Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Cell ; 186(4): 786-802.e28, 2023 02 16.
Artigo em Inglês | MEDLINE | ID: mdl-36754049

RESUMO

Amyotrophic lateral sclerosis (ALS) is a fatal neurodegenerative disease that results from many diverse genetic causes. Although therapeutics specifically targeting known causal mutations may rescue individual types of ALS, these approaches cannot treat most cases since they have unknown genetic etiology. Thus, there is a pressing need for therapeutic strategies that rescue multiple forms of ALS. Here, we show that pharmacological inhibition of PIKFYVE kinase activates an unconventional protein clearance mechanism involving exocytosis of aggregation-prone proteins. Reducing PIKFYVE activity ameliorates ALS pathology and extends survival of animal models and patient-derived motor neurons representing diverse forms of ALS including C9ORF72, TARDBP, FUS, and sporadic. These findings highlight a potential approach for mitigating ALS pathogenesis that does not require stimulating macroautophagy or the ubiquitin-proteosome system.


Assuntos
Esclerose Lateral Amiotrófica , Fosfatidilinositol 3-Quinases , Animais , Esclerose Lateral Amiotrófica/tratamento farmacológico , Esclerose Lateral Amiotrófica/genética , Esclerose Lateral Amiotrófica/metabolismo , Neurônios Motores , Mutação , Proteína FUS de Ligação a RNA/metabolismo , Fosfatidilinositol 3-Quinases/metabolismo , Modelos Animais de Doenças
2.
J Cell Mol Med ; 25(5): 2691-2702, 2021 03.
Artigo em Inglês | MEDLINE | ID: mdl-33496385

RESUMO

Mitochondrial dysfunction contributes to the pathophysiology of acute kidney injury (AKI). Mitophagy selectively degrades damaged mitochondria and thereby regulates cellular homeostasis. RNA-binding proteins (RBPs) regulate RNA processing at multiple levels and thereby control cellular function. In this study, we aimed to understand the role of human antigen R (HuR) in hypoxia-induced mitophagy process in the renal tubular cells. Mitophagy marker expressions (PARKIN, p-PARKIN, PINK1, BNIP3L, BNIP3, LC3) were determined by western blot analysis. Immunofluorescence studies were performed to analyze mitophagosome, mitolysosome, co-localization of p-PARKIN/TOMM20 and BNIP3L/TOMM20. HuR-mediated regulation of PARKIN/BNIP3L expressions was determined by RNA-immunoprecipitation analysis and RNA stability experiments. Hypoxia induced mitochondrial dysfunction by increased ROS, decline in membrane potential and activated mitophagy through up-regulated PARKIN, PINK1, BNIP3 and BNIP3L expressions. HuR knockdown studies revealed that HuR regulates hypoxia-induced mitophagosome and mitolysosome formation. HuR was significantly bound to PARKIN and BNIP3L mRNA under hypoxia and thereby up-regulated their expressions through mRNA stability. Altogether, our data highlight the importance of HuR in mitophagy regulation through up-regulating PARKIN/BNIP3L expressions in renal tubular cells.


Assuntos
Proteína Semelhante a ELAV 1/metabolismo , Células Epiteliais/metabolismo , Hipóxia/genética , Hipóxia/metabolismo , Proteínas de Membrana/genética , Mitofagia/genética , Proteínas Proto-Oncogênicas/genética , Proteínas Supressoras de Tumor/genética , Ubiquitina-Proteína Ligases/genética , Injúria Renal Aguda/etiologia , Injúria Renal Aguda/metabolismo , Injúria Renal Aguda/patologia , Linhagem Celular Tumoral , Expressão Gênica , Técnicas de Silenciamento de Genes , Humanos , Túbulos Renais , Lisossomos/metabolismo , Mitocôndrias/genética , Mitocôndrias/metabolismo , Modelos Biológicos , Fagossomos/metabolismo
3.
Membranes (Basel) ; 10(9)2020 Sep 21.
Artigo em Inglês | MEDLINE | ID: mdl-32967125

RESUMO

In the papermaking industry (reconstituted tobacco), a large number of tobacco stems, dust, and fines are discharged in the wastewater. This high salinity wastewater rich in ionic constituents and nicotine is difficult to be degraded by conventional biological treatment and is a serious threat that needs to be overcome. Electrodialysis (ED) has proved a feasible technique to remove the inorganic components in the papermaking wastewater. However, the fouling in ion exchange membranes causes deterioration of membranes, which causes a decrease in the flux and an increase in the electrical resistance of the membranes. In this study, the fouling potential of the membranes was analyzed by comparing the properties of the pristine and fouled ion exchange membranes. The physical and chemical properties of the ion exchange membranes were investigated in terms of electrical resistance, water content, and ion exchange capacity, as well as studied by infrared spectroscopy (IR) spectra, scanning electron microscopy (SEM), and energy dispersive spectroscopy (EDS) analyses. The results indicated that the membrane fouling is caused by two different mechanisms. For the anion exchange membranes, the fouling is mainly caused by the charged organic anions. For the cation exchange membrane, the fouling is caused by minerals such as Ca2+ and Mg2+. These metal ions reacted with OH- ions generated by water dissociation and precipitated on the membrane surface. The chemical cleaning with alkaline and acid could mitigate the fouling potential of the ion exchange membranes.

4.
Cell Rep ; 27(2): 549-560.e6, 2019 04 09.
Artigo em Inglês | MEDLINE | ID: mdl-30970257

RESUMO

Excessive and unresolved neuroinflammation is a key component of the pathological cascade in brain injuries such as ischemic stroke. Here, we report that TRIM9, a brain-specific tripartite motif (TRIM) protein, was highly expressed in the peri-infarct areas shortly after ischemic insults in mice, but expression was decreased in aged mice, which are known to have increased neuroinflammation after stroke. Mechanistically, TRIM9 sequestered ß-transducin repeat-containing protein (ß-TrCP) from the Skp-Cullin-F-box ubiquitin ligase complex, blocking IκBα degradation and thereby dampening nuclear factor κB (NF-κB)-dependent proinflammatory mediator production and immune cell infiltration to limit neuroinflammation. Consequently, Trim9-deficient mice were highly vulnerable to ischemia, manifesting uncontrolled neuroinflammation and exacerbated neuropathological outcomes. Systemic administration of a recombinant TRIM9 adeno-associated virus that drove brain-wide TRIM9 expression effectively resolved neuroinflammation and alleviated neuronal death, especially in aged mice. These findings reveal that TRIM9 is essential for resolving NF-κB-dependent neuroinflammation to promote recovery and repair after brain injury and may represent an attractive therapeutic target.


Assuntos
Isquemia Encefálica/metabolismo , Proteínas do Tecido Nervoso/biossíntese , Acidente Vascular Cerebral/metabolismo , Proteínas com Motivo Tripartido/biossíntese , Ubiquitina-Proteína Ligases/biossíntese , Animais , Isquemia Encefálica/genética , Isquemia Encefálica/patologia , Células Cultivadas , Feminino , Células HEK293 , Humanos , Inflamação/genética , Inflamação/metabolismo , Inflamação/patologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Proteínas do Tecido Nervoso/genética , Proteínas do Tecido Nervoso/metabolismo , Células-Tronco Neurais/metabolismo , Células-Tronco Neurais/patologia , Neurônios/metabolismo , Neurônios/patologia , Neuroproteção , Fosforilação , Acidente Vascular Cerebral/genética , Proteínas com Motivo Tripartido/genética , Proteínas com Motivo Tripartido/metabolismo , Ubiquitina-Proteína Ligases/genética , Ubiquitina-Proteína Ligases/metabolismo
5.
Anticancer Res ; 38(9): 5159-5164, 2018 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-30194163

RESUMO

BACKGROUND/AIM: Matrix metalloproteinases (MMPs) control the homeostasis of the extracellular matrix and their genetic polymorphisms may contribute to cancer susceptibility. The aim of this study was to reveal the genotypes of MMP8 among the Taiwanese and examine the contribution of MMP8 C-799T, Val436Ala and Lys460Thr polymorphisms to bladder cancer. MATERIALS AND METHODS: MMP8 C-799T, Val436Ala and Lys460Thr polymorphic genotypes were determined in 375 patients with bladder cancer and 375 healthy controls by polymerase chain reaction-restriction fragment length polymorphism methodology. RESULTS: Regarding MMP8 C-799T, there was no significant differential distribution between patient and control groups [p for trend=0.6629]. The odds ratios (ORs) after adjusting for age, gender, smoking and alcohol drinking status for those carrying CT and TT genotypes at MMP8 C-799T were 1.13 (95%CI=0.89-1.44, p=0.3688) and 1.10 (95%CI=0.87-1.52, p=0.9030), respectively, compared to those carrying the wild-type CC genotype. Regarding MMP8 Val436Ala, there was no significant differential distribution between patient and control groups [p for trend=0.8166]. The adjusted OR for those carrying AC and CC genotypes at MMP8 Val436Ala were 0.71 (95%CI=0.31-2.28, p=0.6094) and 1.00 (95%CI=0.21-4.73, p=0.7247), respectively. The polymorphism Lys460Thr at MMP8 was not found among Taiwanese patients. CONCLUSION: MMP8 C-799T, Val436Ala and Lys460Thr may only play an indirect role in determining personal cancer susceptibility for bladder cancer in Taiwan.


Assuntos
Povo Asiático/genética , Metaloproteinase 8 da Matriz/genética , Mutação , Neoplasias da Bexiga Urinária/genética , Feminino , Estudos de Associação Genética , Testes Genéticos , Genótipo , Humanos , Masculino , Razão de Chances , Taiwan
7.
J Neurosci ; 30(46): 15521-34, 2010 Nov 17.
Artigo em Inglês | MEDLINE | ID: mdl-21084607

RESUMO

The anticoagulant factor protein S (PS) protects neurons from hypoxic/ischemic injury. However, molecular mechanisms mediating PS protection in injured neurons remain unknown. Here, we show mouse recombinant PS protects dose-dependently mouse cortical neurons from excitotoxic NMDA-mediated neuritic bead formation and apoptosis by activating the phosphatidylinositol 3-kinase (PI3K)-Akt pathway (EC(50) = 26 ± 4 nm). PS stimulated phosphorylation of Bad and Mdm2, two downstream targets of Akt, which in neurons subjected to pathological overstimulation of NMDA receptors (NMDARs) increased the antiapoptotic Bcl-2 and Bcl-X(L) levels and reduced the proapoptotic p53 and Bax levels. Adenoviral transduction with a kinase-deficient Akt mutant (Ad.Akt(K179A)) resulted in loss of PS-mediated neuronal protection, Akt activation, and Bad and Mdm2 phosphorylation. Using the TAM receptors tyrosine kinases Tyro3-, Axl-, and Mer-deficient neurons, we showed that PS protected neurons lacking Axl and Mer, but not Tyro3, suggesting a requirement of Tyro3 for PS-mediated protection. Consistent with these results, PS dose-dependently phosphorylated Tyro3 on neurons (EC(50) = 25 ± 3 nm). In an in vivo model of NMDA-induced excitotoxic lesions in the striatum, PS dose-dependently reduced the lesion volume in control mice (EC(50) = 22 ± 2 nm) and protected Axl(-/-) and Mer(-/-) transgenic mice, but not Tyro3(-/-) transgenic mice. Using different structural PS analogs, we demonstrated that the C terminus sex hormone-binding globulin-like (SHBG) domain of PS is critical for neuronal protection in vitro and in vivo. Thus, our data show that PS protects neurons by activating the Tyro3-PI3K-Akt pathway via its SHGB domain, suggesting potentially a novel neuroprotective approach for acute brain injury and chronic neurodegenerative disorders associated with excessive activation of NMDARs.


Assuntos
Neurônios/metabolismo , Fosfatidilinositol 3-Quinase/metabolismo , Proteína S/fisiologia , Proteínas Proto-Oncogênicas c-akt/metabolismo , Receptores Proteína Tirosina Quinases/metabolismo , Globulina de Ligação a Hormônio Sexual/metabolismo , Animais , Células Cultivadas , Relação Dose-Resposta a Droga , Agonistas de Aminoácidos Excitatórios/toxicidade , Técnicas de Silenciamento de Genes , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Neurônios/efeitos dos fármacos , Neurônios/patologia , Fármacos Neuroprotetores/farmacologia , Proteínas Proto-Oncogênicas c-akt/fisiologia , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/fisiologia
8.
Blood ; 115(23): 4963-72, 2010 Jun 10.
Artigo em Inglês | MEDLINE | ID: mdl-20348395

RESUMO

The anticoagulant factor protein S (PS) has direct cellular activities. Lack of PS in mice causes lethal coagulopathy, ischemic/thrombotic injuries, vascular dysgenesis, and blood-brain barrier (BBB) disruption with intracerebral hemorrhages. Thus, we hypothesized that PS maintains and/or enhances the BBB integrity. Using a BBB model with human brain endothelial cells, we show PS inhibits time- and dose-dependently (half maximal effective concentration [EC(50)] = 27 +/- 3 nM) oxygen/glucose deprivation-induced BBB breakdown, as demonstrated by measurements of the transmonolayer electrical resistance, permeability of endothelial monolayers to dextran (40 kDa), and rearrangement of F-actin toward the cortical cytoskeletal ring. Using Tyro-3, Axl, and Mer (TAM) receptor, tyrosine kinase silencing through RNA interference, specific N-terminus-blocking antibodies, Tyro3 phosphorylation, and Tyro3-, Axl- and Mer-deficient mouse brain endothelial cells, we show that Tyro3 mediates PS vasculoprotection. After Tyro3 ligation, PS activated sphingosine 1-phosphate receptor (S1P(1)), resulting in Rac1-dependent BBB protection. Using 2-photon in vivo imaging, we show that PS blocks postischemic BBB disruption in Tyro3(+/+), Axl(-/-), and Mer(-/-) mice, but not in Tyro3(-/-) mice or Tyro3(+/+) mice receiving low-dose W146, a S1P(1)-specific antagonist. Our findings indicate that PS protects the BBB integrity via Tyro3 and S1P(1), suggesting potentially novel treatments for neurovascular dysfunction resulting from hypoxic/ischemic BBB damage.


Assuntos
Barreira Hematoencefálica/metabolismo , Isquemia Encefálica/metabolismo , Encéfalo/metabolismo , Células Endoteliais/metabolismo , Proteína S/metabolismo , Receptores Proteína Tirosina Quinases/metabolismo , Receptores de Lisoesfingolipídeo/metabolismo , Animais , Isquemia Encefálica/genética , Humanos , Camundongos , Camundongos Mutantes , Modelos Biológicos , Neuropeptídeos/genética , Neuropeptídeos/metabolismo , Fosforilação/genética , Proteína S/genética , Proteínas Proto-Oncogênicas/genética , Proteínas Proto-Oncogênicas/metabolismo , Receptores Proteína Tirosina Quinases/genética , Receptores de Lisoesfingolipídeo/genética , Receptores de Esfingosina-1-Fosfato , Adulto Jovem , c-Mer Tirosina Quinase , Proteínas rac de Ligação ao GTP/genética , Proteínas rac de Ligação ao GTP/metabolismo , Proteínas rac1 de Ligação ao GTP/genética , Proteínas rac1 de Ligação ao GTP/metabolismo , Receptor Tirosina Quinase Axl
9.
J Cereb Blood Flow Metab ; 30(3): 534-44, 2010 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-19826433

RESUMO

Transplantation of neural cells is a potential approach for stroke treatment, but disruption of tissue architecture may limit transplant efficacy. One strategy for enhancing the ability of transplants to restore brain structure and function is to administer cells together with biomaterial scaffolding. We electrocoagulated the distal middle cerebral artery in adult rats and, 3 weeks later, injected one of the following into the infarct cavity: artificial cerebrospinal fluid, Matrigel scaffolding, human embryonic stem cell-derived neuronal precursor cells, scaffolding plus cells, or cells cultured in and administered together with scaffolding. Five weeks after transplantation, the latter two groups showed approximately 50% and approximately 60% reductions, respectively, in infarct cavity volume. Rats given cells cultured in and administered together with scaffolding also showed (1) survival and neuronal differentiation of transplanted cells shown by immunostaining for neuronal marker proteins and cleaved caspase-3, and by patch-clamp recording, 8 weeks after transplantation and (2) improved outcome on tests of sensorimotor and cognitive functions, 4 to 9 weeks after transplantation. These results indicate that transplantation of human neural cells together with biomaterial scaffolding has the potential to improve the outcome from stroke, even when treatment is delayed for several weeks after the ischemic event.


Assuntos
Isquemia Encefálica/terapia , Colágeno , Laminina , Neurônios/transplante , Proteoglicanas , Transplante de Células-Tronco/métodos , Alicerces Teciduais , Animais , Comportamento Animal/fisiologia , Isquemia Encefálica/patologia , Adesão Celular , Combinação de Medicamentos , Eletrofisiologia , Potenciais Pós-Sinápticos Excitadores/fisiologia , Humanos , Imuno-Histoquímica , Técnicas In Vitro , Infarto da Artéria Cerebral Média/patologia , Infarto da Artéria Cerebral Média/terapia , Masculino , Proteínas do Tecido Nervoso/biossíntese , Técnicas de Patch-Clamp , Desempenho Psicomotor/fisiologia , Ratos , Ratos Sprague-Dawley , Resultado do Tratamento
10.
Eur J Neurosci ; 29(6): 1119-30, 2009 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-19302148

RESUMO

The anticoagulant activated protein C (APC) protects neurons and endothelium via protease activated receptor (PAR)1, PAR3 and endothelial protein C receptor. APC is neuroprotective in stroke models. Bleeding complications may limit the pharmacologic utility of APC. Here, we compared the 3K3A-APC mutant with 80% reduced anticoagulant activity and wild-type (wt)-APC. Murine 3K3A-APC compared with wt-APC protected mouse cortical neurons from N-methyl-D-aspartate-induced apoptosis with twofold greater efficacy and more potently reduced N-methyl-D-aspartate excitotoxic lesions in vivo. Human 3K3A-APC protected human brain endothelial cells (BECs) from oxygen/glucose deprivation with 1.7-fold greater efficacy than wt-APC. 3K3A-APC neuronal protection required PAR1 and PAR3, as shown by using PAR-specific blocking antibodies and PAR1- and PAR3-deficient cells and mice. BEC protection required endothelial protein C receptor and PAR1. In neurons and BECs, 3K3A-APC blocked caspase-9 and -3 activation and induction of p53, and decreased the Bax/Bcl-2 pro-apoptotic ratio. After distal middle cerebral artery occlusion (dMCAO) in mice, murine 3K3A-APC compared with vehicle given 4:00 h after dMCAO improved the functional outcome and reduced the infarction volume by 50% within 3 days. 3K3A-APC compared with wt-APC multi-dosing therapy at 12:00 h, 1, 3, 5 and 7 days after dMCAO significantly improved functional recovery and reduced the infarction volume by 75% and 38%, respectively, within 7 days. The wt-APC, but not 3K3A-APC, significantly increased the risk of intracerebral bleeding as indicated by a 50% increase in hemoglobin levels in the ischemic hemisphere. Thus, 3K3A-APC offers a new approach for safer and more efficacious treatments of neurodegenerative disorders and stroke with APC.


Assuntos
Anticoagulantes/uso terapêutico , Mutação/fisiologia , Fármacos Neuroprotetores/uso terapêutico , Proteína C/genética , Proteína C/uso terapêutico , Análise de Variância , Animais , Anticorpos/farmacologia , Anticoagulantes/metabolismo , Apoptose/efeitos dos fármacos , Encéfalo/citologia , Caspase 3/metabolismo , Caspase 9/metabolismo , Células Cultivadas , Embrião de Mamíferos , Células Endoteliais/efeitos dos fármacos , Inibidores Enzimáticos/farmacologia , Agonistas de Aminoácidos Excitatórios/toxicidade , Feminino , Glucose/deficiência , Hemoglobinas/metabolismo , Humanos , Hipóxia/tratamento farmacológico , Marcação In Situ das Extremidades Cortadas/métodos , Infarto da Artéria Cerebral Média/tratamento farmacológico , Infarto da Artéria Cerebral Média/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , N-Metilaspartato/toxicidade , Neurônios/efeitos dos fármacos , Neurônios/fisiologia , Fármacos Neuroprotetores/metabolismo , Gravidez , Proteína C/química , Proteína C/imunologia , Receptores Ativados por Proteinase/genética , Fatores de Tempo , Proteína Supressora de Tumor p53/metabolismo
11.
J Neurosci ; 27(2): 304-7, 2007 Jan 10.
Artigo em Inglês | MEDLINE | ID: mdl-17215390

RESUMO

We sought genetic evidence for the involvement of neuronal vascular endothelial growth factor (VEGF) in amyotrophic lateral sclerosis (ALS). Mice expressing human ALS mutant superoxide dismutase-1 (SOD1) were crossed with mice that overexpress VEGF in neurons (VEGF+/+). We report that SOD1(G93A)/VEGF+/+ double-transgenic mice show delayed motor neuron loss, delayed motor impairment, and prolonged survival compared with SOD1(G93A) single transgenics. These findings indicate that neuronal VEGF protects against motor neuron degeneration, and may have therapeutic implications for ALS.


Assuntos
Esclerose Lateral Amiotrófica/metabolismo , Regulação da Expressão Gênica/fisiologia , Degeneração Neural/metabolismo , Degeneração Neural/prevenção & controle , Fator A de Crescimento do Endotélio Vascular/biossíntese , Fator A de Crescimento do Endotélio Vascular/genética , Esclerose Lateral Amiotrófica/mortalidade , Esclerose Lateral Amiotrófica/patologia , Animais , Modelos Animais de Doenças , Regulação Enzimológica da Expressão Gênica/fisiologia , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Neurônios Motores/patologia , Degeneração Neural/enzimologia , Degeneração Neural/patologia , Superóxido Dismutase/biossíntese , Superóxido Dismutase/genética , Superóxido Dismutase-1 , Taxa de Sobrevida , Fator A de Crescimento do Endotélio Vascular/fisiologia
12.
J Neurosci Res ; 85(4): 740-7, 2007 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-17243175

RESUMO

New neurons are generated continuously in the subventricular zone and dentate gyrus of the adult brain. Neuropathologic processes, including cerebral ischemia, can enhance neurogenesis, as can growth factors and other physiologic stimuli. Vascular endothelial growth factor (VEGF) is an angiogenic and neuroprotective growth factor that can promote neurogenesis, but it is unknown whether VEGF can enhance migration of newborn neurons toward sites of ischemic injury, where they might be able to replace neurons that undergo ischemic death. In the present study we produced permanent focal cerebral ischemia in transgenic (Tg) mice that overexpress VEGF. Cell proliferation and neurogenesis were assessed with bromodeoxyuridine (Brdu) labeling and immunostaining for cell type-specific markers. In VEGF-Tg mice, brains examined 7-28 days after cerebral ischemia showed markedly increased subventricular zone (SVZ) neurogenesis, chains of neuroblasts extending from the SVZ to the peri-infarct cortex, and an increase in the number of newly generated cortical neurons at 14-28 days after ischemia. In concert with these effects, VEGF overexpression reduced infarct volume and improved postischemic motor function. These findings provide evidence that VEGF increases SVZ neurogenesis and neuromigration, consistent with a possible role in repair. Our data suggest that in addition to its neuroprotective effects, which are associated with improved outcome in the acute phase after cerebral ischemia, VEGF enhances postischemic neurogenesis, which could provide a therapeutic target for more chronic brain repair.


Assuntos
Movimento Celular/genética , Proliferação de Células , Regulação da Expressão Gênica/fisiologia , Infarto da Artéria Cerebral Média/patologia , Neurônios/fisiologia , Fator A de Crescimento do Endotélio Vascular/metabolismo , Animais , Infarto Encefálico/etiologia , Infarto Encefálico/patologia , Bromodesoxiuridina/metabolismo , Contagem de Células/métodos , Modelos Animais de Doenças , Lateralidade Funcional , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Atividade Motora/genética , Neurônios/patologia , Fosfopiruvato Hidratase/metabolismo , Fatores de Tempo
13.
Brain Res ; 1115(1): 186-93, 2006 Oct 18.
Artigo em Inglês | MEDLINE | ID: mdl-16928361

RESUMO

Vascular endothelial growth factor (VEGF) is an angiogenesis factor with neurotrophic, neuroprotective and neuroproliferative effects. Depending on the dose, route and time of administration in relation to focal cerebral ischemia, VEGF can improve histological outcome and sensorimotor function in rodents. However, VEGF also increases vascular permeability, which can lead to brain edema and exacerbate ischemic brain injury. Thus, although VEGF is a candidate therapeutic for stroke and other ischemic disorders, its benefit relative to risk is uncertain. Considering that functional rather than histological measures of outcome are probably most relevant to therapeutic prospects for human stroke, we investigated the effects of VEGF after middle cerebral artery occlusion in rats using a series of behavioral tests. We report that VEGF improves functional outcome in ischemic rats, including both sensorimotor and cognitive deficiencies.


Assuntos
Isquemia Encefálica/tratamento farmacológico , Transtornos Cognitivos/tratamento farmacológico , Transtornos dos Movimentos/tratamento farmacológico , Recuperação de Função Fisiológica/efeitos dos fármacos , Fator A de Crescimento do Endotélio Vascular/farmacologia , Animais , Barreira Hematoencefálica/efeitos dos fármacos , Barreira Hematoencefálica/fisiopatologia , Edema Encefálico/induzido quimicamente , Edema Encefálico/fisiopatologia , Edema Encefálico/prevenção & controle , Isquemia Encefálica/fisiopatologia , Isquemia Encefálica/psicologia , Córtex Cerebral/irrigação sanguínea , Córtex Cerebral/efeitos dos fármacos , Córtex Cerebral/fisiopatologia , Infarto Cerebral/fisiopatologia , Infarto Cerebral/prevenção & controle , Transtornos Cognitivos/fisiopatologia , Modelos Animais de Doenças , Infarto da Artéria Cerebral Média/fisiopatologia , Infarto da Artéria Cerebral Média/prevenção & controle , Masculino , Aprendizagem em Labirinto/efeitos dos fármacos , Aprendizagem em Labirinto/fisiologia , Transtornos dos Movimentos/fisiopatologia , Fármacos Neuroprotetores/farmacologia , Fármacos Neuroprotetores/uso terapêutico , Ratos , Ratos Sprague-Dawley , Recuperação de Função Fisiológica/fisiologia , Resultado do Tratamento , Fator A de Crescimento do Endotélio Vascular/efeitos adversos , Fator A de Crescimento do Endotélio Vascular/uso terapêutico
14.
FASEB J ; 20(8): 1185-7, 2006 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-16641198

RESUMO

Based on its trophic influence on neurons and vascular cells, vascular endothelial growth factor (VEGF) is a promising candidate for stroke treatment. VEGF's survival-promoting effects are purchased at the expense of an increased blood brain barrier permeability, which potentially compromises tissue survival. The mechanisms via which VEGF protects the brain against ischemia remained unknown. We examined signaling pathways underlying VEGF's neuroprotective activity in our transgenic mouse line, which expresses human VEGF165 under a neuron-specific enolase (NSE) promoter. We show that VEGF receptor-2 (Flk-1) is expressed on ischemic neurons and astrocytes and is activated by VEGF. Following 90-min episodes of middle cerebral artery occlusion, VEGF increased phosphorylated (but not total) Akt and ERK-1/-2 and reduced phosphorylated mitogen activated protein kinase/p38 and c-Jun NH2-terminal kinase (JNK)-1/-2 levels, at the same time decreasing inducible NO synthase expression in ischemic neurons. Inhibition of Akt with Wortmannin reversed VEGF's neuroprotective properties, diminished brain swelling, and restored the vascular permeability induced by VEGF to below levels in WT animals. The aggravation of brain injury by Wortmannin was associated with the restitution of p38, but not of JNK-1/-2, ERK-1/-2, or inducible NOS (iNOS). Our data demonstrate that VEGF mediates both neuroprotection and blood brain barrier permeability via the phosphatidylinositol-3 kinase (PI3K)/Akt pathway. Based on our observation that VEGF neuroprotection and vascular leakage depend on PI3K/Akt, which is putatively regulated by VEGF receptor-2, we predict that it may not easily be possible to make use of VEGF's neuroprotective function without accepting its unfavorable consequence, the increased vascular permeability.


Assuntos
Barreira Hematoencefálica/metabolismo , Isquemia Encefálica/enzimologia , Fosfatidilinositol 3-Quinases/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Fator A de Crescimento do Endotélio Vascular/fisiologia , Androstadienos/farmacologia , Animais , Astrócitos/metabolismo , Barreira Hematoencefálica/efeitos dos fármacos , Isquemia Encefálica/sangue , Isquemia Encefálica/metabolismo , Permeabilidade Capilar , Inibidores Enzimáticos/farmacologia , Humanos , Camundongos , Camundongos Transgênicos , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Neurônios/metabolismo , Fármacos Neuroprotetores , Óxido Nítrico Sintase Tipo II/metabolismo , Inibidores de Fosfoinositídeo-3 Quinase , Proteínas Proto-Oncogênicas c-akt/antagonistas & inibidores , Transdução de Sinais/efeitos dos fármacos , Fator A de Crescimento do Endotélio Vascular/antagonistas & inibidores , Fator A de Crescimento do Endotélio Vascular/genética , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/metabolismo , Wortmanina
15.
Brain ; 128(Pt 1): 52-63, 2005 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-15509618

RESUMO

Therapeutic angiogenesis with vascular endothelial growth factor (VEGF) is a clinically promising strategy in ischaemic disease. The pathophysiological consequences of enhanced vessel formation, however, are poorly understood. We established mice overexpressing human VEGF165 under a neuron-specific promoter, which exhibited an increased density of brain vessels under physiological conditions and enhanced angiogenesis after brain ischaemia. Following transient intraluminal middle cerebral artery (MCA) occlusions, VEGF overexpression significantly alleviated neurological deficits and infarct volume, and reduced disseminated neuronal injury and caspase-3 activity, confirming earlier observations that VEGF has neuroprotective properties. Brain swelling was not influenced in VEGF-overexpressing animals, while sodium fluorescein extravasation was moderately increased, suggesting that VEGF induces a mild blood-brain barrier leakage. To elucidate whether enhanced angiogenesis improves regional cerebral blood flow in the ischaemic brain, [14C]iodoantipyrine autoradiography was performed. Autoradiographies revealed that VEGF induces haemodynamic steal phenomena with reduced blood flow in ischaemic areas and increased flow values only outside the MCA territory. Our data demonstrate that VEGF protects neurons from ischaemic cell death by a direct action rather than by promoting angiogenesis, and suggest that strategies aiming at increasing vascular density in the whole brain, e.g. by VEGF overexpression, may worsen rather than improve cerebral haemodynamics after stroke.


Assuntos
Isquemia Encefálica/fisiopatologia , Encéfalo/irrigação sanguínea , Circulação Cerebrovascular/fisiologia , Fator A de Crescimento do Endotélio Vascular/metabolismo , Animais , Barreira Hematoencefálica/fisiopatologia , Encéfalo/metabolismo , Encéfalo/patologia , Isquemia Encefálica/patologia , Capilares/fisiopatologia , Permeabilidade Capilar/fisiologia , Hemodinâmica/fisiologia , Humanos , Camundongos , Camundongos Transgênicos , Artéria Cerebral Média/fisiologia , Necrose , Neovascularização Patológica/patologia , Neovascularização Patológica/fisiopatologia , Neurônios/fisiologia , Acidente Vascular Cerebral/patologia , Acidente Vascular Cerebral/fisiopatologia , Fator A de Crescimento do Endotélio Vascular/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA