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










Base de datos
Intervalo de año de publicación
1.
Phytother Res ; 27(4): 564-71, 2013 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-22678994

RESUMEN

Mitochondrial membrane potential (∆Ψm ) contributes to determining a driving force for calcium to enter the mitochondria. It has been demonstrated that even a small mitochondrial depolarization is sufficient to prevent mitochondrial calcium overload and the subsequent apoptosis. Therefore, mild mitochondrial depolarization has been recently evaluated as a novel mechanism of neuroprotection via inhibiting neurotoxic mitochondrial calcium overload during neuronal insults. In the present study, using both real-time recording and flow cytometric analyses of ∆Ψm , we demonstrated that ethanolic peel extract of Citrus sunki Hort. ex Tanaka (CPE) and its active compounds are capable of inducing a mild mitochondrial depolarization. Polymethoxylated flavones such as nobiletin and tangeretin were found as the active compounds responsible for CPE effects on ∆Ψm . Neuronal viability was significantly increased in a dose-dependent manner by CPE treatment in H2 O2 -stimulated HT-22 cells as an in vitro neuronal insult model. CPE treatment significantly inhibited H2 O2 -induced apoptotic processes such as chromatin condensation, caspase 3 activation and anti-poly (ADP-ribose) polymerase (PARP) cleavage. CPE treatment significantly blocked mitochondrial calcium overload in H2 O2 -stimulated HT-22 neurons as indicated by rhod-2 acetoxymethyl ester. Taken together, our findings suggest that CPE and its active compounds may be considered as promising neuroprotective agents via inducing a mild mitochondrial depolarization.


Asunto(s)
Citrus/química , Potencial de la Membrana Mitocondrial/efectos de los fármacos , Neuronas/efectos de los fármacos , Fármacos Neuroprotectores/farmacología , Extractos Vegetales/farmacología , Apoptosis , Calcio/metabolismo , Línea Celular , Flavonas/farmacología , Citometría de Flujo , Frutas/química , Humanos , Peróxido de Hidrógeno/farmacología , Mitocondrias/efectos de los fármacos
2.
Stroke ; 42(1): 37-43, 2011 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-21164131

RESUMEN

BACKGROUND AND PURPOSE: because brain endothelial cells exist at the neurovascular interface, they may serve as cellular reporters of brain dysfunction by releasing biomarkers into the circulation. METHODS: we used proteomic techniques to screen conditioned media from human brain endothelial cultures subjected to oxidative stress induced by nitric oxide over 24 hours. Plasma samples from human stroke patients were analyzed by enzyme-linked immunosorbent assay. RESULTS: in healthy endothelial cells, interaction mapping demonstrated cross-talk involving secreted factors, membrane receptors, and matrix components. In oxidatively challenged endothelial cells, networks of interacting proteins failed to emerge. Instead, inflammatory markers increased, secreted factors oscillated over time, and endothelial injury repair was manifested as changes in factors related to matrix integrity. Elevated inflammatory markers included heat shock protein, chemokine ligand-1, serum amyloid-A1, annexin-A5, and thrombospondin-1. Neurotrophic factors (prosaposin, nucleobindin-1, and tachykinin precursors) peaked at 12 hours, then rapidly decreased by 24 hours. Basement membrane components (fibronectin, desomoglein, profiling-1) were decreased. Cytoskeletal markers (actin, vimentin, nidogen, and filamin B) increased over time. From this initial analysis, the high-ranking candidate thrombospondin-1 was further explored in human plasma. Acute ischemic stroke patients had significantly higher thrombospondin-1 levels within 8 hours of symptom onset compared to controls with similar clinical risk factors (659 ± 81 vs 1132 ± 98 ng/mL; P<0.05; n=20). CONCLUSIONS: screening of simplified cell culture systems may aid the discovery of novel biomarkers in clinical neurovascular injury. Further collaborative efforts are warranted to discover and validate more candidates of interest.


Asunto(s)
Encéfalo/metabolismo , Células Endoteliales/metabolismo , Endotelio Vascular/metabolismo , Estrés Oxidativo , Proteoma/metabolismo , Accidente Cerebrovascular/metabolismo , Biomarcadores/metabolismo , Encéfalo/patología , Línea Celular , Células Endoteliales/patología , Endotelio Vascular/patología , Femenino , Depuradores de Radicales Libres/farmacología , Humanos , Mediadores de Inflamación/metabolismo , Masculino , Óxido Nítrico/farmacología , Accidente Cerebrovascular/patología , Factores de Tiempo
3.
Biol Pharm Bull ; 33(11): 1814-21, 2010.
Artículo en Inglés | MEDLINE | ID: mdl-21048305

RESUMEN

A growing body of evidence suggests that nobiletin (5,6,7,8,3',4'-hexamethoxy flavone) from the peel of citrus fruits, enhances the damaged cognitive function in disease animal models. However, the neuroprotective mechanism has not been clearly elucidated. Since nobiletin has shown anti-inflammatory effects in several tissues, we investigated whether nobiletin suppresses excessive microglial activation implicated in neurotoxicity in lipopolysaccharide (LPS)-stimulated BV-2 microglia cell culture models. Release of nitric oxide (NO), the major inflammatory mediator in microglia, was markedly suppressed in a dose-dependent manner following nobiletin treatment (1-50 µM) in LPS-stimulated BV-2 microglia cells. The inhibitory effect of nobiletin was similar to that of minocycline, a well-known microglial inactivator. Nobiletin significantly inhibited the release of the pro-inflammatory cytokine tumor necrosis factor (TNF-α) and interleukin-1ß (IL-1ß). LPS-induced phosphorylations of extracellular signal-regulated kinase (ERK), c-Jun NH(2)-terminal kinase (JNK), and p38 mitogen-activated protein kinases (MAPKs) were also significantly inhibited by nobiletin treatment. In addition, nobiletin markedly inhibited the LPS-induced pro-inflammatory transcription factor nuclear factor κB (NF-κB) signaling pathway by suppressing nuclear NF-κB translocation from the cytoplasm and subsequent expression of NF-κB in the nucleus. Taken together, these results may contribute to further exploration of the therapeutic potential and molecular mechanism of nobiletin in relation to neuroinflammation and neurodegenerative diseases.


Asunto(s)
Antiinflamatorios/farmacología , Citrus/química , Inflamación/tratamiento farmacológico , Microglía/efectos de los fármacos , Fármacos Neuroprotectores/farmacología , Extractos Vegetales/farmacología , Animales , Transporte Biológico/efectos de los fármacos , Línea Celular , Núcleo Celular/efectos de los fármacos , Núcleo Celular/metabolismo , Citocinas/antagonistas & inhibidores , Citoplasma/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Frutas , Inflamación/metabolismo , Lipopolisacáridos , Ratones , Microglía/metabolismo , Minociclina/farmacología , Proteínas Quinasas Activadas por Mitógenos/antagonistas & inhibidores , FN-kappa B/antagonistas & inhibidores , Enfermedades Neurodegenerativas/tratamiento farmacológico , Óxido Nítrico/antagonistas & inhibidores , Fosforilación , Fitoterapia , Transducción de Señal/efectos de los fármacos
4.
J Cell Biochem ; 108(2): 476-88, 2009 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-19626665

RESUMEN

In this study, temporal and spatial distribution of three TGF-beta isoforms and their downstream signaling pathways including pSmad2 and p38MAPK were examined during fibrotic wound repair. In normal chick corneas, TGF-beta1, -2, and -3 were weakly detected in Bowman's layer (BL). In healing corneas, TGF-beta1 was primarily deposited in the fibrin clot and the unwounded BL. TGF-beta2 was highly expressed in healing epithelial and endothelial cells, and numerous active fibroblasts/myofibroblasts. TGF-beta3 was mainly detected in the unwound region of basal epithelial cells. alpha-Smooth muscle actin (alpha-SMA) was initially appeared in the posterior region of repairing stroma at day 3, and was detected in the entire healing stroma by day 7. Notably, alpha-SMA was absent in the central region of healing stroma by day 14, and its staining pattern was similar to those of TGF-beta2 and p38MAPK. By contrast, pSmad2 was mainly detected in the fibroblasts. In normal cornea, laminin was mainly detected in both epithelial basement membrane (BM) and Descemet's membrane (DM). By contrast to reconstitution of the BM in the wound region, the DM was not repaired although endothelial layer was regenerated, indicating that high levels of TGF-beta2 were released into the posterior region of healing stroma on day 14. High levels of alpha-SMA staining, shown in cultured repair stromal cells from healing corneas on day 14 and in TGF-beta2 treated normal stromal cells, were significantly reduced by p38MAPK inhibition. Collectively, this study suggests that TGF-beta2-mediated myofibroblast transformation is mediated, at least partly, by the p38MAPK pathway in vivo.


Asunto(s)
Lesiones de la Cornea , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Factor de Crecimiento Transformador beta/metabolismo , Cicatrización de Heridas , Técnicas de Ablación , Actinas/metabolismo , Envejecimiento , Animales , Membrana Basal/metabolismo , Lámina Limitante Anterior/metabolismo , Células Cultivadas , Pollos , Córnea/patología , Córnea/cirugía , Lámina Limitante Posterior/metabolismo , Fibrina/metabolismo , Fibrosis , Laminina/metabolismo , Isoformas de Proteínas , Proteínas Serina-Treonina Quinasas/antagonistas & inhibidores , Receptor Tipo I de Factor de Crecimiento Transformador beta , Receptores de Factores de Crecimiento Transformadores beta/antagonistas & inhibidores , Proteína Smad2/metabolismo , Células del Estroma/metabolismo , Células del Estroma/patología , Factores de Tiempo , Proteínas Quinasas p38 Activadas por Mitógenos/antagonistas & inhibidores , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
5.
J Neurosci Res ; 87(1): 164-70, 2009 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-18711728

RESUMEN

Neuroglobin (Ngb) is a recently discovered tissue globin with a high affinity for oxygen that is widely and specifically expressed in neurons of vertebrate central and peripheral nervous systems. Our laboratory and others have shown Ngb overexpression can protect neurons against hypoxic/ischemic insults, but the underlying mechanisms remain poorly understood. In this study, we examined the effects of Ngb overexpression on mitochondrial function, oxidative stress, and neurotoxicity in primary cortical neurons following hypoxia/reoxygenation (H/R). Ngb-overexpressing transgenic neurons (Ngb-Tg) were significantly protected against H/R-induced cell death. Rates of decline in ATP levels, MTT reduction, and mitochondrial membrane potential were significantly ameliorated in Ngb-Tg neurons. Furthermore, Ngb overexpression reduced superoxide anion generation after H/R, whereas glutathione levels were significantly improved compared with WT controls. Taken together, these data suggest that Ngb is neuroprotective against hypoxia, in part by improving mitochondria function and decreasing oxidative stress.


Asunto(s)
Globinas/metabolismo , Mitocondrias/fisiología , Proteínas del Tejido Nervioso/metabolismo , Neuronas/fisiología , Neuronas/ultraestructura , Estrés Oxidativo/fisiología , Adenosina Trifosfato/metabolismo , Análisis de Varianza , Animales , Células Cultivadas , Corteza Cerebral/citología , Embrión de Mamíferos , Globinas/genética , Glutatión/metabolismo , Oxigenoterapia Hiperbárica/métodos , Hipoxia , L-Lactato Deshidrogenasa/metabolismo , Potencial de la Membrana Mitocondrial/efectos de los fármacos , Potencial de la Membrana Mitocondrial/genética , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Mitocondrias/genética , Proteínas del Tejido Nervioso/genética , Neuroglobina , Estrés Oxidativo/genética , Fenantridinas/metabolismo , Sales de Tetrazolio , Tiazoles
6.
Proc Natl Acad Sci U S A ; 105(21): 7582-7, 2008 May 27.
Artículo en Inglés | MEDLINE | ID: mdl-18495934

RESUMEN

The neurovascular unit is an emerging concept that emphasizes homeostatic interactions between endothelium and cerebral parenchyma. Here, we show that cerebral endothelium are not just inert tubes for delivering blood, but they also secrete trophic factors that can be directly neuroprotective. Conditioned media from cerebral endothelial cells broadly protects neurons against oxygen-glucose deprivation, oxidative damage, endoplasmic reticulum stress, hypoxia, and amyloid neurotoxicity. This phenomenon is largely mediated by endothelial-produced brain-derived neurotrophic factor (BDNF) because filtering endothelial-conditioned media with TrkB-Fc eliminates the neuroprotective effect. Endothelial production of BDNF is sustained by beta-1 integrin and integrin-linked kinase (ILK) signaling. Noncytotoxic levels of oxidative stress disrupts ILK signaling and reduces endothelial levels of neuroprotective BDNF. These data suggest that cerebral endothelium provides a critical source of homeostatic support for neurons. Targeting these signals of matrix and trophic coupling between endothelium and neurons may provide new therapeutic opportunities for stroke and other CNS disorders.


Asunto(s)
Factor Neurotrófico Derivado del Encéfalo/metabolismo , Encéfalo/citología , Encéfalo/metabolismo , Comunicación Celular , Endotelio Vascular/fisiología , Neuronas/fisiología , Animales , Células Cultivadas , Endotelio Vascular/citología , Endotelio Vascular/metabolismo , Integrina beta1/metabolismo , Masculino , Ratones , Ratones Endogámicos , Neuronas/citología , Neuronas/metabolismo , Estrés Oxidativo , Proteínas Serina-Treonina Quinasas/metabolismo
7.
Biosci Biotechnol Biochem ; 71(7): 1781-4, 2007 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-17617702

RESUMEN

Nobiletin contributes to pharmacological activities such as anti-cancer and anti-inflammatory effects, but little is known about its effect on melanogenesis. In this study, we found that nobiletin increased melanin content and tyrosinase activity in murine B16/F10 melanoma cells. Furthermore, inhibition of the extracellular signal-regulated kinase (ERK) pathway with U0216 resulted in inhibition of nobiletin-induced melanin synthesis and tyrosinase expression.


Asunto(s)
Quinasas MAP Reguladas por Señal Extracelular/fisiología , Flavonas/fisiología , Flavonoides/fisiología , Melaninas/biosíntesis , Melanoma Experimental/enzimología , Animales , Línea Celular Tumoral , Melanoma Experimental/etiología , Ratones , Monofenol Monooxigenasa/fisiología
8.
Neurosci Lett ; 417(1): 1-5, 2007 Apr 24.
Artículo en Inglés | MEDLINE | ID: mdl-17386975

RESUMEN

Plasminogen activators are used in thrombolytic stroke therapy. However, it is increasingly recognized that they have other actions besides fibrinolysis. In this study, we assess potential pro-inflammatory effects of tissue-type plasminogen activator (tPA) and urokinase-type plasminogen activator (uPA) in rat cortical astrocytes. Both uPA and tPA induced rapid dose-dependent upregulation in MMP-2 and MMP-9, as demonstrated by zymography of conditioned media. In addition, a multiplex ELISA array demonstrated that patterned responses in chemokines and cytokines were also evoked. Exposure to tPA induced elevations in secreted MIP-2, MCP-1 and GRO/KC. Exposure to uPA induced elevations in secreted IFN-gamma, TNF-alpha, GMCSF, MIP-1alpha, MIP-2, MIP-3alpha, MCP-1, RANTES and fractalkine. These data suggest that plasminogen activators may trigger selected pro-inflammatory responses at the neurovascular interface. Whether these effects influence thrombolytic stroke therapy warrants further investigation.


Asunto(s)
Astrocitos/efectos de los fármacos , Corteza Cerebral/efectos de los fármacos , Encefalitis/inducido químicamente , Mediadores de Inflamación/inmunología , Mediadores de Inflamación/metabolismo , Activadores Plasminogénicos/efectos adversos , Animales , Animales Recién Nacidos , Astrocitos/inmunología , Astrocitos/metabolismo , Biomarcadores/análisis , Biomarcadores/metabolismo , Barrera Hematoencefálica/efectos de los fármacos , Barrera Hematoencefálica/inmunología , Células Cultivadas , Corteza Cerebral/inmunología , Corteza Cerebral/metabolismo , Quimiocinas/inmunología , Quimiocinas/metabolismo , Citocinas/inmunología , Citocinas/metabolismo , Relación Dosis-Respuesta a Droga , Encefalitis/inmunología , Encefalitis/metabolismo , Gliosis/inducido químicamente , Gliosis/inmunología , Gliosis/metabolismo , Trombosis Intracraneal/tratamiento farmacológico , Metaloproteinasa 2 de la Matriz/efectos de los fármacos , Metaloproteinasa 2 de la Matriz/inmunología , Metaloproteinasa 2 de la Matriz/metabolismo , Metaloproteinasa 9 de la Matriz/efectos de los fármacos , Metaloproteinasa 9 de la Matriz/inmunología , Metaloproteinasa 9 de la Matriz/metabolismo , Metaloproteinasas de la Matriz/efectos de los fármacos , Metaloproteinasas de la Matriz/inmunología , Metaloproteinasas de la Matriz/metabolismo , Ratas , Ratas Sprague-Dawley , Regulación hacia Arriba/efectos de los fármacos , Regulación hacia Arriba/inmunología
9.
Brain Res ; 1111(1): 222-6, 2006 Sep 21.
Artículo en Inglés | MEDLINE | ID: mdl-16919608

RESUMEN

Matrix metalloproteinase-9 (MMP-9) may play a role in the inflammatory glial response during Alzheimer's disease (AD). Astrocytes can degrade beta-amyloid (Abeta) and extracellular proteolysis via MMP-9 may be involved. Because Apolipoprotein E (APOE) genotype is an important factor for AD, we ask whether various apoE isoforms can influence Abeta-induced MMP-9 responses in primary rat astrocytes. Our data show that apoE4 significantly dampens Abeta-induced MMP-9 levels, possibly by downregulating the Rho-Rho kinase (ROCK) pathway. Reduction of astrocytic MMP-9 by apoE4 may affect Abeta clearance and promote Abeta deposition in AD.


Asunto(s)
Péptidos beta-Amiloides/metabolismo , Apolipoproteínas E/metabolismo , Astrocitos/enzimología , Gliosis/enzimología , Mediadores de Inflamación/metabolismo , Metaloproteinasa 9 de la Matriz/metabolismo , Enfermedad de Alzheimer/enzimología , Enfermedad de Alzheimer/fisiopatología , Péptidos beta-Amiloides/antagonistas & inhibidores , Animales , Animales Recién Nacidos , Apolipoproteínas E/química , Apolipoproteínas E/farmacología , Astrocitos/efectos de los fármacos , Encéfalo/enzimología , Encéfalo/fisiopatología , Células Cultivadas , Regulación hacia Abajo/efectos de los fármacos , Regulación hacia Abajo/fisiología , Gliosis/inducido químicamente , Gliosis/fisiopatología , Mediadores de Inflamación/antagonistas & inhibidores , Péptidos y Proteínas de Señalización Intracelular/efectos de los fármacos , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Metaloproteinasa 9 de la Matriz/efectos de los fármacos , Placa Amiloide/metabolismo , Isoformas de Proteínas/química , Isoformas de Proteínas/metabolismo , Isoformas de Proteínas/farmacología , Proteínas Serina-Treonina Quinasas/efectos de los fármacos , Proteínas Serina-Treonina Quinasas/metabolismo , Ratas , Ratas Sprague-Dawley , Quinasas Asociadas a rho
10.
Stroke ; 37(7): 1910-2, 2006 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-16741180

RESUMEN

BACKGROUND AND PURPOSE: Hemorrhagic conversion after tissue plasminogen activator (tPA) stroke therapy has been linked with elevations in matrix metalloproteinase-9 (MMP-9) at the neurovascular interface. Here, we test the idea that statins may directly ameliorate tPA-induced MMP-9 dysregulation. METHODS: Recombinant human tPA (5 microg/mL) was added to primary rat cortical astrocytes. Zymography was used to quantify MMP-9 levels in conditioned media. Effects of simvastatin or the Rho kinase inhibitor Y-27632 were assessed by pretreating cells before tPA exposure. RESULTS: Simvastatin (1 to 10 micromol/L) significantly reduced tPA-induced MMP-9 in cortical astrocytes. This effect may be mediated via the Rho kinase pathway because tPA-induced activation of Rho signaling was suppressed by simvastatin, and tPA-induced MMP-9 levels were similarly reduced by the Rho kinase inhibitor Y-27632 (1 to 10 micromol/L). CONCLUSIONS: Statins reduce tPA-induced MMP-9 dysregulation by inhibiting the Rho signaling pathway. Statins may ameliorate tPA-associated MMP imbalances in stroke.


Asunto(s)
Astrocitos/efectos de los fármacos , Inhibidores de Hidroximetilglutaril-CoA Reductasas/farmacología , Metaloproteinasa 9 de la Matriz/biosíntesis , Fármacos Neuroprotectores/farmacología , Transducción de Señal/efectos de los fármacos , Simvastatina/farmacología , Amidas/farmacología , Animales , Astrocitos/enzimología , Células Cultivadas/efectos de los fármacos , Células Cultivadas/enzimología , Hemorragia Cerebral/etiología , Evaluación Preclínica de Medicamentos , Inducción Enzimática/efectos de los fármacos , Péptidos y Proteínas de Señalización Intracelular , Metaloproteinasa 2 de la Matriz/biosíntesis , Metaloproteinasa 2 de la Matriz/genética , Metaloproteinasa 9 de la Matriz/genética , Proteínas Serina-Treonina Quinasas/antagonistas & inhibidores , Piridinas/farmacología , Ratas , Proteínas Recombinantes/farmacología , Accidente Cerebrovascular/complicaciones , Accidente Cerebrovascular/enzimología , Quinasas Asociadas a rho
11.
Stroke ; 36(9): 1954-9, 2005 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-16051896

RESUMEN

BACKGROUND AND PURPOSE: Thrombolytic therapy with tissue plasminogen activator (tPA) in ischemic stroke is limited by increased risks of cerebral hemorrhage and brain injury. In part, these phenomena may be related to neurovascular proteolysis mediated by matrix metalloproteinases (MMPs). Here, we used a combination of pharmacological and genetic approaches to show that tPA promotes MMP-9 levels in stroke in vivo. METHODS: In the first experiment, spontaneously hypertensive rats were subjected to 3 hours of transient focal cerebral ischemia. The effects of tPA (10 mg/kg IV) on ischemic brain MMP-9 levels were assessed by zymography. In the second experiment, wild-type (WT) and tPA knockout mice were subjected to 2 hours of transient focal cerebral ischemia, and MMP-9 levels and brain edema during reperfusion were assessed. Phenotype rescue was performed by administering tPA to the tPA knockout mice. RESULTS: In the first experiment, exogenous tPA did not change infarct size but amplified MMP-9 levels in ischemic rat brain at 24 hours. Coinfusion of the plasmin inhibitor tranexamic acid (300 mg/kg) did not ameliorate this effect, suggesting that it was independent of plasmin. In the second experiment, ischemic MMP-9 levels, infarct size, and brain edema in tPA knockouts were significantly lower than WT mice. Administration of exogenous tPA (10 mg/kg IV) did not alter infarction but reinstated the ischemic MMP-9 response back up to WT levels and correspondingly worsened edema. CONCLUSIONS: These data demonstrate that tPA upregulates brain MMP-9 levels in stroke in vivo, and suggest that combination therapies targeting MMPs may improve tPA therapy.


Asunto(s)
Isquemia Encefálica/metabolismo , Isquemia Encefálica/terapia , Metaloproteinasa 9 de la Matriz/biosíntesis , Activador de Tejido Plasminógeno/metabolismo , Regulación hacia Arriba , Animales , Barrera Hematoencefálica , Encéfalo/patología , Infarto Encefálico/patología , Modelos Animales de Enfermedad , Edema/metabolismo , Edema/patología , Electroforesis en Gel de Poliacrilamida , Fibrinolisina/metabolismo , Humanos , Hidrógeno/metabolismo , Inmunohistoquímica , Isquemia/patología , Metaloproteinasa 2 de la Matriz/biosíntesis , Metaloproteinasa 9 de la Matriz/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Perfusión , Fenotipo , ARN Mensajero/metabolismo , Ratas , Ratas Endogámicas SHR , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transducción de Señal , Factores de Tiempo
12.
J Cell Physiol ; 205(2): 270-7, 2005 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-15887249

RESUMEN

Akt is a key downstream effector of the PI3K signaling pathway and plays a role in cell growth and survival. Expression of a myristoylated constitutively active form of Akt (myr-Akt) in PC12 cells could override cell-growth arrest at G2/M phase and apoptosis that were induced by etoposide treatment. On the other hand, inactivation of Akt by expression of its dominant negative mutant form (km-Akt) inhibited cell proliferation by arresting the cells at G2/M phase. Expression of myr-Akt also led to an increase in the protein and mRNA levels of CDK1 and cyclin B1. Furthermore, EMSA data revealed that expression of myr-Akt promoted the binding of NF-Y to the consensus CCAAT promoter sequence, whereas expression of km-Akt almost completely abolished it. Moreover, the Akt activity was minimal in the cells that were arrested at G2/M phase by nocodazole treatment, but reached to a maximal level as the cells progressed to mitosis and G1 phase upon removal of the drug. Treatment with Akt inhibitors, but not with those of MEK or p70S6K, blocked the release of the cells from the nocodazole-induced G2/M arrest, further revealing that the Akt activity is required for G2/M phase transition. These results suggest that Akt facilitate cell-cycle progression at G2/M phase in PC12 cells and this Akt activity is correlated with upregulation of NF-Y DNA-binding activity and cyclin B1/CDK1 gene expression.


Asunto(s)
Factor de Unión a CCAAT/metabolismo , Ciclo Celular/fisiología , Fase G2 , Mitosis , Proteínas Proto-Oncogénicas c-akt/metabolismo , Regulación hacia Arriba , Animales , Western Blotting , Ensayo de Cambio de Movilidad Electroforética , Activación Enzimática , Células PC12 , Pruebas de Precipitina , Proteínas Proto-Oncogénicas c-akt/análisis , Ratas
13.
Stroke ; 35(11 Suppl 1): 2726-30, 2004 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-15459442

RESUMEN

Reperfusion therapy with tissue plasminogen activator (tPA) is a rational therapy for acute ischemic stroke. Properly titrated use of tPA improves clinical outcomes. However, there is also an associated risk of hemorrhagic transformation after tPA therapy. Emerging data now suggest that some of these potentially neurotoxic side effects of tPA may be due to its signaling actions in the neurovascular unit. Besides its intended role in clot lysis, tPA is also an extracellular protease and signaling molecule in brain. tPA mediates matrix remodeling during brain development and plasticity. By interacting with the NMDA-type glutamate receptor, tPA may amplify potentially excitotoxic calcium currents. At selected concentrations, tPA may be vasoactive. Finally, by augmenting matrix metalloproteinase (MMP) dysregulation after stroke, tPA may degrade extracellular matrix integrity and increase risks of neurovascular cell death, blood-brain barrier leakage, edema, and hemorrhage. Understanding these pleiotropic actions of tPA may reveal new therapeutic opportunities for combination stroke therapy.


Asunto(s)
Encéfalo/efectos de los fármacos , Hemorragia Cerebral/etiología , Activadores Plasminogénicos/farmacología , Accidente Cerebrovascular/tratamiento farmacológico , Terapia Trombolítica , Activador de Tejido Plasminógeno/farmacología , Animales , Matriz Extracelular/metabolismo , Humanos , Metaloproteinasa 9 de la Matriz/metabolismo , Metaloproteinasas de la Matriz/metabolismo , Activadores Plasminogénicos/efectos adversos , Accidente Cerebrovascular/complicaciones , Terapia Trombolítica/efectos adversos , Activador de Tejido Plasminógeno/efectos adversos , Vasoconstricción/efectos de los fármacos
14.
J Cereb Blood Flow Metab ; 24(7): 720-7, 2004 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-15241180

RESUMEN

Matrix metalloproteinases (MMPs) may contribute to the pathophysiology of cerebral ischemia by degrading matrix components in the neurovascular unit. In this study, the authors document a pathway by which MMPs interfere with cell-matrix interactions and trigger caspase-mediated cytotoxicity in brain endothelial cells. Hypoxia-reoxygenation induced endothelial cytotoxicity. Cytoprotection with zDEVD-fmk confirmed that cell death was partly caspase mediated. The temporal profile of caspase-3 activation was matched by elevations in MMP-2 and MMP-9. MMP inhibitors significantly decreased caspase-3 activation and reduced endothelial cell death. Degradation of matrix fibronectin confirmed the presence of extracellular proteolysis. Increasing integrin-linked kinase signaling with the beta1 integrin-activating antibody (8A2) ameliorated endothelial cytotoxicity. The results suggest that MMP-9 and MMP-2 contribute to caspase-mediated brain endothelial cell death after hypoxia-reoxygenation by disrupting cell-matrix interactions and homeostatic integrin signaling.


Asunto(s)
Encéfalo/patología , Caspasas/metabolismo , Células Endoteliales/patología , Hipoxia Encefálica/patología , Metaloproteinasas de la Matriz/fisiología , Daño por Reperfusión/patología , Encéfalo/enzimología , Caspasa 3 , Células Endoteliales/enzimología , Activación Enzimática/efectos de los fármacos , Inhibidores Enzimáticos/farmacología , Matriz Extracelular/metabolismo , Humanos , Integrina beta1/metabolismo , Metaloproteinasa 2 de la Matriz/metabolismo , Metaloproteinasa 9 de la Matriz/metabolismo , Inhibidores de la Metaloproteinasa de la Matriz , Proteínas Recombinantes , Daño por Reperfusión/enzimología , Transducción de Señal
15.
J Neurochem ; 89(1): 232-9, 2004 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-15030407

RESUMEN

Endoplasmic reticulum (ER) stress has increasingly come into focus as a factor contributing to neuronal injury. Although caspase-dependent mechanisms have been implicated in ER stress, the signaling pathways involved remain unclear. In this study, we examined the role of the extracellular signal-regulated kinase (ERK), a mitogen-activated protein (MAP) kinase pathway that is highly conserved in many systems for balancing cell survival and death. Prolonged treatment of the human neuroblastoma cell line SH-SY5Y with thapsigargin, an inducer of ER stress, increased cell death over 24-48 h, as measured by LDH release. Caspases were involved; increased levels of active caspase-3 and cleaved caspase substrate PARP were detected, and treatment with Z-VAD-FMK reduced thapsigargin-induced cytotoxicity. In contrast, inhibition of calpain was not protective, although calpain was activated following thapsigargin treatment. An early and transient phosphorylation of ERK1/2 occurred after thapsigargin-induced ER stress, and targeting this pathway with the MEK inhibitors U0126 or PD98059 significantly reduced cell death. Similar cytoprotection was obtained against brefeldin A, another ER stress agent. However, protection against ER stress via ERK inhibition was not accompanied by amelioration of caspase-3 activation, PARP cleavage, or DNA laddering. These data indicate that ERK may contribute to non-caspase-dependent pathways of injury after ER stress.


Asunto(s)
Retículo Endoplásmico/metabolismo , Proteínas de Choque Térmico , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Chaperonas Moleculares , Neuroblastoma/metabolismo , Estrés Fisiológico/metabolismo , Brefeldino A/farmacología , Proteínas Portadoras/metabolismo , Inhibidores de Caspasas , Caspasas/metabolismo , Muerte Celular/efectos de los fármacos , Línea Celular Tumoral , Retículo Endoplásmico/efectos de los fármacos , Chaperón BiP del Retículo Endoplásmico , Inhibidores Enzimáticos/farmacología , Humanos , Proteínas Quinasas Activadas por Mitógenos/antagonistas & inhibidores , Neuroblastoma/tratamiento farmacológico , Neuroblastoma/enzimología , Inhibidores de la Síntesis de la Proteína/farmacología , Estrés Fisiológico/inducido químicamente , Tapsigargina/farmacología
16.
J Neurosci ; 24(3): 671-8, 2004 Jan 21.
Artículo en Inglés | MEDLINE | ID: mdl-14736853

RESUMEN

Mechanisms of selective neuronal death in the hippocampus after global cerebral ischemia remain to be clarified. Here, we explored a possible role for matrix metalloproteinases (MMPs) in this phenomenon. Although many studies have demonstrated detrimental roles for the gelatinase MMP-9 in focal cerebral ischemia, how dysregulated MMP proteolysis influences global cerebral ischemia is less well understood. In this study, CD-1 mice were subjected to transient global ischemia. Transient occlusions of common carotid arteries for periods between 20 and 40 min led to increasing hippocampal neuronal death after 3 d. Gel zymography showed elevations in gelatinase (MMP-2 and MMP-9) activity. In situ zymography showed that gelatinase activity was mostly colocalized with neuron-specific nuclear protein-stained pyramidal neurons. Mice treated with the broad-spectrum metalloproteinase inhibitor BB-94 (50 mg/kg, i.p.) showed reduced hippocampal gelatinase activity after transient global cerebral ischemia and suffered significantly reduced hippocampal neuronal damage compared with vehicle-treated controls (p < 0.01). Additionally, hippocampal gelatinase activity and neuronal damage after transient global ischemia were also significantly reduced in MMP-9 knock-out mice compared with wild-type mice (p < 0.05). These data indicate a potential deleterious role for MMP-9 in the pathogenesis of delayed neuronal damage in the hippocampus after global cerebral ischemia.


Asunto(s)
Ataque Isquémico Transitorio/enzimología , Metaloproteinasas de la Matriz/metabolismo , Neuronas/enzimología , Fenilalanina/análogos & derivados , Animales , Dipéptidos/farmacología , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Hipocampo/enzimología , Hipocampo/patología , Inmunohistoquímica , Ataque Isquémico Transitorio/patología , Masculino , Metaloproteinasa 2 de la Matriz/metabolismo , Metaloproteinasa 9 de la Matriz/genética , Metaloproteinasa 9 de la Matriz/metabolismo , Inhibidores de la Metaloproteinasa de la Matriz , Metaloproteinasas de la Matriz/genética , Ratones , Ratones Noqueados , Neuronas/patología , Fármacos Neuroprotectores/farmacología , Fenilalanina/farmacología , Inhibidores de Proteasas/farmacología , Tiofenos/farmacología
17.
Neurol Res ; 26(8): 854-61, 2004 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-15727269

RESUMEN

The NINDS Stroke Progress Review Group recommended a shift in emphasis from a purely neurocentric view of cell death towards a more integrative approach whereby responses in all brain cells and matrix are considered. The neurovascular unit (fundamentally comprising endothelium, astrocyte, and neuron) provides a conceptual framework where cell-cell and cell-matrix signaling underlies the overall tissue response to stroke and its treatments. Here, we briefly review recent data on extracellular proteolytic dysfunction in the neurovascular unit after a stroke. The breakdown of neurovascular matrix initiates blood-brain barrier disruption with edema and/or hemorrhage. Endothelial dysfunction amplifies inflammatory responses. Perturbation of cell-matrix homeostasis triggers multiple cell death pathways. Interactions between the major classes of extracellular proteases from the plasminogen and matrix metalloprotease families may underlie processes responsible for some of the hemorrhagic complications of thrombolytic stroke therapy. Targeting the proteolytic imbalance within the neurovascular unit may provide new approaches for improving the safety and efficacy of thrombolytic reperfusion therapy for stroke.


Asunto(s)
Circulación Cerebrovascular/fisiología , Espacio Extracelular/enzimología , Neovascularización Patológica/fisiopatología , Péptido Hidrolasas/fisiología , Accidente Cerebrovascular/fisiopatología , Animales , Barrera Hematoencefálica/fisiopatología , Humanos , Inflamación , Metaloendopeptidasas/fisiología , Modelos Biológicos , Activador de Tejido Plasminógeno/fisiología
18.
Stroke ; 34(11): 2704-9, 2003 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-14551401

RESUMEN

BACKGROUND AND PURPOSE: The emerging concept of the neurovascular unit in stroke reemphasizes the need to focus on endothelial responses in brain. In this study we examined the role of mitogen-activated protein (MAP) kinase signaling in the regulation of hypoxic cell death in cerebral endothelial cells. METHODS: Human cerebral microvascular endothelial cells were exposed to 4 to 12 hours of hypoxia followed by 12 to 24 hours of reoxygenation. Cytotoxicity was measured by quantifying lactate dehydrogenase release. DNA laddering and caspase-3 activity were assessed to document a role for caspase-dependent cell death. zVAD-fmk and zDEVD-fmk were used to inhibit caspases. Activation of extracellular signal-regulated kinase (ERK), p38, and c-Jun N-terminal kinase (JNK) was assessed with Western blotting and kinase activity assays. U0126, SB203580, and SP600125 were used to interrupt the ERK, p38, and JNK pathways, respectively. RESULTS: Endothelial cell death occurred primarily during reoxygenation. DNA laddering and caspase activation were observed, and cytotoxicity was ameliorated by caspase inhibitors (20 micromol/L of zVAD-fmk or zDEVD-fmk). Among the 3 major MAP kinases, only p38 was transiently activated during reoxygenation, and inhibition with 10 micromol/L of SB203580 significantly reduced cytotoxicity. No effects were observed with other MAP kinase inhibitors. Cytoprotection with SB203580 was not accompanied by caspase downregulation. In contrast, cytoprotection with zVAD-fmk was associated with a decrease in p38 activation. Furthermore, cleavage of MEKK1 (an upstream kinase of p38) was significantly reduced by zVAD-fmk. CONCLUSIONS: Cerebral endothelial cell death after hypoxia-reoxygenation is mediated by interactions between caspases and p38 MAP kinase. Surprisingly, p38 pathways lie downstream of caspase mechanisms in this model system.


Asunto(s)
Encéfalo/irrigación sanguínea , Caspasas/metabolismo , Endotelio Vascular/metabolismo , Hipoxia-Isquemia Encefálica/metabolismo , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Encéfalo/citología , Caspasa 3 , Inhibidores de Caspasas , Muerte Celular/efectos de los fármacos , Muerte Celular/fisiología , Hipoxia de la Célula/fisiología , Línea Celular , Endotelio Vascular/citología , Endotelio Vascular/efectos de los fármacos , Inhibidores Enzimáticos/farmacología , Humanos , Proteínas Quinasas JNK Activadas por Mitógenos , Proteínas Quinasas Activadas por Mitógenos/antagonistas & inhibidores , Transducción de Señal/efectos de los fármacos , Transducción de Señal/fisiología , Proteínas Quinasas p38 Activadas por Mitógenos
19.
Nat Med ; 9(10): 1313-7, 2003 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-12960961

RESUMEN

Although thrombolysis with tissue plasminogen activator (tPA) is a stroke therapy approved by the US Food and Drug Administration, its efficacy may be limited by neurotoxic side effects. Recently, proteolytic damage involving matrix metalloproteinases (MMPs) have been implicated. In experimental embolic stroke models, MMP inhibitors decreased cerebral hemorrhage and injury after treatment with tPA. MMPs comprise a family of zinc endopeptidases that can modify several components of the extracellular matrix. In particular, the gelatinases MMP-2 and MMP-9 can degrade neurovascular matrix integrity. MMP-9 promotes neuronal death by disrupting cell-matrix interactions, and MMP-9 knockout mice have reduced blood-brain barrier leakage and infarction after cerebral ischemia. Hence it is possible that tPA upregulates MMPs in the brain, and that subsequent matrix degradation causes brain injury. Here we show that tPA upregulates MMP-9 in cell culture and in vivo. MMP-9 levels were lower in tPA knockouts compared with wild-type mice after focal cerebral ischemia. In human cerebral microvascular endothelial cells, MMP-9 was upregulated when recombinant tPA was added. RNA interference (RNAi) suggested that this response was mediated by the low-density lipoprotein receptor-related protein (LRP), which avidly binds tPA and possesses signaling properties. Targeting the tPA-LRP signaling pathway in brain may offer new approaches for decreasing neurotoxicity and improving stroke therapy.


Asunto(s)
Metaloproteinasa 2 de la Matriz/metabolismo , Metaloproteinasa 9 de la Matriz/metabolismo , Receptores de Lipoproteína/metabolismo , Activador de Tejido Plasminógeno/metabolismo , Animales , Isquemia Encefálica/metabolismo , Células Cultivadas , Endotelio Vascular/citología , Endotelio Vascular/metabolismo , Inducción Enzimática , Fibroblastos/citología , Fibroblastos/fisiología , Humanos , Metaloproteinasa 2 de la Matriz/genética , Metaloproteinasa 9 de la Matriz/genética , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Interferencia de ARN , Receptores de Lipoproteína/genética , Activador de Tejido Plasminógeno/efectos adversos , Activador de Tejido Plasminógeno/uso terapéutico
20.
Glia ; 43(3): 254-64, 2003 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-12898704

RESUMEN

Matrix metalloproteinases (MMPs) contribute to the pathophysiology of brain injury and inflammation but little is known about their regulatory signaling pathways in brain cells. Here we examine the role of mitogen-activated protein (MAP) kinase pathways in MMP-9 regulation in cortical rat astrocytes. The protein kinase C (PKC) activator phorbol 12-myristate 13-acetate (PMA) induced MMP-9 but not MMP-2 secretion as measured by gelatin zymography. Northern blot and RT-PCR analysis showed that MMP-9 responses occurred at the mRNA level. Although PMA increased phosphorylation in all three major MAP kinase pathways (ERK, p38 MAP kinase, and JNK), only inhibition of the ERK pathway by the MEK/ERK inhibitor U0126 (0.1-10 microM) significantly reduced MMP-9 upregulation, even when treatment was delayed for 4 h after PMA exposure. Inhibitors of p38 MAP kinase (SB203580) and JNK (SP600125) had no effect. This PKC pathway was compared to a cytokine response by exposing astrocytes to TNFalpha, which also activated MAP kinase and induced MMP-9 upregulation. But in this case, all three MAP kinase inhibitors (U0126, SB203580, and SP600125) reduced TNFalpha-induced MMP-9 upregulation. Taken together, these results suggest that the ERK MAP kinase is essential for MMP-9 upregulation via PKC and cytokine pathways in astrocytes.


Asunto(s)
Astrocitos/enzimología , Corteza Cerebral/enzimología , Metaloproteinasa 9 de la Matriz/metabolismo , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Animales , Animales Recién Nacidos , Astrocitos/citología , Astrocitos/efectos de los fármacos , Biomarcadores , Células Cultivadas , Corteza Cerebral/citología , Inhibidores Enzimáticos/farmacología , Inmunohistoquímica , Proteínas Quinasas JNK Activadas por Mitógenos , Metaloproteinasa 9 de la Matriz/genética , Proteínas Quinasas Activadas por Mitógenos/efectos de los fármacos , Proteína Quinasa C/efectos de los fármacos , Proteína Quinasa C/metabolismo , ARN Mensajero/efectos de los fármacos , ARN Mensajero/metabolismo , Ratas , Ratas Sprague-Dawley , Acetato de Tetradecanoilforbol/farmacología , Factor de Necrosis Tumoral alfa/metabolismo , Factor de Necrosis Tumoral alfa/farmacología , Regulación hacia Arriba/efectos de los fármacos , Regulación hacia Arriba/fisiología , Proteínas Quinasas p38 Activadas por Mitógenos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...