Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 27
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Sci Rep ; 11(1): 3442, 2021 02 09.
Artículo en Inglés | MEDLINE | ID: mdl-33564058

RESUMEN

Traumatic spinal cord injury (SCI) produces a complex syndrome that is expressed across multiple endpoints ranging from molecular and cellular changes to functional behavioral deficits. Effective therapeutic strategies for CNS injury are therefore likely to manifest multi-factorial effects across a broad range of biological and functional outcome measures. Thus, multivariate analytic approaches are needed to capture the linkage between biological and neurobehavioral outcomes. Injury-induced neuroinflammation (NI) presents a particularly challenging therapeutic target, since NI is involved in both degeneration and repair. Here, we used big-data integration and large-scale analytics to examine a large dataset of preclinical efficacy tests combining five different blinded, fully counter-balanced treatment trials for different acute anti-inflammatory treatments for cervical spinal cord injury in rats. Multi-dimensional discovery, using topological data analysis (TDA) and principal components analysis (PCA) revealed that only one showed consistent multidimensional syndromic benefit: intrathecal application of recombinant soluble TNFα receptor 1 (sTNFR1), which showed an inverse-U dose response efficacy. Using the optimal acute dose, we showed that clinically-relevant 90 min delayed treatment profoundly affected multiple biological indices of NI in the first 48 h after injury, including reduction in pro-inflammatory cytokines and gene expression of a coherent complex of acute inflammatory mediators and receptors. Further, a 90 min delayed bolus dose of sTNFR1 reduced the expression of NI markers in the chronic perilesional spinal cord, and consistently improved neurological function over 6 weeks post SCI. These results provide validation of a novel strategy for precision preclinical drug discovery that is likely to improve translation in the difficult landscape of CNS trauma, and confirm the importance of TNFα signaling as a therapeutic target.


Asunto(s)
Inteligencia Artificial , Modelos Neurológicos , Traumatismos de la Médula Espinal/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo , Animales , Modelos Animales de Enfermedad , Femenino , Inyecciones Espinales , Ratas Long-Evans , Receptores Tipo I de Factores de Necrosis Tumoral/farmacología , Proteínas Recombinantes/farmacología , Traumatismos de la Médula Espinal/tratamiento farmacológico , Traumatismos de la Médula Espinal/patología
2.
Cell Death Dis ; 7: e2214, 2016 05 05.
Artículo en Inglés | MEDLINE | ID: mdl-27148687

RESUMEN

Mitochondrial impairment induced by oxidative stress is a main characteristic of intrinsic cell death pathways in neurons underlying the pathology of neurodegenerative diseases. Therefore, protection of mitochondrial integrity and function is emerging as a promising strategy to prevent neuronal damage. Here, we show that pharmacological inhibition of hypoxia-inducible factor prolyl-4-hydroxylases (HIF-PHDs) by adaptaquin inhibits lipid peroxidation and fully maintains mitochondrial function as indicated by restored mitochondrial membrane potential and ATP production, reduced formation of mitochondrial reactive oxygen species (ROS) and preserved mitochondrial respiration, thereby protecting neuronal HT-22 cells in a model of glutamate-induced oxytosis. Selective reduction of PHD1 protein using CRISPR/Cas9 technology also reduced both lipid peroxidation and mitochondrial impairment, and attenuated glutamate toxicity in the HT-22 cells. Regulation of activating transcription factor 4 (ATF4) expression levels and related target genes may mediate these beneficial effects. Overall, these results expose HIF-PHDs as promising targets to protect mitochondria and, thereby, neurons from oxidative cell death.


Asunto(s)
Hidroxiquinolinas/farmacología , Prolina Dioxigenasas del Factor Inducible por Hipoxia/antagonistas & inhibidores , Neuronas/efectos de los fármacos , Procolágeno-Prolina Dioxigenasa/genética , Inhibidores de Prolil-Hidroxilasa/farmacología , Factor de Transcripción Activador 4/genética , Factor de Transcripción Activador 4/metabolismo , Adenosina Trifosfato/agonistas , Adenosina Trifosfato/biosíntesis , Animales , Apoptosis/efectos de los fármacos , Sistemas CRISPR-Cas , Línea Celular , Regulación de la Expresión Génica , Ácido Glutámico/toxicidad , Hipocampo/citología , Hipocampo/efectos de los fármacos , Hipocampo/metabolismo , Prolina Dioxigenasas del Factor Inducible por Hipoxia/genética , Prolina Dioxigenasas del Factor Inducible por Hipoxia/metabolismo , Peroxidación de Lípido/efectos de los fármacos , Potencial de la Membrana Mitocondrial/efectos de los fármacos , Potencial de la Membrana Mitocondrial/genética , Ratones , Neuronas/citología , Neuronas/metabolismo , Fosforilación Oxidativa/efectos de los fármacos , Estrés Oxidativo , Procolágeno-Prolina Dioxigenasa/deficiencia , Procolágeno-Prolina Dioxigenasa/metabolismo , Especies Reactivas de Oxígeno/antagonistas & inhibidores , Especies Reactivas de Oxígeno/metabolismo , Transducción de Señal
3.
Biochemistry (Mosc) ; 77(10): 1108-19, 2012 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-23157291

RESUMEN

This review describes the catalytic mechanism, substrate specificity, and structural peculiarities of alpha-ketoglutarate dependent nonheme iron dioxygenases catalyzing prolyl hydroxylation of hypoxia-inducible factor (HIF). Distinct localization and regulation of three isoforms of HIF prolyl hydroxylases suggest their different roles in cells. The recent identification of novel substrates other than HIF, namely ß2-adrenergic receptor and the large subunit of RNA polymerase II, places these enzymes in the focus of drug development efforts aimed at development of isoform-specific inhibitors. The challenges and prospects of designing isoform-specific inhibitors are discussed.


Asunto(s)
Factor 1 Inducible por Hipoxia/metabolismo , Procolágeno-Prolina Dioxigenasa/metabolismo , Catálisis , Diseño de Fármacos , Factor 1 Inducible por Hipoxia/química , Ácidos Cetoglutáricos/química , Ácidos Cetoglutáricos/metabolismo , Procolágeno-Prolina Dioxigenasa/química , Isoformas de Proteínas , Especificidad por Sustrato
4.
Ann Neurol ; 49(3): 345-54, 2001 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-11261509

RESUMEN

Global inhibitors of RNA or protein synthesis such as actinomycin D or cycloheximide abrogate neuronal apoptosis induced by numerous pathological stimuli in vitro and in vivo. The clinical application of actinomycin D or cycloheximide to human neurological disease has been limited by the toxicities of these agents. To overcome these toxicities, strategies must be developed to inhibit selectively the expression of deleterious proapoptotic proteins, while leaving the expression of antiapoptotic, proregeneration, and other critical homeostatic proteins unperturbed. Mithramycin A (trade name Plicamycin) is an aureolic acid antibiotic that has been used in humans to treat hypercalcemia and several types of cancers. This class of agents is believed to act, in part, by selectively inhibiting gene expression by displacing transcriptional activators that bind to G-C-rich regions of promoters. Here we demonstrate that mithramycin A and its structural analog chromomycin A3 are potent inhibitors of neuronal apoptosis induced by glutathione depletion-induced oxidative stress or the DNA-damaging agent camptothecin. We correlate the protective effects of mithramycin A with its ability to inhibit enhanced DNA binding of the transcription factors Sp1 and Sp3 to their cognate "G-C" box induced by oxidative stress or DNA damage. The protective effects of mithramycin A cannot be attributed to global inhibition of protein synthesis. Together, these results suggest that mithramycin A and its structural analogs may be effective agents for the treatment of neurological diseases associated with aberrant activation of apoptosis and highlight the potential use of sequence-selective DNA-binding drugs as neurological therapeutics.


Asunto(s)
Apoptosis/efectos de los fármacos , Encéfalo/metabolismo , Cromomicina A3/metabolismo , Daño del ADN/efectos de los fármacos , Enfermedades Neurodegenerativas/metabolismo , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Estrés Oxidativo/efectos de los fármacos , Plicamicina/análogos & derivados , Plicamicina/metabolismo , Animales , Secuencia de Bases , Técnicas de Cultivo de Célula , Electroforesis/métodos , Microscopía de Contraste de Fase , Neuronas/citología , Ratas , Ratas Sprague-Dawley
5.
FEBS Lett ; 491(1-2): 9-15, 2001 Feb 23.
Artículo en Inglés | MEDLINE | ID: mdl-11226409

RESUMEN

Previous studies have shown that certain tumor cell lines which naturally express high levels of the epidermal growth factor receptor (EGFR) undergo apoptosis when exposed to epidermal growth factor. Whether this phenomenon is a direct result of receptor overexpression or some other genetic alteration renders these cells sensitive to apoptosis is yet to be established. We show that experimentally increasing the level of EGFR expression predictably leads to apoptosis in a variety of cell types which requires an active tyrosine kinase but not EGFR autophosphorylation sites. Expression of a dominant negative Ras mutant in EGFR overexpressing cells results in a significant potentiation of EGFR induced apoptosis suggesting that Ras activation is a key survival signal generated by the EGFR. We propose that potentiation of EGFR induced apoptosis by dominant negative Ras results, at least in part, by a block of Akt activation.


Asunto(s)
Apoptosis , Factor de Crecimiento Epidérmico/metabolismo , Receptores ErbB/genética , Proteínas Serina-Treonina Quinasas , Proteínas Proto-Oncogénicas p21(ras)/genética , Western Blotting , Receptores ErbB/metabolismo , Genes Dominantes , Humanos , Mutación , Fosforilación , Proteínas Tirosina Quinasas/genética , Proteínas Tirosina Quinasas/metabolismo , Proteínas Proto-Oncogénicas/genética , Proteínas Proto-Oncogénicas/metabolismo , Proteínas Proto-Oncogénicas c-akt , Proteínas Proto-Oncogénicas p21(ras)/metabolismo , Transducción de Señal , Células Tumorales Cultivadas
6.
J Biol Chem ; 276(12): 8865-74, 2001 Mar 23.
Artículo en Inglés | MEDLINE | ID: mdl-11116146

RESUMEN

The transcription factor nuclear factor-kappaB (NF-kappaB) is activated by a diverse number of stimuli including tumor necrosis factor-alpha, interleukin-1, UV irradiation, viruses, as well as receptor tyrosine kinases such as the epidermal growth factor receptor (EGFR). NF-kappaB activation by the tumor necrosis factor receptor (TNFR) involves the formation of a multiprotein complex termed a signalosome. Although previous studies have shown that the activated EGFR can induce NF-kappaB, the mechanism of this activation remains unknown. In this study, we identify components of the signalosome formed by the activated EGFR required to activate NF-kappaB and show that, although the activated EGFR uses mechanisms similar to the TNFR, it recruits a distinct signalosome. We show the EGFR forms a complex with a TNFR-interacting protein (RIP), which plays a key role in TNFR-induced NF-kappaB activation, but not with TRADD, an adaptor protein which serves to recruit RIP to the TNFR. Furthermore, we show that the EGFR associates with NF-kappaB-inducing kinase (NIK) and provide evidence suggesting multiprotein complex formation between the EGFR, RIP, and NIK. Using a dominant negative NIK mutant, we show that NIK activation is required for EGFR-mediated NF-kappaB induction. We also show that a S32/36 IkappaBalpha mutant blocks EGFR-induced NF-kappaB activation. Our studies also suggest that a high level of EGFR expression, a frequent occurrence in human tumors, is optimal for epidermal growth factor-induced NF-kappaB activation. Finally, although protein kinase B/Akt has been implicated in tumor necrosis factor and PDGF-induced NF-kappaB activation, our studies do not support a role for this protein in EGFR-induced NF-kappaB activation.


Asunto(s)
Receptores ErbB/fisiología , Proteínas I-kappa B , FN-kappa B/metabolismo , Proteínas Serina-Treonina Quinasas/fisiología , Proteínas/fisiología , Secuencia de Bases , Línea Celular , Cartilla de ADN , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/fisiología , Receptores ErbB/antagonistas & inhibidores , Receptores ErbB/metabolismo , Humanos , Mutación , Inhibidor NF-kappaB alfa , Unión Proteica , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas/metabolismo , Proteína Serina-Treonina Quinasas de Interacción con Receptores , Quinasa de Factor Nuclear kappa B
7.
Antioxid Redox Signal ; 2(3): 421-36, 2000.
Artículo en Inglés | MEDLINE | ID: mdl-11229356

RESUMEN

Iron is the most abundant transition metal in the brain, where it functions as an important cofactor in a host of vital metabolic processes and plays an absolutely essential role in cell viability. Free iron is also very toxic when present in high concentrations, thus placing this essential metal at the core of neurotoxic injury in a number of neurological disorders. The pivotal role of iron in cellular homeostasis, including its latent toxicity, necessitates a tight regulation of iron metabolism. Oxygen and iron appear to play an important role in iron homeostasis. They appear to exert their homeostatic role by modulating the proteins involved in a complex interplay between iron sensing, transport, and storage. These key regulatory proteins include ferritin (intracellular storage), transferrin (extracellular transport), transferrin receptor, and iron regulatory protein (sensor of intracellular iron concentration). The interplay of iron and oxygen is most intriguing in the setting of stroke, where hypoxia and free iron appear to interact in causing the subsequent neuronal death.


Asunto(s)
Hipoxia-Isquemia Encefálica , Quelantes del Hierro/farmacología , Neuronas/metabolismo , Fármacos Neuroprotectores/farmacología , Electrones , Humanos , Hierro/metabolismo , Modelos Biológicos , Modelos Químicos , Neuronas/fisiología , Oxidación-Reducción , Accidente Cerebrovascular/metabolismo , Accidente Cerebrovascular/prevención & control
8.
J Neurosci ; 19(22): 9821-30, 1999 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-10559391

RESUMEN

Iron chelators are pluripotent neuronal antiapoptotic agents that have been shown to enhance metabolic recovery in cerebral ischemia models. The precise mechanism(s) by which these agents exert their effects remains unclear. Recent studies have demonstrated that iron chelators activate a hypoxia signal transduction pathway in non-neuronal cells that culminates in the stabilization of the transcriptional activator hypoxia-inducible factor-1 (HIF-1) and increased expression of gene products that mediate hypoxic adaptation. We examined the hypothesis that iron chelators prevent oxidative stress-induced death in cortical neuronal cultures by inducing expression of HIF-1 and its target genes. We report that the structurally distinct iron chelators deferoxamine mesylate and mimosine prevent apoptosis induced by glutathione depletion and oxidative stress in embryonic cortical neuronal cultures. The protective effects of iron chelators are correlated with their ability to enhance DNA binding of HIF-1 and activating transcription factor 1(ATF-1)/cAMP response element-binding protein (CREB) to the hypoxia response element in cortical cultures and the H19-7 hippocampal neuronal cell line. We show that mRNA, protein, and/or activity levels for genes whose expression is known to be regulated by HIF-1, including glycolytic enzymes, p21(waf1/cip1), and erythropoietin, are increased in cortical neuronal cultures in response to iron chelator treatment. Finally, we demonstrate that cobalt chloride, which also activates HIF-1 and ATF-1/CREB in cortical cultures, also prevents oxidative stress-induced death in these cells. Altogether, these results suggest that iron chelators exert their neuroprotective effects, in part, by activating a signal transduction pathway leading to increased expression of genes known to compensate for hypoxic or oxidative stress.


Asunto(s)
Apoptosis/fisiología , Corteza Cerebral/citología , Corteza Cerebral/fisiología , Proteína de Unión a Elemento de Respuesta al AMP Cíclico/farmacología , Ciclinas/genética , Proteínas de Unión al ADN/metabolismo , Deferoxamina/farmacología , Eritropoyetina/genética , Regulación de la Expresión Génica , Neuronas/citología , Neuronas/fisiología , Proteínas Nucleares/metabolismo , Estrés Oxidativo/fisiología , Factores de Transcripción/metabolismo , Factor de Transcripción Activador 1 , Animales , Apoptosis/efectos de los fármacos , Células Cultivadas , Quelantes/farmacología , Cobalto/farmacología , Inhibidor p21 de las Quinasas Dependientes de la Ciclina , Inducción Enzimática/efectos de los fármacos , Feto , Fructosa-Bifosfato Aldolasa/biosíntesis , Fructosa-Bifosfato Aldolasa/genética , Regulación de la Expresión Génica/efectos de los fármacos , Glutatión/metabolismo , Glucólisis , Factor 1 Inducible por Hipoxia , Subunidad alfa del Factor 1 Inducible por Hipoxia , L-Lactato Deshidrogenasa/biosíntesis , L-Lactato Deshidrogenasa/genética , Mimosina/farmacología , Neuronas/efectos de los fármacos , Fármacos Neuroprotectores , Ratas , Ratas Sprague-Dawley
9.
J Biol Chem ; 274(19): 13650-5, 1999 May 07.
Artículo en Inglés | MEDLINE | ID: mdl-10224137

RESUMEN

Infection of many cultured cell types with Sindbis virus (SV), an alphavirus, triggers apoptosis through a commonly utilized caspase activation pathway. However, the upstream signals by which SV activates downstream apoptotic effectors, including caspases, remain unclear. Here we report that in AT-3 prostate carcinoma cells, SV infection decreases superoxide (O-2) levels within minutes of infection as monitored by an aconitase activity assay. This SV-induced decrease in O-2 levels appears to activate or modulate cell death, as a recombinant SV expressing the O-2 scavenging enzyme, copper/zinc superoxide dismutase (SOD), potentiates SV-induced apoptosis. A recombinant SV expressing a mutant form of SOD, which has reduced SOD activity, has no effect. The potentiation of SV-induced apoptosis by wild type SOD is because of its ability to scavenge intracellular O-2 rather than its ability to promote the generation of hydrogen peroxide. Pyruvate, a peroxide scavenger, does not affect the ability of wild type SOD to potentiate cell death; and increasing the intracellular catalase activity via a recombinant SV vector has no effect on SV-induced apoptosis. Moreover, increasing intracellular O-2 by treatment of 3T3 cells with paraquat protects them from SV-induced death. Altogether, our results suggest that SV may activate apoptosis by reducing intracellular superoxide levels and define a novel redox signaling pathway by which viruses can trigger cell death.


Asunto(s)
Apoptosis/fisiología , Virus Sindbis/fisiología , Superóxidos/metabolismo , Células 3T3 , Animales , Peróxido de Hidrógeno/metabolismo , Masculino , Ratones , Paraquat/farmacología , Neoplasias de la Próstata/enzimología , Neoplasias de la Próstata/metabolismo , Neoplasias de la Próstata/patología , Virus Sindbis/efectos de los fármacos , Superóxido Dismutasa/metabolismo , Células Tumorales Cultivadas
10.
Neoplasia ; 1(6): 544-56, 1999 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-10935502

RESUMEN

Epidemiological evidence has suggested an association between diets rich in antioxidants and diminished risks of various types of cancer. Proposed mechanisms for protective effects of antioxidants have involved inhibition of free radical-mediated DNA damage. Recent data suggest that antioxidants may prevent or eliminate cancerous cells through their ability to inhibit proliferation or to induce programmed cell death (PCD). To begin to identify cell cycle and cell death regulatory factors involved in antioxidant-induced growth arrest and PCD, we have studied colorectal carcinoma cells (CRCs) that differ in expression of the tumor suppressor protein p53, and of the cyclin-dependent kinase (CDK) inhibitor p21(Waf1/Cip1). The antioxidants, N-acetylcysteine (NAC) and vitamin E either inhibited proliferation in a p53-independent manner without affecting cell viability or induced cell death. Growth arrest was not associated with upregulation of the CDK inhibitors p21(Waf1/Cip1), p18(ink4c) or p16(ink4a), but was associated with a decrease in reactive oxygen species (ROS). In contrast to previous observations, the absence of p21(Waf1/Cip1) increased susceptibility of CRCs to antioxidant-induced PCD. NAC decreased levels of retinoblastoma protein (Rb) phosphorylation in all cells tested, but Rb was cleaved only in cells which underwent NAC-induced death. Although NAC decreased ROS in all cells studied, cell lines in which PCD occurred had higher baseline levels of ROS than cell lines in which proliferation was blocked. These observations suggest that expression of p21(Waf1/Cip1) and basal levels of ROS are important determinants of outcome after antioxidant treatment.


Asunto(s)
Acetilcisteína/farmacología , Antioxidantes/farmacología , Apoptosis/efectos de los fármacos , Ciclinas/fisiología , Proteína p53 Supresora de Tumor/fisiología , Vitamina E/farmacología , División Celular/efectos de los fármacos , Células Cultivadas , Inhibidor p21 de las Quinasas Dependientes de la Ciclina , Humanos , Fosforilación , Especies Reactivas de Oxígeno/metabolismo , Proteína de Retinoblastoma/metabolismo
11.
Biochem Biophys Res Commun ; 249(2): 325-31, 1998 Aug 19.
Artículo en Inglés | MEDLINE | ID: mdl-9712695

RESUMEN

The calcium-sensing receptor (CaR) is a membrane-bound, G-protein-coupled receptor present on parathyroid cells which monitors the level of extracellular calcium (Ca2+o) and transduces signals involved in serum calcium regulation. Expression of CaR protein in tissues with functions unrelated to systemic calcium homeostasis, including the brain, suggests that extracellular calcium (Ca2+o) may act as a first messenger to regulate diverse cellular functions. To test this hypothesis, we examined the effect of increasing Ca2+o on apoptosis induced by Sindbis Virus in AT-3 prostate carcinoma cells. We found a steep increase in cell survival with between 5 and 7 mM added Ca2+o (EC50 = 6.1 mM). Magnesium, a less potent agonist of the calcium sensing receptor, was also protective (EC50 = 23.4 mM). Northern and immunocytochemical analyses confirmed the presence of the CaR message and protein in AT-3 prostate carcinoma cells. Enforced expression of CaR protein by stable transfection in human embryonic kidney (HEK)-293 cells, which normally don't express the receptor, resulted in resistance to SV-induced apoptosis in the presence of elevated Ca2+o. In addition to preventing SV-induced death, elevated Ca2+o also abrogated apoptosis induced by c-Myc overexpression/serum deprivation in rat 1A fibroblasts, and these fibroblasts were shown to express CaR message and protein. Altogether, these observations suggest that Ca2+o can act with the CaR to prevent apoptosis and define a novel mechanism by which calcium ions can regulate cell survival.


Asunto(s)
Apoptosis/efectos de los fármacos , Calcio/farmacología , Receptores de Superficie Celular/fisiología , Animales , Northern Blotting , Calcio/administración & dosificación , Línea Celular , Embrión de Mamíferos , Expresión Génica , Humanos , Inmunohistoquímica , Riñón , Masculino , Neoplasias de la Próstata , Ratas , Receptores Sensibles al Calcio , Receptores de Superficie Celular/genética , Virus Sindbis/fisiología , Transfección , Células Tumorales Cultivadas
12.
J Cell Biol ; 141(7): 1479-87, 1998 Jun 29.
Artículo en Inglés | MEDLINE | ID: mdl-9647642

RESUMEN

Recent studies have established cell type- specific, proapoptotic, or antiapoptotic functions for the transcription factor NF-kappaB. In each of these studies, inhibitors of NF-kappaB activity have been present before the apoptotic stimulus, and so the role of stimulus- induced NF-kappaB activation in enhancing or inhibiting survival could not be directly assessed. Sindbis virus, an alphavirus, induces NF-kappaB activation and apoptosis in cultured cell lines. To address whether Sindbis virus- induced NF-kappaB activation is required for apoptosis, we used a chimeric Sindbis virus that expresses a superrepressor of NF-kappaB activity. Complete suppression of virus-induced NF-kappaB activity neither prevents nor potentiates Sindbis virus-induced apoptosis. In contrast, inhibition of NF-kappaB activity before infection inhibits Sindbis virus-induced apoptosis. Our results demonstrate that suppression of steady-state, but not stimulus-induced NF-kappaB activity, regulates expression of gene products required for Sindbis virus-induced death. Furthermore, we show that in the same cell line, NF-kappaB can be proapoptotic or antiapoptotic depending on the death stimulus. We propose that the role of NF-kappaB in regulating apoptosis is determined by the death stimulus and by the timing of modulating NF-kappaB activity relative to the death stimulus.


Asunto(s)
Apoptosis , Proteínas de Unión al ADN/metabolismo , Proteínas I-kappa B , FN-kappa B/metabolismo , Virus Sindbis/fisiología , Células 3T3 , Acetilcisteína/farmacología , Animales , Antivirales/farmacología , Proteínas de Unión al ADN/genética , Vectores Genéticos , Peróxido de Hidrógeno/toxicidad , Ratones , Inhibidor NF-kappaB alfa , Subunidad p50 de NF-kappa B , Ratas , Estaurosporina/toxicidad , Factores de Tiempo , Factor de Transcripción ReIA , Células Tumorales Cultivadas , Factor de Necrosis Tumoral alfa/toxicidad
13.
J Neurosci ; 18(11): 4083-95, 1998 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-9592089

RESUMEN

Catalase is an antioxidant enzyme that has been shown to inhibit apoptotic or necrotic neuronal death induced by hydrogen peroxide. We report the purification of a contaminating antiapoptotic activity from a commercial bovine liver catalase preparation by following its ability to inhibit apoptosis when applied extracellularly in multiple death paradigms. The antiapoptotic activity was identified by protein microsequencing as arginase, a urea cycle and nitric oxide synthase-regulating enzyme, and confirmed by demonstrating the presence of antiapoptotic activity in a >97% pure preparation of recombinant arginase. The pluripotency of recombinant arginase was demonstrated by its ability to inhibit apoptosis in multiple paradigms including rat cortical neurons induced to die by glutathione depletion and oxidative stress, by 100 nM staurosporine treatment, or by Sindbis virus infection. The protective effects of arginase in these apoptotic paradigms, in contrast to previous studies on excitotoxic neuronal necrosis, are independent of nitric oxide synthase inhibition. Rather, arginase-induced depletion of arginine leads to inhibition of protein synthesis, resulting in cell survival. Because inhibitors of nitric oxide synthesis and of protein synthesis have been shown to decrease necrotic and apoptotic death, respectively, in animal models of stroke and spinal cord injury, arginine-depleting enzymes, capable of simultaneously inhibiting protein synthesis and nitric oxide generation, may be propitious therapeutic agents for acute neurological diseases. Furthermore, our results suggest caution in attributing the cytoprotective effects of some catalase preparations to catalase.


Asunto(s)
Apoptosis/fisiología , Arginasa/metabolismo , Hígado/enzimología , Neuronas/citología , Óxido Nítrico/metabolismo , Secuencia de Aminoácidos , Aminoácidos/metabolismo , Animales , Apoptosis/efectos de los fármacos , Arginasa/genética , Arginasa/farmacología , Catalasa/genética , Catalasa/metabolismo , Catalasa/farmacología , Bovinos , Supervivencia Celular/efectos de los fármacos , Células Cultivadas , Corteza Cerebral/citología , Inhibidores Enzimáticos/farmacología , Feto/citología , Glutatión/metabolismo , Datos de Secuencia Molecular , Fármacos Neuroprotectores/farmacología , Estrés Oxidativo/fisiología , Ratas , Ratas Sprague-Dawley , Estaurosporina/farmacología
14.
J Biol Chem ; 273(12): 6885-91, 1998 Mar 20.
Artículo en Inglés | MEDLINE | ID: mdl-9506992

RESUMEN

Receptor tyrosine kinases are classified into subfamilies, which are believed to function independently, with heterodimerization occurring only within the same subfamily. In this study, we present evidence suggesting a direct interaction between the epidermal growth factor (EGF) receptor (EGFR) and the platelet-derived growth factor beta (PDGFbeta) receptor (PDGFbetaR), members of different receptor tyrosine kinase subfamilies. We find that the addition of EGF to COS-7 cells and to human foreskin Hs27 fibroblasts results in a rapid tyrosine phosphorylation of the PDGFbetaR and results in the recruitment of phosphatidylinositol 3-kinase to the PDGFbetaR. In R1hER cells, which overexpress the EGFR, we find ligand-independent tyrosine phosphorylation of the PDGFbetaR and the constitutive binding of a substantial amount of PI-3 kinase activity to it, mimicking the effect of ligand in untransfected cells. In support of the possibility that this may be a direct interaction, we show that the two receptors can be coimmunoprecipitated from untransfected Hs27 fibroblasts and from COS-7 cells. This association can be reconstituted by introducing the two receptors into 293 EBNA cells. The EGFR/PDGFbetaR association is ligand-independent in all cell lines tested. We also demonstrate that the fraction of PDGFbetaR bound to the EGFR in R1hER cells undergoes an EGF-induced mobility shift on Western blots indicative of phosphorylation. Our findings indicate that direct interactions between receptor tyrosine kinases classified under different subfamilies may be more widespread than previously believed.


Asunto(s)
Receptores ErbB/metabolismo , Fosfatidilinositol 3-Quinasas/metabolismo , Receptores del Factor de Crecimiento Derivado de Plaquetas/metabolismo , Animales , Células COS , Línea Celular , Humanos , Pruebas de Precipitina , Unión Proteica , Receptor beta de Factor de Crecimiento Derivado de Plaquetas , Transducción de Señal , Transfección
15.
Cell Death Differ ; 5(7): 577-83, 1998 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-10200512

RESUMEN

We previously established that NF-kappaB DNA binding activity is required for Sindbis Virus (SV)-induced apoptosis. To investigate whether SV induces nuclear translocation of NF-kappaB via the proteasomal degradation pathway, we utilized MG132, a peptide aldehyde inhibitor of the catalytic subunit of the proteasome. 20 microM MG132 completely abrogated SV-induced NF-kappaB nuclear activity at early time points after infection. Parallel measures of cell viability 48 h after SV infection revealed that 20 microM MG132 induced apoptosis in uninfected cells. In contrast, a lower concentration of MG132 (200 nM) resulted in partial inhibition of SV-induced nuclear NF-kappaB activity and inhibition of SV-induced apoptosis without inducing toxicity in uninfected cells. The specific proteasomal inhibitor, lactacystin, also inhibited SV-induced death. Taken together, these results suggest that the pro-apoptotic and anti-apoptotic functions of peptide aldehyde proteasome inhibitors such as MG-132 depend on the concentration of inhibitor utilized and expand the list of stimuli requiring proteasomal activation to induce apoptosis to include viruses.


Asunto(s)
Apoptosis , Cisteína Endopeptidasas/metabolismo , Inhibidores de Cisteína Proteinasa/farmacología , Leupeptinas/farmacología , Complejos Multienzimáticos/metabolismo , Relación Dosis-Respuesta a Droga , FN-kappa B/metabolismo , Complejo de la Endopetidasa Proteasomal , Virus Sindbis/fisiología
16.
Proc Natl Acad Sci U S A ; 94(6): 2676-80, 1997 Mar 18.
Artículo en Inglés | MEDLINE | ID: mdl-9122255

RESUMEN

Nitric oxide subserves diverse physiologic roles in the nervous system. NO is produced from at least three different NO synthase (NOS) isoforms: neuronal NOS (nNOS), endothelial NOS, and immunologic NOS (iNOS). We show that nNOS is the predominant isoform constitutively expressed in glia. NO derived from nNOS in glia inhibits the transcription factor nuclear factor kappaB (NF kappaB) as NOS inhibitors enhance basal NF kappaB activation. Pyrrolidine dithiocarbamate (PDTC) is an inhibitor of NF kappaB in most cells; however, we show that PDTC is also a potent scavenger of NO through formation of mononitrosyl iron complexes with PDTC. In Jurkat cells, a human T-cell lymphoma cell line, tumor necrosis factor-alpha (TNF-alpha) induces NF kappaB activation that is inhibited by PDTC. Contrary to the results in Jurkat cells, PDTC did not inhibit tumor necrosis factor-alpha-induced NF kappaB activation in astrocytes; instead PDTC itself induces NF kappaB activation in astrocytes, and this may be related to scavenging of endogenously produced NO by the PDTC iron complex. In astrocytes PDTC also dramatically induces the NF kappaB-dependent enzyme, iNOS, supporting the physiologic relevance of endogenous NO regulation of NF kappaB. NF kappaB activation in glia from mice lacking nNOS responds more rapidly to PDTC compared with astrocytes from wild-type mice. Our data suggest that nNOS in astrocytes regulates NF kappaB activity and iNOS expression, and indicate a novel regulatory role for nNOS in tonically suppressing central nervous system, NF kappaB-regulated genes.


Asunto(s)
Corteza Cerebral/metabolismo , Isoenzimas/metabolismo , FN-kappa B/biosíntesis , Neuronas/metabolismo , Óxido Nítrico Sintasa/metabolismo , Animales , Animales Recién Nacidos , Antioxidantes/farmacología , Astrocitos/efectos de los fármacos , Astrocitos/metabolismo , Humanos , Isoenzimas/biosíntesis , Células Jurkat , Cinética , Linfocitos/efectos de los fármacos , Linfocitos/metabolismo , Ratones , FN-kappa B/aislamiento & purificación , Neuronas/efectos de los fármacos , Óxido Nítrico/fisiología , Óxido Nítrico Sintasa/biosíntesis , Óxido Nítrico Sintasa de Tipo I , Nitritos/metabolismo , Pirrolidinas/farmacología , Ratas , Ratas Endogámicas Lew , Tiocarbamatos/farmacología , omega-N-Metilarginina/farmacología
17.
Neurochem Int ; 29(2): 153-7, 1996 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-8837044

RESUMEN

We have previously shown that glutamate-induced cystine deprivation of embryonic cortical neurons leads to intracellular depletion of the antioxidant glutathione, consequent oxidative stress and apoptotic cell death. To test the hypothesis that glutathione depletion and oxidative stress represent a common pathway of neuronal apoptosis, we examined the effect of a variety of antioxidants on serum deprivation-induced death in embryonic cortical neurons. A host of antioxidant agents, capable of abrogating glutathione depletion-induced apoptosis in cortical neurons, were unable to inhibit serum deprivation-induced death in these cells. To test whether serum deprivation and glutathione depletion involve different or antagonistic pathways, we serum-deprived cortical neurons at the time of induction of glutathione depletion. Surprisingly, we found that serum deprivation diminished glutathione depletion-induced death as compared to cultures treated with growth factors or serum. These observations suggest that serum deprivation antagonizes the cell death signaling pathway activated by glutathione depletion and that serum and growth factors can enhance susceptibility to oxidative stress. Consistent with these conclusions, we show that growth factors or serum added in combination with antioxidants possess superior survival promoting effects as compared to either agent alone.


Asunto(s)
Apoptosis , Corteza Cerebral/citología , Corteza Cerebral/embriología , Medio de Cultivo Libre de Suero , Glutatión/administración & dosificación , Estrés Oxidativo , Acetilcisteína/farmacología , Animales , Antioxidantes/farmacología , Factor Neurotrófico Derivado del Encéfalo/farmacología , Hidroxianisol Butilado/farmacología , Células Cultivadas , Cistina/farmacología , Deferoxamina/farmacología , Insulina/farmacología , Neuronas/efectos de los fármacos , Neuronas/fisiología , Ratas , Ratas Sprague-Dawley
18.
J Cell Biol ; 131(5): 1149-61, 1995 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-8522579

RESUMEN

Oxidative stress has been proposed as a common mediator of apoptotic death. To investigate further the role of oxidants in this process we have studied the effects of antioxidants on Sindbis virus (SV)-induced apoptosis in two cell lines, AT-3 (a prostate carcinoma line) and N18 (a neuroblastoma line). The thiol antioxidant, N-acetylcysteine (NAC), at concentrations above 30 mM, completely abrogates SV-induced apoptosis in AT-3 and N18 cells. The effects of NAC cannot be attributed to inhibition of viral entry or viral replication, changes in extracellular osmolarity or to increases in cellular glutathione levels, nor can they be mimicked by chelators of trace metals, inhibitors of lipid peroxidation or peroxide scavengers. In contrast, other thiol agents including pyrrolidine dithiocarbamate (PDTC, 75 microM) are protective. Because NAC and PDTC are among the most effective inhibitors of the transcription factor NF-kappa B, we examined SV's ability to activate NF-kappa B before the onset of morphologic or biochemical evidence of apoptosis. Within hours of infection, SV induced a robust increase in nuclear NF-kappa B activity in AT-3 and N18 cells; this activation was suppressible by NAC and PDTC. Over-expression of bcl-2 in AT-3 cells, which has been shown to inhibit SV-induced apoptosis, also inhibits SV-induced NF-kappa B activation. To determine if NF-kappa B activation is necessary for SV-induced apoptosis in these cells, we used double stranded oligonucleotides with consensus NF-kappa B sequences as transcription factor decoys (TFDs) to inhibit NF-kappa B binding to native DNA sites. Wild-type, but not mutant, TFDs inhibit SV-induced apoptosis in AT-3 cells. In contrast, TFD inhibition of NF-kappa B nuclear activity in N18 cells did not prevent SV-induced apoptosis. Taken together, these observations define a cell type-specific, transcription factor signaling pathway necessary for SV-induced apoptosis. Understanding the precise mechanism by which Bcl-2 and thiol agents inhibit SV-induced nuclear NF-kappa B activity in AT-3 cells may provide insights into the pluripotent antiapoptotic actions of these agents.


Asunto(s)
Apoptosis , FN-kappa B/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , Virus Sindbis/fisiología , Reactivos de Sulfhidrilo/farmacología , Acetilcisteína/farmacología , Animales , Secuencia de Bases , Núcleo Celular/metabolismo , Quelantes/farmacología , ADN , Ditiotreitol/farmacología , Hierro , Peroxidación de Lípido/efectos de los fármacos , Masculino , Mercaptoetanol/farmacología , Ratones , Datos de Secuencia Molecular , FN-kappa B/antagonistas & inhibidores , Neuroblastoma/patología , Neoplasias de la Próstata/patología , Proteínas Proto-Oncogénicas c-bcl-2 , Pirrolidinas/farmacología , Ratas , Tiocarbamatos/farmacología , Células Tumorales Cultivadas
19.
Clin Exp Pharmacol Physiol ; 22(4): 309-10, 1995 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-7671450

RESUMEN

1. Oxidative stress is believed to be an important mediator of neuronal cell death but the precise mechanism by which this occurs is unknown. 2. We have developed an in vitro model of neuronal oxidative stress to study the pathways by which free radicals kill neurones. 3. We have shown that oxidative stress, cystine deprivation and glutathione depletion results in cell death with the morphological and biochemical features of apoptosis. 4. Neuronal apoptosis induced by oxidative stress can be inhibited by macromolecular synthesis inhibitors. 5. This in vitro model will be a valuable tool for defining the molecular targets of toxic free radicals in neurones and, in turn, in designing rational new therapies for free radical mediated diseases.


Asunto(s)
Apoptosis , Neuronas/efectos de los fármacos , Estrés Oxidativo , Especies Reactivas de Oxígeno/toxicidad , Células Cultivadas , Enfermedades del Sistema Nervioso Central/etiología , Cistina/metabolismo , Radicales Libres , Ácido Glutámico/toxicidad , Glutatión/metabolismo , Humanos , Neuronas/citología , Neuronas/patología
20.
J Neurosci ; 14(7): 4385-92, 1994 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-8027786

RESUMEN

Although macromolecular synthesis inhibitors have been demonstrated to prevent neuronal apoptosis in a number of paradigms, their mechanisms of protection remains unclear. Recently, we found that neuronal death resulting from cystine deprivation, glutathione loss, and oxidative stress is apoptotic and is prevented by inhibitors of macromolecular synthesis. We now report that protection is associated with enhanced availability of acid-soluble cyst(e)ine and restoration of cellular glutathione levels. N-acetylcysteine, an agent that delivers exogenous cysteine intracellularly and raises glutathione, is also protective, while buthionine sulfoximine, an inhibitor of glutathione synthesis, prevents protection by inhibitors of macromolecular synthesis. These results suggest that protection provided by these agents, in this paradigm, derives from shunting of the amino acid cysteine from global protein synthesis into the formation of the antioxidant glutathione.


Asunto(s)
Apoptosis/efectos de los fármacos , Cicloheximida/farmacología , Cisteína/metabolismo , Glutatión/metabolismo , Neuronas/efectos de los fármacos , Oxidantes/farmacología , Acetilcisteína/farmacología , Animales , Butionina Sulfoximina , Células Cultivadas , Corteza Cerebral/citología , Corteza Cerebral/embriología , Glutamatos/envenenamiento , Glutatión/antagonistas & inhibidores , Homocisteína/análogos & derivados , Homocisteína/farmacología , Sustancias Macromoleculares , Metionina Sulfoximina/análogos & derivados , Metionina Sulfoximina/farmacología , Neuronas/fisiología , Biosíntesis de Proteínas , Proteínas/antagonistas & inhibidores , Ratas
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA