical hypoxic stress suppressed OPC differentiation in vitro. Radical scavenging with edaravone ameliorated these effects. After 28 days of cerebral hypoperfusion in vivo, reactive oxygen species levels were increased in damaged white matter, along with the suppression of OPC-to-oligodendrocyte differentiation and loss of myelin staining. Concomitantly, mice showed functional deficits in working memory. Radical scavenging with edaravone rescued OPC differentiation, ameliorated myelin loss, and restored working memory function. CONCLUSIONS: Our proof-of-concept study demonstrates that after prolonged cerebral hypoperfusion, oxidative stress interferes with white matter repair by disrupting OPC renewal mechanisms. Radical scavengers may provide a potential therapeutic approach for white matter injury in vascular dementia and stroke.


Assuntos
Isquemia Encefálica/fisiopatologia , Fibras Nervosas Mielinizadas/patologia , Células-Tronco Neurais/fisiologia , Neurogênese/fisiologia , Oligodendroglia/fisiologia , Estresse Oxidativo/fisiologia , Animais , Antipirina/análogos & derivados , Antipirina/farmacologia , Isquemia Encefálica/metabolismo , Isquemia Encefálica/patologia , Modelos Animais de Doenças , Edaravone , Sequestradores de Radicais Livres/farmacologia , Camundongos , Fibras Nervosas Mielinizadas/efeitos dos fármacos , Fibras Nervosas Mielinizadas/fisiologia , Células-Tronco Neurais/efeitos dos fármacos , Células-Tronco Neurais/patologia , Oligodendroglia/efeitos dos fármacos , Oligodendroglia/patologia , Estresse Oxidativo/efeitos dos fármacos , Espécies Reativas de Oxigênio/metabolismo
5.
Stroke ; 44(9): 2573-8, 2013 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-23881957

RESUMO

BACKGROUND AND PURPOSE: Aging is one of the major risk factors for white matter injury in cerebrovascular disease. However, the effects of age on the mechanisms of injury/repair in white matter remain to be fully elucidated. Here, we ask whether, compared with young brains, white matter regions in older brains may be more vulnerable in part because of decreased rates of compensatory oligodendrogenesis after injury. METHODS: A mouse model of prolonged cerebral hypoperfusion was prepared by bilateral common carotid artery stenosis in 2-month and 8-month-old mice. Matching in vitro studies were performed by subjecting oligodendrocyte precursor cells to sublethal 7-day CoCl2 treatment to induce chemical hypoxic stress. RESULTS: Baseline myelin density in the corpus callosum was similar in 2-month and 8-month-old mice. But after induction of prolonged cerebral hypoperfusion, older mice showed more severe white matter injury together with worse deficits in working memory. The numbers of newborn oligodendrocytes and their precursors were increased by cerebral hypoperfusion in young mice, whereas these endogenous responses were significantly dampened in older mice. Defects in cyclic AMP response element-binding protein signaling may be involved because activating cyclic AMP response element-binding protein with the type-III phosphodiesterase inhibitor cilostazol in older mice restored the differentiation of oligodendrocyte precursor cells, alleviated myelin loss, and improved cognitive dysfunction during cerebral hypoperfusion. Cell culture systems confirmed that cilostazol promoted the differentiation of oligodendrocyte precursor cells. CONCLUSIONS: An age-related decline in cyclic AMP response element-binding protein-mediated oligodendrogenesis may compromise endogenous white matter repair mechanisms, and therefore, drugs that activate cyclic AMP response element-binding protein signaling provide a potential therapeutic approach for treating white matter injury in aging brains.


Assuntos
Envelhecimento/fisiologia , Leucoencefalopatias/tratamento farmacológico , Células-Tronco Neurais , Neurogênese/fisiologia , Fármacos Neuroprotetores/farmacologia , Envelhecimento/patologia , Animais , Antimutagênicos/administração & dosagem , Antimutagênicos/toxicidade , Células Cultivadas , Cobalto/administração & dosagem , Cobalto/toxicidade , Transtornos Cognitivos/tratamento farmacológico , Transtornos Cognitivos/etiologia , Transtornos Cognitivos/patologia , Corpo Caloso/patologia , Proteína de Ligação ao Elemento de Resposta ao AMP Cíclico/efeitos dos fármacos , Proteína de Ligação ao Elemento de Resposta ao AMP Cíclico/fisiologia , Modelos Animais de Doenças , Hipóxia Encefálica/induzido quimicamente , Hipóxia Encefálica/complicações , Leucoencefalopatias/complicações , Leucoencefalopatias/etiologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Bainha de Mielina/efeitos dos fármacos , Bainha de Mielina/patologia , Células-Tronco Neurais/efeitos dos fármacos , Células-Tronco Neurais/patologia , Neurogênese/efeitos dos fármacos
6.
J Neurochem ; 125(2): 273-80, 2013 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-23227954

RESUMO

High-mobility group box 1 (HMGB1) was initially described as a damage-associated-molecular-pattern (DAMP) mediator that worsens acute brain injury after stroke. But, recent findings suggest that HMGB1 can play a surprisingly beneficial role during stroke recovery by promoting endothelial progenitor cell (EPC) function and vascular remodeling in cortical gray matter. Here, we ask whether HMGB1 may also influence EPC responses in white matter injury. The standard lysophosphatidylcholine (LPC) injection model was used to induce focal demyelination in the corpus callosum of mice. Immunostaining showed that within the focal white matter lesions, HMGB1 was up-regulated in GFAP-positive reactive astrocytes, along with the accumulation of Flk1/CD34-double-positive EPCs that expressed pro-recovery mediators such as brain-derived neurotrophic factor and basic fibroblast growth factor. Astrocyte-EPC signaling required the HMGB1 receptor RAGE as treatment with RAGE-neutralizing antibody significantly decreased EPC accumulation. Moreover, suppression of HMGB1 with siRNA in vivo significantly decreased EPC numbers in damaged white matter as well as proliferated endothelial cell numbers. Finally, in vitro cell culture systems confirmed that HMGB1 directly affected EPC function such as migration and tube formation. Taken together, our findings suggest that HMGB1 from reactive astrocytes may attract EPCs to promote recovery after white matter injury.


Assuntos
Astrócitos/metabolismo , Células Progenitoras Endoteliais/metabolismo , Proteína HMGB1/metabolismo , Substância Branca/lesões , Animais , Western Blotting , Lesões Encefálicas/metabolismo , Modelos Animais de Doenças , Citometria de Fluxo , Humanos , Imuno-Histoquímica , Masculino , Camundongos , Camundongos Endogâmicos C57BL , RNA Interferente Pequeno , Ratos , Substância Branca/metabolismo
7.
Acta Neurochir Suppl ; 118: 31-8, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23564100

RESUMO

Stroke induces a highly complex web of pathophysiology that usually leads to serious long-term -disability. Molecules from the damage-associated molecular pattern (DAMP) family immediately increase after stroke. DAMPs are known to cause massive inflammation and brain damage. Thus, they may be targets for neuroprotection. However, emerging data now suggest that DAMPs may not always be detrimental. The high-mobility group box1 (HMGB1) protein is discussed as an example of this idea. During the acute phase after stroke, HMGB1 amplifies neuroinflammation. But during the brain remodeling phase of stroke recovery, HMGB1 can mediate beneficial plasticity and enhance stem and progenitor cell recruitment, proliferation, and differentiation within damaged brain. These emerging findings support the hypothesis that HMGB1 might be an important molecule for regulating stem and progenitor cell therapies in stroke patients.


Assuntos
Encéfalo/patologia , Proteína HMGB1/metabolismo , Células-Tronco/fisiologia , Acidente Vascular Cerebral/patologia , Animais , Diferenciação Celular , Proliferação de Células , Humanos , Acidente Vascular Cerebral/terapia
8.
J Neurosci ; 31(29): 10666-70, 2011 Jul 20.
Artigo em Inglês | MEDLINE | ID: mdl-21775609

RESUMO

Originally identified as an angiogenic factor, vascular endothelial growth factor (VEGF-A) is now known to play multiple roles in the CNS, including the direct regulation of neuronal and astrocytic functions. Here, we ask whether VEGF-A can also have a novel role in white matter by modulating oligodendrocyte precursor cells (OPCs). OPCs were cultured from rat neonatal cortex. Expression of VEGF-receptor2/KDR/Flk-1 was confirmed with Western blot and immunostaining. VEGF-A did not affect proliferation or differentiation in OPC cultures, but VEGF-A promoted OPC migration in a concentration-dependent manner. Consistent with this migration phenotype, VEGF-A-treated OPCs showed reorganization of actin cytoskeleton in leading-edge processes. VEGF-A-induced migration and actin reorganization were inhibited by an anti-Flk-1 receptor-blocking antibody. Mechanistically, VEGF-A induced binding of focal adhesion kinase (FAK) with paxillin. The FAK inhibitor PF573228 reduced VEGF-A-induced OPC migration. VEGF-A signaling also evoked a transient rise in reactive oxygen species (ROS), and OPC migration was increased when antioxidants were removed from the culture media. Our findings demonstrate that VEGF-A can induce OPC migration via an ROS- and FAK-dependent mechanism, and suggest a novel role for VEGF-A in white-matter maintenance and homeostasis.


Assuntos
Movimento Celular/genética , Oligodendroglia/efeitos dos fármacos , Células-Tronco/efeitos dos fármacos , Fator A de Crescimento do Endotélio Vascular/farmacologia , Actinas/metabolismo , Animais , Animais Recém-Nascidos , Antígenos/metabolismo , Antioxidantes/farmacologia , Contagem de Células , Diferenciação Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Células Cultivadas , Córtex Cerebral/citologia , Relação Dose-Resposta a Droga , Inibidores Enzimáticos/farmacologia , Feminino , Gangliosídeos/metabolismo , Masculino , Oligodendroglia/fisiologia , Paxilina/farmacocinética , Ligação Proteica/efeitos dos fármacos , Proteoglicanas/metabolismo , Quinolonas/farmacologia , Ratos , Ratos Sprague-Dawley , Células-Tronco/fisiologia , Sulfonas/farmacologia , Receptor 1 de Fatores de Crescimento do Endotélio Vascular/metabolismo
9.
Glia ; 60(6): 875-81, 2012 May.
Artigo em Inglês | MEDLINE | ID: mdl-22392631

RESUMO

After stroke and brain injury, cortical gray matter recovery involves mechanisms of neurovascular matrix remodeling. In white matter, however, the mechanisms of recovery remain unclear. In this study, we demonstrate that oligodendrocytes secrete matrix metalloproteinase-9 (MMP-9), which accelerates the angiogenic response after white matter injury. In primary oligodendrocyte cultures, treatment with the proinflammatory cytokine interleukin-1ß (IL-1ß) induced an upregulation and secretion of MMP-9. Conditioned media from IL-1ß-stimulated oligodendrocytes significantly amplified matrigel tube formation in brain endothelial cells, indicating that MMP-9 from oligodendrocytes can promote angiogenesis in vitro. Next, we asked whether similar signals and substrates operate after white matter injury in vivo. Focal white matter injury and demyelination was induced in mice via stereotactic injection of lysophosphatidylcholine into corpus callosum. Western blot analysis showed that IL-1ß expression was increased in damaged white matter. Immunostaining demonstrated MMP-9 signals in myelin-associated oligodendrocytic basic protein-positive oligodendrocytes. Treatment with an IL-1ß-neutralizing antibody suppressed the MMP-9 response in oligodendrocytes. Finally, we confirmed that the broad spectrum MMP inhibitor GM6001 inhibited angiogenesis around the injury area in this white matter injury model. In gray matter, a neurovascular niche promotes cortical recovery after brain injury. Our study suggests that an analogous oligovascular niche may mediate recovery in white matter.


Assuntos
Lesões Encefálicas/fisiopatologia , Comunicação Celular/fisiologia , Córtex Cerebral/citologia , Endotélio/fisiologia , Oligodendroglia/fisiologia , Animais , Animais Recém-Nascidos , Fatores de Transcrição Hélice-Alça-Hélice Básicos/metabolismo , Lesões Encefálicas/patologia , Comunicação Celular/efeitos dos fármacos , Células Cultivadas , Meios de Cultivo Condicionados/farmacologia , Dipeptídeos/farmacologia , Modelos Animais de Doenças , Endotélio/efeitos dos fármacos , Técnicas In Vitro , Interleucina-1beta/metabolismo , L-Lactato Desidrogenase/metabolismo , Masculino , Proteína Básica da Mielina/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Fator de Transcrição 2 de Oligodendrócitos , Oligodendroglia/química , Oligodendroglia/efeitos dos fármacos , Molécula-1 de Adesão Celular Endotelial a Plaquetas/metabolismo , Inibidores de Proteases/farmacologia , Ratos , Ratos Sprague-Dawley
10.
Neurology ; 93(13): e1288-e1298, 2019 09 24.
Artigo em Inglês | MEDLINE | ID: mdl-31455665

RESUMO

OBJECTIVES: To assess whether plasma biomarkers of oxidative stress predict diffusion-perfusion mismatch in patients with acute ischemic stroke (AIS). METHODS: We measured plasma levels of oxidative stress biomarkers such as F2-isoprostanes (F2-isoPs), total and perchloric acid Oxygen Radical Absorbance Capacity (ORACTOT and ORACPCA), urinary levels of 8-oxo-7,8-dihydro-2'-deoxyguoanosine, and inflammatory and tissue-damage biomarkers (high-sensitivity C-reactive protein, matrix metalloproteinase-2 and -9) in a prospective study of patients with AIS presenting within 9 hours of symptom onset. Diffusion-weighted (DWI) and perfusion-weighted (PWI) MRI sequences were analyzed with a semiautomated volumetric method. Mismatch was defined as baseline mean transit time volume minus DWI volume. A percent mismatch cutoff of >20% was considered clinically significant. A stricter definition of mismatch was also used. Mismatch salvage was the region free of overlap by final infarction. RESULTS: Mismatch >20% was present in 153 of 216 (70.8%) patients (mean [±SD] age 69.2 ± 14.3 years, 41.2% women). Patients with mismatch >20% were more likely to have higher baseline plasma levels of ORACPCA (p = 0.020) and F2-isoPs (p = 0.145). Multivariate binary logistic regression demonstrated that lnF2-isoP (odds ratio [OR] 2.44, 95% confidence interval [CI] 1.19-4.98, p = 0.014) and lnORACPCA (OR 4.18, 95% CI 1.41-12.41, p = 0.010) were independent predictors of >20% PWI-DWI mismatch and the stricter mismatch definition, respectively. lnORACTOT significantly predicted mismatch salvage volume (>20% mismatch p = 0.010, stricter mismatch definition p = 0.003). CONCLUSIONS: Elevated hyperacute plasma levels of F2-isoP and ORAC are associated with radiographic evidence of mismatch and mismatch salvage in patients with AIS. If validated, these findings may add to our understanding of the role of oxidative stress in cerebral tissue fate during acute ischemia.


Assuntos
Biomarcadores/análise , Isquemia Encefálica/metabolismo , Estresse Oxidativo/fisiologia , Acidente Vascular Cerebral/metabolismo , Adulto , Idoso , Isquemia Encefálica/diagnóstico , Isquemia Encefálica/fisiopatologia , Circulação Cerebrovascular/fisiologia , Imagem de Difusão por Ressonância Magnética/métodos , Feminino , Humanos , Processamento de Imagem Assistida por Computador/métodos , Isquemia/metabolismo , Isquemia/fisiopatologia , Imageamento por Ressonância Magnética/métodos , Masculino , Pessoa de Meia-Idade , Estudos Prospectivos , Acidente Vascular Cerebral/diagnóstico , Acidente Vascular Cerebral/fisiopatologia
11.
Neurosci Lett ; 668: 120-125, 2018 03 06.
Artigo em Inglês | MEDLINE | ID: mdl-29337010

RESUMO

Oligodendrocyte precursor cells (OPCs) play critical roles in maintaining the number of oligodendrocytes in white matter. Previously, we have shown that oxidative stress dampens oligodendrocyte regeneration after white matter damage, while a clinically proven radical scavenger, edaravone, supports oligodendrocyte repopulation. However, it is not known how edaravone exerts this beneficial effect against oxidative stress. Using in vivo and in vitro experiments, we have examined whether edaravone exhibits direct OPC-protective effects. For in vivo experiments, prolonged cerebral hypoperfusion was induced by bilateral common carotid artery stenosis in mice. OPC damage was observed on day 14 after the onset of cerebral hypoperfusion, and edaravone was demonstrated to decrease OPC death in cerebral white matter. In vitro experiments also confirmed that edaravone reduced oxidative-stress-induced OPC death. Because white matter damage is a major hallmark of many neurological diseases, and OPCs are instrumental in white matter repair after injury, our current study supports the idea that radical scavengers may provide a potential therapeutic approach for white matter related diseases.


Assuntos
Antipirina/análogos & derivados , Isquemia Encefálica/tratamento farmacológico , Sequestradores de Radicais Livres/farmacologia , Fármacos Neuroprotetores/farmacologia , Células Precursoras de Oligodendrócitos/efeitos dos fármacos , Estresse Oxidativo/efeitos dos fármacos , Substância Branca/efeitos dos fármacos , Substância Branca/lesões , Animais , Antipirina/farmacologia , Edaravone , Masculino , Camundongos , Camundongos Endogâmicos C57BL
12.
Neurosci Lett ; 633: 7-13, 2016 10 28.
Artigo em Inglês | MEDLINE | ID: mdl-27589890

RESUMO

A free radical scavenger edaravone is clinically used in Japan for acute stroke, and several basic researches have carefully examined the mechanisms of edaravone's protective effects. However, its actions on pro-inflammatory responses under stroke are still understudied. In this study, we subjected adult male Sprague-Dawley rats to 90-min middle cerebral artery (MCA) occlusion followed by reperfusion. Edaravone was treated twice via tail vein; after MCA occlusion and after reperfusion. As expected, edaravone-treated group showed less infarct volume and edema formation compared with control group at 24-h after an ischemic onset. Furthermore, edaravone reduced the levels of plasma interleukin (IL)-1ß and matrix metalloproteinase-9 at 3-h after ischemic onset. Several molecules besides IL-1ß and MMP-9 are involved in inflammatory responses under stroke conditions. Therefore, we also examined whether edaravone treatment could decrease a wide range of pro-inflammatory cytokines/chemokines by testing rat plasma samples with a rat cytokine array. MCAO rats showed elevations in plasma levels of CINC-1, Fractalkine, IL-1α, IL-1ra, IL-6, IL-10, IP-10, MIG, MIP-1α, and MIP-3α, and all these increases were reduced by edaravone treatment. These data suggest that free radical scavengers may reduce systemic inflammatory responses under acute stroke conditions, and therefore, oxidative stress can be still a viable target for acute stroke therapy.


Assuntos
Antipirina/análogos & derivados , Citocinas/sangue , Sequestradores de Radicais Livres/uso terapêutico , Ataque Isquêmico Transitório/tratamento farmacológico , Fármacos Neuroprotetores/uso terapêutico , Animais , Antipirina/uso terapêutico , Edema Encefálico/tratamento farmacológico , Infarto Cerebral/tratamento farmacológico , Infarto Cerebral/patologia , Edaravone , Inflamação/tratamento farmacológico , Interleucina-1beta/sangue , Ataque Isquêmico Transitório/metabolismo , Ataque Isquêmico Transitório/patologia , Masculino , Metaloproteinase 9 da Matriz/sangue , Ratos Sprague-Dawley , Regulação para Cima
13.
J Cereb Blood Flow Metab ; 36(4): 781-93, 2016 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-26661156

RESUMO

There are numerous barriers to white matter repair after central nervous system injury and the underlying mechanisms remain to be fully understood. In this study, we propose the hypothesis that inflammatory macrophages in damaged white matter attack oligodendrocyte precursor cells via toll-like receptor 4 signaling thus interfering with this endogenous progenitor recovery mechanism. Primary cell culture experiments demonstrate that peritoneal macrophages can attack and digest oligodendrocyte precursor cells via toll-like receptor 4 signaling, and this phagocytosis of oligodendrocyte precursor cells can be inhibited by using CD200-Fc to downregulate toll-like receptor 4. In an in vivo model of white matter ischemia induced by endothelin-1, treatment with CD200-Fc suppressed toll-like receptor 4 expression in peripherally circulating macrophages, thus restraining macrophage phagocytosis of oligodendrocyte precursor cells and leading to improved myelination. Taken together, these findings suggest that deleterious macrophage effects may occur after white matter ischemia, whereby macrophages attack oligodendrocyte precursor cells and interfere with endogenous recovery responses. Targeting this pathway with CD200 may offer a novel therapeutic approach to amplify endogenous oligodendrocyte precursor cell-mediated repair of white matter damage in mammalian brain.


Assuntos
Antígenos CD/farmacologia , Macrófagos/efeitos dos fármacos , Células-Tronco Neurais/efeitos dos fármacos , Oligodendroglia/efeitos dos fármacos , Receptor 4 Toll-Like/biossíntese , Animais , Isquemia Encefálica/patologia , Regulação para Baixo/efeitos dos fármacos , L-Lactato Desidrogenase/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Bainha de Mielina/metabolismo , Fagocitose/efeitos dos fármacos , Receptor 4 Toll-Like/genética , Substância Branca/patologia
14.
Clin Cancer Res ; 10(1 Pt 1): 228-33, 2004 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-14734474

RESUMO

PURPOSE: Although the genetic alterations in glioblastoma have been well characterized, reports regarding their prognostic effects have been inconsistent. EXPERIMENTAL DESIGN: In this series of 140 consecutive cases of glioblastoma treated at a single center, we analyzed the frequency, age dependency and prognostic effects of TP53 mutation, CDKN2A/p16 deletion, EGFR amplification, as well as loss of chromosome 1p, chromosome 10q, and chromosome 19q. The complete set of genetic alterations was available on 60 of 140 patients. RESULTS: In this cohort of glioblastoma cases, TP53 mutation was significantly associated with patient age. The prognostic effects of TP53 mutation, EGFR amplification, CDKN2A/p16 alterations, and loss of chromosome 1p were dependent on the age of the patient. CONCLUSIONS: This is the first observation that the prognostic effects of TP53, 1p, and CDKN2A/p16 alterations are dependent on patient age. These observations concerning the interactions of age and genetic changes in glioblastoma suggest that tumorigenic pathways to glioblastoma vary with the age of the patient and that future molecular marker studies should carefully evaluate the potential age-dependent prognostic effects of these biological variables. The inconsistent or negative prognostic effects of molecular markers reported in prior studies of glioblastoma may be because different effects at different ages may have resulted in a cancellation of an overall effect in the entire cohort.


Assuntos
Inibidor p16 de Quinase Dependente de Ciclina/genética , Receptores ErbB/genética , Amplificação de Genes , Deleção de Genes , Glioblastoma/genética , Mutação/genética , Proteína Supressora de Tumor p53/genética , Adolescente , Adulto , Distribuição por Idade , Idoso , Idoso de 80 Anos ou mais , Neoplasias Encefálicas/genética , Cromossomos Humanos Par 1/genética , Cromossomos Humanos Par 10/genética , Cromossomos Humanos Par 19/genética , Estudos de Coortes , DNA de Neoplasias/genética , Feminino , Frequência do Gene , Humanos , Perda de Heterozigosidade , Masculino , Pessoa de Meia-Idade , Reação em Cadeia da Polimerase , Polimorfismo Conformacional de Fita Simples , Prognóstico , Taxa de Sobrevida
15.
Stem Cell Res ; 12(2): 531-8, 2014 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-24480450

RESUMO

Endothelial progenitor cells (EPCs) may contribute to neurovascular repair after stroke and neurodegeneration. A key step in this process should involve adhesive interactions between EPCs and the targeted cerebral endothelium. Here, we tested the hypothesis that reactive astrocytes may play a critical role in enhancing adhesive interactions and transmigration of EPCs across cerebral endothelial cells. Transiently seeding EPCs onto a monolayer of RBE.4 rat brain endothelial cells resulted in a time-dependent adherence between the two cell types. Blocking ß2 integrins on EPCs or blocking the receptor for advanced glycation endproducts (RAGE) on endothelial cells significantly decreased EPC-endothelial adherence. Next, we tested whether reactive astrocytes can enhance this process by growing EPCs, brain endothelial cells and astrocytes together in a transwell co-culture system. The presence of reactive astrocytes in the lower chamber significantly promoted adherence between EPCs and endothelial cells in the upper chamber. This process involved the release of soluble HMGB1 from reactive astrocytes that then upregulated endothelial expression of RAGE via Egr1 signaling. Directly adding HMGB1 to the transwell system also promoted EPC-endothelial adhesion and accelerated EPC transmigration into the lower chamber. These initial findings provide proof-of-concept that reactive astrocytes promote crosstalk between cerebral endothelium and EPCs. Further investigation of this phenomenon may lead to a better understanding of cell-cell interactions required for neurovascular recovery after stroke.


Assuntos
Astrócitos/citologia , Antígenos CD18/metabolismo , Comunicação Celular/fisiologia , Células Endoteliais/citologia , Proteína HMGB1/metabolismo , Células-Tronco/citologia , Animais , Astrócitos/metabolismo , Adesão Celular/fisiologia , Processos de Crescimento Celular/fisiologia , Células Cultivadas , Células Endoteliais/metabolismo , Ratos , Transdução de Sinais , Células-Tronco/metabolismo
16.
Neurosci Lett ; 573: 40-45, 2014 Jun 24.
Artigo em Inglês | MEDLINE | ID: mdl-24820542

RESUMO

Matrix metalloproteinase-9 (MMP-9) plays key roles in the brain pathophysiology, especially in blood-brain barrier (BBB) breakdown. Therefore, inhibiting MMP-9 activity may be a promising therapy for protecting brains in cerebrovascular diseases. Here we show that in a mouse prolonged cerebral hypoperfusion model, a clinically proven radical scavenger edaravone suppressed MMP-9 and reduced BBB damage in cerebral white matter. Prolonged cerebral hypoperfusion was induced by bilateral common carotid artery stenosis in male adult C57BL/6J mice (10 weeks old). After 7 days of cerebral hypoperfusion, white matter region (e.g. corpus callosum) exhibited significant BBB leakage, assessed by IgG staining. Correspondingly, immunostaining and western blotting showed that MMP-9 was upregulated in the white matter. Edaravone treatment (3mg/kg, i.p. at days 0 and 3) inhibited both BBB leakage and MMP-9 increase. Under the early phase of cerebral hypoperfusion conditions, oligodendrocyte precursor cells (OPCs) mainly contribute to the MMP-9 increase, but our immunostaining data showed that very little OPCs expressed MMP-9 in the edaravone-treated animals at day 7. Therefore, in vitro studies with primary rat OPCs were conducted to examine whether edaravone would directly suppressed MMP-9 expressions in OPCs. OPC cultures were exposed to sub-lethal CoCl2 for 7 days to induce prolonged chemical hypoxic stress. Prolonged chemical hypoxic stress increased MMP-9 expression in OPCs, and radical scavenging with edaravone (10µM for 7 days) ameliorated the increase. Taken together, our proof-of-concept study demonstrates that radical scavengers may provide a potential therapeutic approach for white matter injury by suppressing BBB damage.


Assuntos
Antipirina/análogos & derivados , Barreira Hematoencefálica/efeitos dos fármacos , Córtex Cerebral/efeitos dos fármacos , Sequestradores de Radicais Livres/farmacologia , Hipóxia Encefálica/metabolismo , Metaloproteinase 9 da Matriz/metabolismo , Inibidores de Metaloproteinases de Matriz/farmacologia , Animais , Antipirina/farmacologia , Barreira Hematoencefálica/metabolismo , Artéria Carótida Primitiva , Estenose das Carótidas/complicações , Estenose das Carótidas/metabolismo , Estenose das Carótidas/fisiopatologia , Morte Celular , Sobrevivência Celular , Córtex Cerebral/irrigação sanguínea , Córtex Cerebral/metabolismo , Circulação Cerebrovascular/efeitos dos fármacos , Edaravone , Hipóxia Encefálica/etiologia , Hipóxia Encefálica/patologia , Hipóxia Encefálica/fisiopatologia , Masculino , Camundongos Endogâmicos C57BL , Oligodendroglia/efeitos dos fármacos , Oligodendroglia/metabolismo , Oligodendroglia/patologia , Regulação para Cima , Substância Branca/irrigação sanguínea , Substância Branca/efeitos dos fármacos , Substância Branca/metabolismo
17.
PLoS One ; 9(7): e103174, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25078775

RESUMO

Trophic coupling between cerebral endothelium and their neighboring cells is required for the development and maintenance of blood-brain barrier (BBB) function. Here we report that oligodendrocyte precursor cells (OPCs) secrete soluble factor TGF-ß1 to support BBB integrity. Firstly, we prepared conditioned media from OPC cultures and added them to cerebral endothelial cultures. Our pharmacological experiments showed that OPC-conditioned media increased expressions of tight-junction proteins and decreased in vitro BBB permeability by activating TGB-ß-receptor-MEK/ERK signaling pathway. Secondly, our immuno-electron microscopic observation revealed that in neonatal mouse brains, OPCs attach to cerebral endothelial cells via basal lamina. And finally, we developed a novel transgenic mouse line that TGF-ß1 is knocked down specifically in OPCs. Neonates of these OPC-specific TGF-ß1 deficient mice (OPC-specific TGF-ß1 partial KO mice: PdgfraCre/Tgfb1flox/wt mice or OPC-specific TGF-ß1 total KO mice: PdgfraCre/Tgfb1flox/flox mice) exhibited cerebral hemorrhage and loss of BBB function. Taken together, our current study demonstrates that OPCs increase BBB tightness by upregulating tight junction proteins via TGF-ß signaling. Although astrocytes and pericytes are well-known regulators of BBB maturation and maintenance, these findings indicate that OPCs also play a pivotal role in promoting BBB integrity.


Assuntos
Barreira Hematoencefálica , Células-Tronco Neurais/citologia , Oligodendroglia/citologia , Transdução de Sinais , Fator de Crescimento Transformador beta/metabolismo , Animais , Ratos , Ratos Sprague-Dawley
18.
CNS Neurol Disord Drug Targets ; 12(3): 302-15, 2013 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-23469847

RESUMO

In the past decade, evidence has emerged that there is a variety of bidirectional cell-cell and/or cell-extracellular matrix interactions within the neurovascular unit (NVU), which is composed of neuronal, glial, and vascular cells along with extracellular matrix. Many central nervous system diseases, which lead to NVU dysfunction, have common features such as glial activation/transformation and vascular/blood-brain-barrier alteration. These phenomena show dual opposite roles, harmful at acute phase and beneficial at chronic phase. This diverse heterogeneity may induce biphasic clinical courses, i.e. degenerative and regenerative processes in the context of dynamically coordinated cellcell/ cell-matrix interactions in the NVU. A deeper understanding of the seemingly contradictory actions in cellular levels is essential for NVU protection or regeneration to suppress the deleterious inflammatory reactions and promote adaptive remodeling after central nervous system injury. This mini-review will present an overview of recent progress in the biphasic roles of the NVU and discuss the clinical relevance of NVU responses associated with central nervous system diseases, such as stroke and other chronic neurodegenerative diseases.


Assuntos
Comunicação Celular/fisiologia , Doenças do Sistema Nervoso Central/fisiopatologia , Células Endoteliais/fisiologia , Regeneração Nervosa/fisiologia , Neuroglia/fisiologia , Neurônios/fisiologia , Humanos
19.
J Clin Invest ; 123(2): 782-6, 2013 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-23281396

RESUMO

Oligodendrocyte precursor cells (OPCs) are thought to maintain homeostasis and contribute to long-term repair in adult white matter; however, their roles in the acute phase after brain injury remain unclear. Mice that were subjected to prolonged cerebral hypoperfusion stress developed white matter demyelination over time. Prior to demyelination, we detected increased MMP9 expression, blood-brain barrier (BBB) leakage, and neutrophil infiltration in damaged white matter. Notably, at this early stage, OPCs made up the majority of MMP9-expressing cells. The standard MMP inhibitor GM6001 reduced the early BBB leakage and neutrophil infiltration, indicating that OPC-derived MMP9 induced early BBB disruption after white matter injury. Cell-culture experiments confirmed that OPCs secreted MMP9 under pathological conditions, and conditioned medium prepared from the stressed OPCs weakened endothelial barrier tightness in vitro. Our study reveals that OPCs can rapidly respond to white matter injury and produce MMP9 that disrupts the BBB, indicating that OPCs may mediate injury in white matter under disease conditions.


Assuntos
Barreira Hematoencefálica/patologia , Barreira Hematoencefálica/fisiopatologia , Lesões Encefálicas/patologia , Lesões Encefálicas/fisiopatologia , Oligodendroglia/patologia , Oligodendroglia/fisiologia , Células-Tronco Adultas/patologia , Células-Tronco Adultas/fisiologia , Animais , Barreira Hematoencefálica/efeitos dos fármacos , Dipeptídeos/farmacologia , Modelos Animais de Doenças , Masculino , Metaloproteinase 9 da Matriz/biossíntese , Metaloproteinases da Matriz Secretadas/antagonistas & inibidores , Camundongos , Camundongos Endogâmicos C57BL , Células-Tronco Neurais/patologia , Células-Tronco Neurais/fisiologia , Infiltração de Neutrófilos/efeitos dos fármacos , Infiltração de Neutrófilos/fisiologia , Inibidores de Proteases/farmacologia
20.
Neurosci Lett ; 513(1): 42-6, 2012 Mar 28.
Artigo em Inglês | MEDLINE | ID: mdl-22342913

RESUMO

In gray matter, cerebral endothelium is known to provide trophic support for neighboring cells such as neurons. However, signaling from cerebral endothelium to white matter cells remains to be elucidated. Here, we show that vascular endothelial growth factor (VEGF-A) secreted from cerebral endothelial cells promotes the migration but not the proliferation of oligodendrocyte precursor cells (OPCs). Cultured OPCs were obtained from newborn rat cortex, and treatment with conditioned culture media of cerebral endothelial cells increased the OPC proliferation and migration. Importantly, co-treatment with anti-neutralizing antibody for Flk-1 (VEGF-receptor2) inhibited OPC movement but did not affect OPC propagation. Western blot and flow cytometry analyses confirmed that our cultured cerebral endothelial cells produced VEGF-A and our cultured OPCs expressed Flk-1. Taken together, our current data suggest that cerebral endothelium is supportive for oligodendrocyte lineage cells and VEGF-A may participate in the endothelium-OPC cell-cell signaling. This phenomenon may be important for white matter homeostasis.


Assuntos
Movimento Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Células-Tronco Neurais/efeitos dos fármacos , Oligodendroglia/efeitos dos fármacos , Fator A de Crescimento do Endotélio Vascular/farmacologia , Animais , Biomarcadores/metabolismo , Western Blotting , Meios de Cultivo Condicionados , Células Endoteliais/efeitos dos fármacos , Citometria de Fluxo , Imuno-Histoquímica , Ratos , Ratos Sprague-Dawley , Receptores de Fatores de Crescimento do Endotélio Vascular/metabolismo , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/farmacologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA