Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
1.
Stroke ; 51(12): 3701-3712, 2020 12.
Artigo em Inglês | MEDLINE | ID: mdl-33138691

RESUMO

BACKGROUND AND PURPOSE: Cerebral endothelial cells (CECs) and axons of neurons interact to maintain vascular and neuronal homeostasis and axonal remodeling in normal and ischemic brain, respectively. However, the role of exosomes in the interaction of CECs and axons in brain under normal conditions and after stroke is unknown. METHODS: Exosomes were isolated from CECs of nonischemic rats and is chemic rats (nCEC-exos and isCEC-exos), respectively. A multicompartmental cell culture system was used to separate axons from neuronal cell bodies. RESULTS: Axonal application of nCEC-exos promotes axonal growth of cortical neurons, whereas isCEC-exos further enhance axonal growth than nCEC-exos. Ultrastructural analysis revealed that CEC-exos applied into distal axons were internalized by axons and reached to their parent somata. Bioinformatic analysis revealed that both nCEC-exos and isCEC-exos contain abundant mature miRNAs; however, isCEC-exos exhibit more robust elevation of select miRNAs than nCEC-exos. Mechanistically, axonal application of nCEC-exos and isCEC-exos significantly elevated miRNAs and reduced proteins in distal axons and their parent somata that are involved in inhibiting axonal outgrowth. Blockage of axonal transport suppressed isCEC-exo-altered miRNAs and proteins in somata but not in distal axons. CONCLUSIONS: nCEC-exos and isCEC-exos facilitate axonal growth by altering miRNAs and their target protein profiles in recipient neurons.


Assuntos
Axônios/metabolismo , Isquemia Encefálica/metabolismo , Corpo Celular/metabolismo , Células Endoteliais/metabolismo , Exossomos/metabolismo , MicroRNAs/metabolismo , Crescimento Neuronal , Neurônios/metabolismo , Animais , Axônios/ultraestrutura , Corpo Celular/ultraestrutura , Técnicas de Cultura de Células , Córtex Cerebral/citologia , Dispositivos Lab-On-A-Chip , Masculino , Neovascularização Fisiológica , Neurônios/ultraestrutura , Cultura Primária de Células , Ratos
2.
Int J Mol Sci ; 21(17)2020 Aug 27.
Artigo em Inglês | MEDLINE | ID: mdl-32867041

RESUMO

Stroke remains the leading cause of adult disability. Post-stroke neurogenesis contributes to functional recovery. As an intrinsic neurorestorative process, it is important to elucidate the molecular mechanism underlying stroke-induced neurogenesis and to develop therapies designed specifically to augment neurogenesis. Epigenetic mechanisms include DNA methylation, histone modification and its mediation by microRNAs and long-non-coding RNAs. In this review, we highlight how epigenetic factors including DNA methylation, histone modification, microRNAs and long-non-coding RNAs mediate stroke-induced neurogenesis including neural stem cell self-renewal and cell fate determination. We also summarize therapies targeting these mechanisms in the treatment of stroke.


Assuntos
Epigênese Genética , Células-Tronco Neurais/citologia , Acidente Vascular Cerebral/genética , Diferenciação Celular , Autorrenovação Celular , Metilação de DNA , Código das Histonas , Humanos , MicroRNAs/genética , Neurogênese , RNA Longo não Codificante/genética
3.
Front Neurosci ; 18: 1408205, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-39050669

RESUMO

Background: Vascular dementia (VaD) is a complex neurodegenerative disorder. We previously found that treatment of VaD in middle-aged male rats subjected to multiple microinfarction (MMI) with AV-001, a Tie2 receptor agonist, significantly improves cognitive function. Age and sex affect the development and response of VaD to therapeutic intervention. Thus, the present study investigated the therapeutic effect of AV-001 on VaD in aged female rats subjected to MMI. Methods: Female 18-month-old Wistar rats were subjected to MMI by injecting either 1,000 (low dose, LD-MMI) or 6,000 (high dose, HD-MMI) cholesterol crystals of size 70-100 µm into the right internal carotid artery. AV-001 (1 µg/Kg, i.p.) was administered once daily after MMI for 1 month, with treatment initiated 1 day after MMI. A battery of behavioral tests to examine sensorimotor and cognitive functions was performed at 21-28 days after MMI. All rats were sacrificed at 1 month after MMI. Results: Aged female rats subjected to LD-MMI exhibit severe neurological deficits, memory impairment, and significant white matter (WM) and oligodendrogenesis injury in the corpus callosum compared with control rats. HD-MMI in aged female rats induces significant anxiety- and depression-like behaviors, which were not detected in LD-MMI aged female rats. Also, HD-MMI induces significantly increased WM injury compared to LD-MMI. AV-001 treatment of LD-MMI and HD-MMI increases oligodendrogenesis, myelin and axon density in the corpus callosum and striatal WM bundles, promotes WM integrity and attenuates neurological and cognitive deficits. Additionally, both LD-MMI and HD-MMI rats exhibit a significant increase, while AV-001 significantly decreases the levels of inflammatory factors in the cerebrospinal fluid (CSF). Conclusion: MMI reduces oligodendrogenesis, and induces demyelination, axonal injury and WM injury, and causes memory impairment, while HD-MMI induces increased WM injury and further depression-like behaviors compared to LD-MMI rats. AV-001 has a therapeutic effect on aged female rats with MMI by reducing WM damage and improving neuro-cognitive outcomes.

4.
Arterioscler Thromb Vasc Biol ; 32(8): 1856-64, 2012 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-22723435

RESUMO

OBJECTIVE: Activation of the toll-like receptor (TLR) signaling pathway exacerbates ischemic brain damage. The present study tested the hypothesis that combination treatment with VELCADE and tissue plasminogen activator (tPA) modulates the TLR signaling pathway on cerebral vasculature, which leads to neuroprotection in aged rats after stroke. METHODS AND RESULTS: Focal cerebral ischemia acutely increased TLR2, TLR4, and interleukin-1 receptor-activated kinases 1 immunoreactivity on fibrin/fibrinogen-positive vessels in aged rats. Monotherapy of tPA further amplified these signals. However, VELCADE in combination with tPA-blocked stroke- and tPA-potentiated vascular TLR signals, leading to robust reduction of infarct volume compared with respective monotherapies. Quantitative reverse transcription polymerase chain reaction analysis of cerebral endothelial cells isolated by laser capture microdissection revealed that the combination treatment increased miR-l46a levels, which was inversely associated with the reduction of vascular interleukin-1 receptor-activated kinases 1 immunoreactivity. In vitro, fibrin upregulated interleukin-1 receptor-activated kinases 1 and TLR4 expression and downregulated miR-146a on primary human cerebral endothelial cells. VELCADE elevated miR-146 levels and abolished fibrin-increased interleukin-1 receptor activated kinases 1 proteins. CONCLUSIONS: Stroke acutely activates the TLR signaling pathway on cerebral vasculature. Upregulation of miR-146a and inactivation of ischemia and tPA-potentiated TLR signaling pathway by VELCADE may play an important role in the neuroprotective effect of the combination therapy of VELCADE and tPA for acute stroke.


Assuntos
Ácidos Borônicos/administração & dosagem , MicroRNAs/fisiologia , Fármacos Neuroprotetores/administração & dosagem , Pirazinas/administração & dosagem , Transdução de Sinais/efeitos dos fármacos , Acidente Vascular Cerebral/tratamento farmacológico , Ativador de Plasminogênio Tecidual/administração & dosagem , Receptores Toll-Like/fisiologia , Envelhecimento , Animais , Bortezomib , Células Cultivadas , Regulação para Baixo , Quimioterapia Combinada , Humanos , Masculino , NF-kappa B/metabolismo , Ratos , Ratos Wistar
5.
Micromachines (Basel) ; 14(3)2023 Mar 22.
Artigo em Inglês | MEDLINE | ID: mdl-36985111

RESUMO

The self-cleaning ability of superhydrophobic metal surfaces has attracted extensive attention. The preparation of superhydrophobic material using the coating method is a common processing method. In this experiment, aluminum fins were processed by laser etching and perfluorinated two-step coating. The aluminum surface was modified using a femtosecond laser and 1H,1H,2H,2H- perfluorooctane triethoxysilane (PFOTES). A superhydrophobic aluminum surface with excellent mechanical stability and self-cleaning properties was obtained with the superhydrophobic contact angle (WCA) of 152.8° and the rolling angle (SA) of 0.6°. The results show that the superhydrophobic surface has an excellent cleaning effect compared with an ordinary surface in unit time. Then, a wear resistance test of the superhydrophobic surface was carried out by using the physical wear method. The results show that physical wear had a low influence on the hydrophobic property of the specimen surface. Finally, the Vickers hardness analysis found that the superhydrophobic surface hardness was significantly better than the ordinary surface hardness compared with the superhydrophobic surface hardness. Based on the excellent self-cleaning properties, wear resistance, and robustness of superhydrophobic materials, the laser-etched and perfluorinated superhydrophobic aluminum fins designed and manufactured in this study have broad application prospects in improving the heat transfer efficiency of finned heat exchangers.

6.
Stroke ; 41(5): 1001-7, 2010 May.
Artigo em Inglês | MEDLINE | ID: mdl-20203318

RESUMO

BACKGROUND AND PURPOSE: Treatment with a selective proteasome inhibitor, VELCADE, in combination with tissue plasminogen activator (tPA) extended the therapeutic window to 6 hours in young rats after stroke. However, stroke is a major cause of death and disability in the elderly. The present study investigated the effect of VELCADE in combination with a low-dose tPA on aged rats after embolic stroke. METHODS: Male Wistar rats at the age of 18 to 20 months were treated with VELCADE (0.2 mg/kg) alone, a low-dose tPA (5 mg/kg) alone, combination of VELCADE and tPA, or saline 2 hours after embolic middle cerebral artery occlusion. To test the contribution of endothelial nitric oxide synthase to VELCADE-mediated neuroprotection, endothelial nitric oxide synthase knockout and wild-type mice were treated with VELCADE (0.5 mg/kg) 2 hours after embolic stroke. RESULTS: Treatment with VELCADE significantly reduced infarct volume, whereas tPA alone did not reduce infarct volume and aggravated blood-brain barrier disruption in aged rats compared with saline-treated rats. However, the combination treatment significantly enhanced the reduction of infarct volume, which was associated with an increase in endothelial nitric oxide synthase activity compared with saline-treated rats. Additionally, the combination treatment promoted thrombolysis and did not increase the incidence of hemorrhage transformation. VELCADE significantly reduced lesion volume in wild-type mice but failed to significantly reduce lesion volume in endothelial nitric oxide synthase knockout mice. CONCLUSIONS: Treatment with VELCADE exerts a neuroprotective effect in aged rats after stroke. The combination of VELCADE with the low-dose tPA further amplifies the neuroprotective effect. Endothelial nitric oxide synthase at least partly contributes to VELCADE-mediated neuroprotection after stroke.


Assuntos
Envelhecimento/efeitos dos fármacos , Ácidos Borônicos/administração & dosagem , Isquemia Encefálica/prevenção & controle , Embolia Intracraniana/prevenção & controle , Fármacos Neuroprotetores/administração & dosagem , Pirazinas/administração & dosagem , Ativador de Plasminogênio Tecidual/administração & dosagem , Envelhecimento/patologia , Animais , Bortezomib , Isquemia Encefálica/enzimologia , Isquemia Encefálica/patologia , Esquema de Medicação , Quimioterapia Combinada , Embolia Intracraniana/enzimologia , Embolia Intracraniana/patologia , Masculino , Inibidores de Proteases/administração & dosagem , Ratos , Ratos Wistar
7.
Arthritis Res Ther ; 20(1): 45, 2018 03 15.
Artigo em Inglês | MEDLINE | ID: mdl-29544526

RESUMO

BACKGROUND: MicroRNAs (miRNAs) serve as important regulators of inflammatory and immune responses and are implicated in several immune disorders including gouty arthritis. The expression of miR-146a is upregulated in the peripheral blood mononuclear cells of patients with inter-critical gout when compared to normouricemic and hyperuricemic controls and those patients with acute gout flares. However, the role of miR-146a in the development of gout remains unknown. Here, we used miR-146a knockout (KO) mice to test miR-146a function in a monosodium urate (MSU)-induced gouty arthritis model. METHODS: The footpad or ankle joint of miR-146a KO and wild-type (WT) mice were injected with an MSU suspension to induce acute gouty arthritis. Bone marrow-derived macrophages (BMDMs) were stimulated with MSU and the gene expression of miR-146a; interleukin 1 beta (IL-1ß); tumor necrosis factor-α (TNF-α); and the NACHT, LRR and PYD domains-containing protein 3 (NALP3) inflammasome was evaluated. TNF-α and IL-1ß protein levels in BMDMs were assessed by fluorescence-activated cell sorting and western blot analyses. Gene and protein levels of TNF receptor-associated factor 6 (TRAF6) and IL-1 receptor-associated kinase (IRAK1), the targets of miR-146a, were also measured. RESULTS: Significantly increased paw swelling and index and ankle joint swelling were observed in miR-146a KO mice compared to WT controls after MSU treatment. MiR-146a expression in BMDMs from WT mice was dramatically upregulated at 4 h following MSU stimulation. Additionally, the expression of IL-1ß, TNF-α, and NALP3 was higher in BMDMs from miR-146a KO mice after exposure to MSU crystals compared to those from WT mice. Consistent with the observed gene expression, the IL-1ß and TNF-α proteins were upregulated in miR-146a KO mice. Additionally quantitative RT-PCR and western blot demonstrated that TRAF6 and IRAK1 were dramatically upregulated in BMDMs from miR-146 KO mice compared to those from WT mice. CONCLUSIONS: Collectively, these observations suggest that miR-146a provides negative feedback regulation of gouty arthritis development and lack of miR-146a enhances gouty arthritis via upregulation of TRAK6, IRAK-1, and the NALP3 inflammasome function.


Assuntos
Artrite Gotosa/metabolismo , Quinases Associadas a Receptores de Interleucina-1/biossíntese , MicroRNAs/metabolismo , Proteína 3 que Contém Domínio de Pirina da Família NLR/biossíntese , Índice de Gravidade de Doença , Fator 6 Associado a Receptor de TNF/biossíntese , Animais , Artrite Gotosa/patologia , Células Cultivadas , Macrófagos/metabolismo , Macrófagos/patologia , Camundongos , Camundongos Knockout
8.
Circulation ; 112(22): 3486-94, 2005 Nov 29.
Artigo em Inglês | MEDLINE | ID: mdl-16316970

RESUMO

BACKGROUND: Microvascular dysfunction posttreatment of stroke with recombinant human tissue-type plasminogen activator (rht-PA) constrains the therapeutic window to 3 hours. Statins (3-hydroxy-3-methylglutaryl coenzyme A reductase inhibitors) promote vascular thrombolysis and reduce the inflammation response. We therefore investigated the neuroprotective effects of a combination of atorvastatin and delayed rht-PA treatment in a rat model of embolic stroke. METHODS AND RESULTS: Rats subjected to embolic middle cerebral artery occlusion were treated with atorvastatin in combination with rht-PA 4 hours after stroke. Magnetic resonance imaging measurements revealed that combination treatment with atorvastatin and rht-PA blocked the expansion of the ischemic lesion, which improved neurological function compared with saline-treated rats. Real-time reverse transcription-polymerase chain reaction analysis of single endothelial cells isolated by laser-capture microdissection from brain tissue and immunostaining showed that combination treatment downregulated expression of tissue factor, von Willebrand factor, protease-activated receptor-1, intercellular adhesion molecule-1, and matrix metalloproteinase-9, which concomitantly reduced cerebral microvascular thrombosis and enhanced microvascular integrity. Combination treatment did not increase cerebrovascular endothelial nitric oxide synthase (eNOS) levels or eNOS activity, and inhibition of NOS activity with N-nitro-L-arginine methyl ester did not block the beneficial effects of combination treatment on stroke. Furthermore, combination treatment compared with thrombolytic monotherapy increased cerebral blood flow and reduced infarct volume in eNOS-null mice. CONCLUSIONS: These data demonstrate that combination treatment with atorvastatin and rht-PA exerts a neuroprotective effect when administered 4 hours after stroke and that the therapeutic benefits are likely attributed to its multitargeted effects on cerebrovascular patency and integrity.


Assuntos
Inibidores de Hidroximetilglutaril-CoA Redutases/uso terapêutico , Acidente Vascular Cerebral/tratamento farmacológico , Terapia Trombolítica/métodos , Ativador de Plasminogênio Tecidual/uso terapêutico , Animais , Animais Geneticamente Modificados , Encéfalo/citologia , Modelos Animais de Doenças , Quimioterapia Combinada , Células Endoteliais/metabolismo , Regulação da Expressão Gênica/efeitos dos fármacos , Inibidores de Hidroximetilglutaril-CoA Redutases/farmacologia , Infarto da Artéria Cerebral Média/tratamento farmacológico , Imageamento por Ressonância Magnética , Microcirculação/efeitos dos fármacos , Fármacos Neuroprotetores/farmacologia , Fármacos Neuroprotetores/uso terapêutico , Óxido Nítrico Sintase Tipo III/análise , Ratos , Proteínas Recombinantes , Fatores de Tempo , Ativador de Plasminogênio Tecidual/farmacologia , Grau de Desobstrução Vascular/efeitos dos fármacos
9.
Thromb Haemost ; 95(1): 166-73, 2006 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-16543976

RESUMO

Stroke elicits a progressive vascular dysfunction, which contributes to the evolution of brain injury. Thrombolysis with tissue plasminogen activator (tPA) promotes adverse vascular events that limit the therapeutic window of stroke to three hours. Proteasome inhibitors reduce vascular thrombotic and inflammatory events, and consequently protect vascular function. The present study evaluated the neuroprotective effect of bortezomib, a potent and selective inhibitor of the proteasome, alone and in combination with delayed thrombolytic therapy on a rat model of embolic focal cerebral ischemia. Treatment with bortezomib reduces adverse cerebrovascular events including secondary thrombosis, inflammatory responses, and blood brain barrier (BBB) disruption, and hence reduces infarct volume and neurological functional deficit when administrated within 4 h after stroke onset. Combination of bortezomib and tPA extends the thrombolytic window for stroke to 6 h, which is associated with the improvement of vascular patency and integrity. Real time RT-PCR of endothelial cells isolated by laser-capture microdissection from brain tissue and Western blot analysis showed that bortezomib upregulates endothelial nitric oxide synthase (eNOS) expression and blocks NF-kappaB activation. These results demonstrate that bortezomib promotes eNOS dependent vascular protection, and reduces NF-kappaB dependent vascular disruption, all of which may contribute to neuroprotection after stroke.


Assuntos
Anti-Inflamatórios/uso terapêutico , Ácidos Borônicos/uso terapêutico , Fibrinolíticos/uso terapêutico , Inibidores de Proteases/uso terapêutico , Pirazinas/uso terapêutico , Acidente Vascular Cerebral/tratamento farmacológico , Ativador de Plasminogênio Tecidual/uso terapêutico , Animais , Anti-Inflamatórios/farmacologia , Ácidos Borônicos/farmacologia , Bortezomib , Encéfalo/irrigação sanguínea , Modelos Animais de Doenças , Quimioterapia Combinada , Endotélio Vascular/efeitos dos fármacos , Endotélio Vascular/enzimologia , Fibrinolíticos/farmacologia , Humanos , Infarto da Artéria Cerebral Média/tratamento farmacológico , Infarto da Artéria Cerebral Média/enzimologia , Infarto da Artéria Cerebral Média/patologia , Embolia Intracraniana/tratamento farmacológico , Embolia Intracraniana/enzimologia , Embolia Intracraniana/patologia , Masculino , NF-kappa B/metabolismo , Óxido Nítrico Sintase Tipo III/metabolismo , Agregação Plaquetária/efeitos dos fármacos , Inibidores de Proteases/farmacologia , Pirazinas/farmacologia , Ratos , Ratos Wistar , Proteínas Recombinantes/farmacologia , Proteínas Recombinantes/uso terapêutico , Acidente Vascular Cerebral/enzimologia , Acidente Vascular Cerebral/patologia , Fatores de Tempo , Ativador de Plasminogênio Tecidual/genética , Ativador de Plasminogênio Tecidual/farmacologia , Grau de Desobstrução Vascular/efeitos dos fármacos
10.
Front Neurol ; 12: 662373, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33959091
11.
Neurochem Int ; 96: 24-31, 2016 06.
Artigo em Inglês | MEDLINE | ID: mdl-27103167

RESUMO

We have previously demonstrated that stroke induces nuclear shuttling of class IIa histone deacetylase 4 (HDAC4). Stroke-induced nuclear shuttling of HDAC4 is positively and significantly correlated with improved indices of neuronal remodeling in the peri-infarct cortex. In this study, using a rat model for middle cerebral artery occlusion (MCAO), we tested the effects of selective inhibition of class IIa HDACs on functional recovery and neuronal remodeling when administered 24hr after stroke. Adult male Wistar rats (n = 15-17/group) were subjected to 2 h MCAO and orally gavaged with MC1568 (a selective class IIa HDAC inhibitor), SAHA (a non-selective HDAC inhibitor), or vehicle-control for 7 days starting 24 h after MCAO. A battery of behavioral tests was performed. Lesion volume measurement and immunohistochemistry were performed 28 days after MCAO. We found that stroke increased total HDAC activity in the ipsilateral hemisphere compared to the contralateral hemisphere. Stroke-increased HDAC activity was significantly decreased by the administration of SAHA as well as by MC1568. However, SAHA significantly improved functional outcome compared to vehicle control, whereas selective class IIa inhibition with MC1568 increased mortality and lesion volume and did not improve functional outcome. In addition, MC1568 decreased microtubule associated protein 2 (MAP2, dendrites), phosphorylated neurofilament heavy chain (pNFH, axons) and myelin basic protein (MBP, myelination) immunoreactivity in the peri-infarct cortex. Quantitative RT-PCR of cortical neurons isolated by laser capture microdissection revealed that MC1568, but not SAHA, downregulated CREB and c-fos expression. Additionally, MC1568 decreased the expression of phosphorylated CREB (active) in neurons. Taken together, these findings demonstrate that selective inhibition of class IIa HDACs impairs neuronal remodeling and neurological outcome. Inactivation of CREB and c-fos by MC1568 likely contributes to this detrimental effect.


Assuntos
Histona Desacetilase 2/metabolismo , Inibidores de Histona Desacetilases/farmacologia , Plasticidade Neuronal/fisiologia , Neurônios/enzimologia , Recuperação de Função Fisiológica/fisiologia , Acidente Vascular Cerebral/enzimologia , Animais , Histona Desacetilase 2/antagonistas & inibidores , Masculino , Plasticidade Neuronal/efeitos dos fármacos , Neurônios/efeitos dos fármacos , Neurônios/patologia , Distribuição Aleatória , Ratos , Ratos Wistar , Recuperação de Função Fisiológica/efeitos dos fármacos , Acidente Vascular Cerebral/patologia
12.
PLoS One ; 9(12): e113972, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25437857

RESUMO

The unique cellular and vascular architecture of the adult ventricular-subventricular zone (V/SVZ) neurogenic niche plays an important role in regulating neural stem cell function. However, the in vivo identification of neural stem cells and their relationship to blood vessels within this niche in response to stroke remain largely unknown. Using whole-mount preparation of the lateral ventricle wall, we examined the architecture of neural stem cells and blood vessels in the V/SVZ of adult mouse over the course of 3 months after onset of focal cerebral ischemia. Stroke substantially increased the number of glial fibrillary acidic protein (GFAP) positive neural stem cells that are in contact with the cerebrospinal fluid (CSF) via their apical processes at the center of pinwheel structures formed by ependymal cells residing in the lateral ventricle. Long basal processes of these cells extended to blood vessels beneath the ependymal layer. Moreover, stroke increased V/SVZ endothelial cell proliferation from 2% in non-ischemic mice to 12 and 15% at 7 and 14 days after stroke, respectively. Vascular volume in the V/SVZ was augmented from 3% of the total volume prior to stroke to 6% at 90 days after stroke. Stroke-increased angiogenesis was closely associated with neuroblasts that expanded to nearly encompass the entire lateral ventricular wall in the V/SVZ. These data indicate that stroke induces long-term alterations of the neural stem cell and vascular architecture of the adult V/SVZ neurogenic niche. These post-stroke structural changes may provide insight into neural stem cell mediation of stroke-induced neurogenesis through the interaction of neural stem cells with proteins in the CSF and their sub-ependymal neurovascular interaction.


Assuntos
Isquemia Encefálica/patologia , Encéfalo/irrigação sanguínea , Proteínas do Tecido Nervoso/metabolismo , Células-Tronco Neurais/metabolismo , Acidente Vascular Cerebral/patologia , Animais , Proliferação de Células , Células Cultivadas , Modelos Animais de Doenças , Células Endoteliais/citologia , Proteína Glial Fibrilar Ácida , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Neurogênese , Acidente Vascular Cerebral/etiologia
13.
PLoS One ; 7(10): e48141, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-23118941

RESUMO

Adult neural stem cells give rise to neurons, oligodendrocytes and astrocytes. Aging reduces neural stem cells. Using an inducible nestin-CreER(T2)/R26R-yellow fluorescent protein (YFP) mouse, we investigated the effect of Sildenafil, a phosphodiesterase type 5 (PDE5) inhibitor, on nestin lineage neural stem cells and their progeny in the ischemic brain of the middle-aged mouse. We showed that focal cerebral ischemia induced nestin lineage neural stem cells in the subventricular zone (SVZ) of the lateral ventricles and nestin expressing NeuN positive neurons and adenomatous polyposis coli (APC) positive mature oligodendrocytes in the ischemic striatum and corpus callosum in the aged mouse. Treatment of the ischemic middle-aged mouse with Sildenafil increased nestin expressing neural stem cells, mature neurons, and oligodendrocytes by 33, 75, and 30%, respectively, in the ischemic brain. These data indicate that Sildenafil amplifies nestin expressing neural stem cells and their neuronal and oligodendrocyte progeny in the ischemic brain of the middle-aged mouse.


Assuntos
Infarto da Artéria Cerebral Média/tratamento farmacológico , Neurogênese/efeitos dos fármacos , Oligodendroglia/efeitos dos fármacos , Inibidores da Fosfodiesterase 5/farmacologia , Piperazinas/farmacologia , Sulfonas/farmacologia , Animais , Proteínas de Bactérias/biossíntese , Proteínas de Bactérias/genética , Rastreamento de Células , Corpo Caloso/efeitos dos fármacos , Corpo Caloso/metabolismo , Corpo Caloso/patologia , Proteínas de Ligação a DNA , Infarto da Artéria Cerebral Média/patologia , Proteínas de Filamentos Intermediários/metabolismo , Ventrículos Laterais/efeitos dos fármacos , Ventrículos Laterais/metabolismo , Ventrículos Laterais/patologia , Proteínas Luminescentes/biossíntese , Proteínas Luminescentes/genética , Masculino , Camundongos , Camundongos Transgênicos , Proteínas do Tecido Nervoso/metabolismo , Nestina , Células-Tronco Neurais/efeitos dos fármacos , Células-Tronco Neurais/metabolismo , Células-Tronco Neurais/fisiologia , Proteínas Nucleares/metabolismo , Oligodendroglia/metabolismo , Oligodendroglia/fisiologia , Inibidores da Fosfodiesterase 5/uso terapêutico , Piperazinas/uso terapêutico , Purinas/farmacologia , Purinas/uso terapêutico , Citrato de Sildenafila , Sulfonas/uso terapêutico
14.
J Cereb Blood Flow Metab ; 31(11): 2181-8, 2011 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-21829213

RESUMO

Multipotent mesenchymal stromal cells (MSCs) increase tissue plasminogen activator (tPA) activity in astrocytes of the ischemic boundary zone, leading to increased neurite outgrowth in the brain. To probe the mechanisms that underlie MSC-mediated activation of tPA, we investigated the morphogenetic gene, sonic hedgehog (Shh) pathway. In vitro oxygen and glucose deprivation and coculture of astrocytes and MSCs were used to mimic an in vivo ischemic condition. Both real-time-PCR and western blot showed that MSC coculture significantly increased the Shh level and concomitantly increased tPA and decreased plasminogen activator inhibitor 1 (PAI-1) levels in astrocytes. Inhibiting the Shh signaling pathway with cyclopamine blocked the increase of tPA and the decrease of PAI-1 expression in astrocytes subjected to MSC coculture or recombinant mouse Shh (rm-Shh) treatment. Both MSCs and rm-Shh decreased the transforming growth factor-ß1 level in astrocytes, and the Shh pathway inhibitor cyclopamine reversed these decreases. Both Shh-small-interfering RNA (siRNA) and Glil-siRNA downregulated Shh and Gli1 (a key mediator of the Shh transduction pathway) expression in cultured astrocytes and concomitantly decreased tPA expression and increased PAI-1 expression in these astrocytes after MSC or rm-Shh treatment. Our data indicate that MSCs increase astrocytic Shh, which subsequently increases tPA expression and decreases PAI-1 expression after ischemia.


Assuntos
Astrócitos/metabolismo , Proteínas Hedgehog/metabolismo , Células-Tronco Mesenquimais/fisiologia , Células-Tronco Multipotentes/fisiologia , Serpina E2/biossíntese , Acidente Vascular Cerebral , Ativador de Plasminogênio Tecidual/biossíntese , Animais , Astrócitos/citologia , Astrócitos/patologia , Técnicas de Cultura de Células , Hipóxia Celular , Técnicas de Cocultura , Meios de Cultura , Regulação para Baixo , Glucose/metabolismo , Células-Tronco Mesenquimais/citologia , Células-Tronco Mesenquimais/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Células-Tronco Multipotentes/citologia , Células-Tronco Multipotentes/metabolismo , Oxigênio/metabolismo , Receptor Cross-Talk , Transdução de Sinais , Acidente Vascular Cerebral/metabolismo , Acidente Vascular Cerebral/patologia , Regulação para Cima
15.
FEBS J ; 277(20): 4299-307, 2010 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-20840605

RESUMO

MicroRNAs are small RNAs that attenuate protein expression by complementary binding to the 3'-UTR of a target mRNA. Currently, very little is known about microRNAs after cerebral ischemia. In particular, microRNA-21 (miR-21) is a strong antiapoptotic factor in some biological systems. We investigated the role of miR-21 after stroke in the rat. We employed in situ hybridization and laser capture microdissection in combination with real-time RT-PCR to investigate the expression of miR-21 after stroke. In situ hybridization revealed that miR-21 expression was upregulated in neurons of the ischemic boundary zone, and quantitative real-time RT-PCR analysis revealed that stroke increased mature miR-21 levels by approximately threefold in neurons isolated from the ischemic boundary zone by laser capture microdissection as compared with homologous contralateral neurons 2 days (n = 4; P < 0.05) and 7 days (n = 3; P < 0.05) after stroke. In vitro, overexpression of miR-21 in cultured cortical neurons substantially suppressed oxygen and glucose deprivation-induced apoptotic cell death, whereas attenuation of endogenous miR-21 by antisense inhibition exacerbated cell death after oxygen and glucose deprivation. Moreover, overexpression of miR-21 in neurons significantly reduced FASLG levels, and introduction of an miR-21 mimic into 293-HEK cells substantially reduced luciferase activity in a reporter system containing the 3'-UTR of Faslg. Our data indicate that overexpression of miR-21 protects against ischemic neuronal death, and that downregulation of FASLG, a tumor necrosis factor-α family member and an important cell death-inducing ligand whose gene is targeted by miR-21, probably mediates the neuroprotective effect. These novel findings suggest that miR-21 may be an attractive therapeutic molecule for treatment of stroke.


Assuntos
Proteína Ligante Fas/fisiologia , Isquemia/patologia , MicroRNAs/fisiologia , Neurônios/patologia , Animais , Morte Celular , Proteína Ligante Fas/genética , Regulação da Expressão Gênica , MicroRNAs/análise , MicroRNAs/genética , Substâncias Protetoras , Ratos , Ratos Wistar , Acidente Vascular Cerebral
16.
PLoS One ; 5(2): e9027, 2010 Feb 03.
Artigo em Inglês | MEDLINE | ID: mdl-20140248

RESUMO

We demonstrate that tissue plasminogen activator (tPA) and its inhibitors contribute to neurite outgrowth in the central nervous system (CNS) after treatment of stroke with multipotent mesenchymal stromal cells (MSCs). In vivo, administration of MSCs to mice subjected to middle cerebral artery occlusion (MCAo) significantly increased activation of tPA and downregulated PAI-1 levels in the ischemic boundary zone (IBZ) compared with control PBS treated mice, concurrently with increases of myelinated axons and synaptophysin. In vitro, MSCs significantly increased tPA levels and concomitantly reduced plasminogen activator inhibitor 1 (PAI-1) expression in astrocytes under normal and oxygen and glucose deprivation (OGD) conditions. ELISA analysis of conditioned medium revealed that MSCs stimulated astrocytes to secrete tPA. When primary cortical neurons were cultured in the conditioned medium from MSC co-cultured astrocytes, these neurons exhibited a significant increase in neurite outgrowth compared to conditioned medium from astrocytes alone. Blockage of tPA with a neutralizing antibody or knock-down of tPA with siRNA significantly attenuated the effect of the conditioned medium on neurite outgrowth. Addition of recombinant human tPA into cortical neuronal cultures also substantially enhanced neurite outgrowth. Collectively, these in vivo and in vitro data suggest that the MSC mediated increased activation of tPA in astrocytes promotes neurite outgrowth after stroke.


Assuntos
Astrócitos/citologia , Células-Tronco Multipotentes/citologia , Neuritos/fisiologia , Células Estromais/citologia , Ativador de Plasminogênio Tecidual/metabolismo , Animais , Apoptose/efeitos dos fármacos , Astrócitos/metabolismo , Western Blotting , Encéfalo/efeitos dos fármacos , Encéfalo/metabolismo , Encéfalo/patologia , Células Cultivadas , Técnicas de Cocultura , Meios de Cultivo Condicionados/farmacologia , Expressão Gênica/efeitos dos fármacos , Glucose/farmacologia , Humanos , Marcação In Situ das Extremidades Cortadas , Mesoderma/citologia , Camundongos , Camundongos Endogâmicos C57BL , Células-Tronco Multipotentes/metabolismo , Neuritos/efeitos dos fármacos , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Neurônios/patologia , Inibidor 1 de Ativador de Plasminogênio/genética , Inibidor 1 de Ativador de Plasminogênio/metabolismo , Interferência de RNA , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Acidente Vascular Cerebral/fisiopatologia , Células Estromais/metabolismo , Sinaptofisina/metabolismo , Ativador de Plasminogênio Tecidual/genética
17.
J Biol Chem ; 282(44): 32462-70, 2007 Nov 02.
Artigo em Inglês | MEDLINE | ID: mdl-17804404

RESUMO

Carbamylated erythropoietin (CEPO), a well characterized erythropoietin (EPO) derivative, does not bind to the classical EPO receptor and does not stimulate erythropoiesis. Using neural progenitor cells derived from the subventricular zone of the adult mouse, we investigated the effect of CEPO on neurogenesis and the associated signaling pathways in vitro. We found that CEPO significantly increased neural progenitor cell proliferation and promoted neural progenitor cell differentiation into neurons, which was associated with up-regulation of Sonic hedgehog (Shh), its receptor ptc, and mammalian achaete-scute homolog 1 (Mash1), a pro-neuron basic helix-loop-helix protein transcription factor. Blockage of the Shh signaling pathway with a pharmacological inhibitor, cyclopamine, abolished the CEPO-induced neurogenesis. Attenuation of endogenous Mash1 expression by short-interfering RNA blocked CEPO-promoted neuronal differentiation. In addition, recombinant mouse Shh up-regulated Mash1 expression in neural progenitor cells. These results demonstrate that the Shh signaling pathway mediates CEPO-enhanced neurogenesis and Mash1 is a downstream target of the Shh signaling pathway that regulates CEPO-enhanced neuronal differentiation.


Assuntos
Células-Tronco Adultas , Diferenciação Celular , Proliferação de Células , Eritropoetina/análogos & derivados , Proteínas Hedgehog/metabolismo , Transdução de Sinais , Animais , Sequência de Bases , Fatores de Transcrição Hélice-Alça-Hélice Básicos/metabolismo , Eritropoetina/metabolismo , Masculino , Camundongos , Receptores da Eritropoetina/metabolismo , Regulação para Cima
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA