Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
1.
J Neurosci ; 32(39): 13587-96, 2012 Sep 26.
Artículo en Inglés | MEDLINE | ID: mdl-23015448

RESUMEN

The bcl-x gene appears to play a critical role in regulating apoptosis in the developing and mature CNS and following CNS injury. Two isoforms of Bcl-x are produced as a result of alternative pre-mRNA splicing: Bcl-x(L) (the long form) is anti-apoptotic, while Bcl-x(S) (short form) is pro-apoptotic. Despite the antagonistic activities of these two isoforms, little is known about how regulation of alternative splicing of bcl-x may mediate neural cell apoptosis. Here, we report that apoptotic stimuli (staurosporine or C2-ceramide) reciprocally altered Bcl-x splicing in neural cells, decreasing Bcl-x(L) while increasing Bcl-x(S). Specific knockdown of Bcl-x(S) attenuated apoptosis. To further define regulatory elements that influenced Bcl-x splicing, a Bcl-x minigene was constructed. Deletional analysis revealed several consensus sequences within intron 2 that altered splicing. We found that the splicing factor, CUG-binding-protein-1 (CUGBP1), bound to a consensus sequence close to the Bcl-x(L) 5' splice site, altering the Bcl-x(L)/Bcl-x(S) ratio and influencing cell death. In vivo, neonatal hypoxia-ischemia reciprocally altered Bcl-x pre-mRNA splicing, similar to the in vitro studies. Manipulation of the splice isoforms using viral gene transfer of Bcl-x(S) shRNA into the hippocampus of rats before neonatal hypoxia-ischemia decreased vulnerability to injury. Moreover, alterations in nuclear CUGBP1 preceded Bcl-x splicing changes. These results suggest that alternative pre-mRNA splicing may be an important regulatory mechanism for cell death after acute neurological injury and may potentially provide novel targets for intervention.


Asunto(s)
Empalme Alternativo/genética , Lesiones Encefálicas/etiología , Hipoxia-Isquemia Encefálica/complicaciones , Precursores del ARN/metabolismo , Proteína bcl-X/metabolismo , Análisis de Varianza , Animales , Animales Recién Nacidos , Apoptosis/efectos de los fármacos , Apoptosis/genética , Proteínas CELF1 , Células Cultivadas , Corteza Cerebral/citología , Relación Dosis-Respuesta a Droga , Embrión de Mamíferos , Inhibidores Enzimáticos/farmacología , Femenino , Lateralidad Funcional , Regulación de la Expresión Génica/efectos de los fármacos , Regulación de la Expresión Génica/genética , Proteínas Fluorescentes Verdes/genética , Humanos , Etiquetado Corte-Fin in Situ , L-Lactato Deshidrogenasa/metabolismo , Masculino , Ratones , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Embarazo , Precursores del ARN/genética , ARN Interferente Pequeño/metabolismo , Proteínas de Unión al ARN/metabolismo , Ratas , Esfingosina/análogos & derivados , Esfingosina/farmacología , Estaurosporina/farmacología , Factores de Tiempo , Transfección/métodos , Proteína bcl-X/genética
2.
J Neurosci ; 30(28): 9621-30, 2010 Jul 14.
Artículo en Inglés | MEDLINE | ID: mdl-20631191

RESUMEN

Glucocorticoids are commonly used in treating diseases with white matter lesions, including demyelinating diseases and spinal cord injury (SCI). However, glucocorticoids are ineffective in gray matter injuries, such as head injury and stroke. The differential glucocorticoid effects in white and gray matter injuries are unclear. We report here a novel mechanism of methylprednisolone (MP), a synthetic glucocorticoid widely used for treating multiple sclerosis and SCI, in protecting oligodendrocytes (OLGs) against AMPA-induced excitotoxicity, which has been implicated in the white matter injuries and diseases. The cytoprotective action of MP in OLGs is causally related to its upregulation of a neuroprotective cytokine erythropoietin (Epo). MP transactivation of Epo expression involves dual transcription factors: glucocorticoid receptor (GR) and hypoxia-inducible factor-1alpha (HIF-1alpha). Coimmunoprecipitation, chromatin immunoprecipitation analysis, yeast two-hybrid analysis, and structure modeling of three-dimensional protein-protein interactions confirm that MP induces interaction between GR DNA binding domain and HIF-1alpha PAS domain, with subsequent recruitment of HIF-1beta to transactivate Epo expression in OLGs. In contrast, MP activates GR but does not induce GR-HIF-1alpha interaction, HIF-1alpha binding to Epo enhancer/promoter, or Epo expression in cultured cortical neurons. The OLG-specific GR-HIF-1alpha transactivation of Epo provides novel insights into the development of more effective therapies for diseases affecting the white matter.


Asunto(s)
Muerte Celular/efectos de los fármacos , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Metilprednisolona/farmacología , Oligodendroglía/efectos de los fármacos , Ácido alfa-Amino-3-hidroxi-5-metil-4-isoxazol Propiónico/toxicidad , Análisis de Varianza , Animales , Células Cultivadas , Inmunoprecipitación de Cromatina , Simulación por Computador , Eritropoyetina/genética , Eritropoyetina/metabolismo , Subunidad alfa del Factor 1 Inducible por Hipoxia/genética , Inmunoprecipitación , Oligodendroglía/metabolismo , Ratas , Ratas Sprague-Dawley , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
3.
J Neurosci ; 29(7): 2022-6, 2009 Feb 18.
Artículo en Inglés | MEDLINE | ID: mdl-19228956

RESUMEN

Methylprednisolone (MP), a synthetic glucocorticoid agonist, is widely used for the clinical therapy of white matter diseases in the nervous system, such as spinal cord injury and multiple sclerosis. In addition to its potent anti-inflammatory and antioxidant properties, we recently discovered a selective antiapoptotic effect of MP on oligodendrocytes via the activation of the glucocorticoid receptor (GR) and the upregulation of bcl-X(L), a splicing isoform of the bcl-x gene. Based on published findings of the functional interactions between GR and STAT5, a transcription factor from the family of signal transducers and activators of transcription (STAT), we examined whether the glucocorticoid signaling pathway interacts with STAT5 to upregulate bcl-X(L) and protect oligodendrocytes. We show herein that (1) the GR and STAT5 complex is present on the STAT5-binding site of the bcl-x promoter region in oligodendrocytes; (2) the overexpression of an activated form of STAT5 prevents alpha-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid-induced oligodendrocyte cell death; and (3) this prevention is lost when the STAT5 gene is knocked down. Thus, our results provide one molecular mechanism underlying the postinjury protective effects of oligodendrocytes by stress hormones.


Asunto(s)
Apoptosis/efectos de los fármacos , Metilprednisolona/farmacología , Oligodendroglía/efectos de los fármacos , Oligodendroglía/metabolismo , Factor de Transcripción STAT5/efectos de los fármacos , Factor de Transcripción STAT5/metabolismo , Animales , Animales Recién Nacidos , Apoptosis/genética , Sitios de Unión/efectos de los fármacos , Sitios de Unión/genética , Células Cultivadas , Citoprotección/efectos de los fármacos , Citoprotección/genética , Regulación hacia Abajo/genética , Esclerosis Múltiple/tratamiento farmacológico , Esclerosis Múltiple/genética , Esclerosis Múltiple/metabolismo , Fármacos Neuroprotectores/farmacología , Regiones Promotoras Genéticas/efectos de los fármacos , Regiones Promotoras Genéticas/genética , Interferencia de ARN , Ratas , Receptores de Glucocorticoides/efectos de los fármacos , Receptores de Glucocorticoides/metabolismo , Factor de Transcripción STAT5/genética , Traumatismos de la Médula Espinal/tratamiento farmacológico , Traumatismos de la Médula Espinal/genética , Traumatismos de la Médula Espinal/metabolismo , Regulación hacia Arriba/efectos de los fármacos , Regulación hacia Arriba/fisiología , Proteína bcl-X/efectos de los fármacos , Proteína bcl-X/metabolismo
4.
Biochim Biophys Acta ; 1793(5): 764-71, 2009 May.
Artículo en Inglés | MEDLINE | ID: mdl-19339209

RESUMEN

Dexamethasone (DX) induces apoptosis resistance in most solid malignant tumors during co-treatment with chemotherapy agents, such as camptothecin (CAM). In this study, we investigated the mechanism by which DX reduces chemotherapy efficiency in C6-glioma. DX reduced CAM-increased DNA fragmentation and caspase-3 activation. The DX's protection was negated by RU486, an antagonist of glucocorticoid receptor (GR). DX itself increased anti-apoptotic gene, Bcl-xL expression, and its transcription factor, signaling transducer and activator of transcription 5 (Stat5), DNA binding activity and phospho-Stat5 expression. DX blocked the CAM-decreased Bcl-xL and phospho-Stat5 expression, and Stat5 binding activity. RU486 negated DX's actions. To determine whether Stat5 regulates Bcl-xL expression in CAM-induced cell death, C6-glioma was infected with an adenovirus containing a constitutively activated Stat5-GFP (Ad-Stat5ca). Overexpression of Stat5ca increased Bcl-xL and decreased CAM-induced cell death compared to control adenovirus infected cells; whereas Stat5 siRNA decreased DX-induced Bcl-xL and increased cell death. Phospho-Stat5 expression was observed in the nuclear extract by co-immunoprecipitation with an anti-GR antibody, indicating that Stat5 and GR were interactive and formed a complex in the nuclei. These results suggest that DX's prevention from CAM-induced apoptosis and RU486's antagonism of DX's protection may be through Stat5/Bcl-xL signal pathway regulated by a GR.


Asunto(s)
Antineoplásicos/farmacología , Apoptosis/efectos de los fármacos , Camptotecina/farmacología , Línea Celular Tumoral/efectos de los fármacos , Dexametasona/farmacología , Factor de Transcripción STAT5/metabolismo , Proteína bcl-X/metabolismo , Animales , Fragmentación del ADN , Glioma/metabolismo , Interferencia de ARN , Ratas , Receptores de Glucocorticoides/metabolismo , Factor de Transcripción STAT5/genética , Proteína bcl-X/genética
5.
J Neurosci ; 28(12): 3141-9, 2008 Mar 19.
Artículo en Inglés | MEDLINE | ID: mdl-18354017

RESUMEN

Methylprednisolone (MP) is used to treat a variety of neurological disorders involving white matter injury, including multiple sclerosis, acute disseminated encephalomyelitis, and spinal cord injury (SCI). Although its mechanism of action has been attributed to anti-inflammatory or antioxidant properties, we examined the possibility that MP may have direct neuroprotective activities. Neurons and oligodendrocytes treated with AMPA or staurosporine died within 24 h after treatment. MP attenuated oligodendrocyte death in a dose-dependent manner; however, neurons were not rescued by the same doses of MP. This protective effect was reversed by the glucocorticoid receptor (GR) antagonist (11, 17)-11-[4-(dimethylamino)phenyl]-17-hydroxy-17-(1-propynyl)estra-4,9-dien-3-one (RU486) and small interfering RNA directed against GR, suggesting a receptor-dependent mechanism. MP reversed AMPA-induced decreases in the expression of anti-apoptotic Bcl-x(L), caspase-3 activation, and DNA laddering, suggesting anti-apoptotic activity in oligodendrocytes. To examine whether MP demonstrated this selective protection in vivo, neuronal and oligodendrocyte survival was assessed in rats subjected to spinal cord injury (SCI); groups of rats were treated with or without MP in the presence or absence of RU486. Eight days after SCI, MP significantly increased oligodendrocytes (CC-1-immunoreactive cells) after SCI, but neuronal (neuronal-specific nuclear protein-immunoreactive cells) number remained unchanged; RU486 reversed this protective effect. MP also inhibited SCI-induced decreases in Bcl-x(L) and caspase-3 activation. Consistent with these findings, the volume of demyelination, assessed by Luxol fast blue staining, was attenuated by MP and reversed by RU486. These results suggest that MP selectively inhibits oligodendrocyte but not neuronal cell death via a receptor-mediated action and may be a mechanism for its limited protective effect after SCI.


Asunto(s)
Metilprednisolona/farmacología , Neuronas/efectos de los fármacos , Fármacos Neuroprotectores/farmacología , Oligodendroglía/efectos de los fármacos , Traumatismos de la Médula Espinal/patología , Análisis de Varianza , Animales , Benzotiadiazinas/farmacología , Células Cultivadas , Corteza Cerebral/citología , Fragmentación del ADN/efectos de los fármacos , Modelos Animales de Enfermedad , Relación Dosis-Respuesta a Droga , Embrión de Mamíferos , Inhibidores Enzimáticos/farmacología , Ensayo de Inmunoadsorción Enzimática/métodos , Femenino , L-Lactato Deshidrogenasa/metabolismo , Proteína Básica de Mielina/metabolismo , Fosfopiruvato Hidratasa/metabolismo , ARN Interferente Pequeño/farmacología , Ratas , Ratas Long-Evans
6.
J Cell Biol ; 164(1): 123-31, 2004 Jan 05.
Artículo en Inglés | MEDLINE | ID: mdl-14709545

RESUMEN

Amyloid-beta peptide (Abeta) accumulation in senile plaques, a pathological hallmark of Alzheimer's disease (AD), has been implicated in neuronal degeneration. We have recently demonstrated that Abeta induced oligodendrocyte (OLG) apoptosis, suggesting a role in white matter pathology in AD. Here, we explore the molecular mechanisms involved in Abeta-induced OLG death, examining the potential role of ceramide, a known apoptogenic mediator. Both Abeta and ceramide induced OLG death. In addition, Abeta activated neutral sphingomyelinase (nSMase), but not acidic sphingomyelinase, resulting in increased ceramide generation. Blocking ceramide degradation with N-oleoyl-ethanolamine exacerbated Abeta cytotoxicity; and addition of bacterial sphingomyelinase (mimicking cellular nSMase activity) induced OLG death. Furthermore, nSMase inhibition by 3-O-methyl-sphingomyelin or by gene knockdown using antisense oligonucleotides attenuated Abeta-induced OLG death. Glutathione (GSH) precursors inhibited Abeta activation of nSMase and prevented OLG death, whereas GSH depletors increased nSMase activity and Abeta-induced death. These results suggest that Abeta induces OLG death by activating the nSMase-ceramide cascade via an oxidative mechanism.


Asunto(s)
Péptidos beta-Amiloides/metabolismo , Ceramidas/metabolismo , Oligodendroglía/metabolismo , Transducción de Señal/fisiología , Esfingomielina Fosfodiesterasa/metabolismo , Enfermedad de Alzheimer/metabolismo , Enfermedad de Alzheimer/fisiopatología , Péptidos beta-Amiloides/farmacología , Animales , Muerte Celular/efectos de los fármacos , Muerte Celular/fisiología , Células Cultivadas , Ceramidas/farmacología , Endocannabinoides , Etanolaminas/farmacología , Glutatión/metabolismo , Degeneración Nerviosa/metabolismo , Degeneración Nerviosa/fisiopatología , Ácidos Oléicos , Oligodendroglía/citología , Oligodendroglía/efectos de los fármacos , Estrés Oxidativo/efectos de los fármacos , Estrés Oxidativo/fisiología , Ratas , Esferoides Celulares/citología , Esfingomielinas/farmacología , Regulación hacia Arriba/efectos de los fármacos , Regulación hacia Arriba/fisiología
7.
J Neurosci ; 26(8): 2290-9, 2006 Feb 22.
Artículo en Inglés | MEDLINE | ID: mdl-16495456

RESUMEN

Amyloid-beta peptide (Abeta)-induced death in cerebral endothelial cells (CECs) is preceded by mitochondrial dysfunction and signaling events characteristic of apoptosis. Mitochondria-dependent apoptosis engages Bcl-2 family proteins, especially the BH3-only homologues, which play a key role in initiating the apoptotic cascade. Here, we report that the expression of bim, but not other BH3-only members, was selectively increased in cerebral microvessels isolated from 18-month-old APPsw (Tg2576) mice, a model of cerebral amyloid angiopathy (CAA), suggesting a pivotal role for Bim in Abeta-induced cerebrovascular degeneration in vivo. A similar expression profile was observed in Abeta-treated CECs. Furthermore, Abeta induction of bim expression involved a pro-apoptotic transcription factor, FKHRL1. FKHRL1 bound to a consensus sequence in the bim promoter region and was activated by Abeta before bim expression. FKHRL1 activity was negatively regulated by phosphorylation catalyzed by Akt, an anti-apoptotic kinase. Akt upregulation by adenoviral gene transfer inhibited Abeta-induced FKHRL1 activation and bim induction. In addition, Abeta increased the activity of protein phosphatase 2A (PP2A), a ceramide-activated protein phosphatase. Suppression of PP2A activity by RNA interference or a specific inhibitor, okadaic acid, effectively suppressed Abeta-induced Akt inactivation and FKHRL1 activation, leading to an attenuation of bim expression and cell death in CECs. Coimmunoprecipitation experiments revealed that Abeta enhanced the binding of the PP2A regulatory subunit PP2ACalphabeta to Akt. These results implicate PP2A as an early regulator of Abeta-induced bim expression and CEC apoptosis via the Akt/FKHRL1 signaling pathway. We raise the possibility that this pathway may play a role in cerebrovascular degeneration in CAA.


Asunto(s)
Péptidos beta-Amiloides/administración & dosificación , Circulación Cerebrovascular/fisiología , Células Endoteliales/fisiología , Factores de Transcripción Forkhead/metabolismo , Fragmentos de Péptidos/administración & dosificación , Fosfoproteínas Fosfatasas/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Animales , Apoptosis/efectos de los fármacos , Apoptosis/fisiología , Células Cultivadas , Circulación Cerebrovascular/efectos de los fármacos , Células Endoteliales/efectos de los fármacos , Proteína Forkhead Box O3 , Regulación de la Expresión Génica/efectos de los fármacos , Regulación de la Expresión Génica/fisiología , Ratones , Proteína Fosfatasa 2 , Transducción de Señal/efectos de los fármacos , Transducción de Señal/fisiología
8.
J Neurosci ; 26(43): 10939-48, 2006 Oct 25.
Artículo en Inglés | MEDLINE | ID: mdl-17065436

RESUMEN

It has been postulated that the development of amyloid plaques in Alzheimer's disease (AD) may result from an imbalance between the generation and clearance of the amyloid-beta peptide (Abeta). Although familial AD appears to be caused by Abeta overproduction, sporadic AD (the most prevalent form) may result from impairment in clearance. Recent evidence suggests that several proteases may contribute to the degradation of Abeta. Furthermore, astrocytes have recently been implicated as a potential cellular mediator of Abeta degradation. In this study, we examined the possibility that matrix metalloproteinases (MMPs), proteases known to be expressed and secreted by astrocytes, could play a role in extracellular Abeta degradation. We found that astrocytes surrounding amyloid plaques showed enhanced expression of MMP-2 and MMP-9 in aged amyloid precursor protein (APP)/presenilin 1 mice. Moreover, astrocyte-conditioned medium (ACM) degraded Abeta, lowering levels and producing several fragments after incubation with synthetic human Abeta(1-40) and Abeta(1-42). This activity was attenuated with specific inhibitors of MMP-2 and -9, as well as in ACM derived from mmp-2 or -9 knock-out (KO) mice. In vivo, significant increases in the steady-state levels of Abeta were found in the brains of mmp-2 and -9 KO mice compared with wild-type controls. Furthermore, pharmacological inhibition of the MMPs with N-[(2R)-2-(hydroxamidocarbonylmethyl)-4-methylpentanoyl]-L-tryptophan methylamide (GM 6001) increased brain interstitial fluid Abeta levels and elimination of half-life in APPsw mice. These results suggest that MMP-2 and -9 may contribute to extracellular brain Abeta clearance by promoting Abeta catabolism.


Asunto(s)
Péptidos beta-Amiloides/fisiología , Astrocitos/enzimología , Líquido Extracelular/enzimología , Regulación Enzimológica de la Expresión Génica/fisiología , Metaloproteinasa 2 de la Matriz/biosíntesis , Metaloproteinasa 2 de la Matriz/genética , Metaloproteinasa 9 de la Matriz/biosíntesis , Metaloproteinasa 9 de la Matriz/genética , Péptidos beta-Amiloides/genética , Animales , Células Cultivadas , Líquido Extracelular/metabolismo , Células HeLa , Humanos , Metaloproteinasa 2 de la Matriz/deficiencia , Metaloproteinasa 2 de la Matriz/fisiología , Metaloproteinasa 9 de la Matriz/deficiencia , Metaloproteinasa 9 de la Matriz/fisiología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Transporte de Proteínas/genética
9.
Biochem J ; 388(Pt 3): 913-20, 2005 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-15702971

RESUMEN

VEGI (vascular endothelial growth inhibitor), a member of the tumour necrosis factor superfamily, has been reported to inhibit endothelial cell proliferation, angiogenesis and tumour growth. We identified and cloned approx. 2.2 kb of the VEGI promoter from mouse cerebral endothelial cells. The promoter contained an atypical TATA-box-binding protein sequence TAAAAAA residing at -32/-26 relative to the transcription initiation site (+1), 83 bp upstream from the ATG start codon. To investigate critical sequences in the VEGI promoter, a series of deleted and truncated segments were constructed from a 2300 bp promoter construct (-2201/+96) linked to a luciferase reporter gene. Transient transfection of cerebral microvascular cells (bEND.3) and rat C6 glioma cells demonstrated that a 1700 bp deletion from the -2201 to -501 did not significantly affect promoter activity; however, a truncated construct (-501/+96) lacking the region between -312 and -57 resulted in nearly 90% loss of promoter activity. A consensus NF-kappaB (nuclear factor kappaB) and several SP1 (specificity protein-1)-binding sequences were identified within the deleted segment. Supershift analysis revealed that NF-kappaB subunits, p50 and p65, interacted with the VEGI promoter. Exposure of cerebral endothermic cells to the pro-inflammatory cytokine, tumour necrosis factor-alpha, increased VEGI mRNA levels and DNA-binding activities, whereas an NF-kappaB inhibitor attenuated this increase. In addition, p65 overexpression enhanced, whereas p50 overexpression decreased, the luciferase activity. Furthermore, mutation of the NF-kappaB DNA binding site blocked this p65- and tumour necrosis factor-alpha-induced luciferase activity. These findings suggest that the transcription factor NF-kappaB plays an important role in the regulation of VEGI expression.


Asunto(s)
Regulación de la Expresión Génica/genética , Proteínas de la Membrana/genética , Regiones Promotoras Genéticas/genética , Animales , Secuencia de Bases , Células Cultivadas , Ensayo de Cambio de Movilidad Electroforética , Células Endoteliales/efectos de los fármacos , Células Endoteliales/metabolismo , Exones/genética , Regulación de la Expresión Génica/efectos de los fármacos , Intrones/genética , Ratones , Datos de Secuencia Molecular , Mutación/genética , FN-kappa B/metabolismo , Ratas , Elementos de Respuesta/genética , Análisis de Secuencia de ADN , Telencéfalo/citología , Factores de Transcripción/metabolismo , Factor de Necrosis Tumoral alfa/farmacología
10.
J Cereb Blood Flow Metab ; 25(11): 1445-55, 2005 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-15973355

RESUMEN

The amyloid-beta peptide (Abeta) induces apoptosis in cerebrovascular endothelial cells (CECs), contributing to the pathogenesis of cerebral amyloid angiopathy. We have previously shown that Abeta induces apoptosis in CECs. In the present study, we report that Abeta25-35-induced CEC apoptosis involves the inactivation of Akt, a signaling kinase important in maintaining cell viability. Akt prevents the activation of death-signaling events by facilitating the inactivation of proapoptotic proteins such as Bad. We applied three strategies to show that Abeta25-35 inactivation of Akt is causally related to Abeta25-35-induced CEC death by preventing Bad activation and subsequent mitochondrial dysfunction (reflected by the release of endonuclease G and Smac, two proapoptotic intermembranous proteins of the mitochondria). Wortmannin, a PI3-kinase inhibitor, enhanced Abeta25-35-induced Bad activation, mitochondrial dysfunction and CEC death. Enhancement of Akt activity by a Tat-Akt fusion protein, or by viral gene transfer of a constitutively active mutant of akt, reduced Bad activation, mitochondrial dysfunction, and CEC death. Using a siRNA strategy to knock down the bad gene, we showed that Bad activation is causally related to Abeta25-35-induced mitochondrial dysfunction and CEC death. Together, these results establish that the Akt-Bad cascade is altered by Abeta25-35, resulting in CEC apoptosis.


Asunto(s)
Péptidos beta-Amiloides/farmacología , Apoptosis/efectos de los fármacos , Encéfalo/metabolismo , Circulación Cerebrovascular , Células Endoteliales/metabolismo , Fragmentos de Péptidos/farmacología , Transducción de Señal/efectos de los fármacos , Androstadienos/farmacología , Animales , Proteínas Reguladoras de la Apoptosis , Encéfalo/irrigación sanguínea , Encéfalo/patología , Células Cultivadas , Angiopatía Amiloide Cerebral/metabolismo , Angiopatía Amiloide Cerebral/patología , Endodesoxirribonucleasas/metabolismo , Células Endoteliales/patología , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Ratones , Mitocondrias/metabolismo , Mitocondrias/patología , Proteínas Mitocondriales/metabolismo , Fosfatidilinositol 3-Quinasas/metabolismo , Inhibidores de las Quinasa Fosfoinosítidos-3 , Inhibidores de Proteínas Quinasas/farmacología , Transporte de Proteínas/efectos de los fármacos , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/metabolismo , ARN Interferente Pequeño/farmacología , Wortmanina
11.
Ann N Y Acad Sci ; 1042: 439-47, 2005 May.
Artículo en Inglés | MEDLINE | ID: mdl-15965090

RESUMEN

We test whether inhibition of inducible nitric oxide synthase (iNOS) can exert a cytoprotective effect on cerebral endothelial cells upon stimulation by pro-inflammatory cytokines. Mouse brain endothelial cells were stably transfected to express an antisense RNA against iNOS driven by an endothelium-specific von Willebrand factor (vWF) promoter. Upon stimulation with tumor necrosis factor-alpha (TNF-alpha) plus interferon-gamma (IFN-gamma), antisense transfectants showed less iNOS enzymatic activity with less nitric oxide (NO) when compared to the sense control cells. Correspondingly, the antisense cells showed a reduced LDH release and less cytosolic content of oligonucleosomes. These findings establish a cell-specific antisense strategy and confirm the cytotoxic role of iNOS expression in cultured cerebral endothelial cells.


Asunto(s)
Encéfalo/citología , Encéfalo/metabolismo , Citocinas/farmacología , Células Endoteliales/citología , Células Endoteliales/metabolismo , Óxido Nítrico Sintasa de Tipo II/metabolismo , ARN sin Sentido/genética , Animales , Encéfalo/efectos de los fármacos , Muerte Celular/efectos de los fármacos , Línea Celular , Células Endoteliales/efectos de los fármacos , Ratones , Óxido Nítrico Sintasa de Tipo II/genética
12.
Ann N Y Acad Sci ; 1042: 357-64, 2005 May.
Artículo en Inglés | MEDLINE | ID: mdl-15965081

RESUMEN

Ceramide is a pro-apoptotic lipid messenger that induces oxidative stress and may mediate apoptosis in cerebral endothelial cells (CECs) induced by TNF-alpha/cycloheximide, lipopolysaccharide, oxidized LDL, IL-1, and amyloid peptide. Exposure of CECs to C2 ceramide for 12 h caused cell death in a concentration-dependent manner, with a LC50 of 30 microM. Statins are inhibitors of 3-hydroxyl-3-methyl coenzyme A reductase which is the rate-limiting enzyme for cholesterol biosynthesis. Pretreatment with pravastatin at 20 microM for 16 h substantially attenuated ceramide cytotoxicity in mouse CECs. Increases in vascular endothelial growth factor (VEGF) expression were detected within 1-3 h after pravastatin treatment. This pravastatin action was accompanied by the activation of hypoxia-inducible factor-1 (HIF-1), a transcription factor known to activate VEGF expression. These results raise the possibility that pravastatin may protect CECs against ceramide-induced death via the HIF-VEGF cascade.


Asunto(s)
Encéfalo/efectos de los fármacos , Ceramidas/toxicidad , Células Endoteliales/efectos de los fármacos , Células Endoteliales/metabolismo , Factor 1 Inducible por Hipoxia/metabolismo , Pravastatina/farmacología , Factor A de Crecimiento Endotelial Vascular/metabolismo , Animales , Encéfalo/metabolismo , Supervivencia Celular/efectos de los fármacos , Células Cultivadas , ADN/metabolismo , Ratones , ARN Mensajero/genética , Regulación hacia Arriba/efectos de los fármacos , Factor A de Crecimiento Endotelial Vascular/genética
13.
Stroke ; 33(10): 2471-7, 2002 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-12364740

RESUMEN

BACKGROUND AND PURPOSE: Cells lacking the ATM (ataxia telangectasia mutated) gene are hypersensitive to DNA damage caused by a variety of insults. ATM may regulate oxidative stress-induced signaling cascades involving nuclear factor-kappaB (NF-kappaB), a transcription factor that is upstream of a wide variety of stress-responsive genes. We investigated the potential interaction of ATM and NF-kappaB after oxygen-glucose deprivation (OGD) in cerebral endothelial cells (CECs). METHODS: Primary cultures of mouse CECs were subjected to OGD in the absence or presence of ATM antisense oligonucleotides or the NF-kappaB inhibitor SN50. ATM expression was determined with the use of reverse transcription-polymerase chain reaction and Western blot, and NF-kappaB activity was assessed by electrophoretic mobility shift assay. Cells were assessed for mitochondrial DNA damage with the use of long polymerase chain reaction and were assessed for caspase-3 and caspase-8 activity with the use of fluorogenic substrates. Cell death was determined by 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyl-tetrazolium bromide and LDH release. RESULTS: OGD stimulated ATM gene expression at the mRNA and protein level in CECs as early as 1 hour after OGD initiation. ATM gene knockdown with the use of an antisense oligonucleotide suppressed OGD-induced ATM protein expression, which was accompanied by an attenuation of NF-kappaB activation and the subsequent expression of downstream genes, including the antiapoptotic gene c-IAP2. ATM knockdown also accentuated OGD-induced mitochondrial DNA damage and the activation of caspase-3 and caspase-8, leading to enhanced CEC death. The specific NF-kappaB inhibitor SN50 mimicked the effects of ATM knockdown. CONCLUSIONS: We conclude that ATM may play a cytoprotective role in OGD-induced CEC death via a NF-kappaB-dependent signaling pathway.


Asunto(s)
Encéfalo/irrigación sanguínea , Endotelio Vascular/metabolismo , Glucosa/metabolismo , Hipoxia Encefálica/metabolismo , FN-kappa B/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Animales , Apoptosis/fisiología , Proteínas de la Ataxia Telangiectasia Mutada , Caspasas/metabolismo , Proteínas de Ciclo Celular , Hipoxia de la Célula , Células Cultivadas , Citoprotección/efectos de los fármacos , Citoprotección/fisiología , ADN/metabolismo , Daño del ADN/efectos de los fármacos , ADN Mitocondrial/metabolismo , Proteínas de Unión al ADN , Endotelio Vascular/citología , Endotelio Vascular/efectos de los fármacos , Activación Enzimática/efectos de los fármacos , Glucosa/deficiencia , Ratones , FN-kappa B/antagonistas & inhibidores , Oligonucleótidos Antisentido/farmacología , Péptidos/farmacología , Proteínas Serina-Treonina Quinasas/antagonistas & inhibidores , Proteínas Serina-Treonina Quinasas/genética , Proteínas/metabolismo , ARN Mensajero/metabolismo , Transducción de Señal/efectos de los fármacos , Transducción de Señal/fisiología , Proteínas Supresoras de Tumor , Regulación hacia Arriba/efectos de los fármacos , Regulación hacia Arriba/fisiología
14.
Brain Res Bull ; 88(4): 354-8, 2012 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-22498308

RESUMEN

The deposition of ß-amyloid protein(Aß) and loss of neurons within the brain are the pathologic hallmarks of Alzheimer's disease (AD). Apoptosis is a crucial pathway in neuronal loss in AD. Tanshinone IIA (tanIIA) is one of ingredients of tanshinone which is the major component of the traditional Chinese herb Danshen. The present study explores the effects of tanIIA on Aß(1-42)-induced cytotoxicity. Cultured cortical neurons that were treated with 4 µM Aß(1-42) showed shrunken perikaryon with loss of neurite processes; the survival rate of neurons decreased almost to 57% and the apoptotic rate of neurons increased to 47%. In addition, the level of gene bcl-xl mRNA and Bcl-xL protein decreased significantly. These changes, however, were prevented by pretreatment of neurons with tanIIA for 24h before Aß(1-42), which markedly increased neuron survival rate compared to neurons treated with Aß(1-42) alone; the apoptotic rate of neurons decreased to 15%, and the decrease in level of gene bcl-xl mRNA and Bcl-xL protein in Aß-treated neurons, were prevented. Thus, we conclude that tanIIA might serve as an obvious neuroprotection. TanIIA protected neurons against the Aß-induced cytotoxicity most likely via activation of the Bcl-xL pathway.


Asunto(s)
Abietanos/farmacología , Apoptosis/efectos de los fármacos , Neuronas/efectos de los fármacos , Fármacos Neuroprotectores/farmacología , Proteína bcl-X/metabolismo , Péptidos beta-Amiloides/toxicidad , Animales , Apoptosis/fisiología , Western Blotting , Ratones , Neuronas/metabolismo , Fragmentos de Péptidos/toxicidad , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
15.
J Neurochem ; 97(3): 631-40, 2006 May.
Artículo en Inglés | MEDLINE | ID: mdl-16524368

RESUMEN

There is growing recognition that white matter pathology is a common feature in Alzheimer's disease. We have previously reported that the amyloid beta peptide (Abeta) induces apoptosis in oligodendrocytes (OLG), via activation of neutral sphingomyelinase (nSMase) and resultant generation of ceramide. In the current study, we report that both Abeta and ceramide increased expression of the proapoptotic protein DP5/Hrk (DP5), and release of cytochrome C from mitochondria to cytoplasm in OLGs. We provide evidence that the Jun N-terminal kinase (JNK) signaling pathway mediates Abeta- and ceramide-induced apoptosis: Both Abeta and ceramide activated JNK phosphorylation, and subsequent AP-1 DNA binding activity; JNK siRNA decreased AP-1 DNA binding, DP5 expression and reduced cell death. Furthermore, inhibition of nSMase attenuated Abeta-induced JNK phosphorylation, AP-1 DNA binding activity, DP5 expression, and cytochrome C release. Collectively, these results suggest that Abeta-induced apoptosis involves the sequential activation of nSMase with ceramide generation, JNK activation, AP-1 DNA binding, and DP5 expression.


Asunto(s)
Péptidos beta-Amiloides/farmacología , Proteínas Reguladoras de la Apoptosis/metabolismo , MAP Quinasa Quinasa 4/metabolismo , Neuropéptidos/metabolismo , Oligodendroglía/efectos de los fármacos , Esfingomielina Fosfodiesterasa/metabolismo , Animales , Proteínas Reguladoras de la Apoptosis/genética , Western Blotting/métodos , Encéfalo/citología , Muerte Celular/efectos de los fármacos , Fraccionamiento Celular/métodos , Núcleo Celular/efectos de los fármacos , Citosol/efectos de los fármacos , Embrión de Mamíferos , Activación Enzimática/efectos de los fármacos , Inhibidores Enzimáticos/farmacología , MAP Quinasa Quinasa 4/química , Neuropéptidos/genética , Fosforilación/efectos de los fármacos , ARN Mensajero/metabolismo , ARN Interferente Pequeño/farmacología , Ratas , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa/métodos , Esfingosina/análogos & derivados , Esfingosina/farmacología , Sales de Tetrazolio , Tiazoles , Factores de Tiempo , Factor de Transcripción AP-1/metabolismo
16.
J Biol Chem ; 281(34): 24566-74, 2006 Aug 25.
Artículo en Inglés | MEDLINE | ID: mdl-16787929

RESUMEN

The pathological hallmark of Alzheimer disease is the senile plaque principally composed of tightly aggregated amyloid-beta fibrils (fAbeta), which are thought to be resistant to degradation and clearance. In this study, we explored whether proteases capable of degrading soluble Abeta (sAbeta) could degrade fAbeta as well. We demonstrate that matrix metalloproteinase-9 (MMP-9) can degrade fAbeta and that this ability is not shared by other sAbeta-degrading enzymes examined, including endothelin-converting enzyme, insulin-degrading enzyme, and neprilysin. fAbeta was decreased in samples incubated with MMP-9 compared with other proteases, assessed using thioflavin-T. Furthermore, fAbeta breakdown with MMP-9 but not with other proteases was demonstrated by transmission electron microscopy. Proteolytic digests of purified fAbeta were analyzed with matrix-assisted laser desorption ionization time-of-flight mass spectrometry to identify sites of Abeta that are cleaved during its degradation. Only MMP-9 digests contained fragments (Abeta(1-20) and Abeta(1-30)) from fAbeta(1-42) substrate; the corresponding cleavage sites are thought to be important for beta-pleated sheet formation. To determine whether MMP-9 can degrade plaques formed in vivo, fresh brain slices from aged APP/PS1 mice were incubated with proteases. MMP-9 digestion resulted in a decrease in thioflavin-S (ThS) staining. Consistent with a role for endogenous MMP-9 in this process in vivo, MMP-9 immunoreactivity was detected in astrocytes surrounding amyloid plaques in the brains of aged APP/PS1 and APPsw mice, and increased MMP activity was selectively observed in compact ThS-positive plaques. These findings suggest that MMP-9 can degrade fAbeta and may contribute to ongoing clearance of plaques from amyloid-laden brains.


Asunto(s)
Péptidos beta-Amiloides/metabolismo , Metaloproteinasa 9 de la Matriz/metabolismo , Envejecimiento/metabolismo , Enfermedad de Alzheimer/metabolismo , Péptidos beta-Amiloides/química , Animales , Encéfalo/enzimología , Humanos , Hidrólisis , Inmunohistoquímica , Espectrometría de Masas , Ratones , Ratones Transgénicos , Placa Amiloide/metabolismo , Especificidad por Sustrato
17.
Neurobiol Dis ; 20(3): 881-9, 2005 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-16005241

RESUMEN

Growing evidence suggests that cells undergo apoptosis after spinal cord injury (SCI). However, little is known about the early events that trigger apoptosis in the contused cord. The BH3-only subfamily of pro-apoptotic regulators (e.g., bim, bad, and dp5) is recognized as initiators of the apoptotic cascade, and is subject to stringent control, both at the transcriptional and post-translational level. In the current study, we studied upstream events regulating trauma-induced apoptosis in the spinal cord. Within 1 h after SCI in rats, DP5 was induced, while Bim and Bad levels remained unchanged. In parallel, SCI also activated the stress-induced c-Jun N-terminal kinase (JNK), leading to the phosphorylation of c-Jun, with a similar temporal profile. Immunohistochemical analysis revealed that p-JNK and DP5 colocalized to neurons and oligodendrocytes undergoing apoptosis in the injured cord, but were absent in uninjured spinal cord. Furthermore, inhibition of JNK activity with in vivo delivery of SP600125 or a jnk1 antisense oligodeoxynucleotide (ODN) attenuated DP5 induction and caspase-3 activation. These results suggest that JNK activation contributes to trauma-induced DP5 expression and subsequent apoptosis in SCI.


Asunto(s)
Proteínas Reguladoras de la Apoptosis/metabolismo , Apoptosis/fisiología , Proteínas Quinasas JNK Activadas por Mitógenos/metabolismo , Degeneración Nerviosa/enzimología , Neuropéptidos/metabolismo , Traumatismos de la Médula Espinal/enzimología , Médula Espinal/enzimología , Animales , Apoptosis/efectos de los fármacos , Caspasa 3 , Caspasas/metabolismo , Modelos Animales de Enfermedad , Activación Enzimática/efectos de los fármacos , Activación Enzimática/fisiología , Inhibidores Enzimáticos/farmacología , Femenino , Proteínas Quinasas JNK Activadas por Mitógenos/antagonistas & inhibidores , Degeneración Nerviosa/patología , Degeneración Nerviosa/fisiopatología , Neuronas/enzimología , Neuronas/patología , Oligonucleótidos Antisentido/farmacología , Fosforilación/efectos de los fármacos , Proteínas Proto-Oncogénicas c-jun/metabolismo , Ratas , Ratas Long-Evans , Médula Espinal/patología , Médula Espinal/fisiopatología , Traumatismos de la Médula Espinal/patología , Traumatismos de la Médula Espinal/fisiopatología
18.
Neurobiol Dis ; 17(1): 99-107, 2004 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-15350970

RESUMEN

We have explored the molecular mechanism underlying amyloid beta-peptide (Abeta)-mediated cytotoxicity in vitro. Exposure of murine cerebral endothelial cells (CECs) or C6 glioma cells to Abeta25-35 resulted in dose-dependent cell death. Ceramide is a pro-apoptotic lipid mediator. Forced elevation of cellular ceramide levels, either by application of an exogenous C2 ceramide analogue or bacterial sphingomyelinase that induces endogenous ceramide release from sphingomyelin, mimicked Abeta25-35 cytotoxicity in both CECs and C6 glioma cells. Abeta25-35-induced synthesis of ceramide was selectively mediated by activation of neutral sphingomyelinase (nSMase), but not acidic sphingomyelinase (aSMase) or ceramide synthase. Both 3-O-Me-SM and N-acetyl-L-cysteine, the selective and nonselective pharmacological inhibitors of nSMase, respectively, suppressed nSMase activation, ceramide production, and cytotoxic action induced by Abeta25-35 in CECs. Furthermore, genetic knockdown of nSMase by an antisense strategy rendered C6 glioma cells specifically resistant to Abeta25-35 cytotoxicity without affecting their vulnerability to serum deprivation. Together, nSMase activation with subsequent ceramide production may contribute, at least partially, to Abeta25-35 cytotoxicity in cell types with cerebral endothelial and glial lineage.


Asunto(s)
Péptidos beta-Amiloides/toxicidad , Células Endoteliales/efectos de los fármacos , Neuroglía/efectos de los fármacos , Fragmentos de Péptidos/toxicidad , Esfingomielina Fosfodiesterasa/metabolismo , Animales , Muerte Celular/efectos de los fármacos , Muerte Celular/fisiología , Línea Celular , Células Endoteliales/enzimología , Activación Enzimática/efectos de los fármacos , Activación Enzimática/fisiología , Ratones , Neuroglía/enzimología , Inhibidores de Fosfodiesterasa/farmacología , Ratas , Esfingomielina Fosfodiesterasa/antagonistas & inhibidores
19.
J Neurochem ; 90(3): 637-45, 2004 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-15255941

RESUMEN

Although mRNA expression of group IIA secretory phospholipase A2 (sPLA2-IIA) has been implicated in responses to injury in the CNS, information on protein expression remains unclear. In this study, we investigated temporal and spatial expression of sPLA2-IIA mRNA and immunoreactivity in transient focal cerebral ischemia induced in rats by occlusion of the middle cerebral artery. Northern blot analysis showed a biphasic increase in sPLA2-IIA mRNA expression following 60-min of ischemia-reperfusion: an early phase at 30 min and a second increase at a late phase ranging from 12 h to 14 days. In situ hybridization localized the early-phase increase in sPLA2-IIA mRNA to the affected ischemic cortex and the late-phase increase to the penumbral area. Besides sPLA2-IIA mRNA, glial fibrillary acidic protein (GFAP) and cyclo-oxygenase-2 mRNAs, but not cytosolic PLA2, also showed an increase in the penumbral area at 3 days after ischemia-reperfusion. Immunohistochemistry of sPLA2-IIA indicated positive cells in the penumbral area similar to the GFAP-positive astrocytes but different from the isolectin B4-positive microglial cells. Confocal microscopy further confirmed immunoreactivity of sPLA2-IIA in reactive astrocytes but not in microglial cells. Taken together, these results demonstrate for the first time an up-regulation of the inflammatory sPLA2-IIA in reactive astrocytes in response to cerebral ischemia-reperfusion.


Asunto(s)
Astrocitos/enzimología , Encéfalo/enzimología , Ataque Isquémico Transitorio/enzimología , Fosfolipasas A/biosíntesis , Animales , Astrocitos/patología , Encéfalo/irrigación sanguínea , Encéfalo/patología , Ciclooxigenasa 2 , Citosol/enzimología , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Inducción Enzimática , Proteína Ácida Fibrilar de la Glía/genética , Fosfolipasas A2 Grupo II , Inmunohistoquímica , Ataque Isquémico Transitorio/patología , Isoenzimas/genética , Masculino , Neuronas/enzimología , Neuronas/patología , Fosfolipasas A/genética , Fosfolipasas A2 , Prostaglandina-Endoperóxido Sintasas/genética , ARN Mensajero/biosíntesis , Ratas , Ratas Long-Evans
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA