Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 58
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Cell Physiol Biochem ; 45(5): 2095-2106, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29533931

RESUMEN

BACKGROUND/AIMS: Our previous study suggested the anti-tumor activity of sepia ink oligopeptide (SIO). Here we sought to investigate the underlying molecular mechanism. METHODS: Cell proliferation was evaluated by cell counting kit-8 (CCK-8) assay. Cell apoptosis was determined by Annexin V/Propidium Iodide (PI) staining. The mitochondria pathway was characterized by quantification of Bcl-2, Bax, Caspase-9 and Cyto-C. The death receptor pathway was analyzed by determinement of Fas, Caspase-8 and NIK. The endoplasmic reticulum (ER)-dependent pathway was determined by measurement the expression of CHOP, Caspase-12, GRP78 and Calpain. The associated gene expression was quantified by RT-PCR and protein level was determined by immunoblotting. RESULTS: We demonstrated treatment with structurally modified SIO (CSIO, 5 µM) significantly inhibited cell proliferation and induced apoptosis in lung cancer cell line A549. The mitochondrial pathway, death receptor pathway and ER stress induced apoptosis were stimulated upon CSIO treatment. The administration with respective inhibitors including midiv-1 (50 µM for 2 h), PDTC (20 µM PDTC for 30 min) and ALLN (20 mM ALLN for 5 h) readily reversed the apoptosis inducing effect of CSIO. CONCLUSION: Our data demonstrates that CSIO is capable of induction apoptosis in lung cancer cell line, which is mediated by all three classical apoptotic pathways. Our results warrant further in vivo investigations of the anti-tumor potential of CSIO.


Asunto(s)
Apoptosis/efectos de los fármacos , Mitocondrias/efectos de los fármacos , Oligopéptidos/toxicidad , Sepia/metabolismo , Células A549 , Animales , Calpaína/genética , Calpaína/metabolismo , Caspasa 12/genética , Caspasa 12/metabolismo , Caspasa 8/genética , Caspasa 8/metabolismo , Caspasa 9/genética , Caspasa 9/metabolismo , Proliferación Celular/efectos de los fármacos , Retículo Endoplásmico/metabolismo , Chaperón BiP del Retículo Endoplásmico , Estrés del Retículo Endoplásmico/efectos de los fármacos , Humanos , Tinta , Leupeptinas/toxicidad , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/patología , Mitocondrias/metabolismo , Prolina/análogos & derivados , Prolina/toxicidad , Proteínas Proto-Oncogénicas c-bcl-2/genética , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , Tiocarbamatos/toxicidad , Factor de Transcripción CHOP/genética , Factor de Transcripción CHOP/metabolismo , Proteína X Asociada a bcl-2/genética , Proteína X Asociada a bcl-2/metabolismo , Receptor fas/genética , Receptor fas/metabolismo
2.
J Nutr Biochem ; 51: 69-79, 2018 01.
Artículo en Inglés | MEDLINE | ID: mdl-29096318

RESUMEN

Proteasomes are known to degrade proteins involved in various processes like metabolism, signal transduction, cell-cycle regulation, inflammation, and apoptosis. Evidence showed that protein degradation has a strong influence on developing neurons as well as synaptic plasticity. Here, we have shown that sulforaphane (SFN) could prevent the deleterious effects of postnatal proteasomal inhibition on spatial reference and working memory of adult mice. One day old Balb/c mice received intracerebroventricular injections of MG132 and SFN. Sham received an equal volume of aCSF. We observed that SFN pre-administration could attenuate MG132 mediated decrease in proteasome and calpain activities. In vitro findings revealed that SFN could induce proteasomal activity by enhancing the expression of catalytic subunit-ß5. SFN pre-administration prevented the hippocampus based spatial memory impairments during adulthood, mediated by postnatal MG132 exposure. Histological examination showed deleterious effects of MG132 on pyramidal neurons and granule cell neurons in DG and CA3 sub-regions respectively. Furthermore, SFN pre-administration has shown to attenuate the effect of MG132 on proteasome subunit-ß5 expression and also induce the Nrf2 nuclear translocation. In addition, SFN pre-administered mice have also shown to induce expression of pCaMKII, pCreb, and mature/pro-Bdnf, molecules which play a crucial role in spatial learning and memory consolidation. Our findings have shown that proteasomes play an important role in hippocampal synaptic plasticity during the early postnatal period and SFN pre-administration could enhance the proteasomal activity as well as improve spatial learning and memory consolidation.


Asunto(s)
Hipocampo/efectos de los fármacos , Isotiocianatos/uso terapéutico , Neuronas/efectos de los fármacos , Fármacos Neuroprotectores/uso terapéutico , Síndromes de Neurotoxicidad/prevención & control , Inhibidores de Proteasoma/toxicidad , Aprendizaje Espacial/efectos de los fármacos , Animales , Animales Recién Nacidos , Región CA3 Hipocampal/efectos de los fármacos , Región CA3 Hipocampal/metabolismo , Región CA3 Hipocampal/patología , Línea Celular Tumoral , Giro Dentado/efectos de los fármacos , Giro Dentado/metabolismo , Giro Dentado/patología , Regulación del Desarrollo de la Expresión Génica/efectos de los fármacos , Hipocampo/metabolismo , Hipocampo/patología , Humanos , Inyecciones Intraventriculares , Isotiocianatos/administración & dosificación , Leupeptinas/administración & dosificación , Leupeptinas/toxicidad , Trastornos de la Memoria/etiología , Trastornos de la Memoria/prevención & control , Ratones Endogámicos BALB C , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/metabolismo , Plasticidad Neuronal/efectos de los fármacos , Neuronas/metabolismo , Neuronas/patología , Fármacos Neuroprotectores/administración & dosificación , Fármacos Neuroprotectores/farmacología , Síndromes de Neurotoxicidad/metabolismo , Síndromes de Neurotoxicidad/patología , Síndromes de Neurotoxicidad/fisiopatología , Complejo de la Endopetidasa Proteasomal/genética , Complejo de la Endopetidasa Proteasomal/metabolismo , Inhibidores de Proteasoma/administración & dosificación , Células Piramidales/efectos de los fármacos , Células Piramidales/metabolismo , Células Piramidales/patología , Sulfóxidos
3.
J Cell Biochem ; 119(1): 1062-1073, 2018 01.
Artículo en Inglés | MEDLINE | ID: mdl-28688199

RESUMEN

Mutations in PARK2 (or parkin) are responsible for 50% of cases of autosomal-recessive juvenile-onset Parkinson's disease (PD). To date, 21 alternative splice variants of the human gene have been cloned. Yet most studies have focused on the full-length protein, whereas the spectrum of the parkin isoforms expressed in PD has never been investigated. In this study, the role of parkin proteins in PD neurodegeneration was explored for the first time by analyzing their expression profile in an in vitro model of PD. To do so, undifferentiated and all-trans-retinoic-acid (RA)-differentiated SH-SY5Y cells (which thereby acquire a PD-like phenotype) were exposed to PD-mimicking neurotoxins: 1-methyl-4-phenylpyridinium (MPP+ ) and 6-hydroxydopamine (6-OHDA) are widely used in PD models, whereas carbonyl cyanide m-chlorophenyl hydrazone (CCCP) and carbobenzoxy-Leu-Leu-leucinal (MG132) interfere, respectively, with mitochondrial mitophagy and proteasomal degradation. Following treatment with each neurotoxin H1, the first parkin isoform to be cloned, was down-regulated compared to the respective controls both in undifferentiated and RA-differentiated cells. In contrast, the expression pattern of the minor splice isoforms varied as a function of the compound used: it was largely unchanged in both cell cultures (eg, H21-H6, H12, XP isoform) or it showed virtually opposite alterations in undifferentiated and RA-differentiated cells (eg, H20 and H3 isoform). This complex picture suggests that up- or down-regulation may be a direct effect of toxin exposure, and that the different isoforms may exert different actions in neurodegeneration via modulation of different molecular pathways.


Asunto(s)
Empalme Alternativo/efectos de los fármacos , Modelos Biológicos , Neurotoxinas/toxicidad , Enfermedad de Parkinson/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo , 1-Metil-4-fenilpiridinio/toxicidad , Carbonil Cianuro m-Clorofenil Hidrazona/toxicidad , Diferenciación Celular , Línea Celular , Supervivencia Celular/efectos de los fármacos , Regulación hacia Abajo , Humanos , Técnicas In Vitro , Leupeptinas/toxicidad , Oxidopamina/toxicidad , Enfermedad de Parkinson/etiología , Enfermedad de Parkinson/genética , Isoformas de Proteínas/metabolismo , Transducción de Señal , Tretinoina/farmacología , Ubiquitina-Proteína Ligasas/genética
4.
Neuroscience ; 367: 47-59, 2017 Dec 26.
Artículo en Inglés | MEDLINE | ID: mdl-29080716

RESUMEN

Ubiquitin-proteasome system (UPS) has emerged as major molecular mechanism which modulates synaptic plasticity. However, very little is known about what happens if this system fails during postnatal brain development. In the present study, MG132 was administered intracerebroventricularly in BALB/c mice pups at postnatal day one (P1), a very crucial period for synaptogenesis. Both 20S proteasome and calpain activities were found to be reduced in the mid brain of MG132-administered pups after 24 h. Mice (P40) which received MG132 on P1 were subjected to Morris water maze (MWM) training. Analysis showed spatial learning and memory of MG132 mice was significantly impaired when compared to age-matched controls. Hematoxylin and eosin as well as Cresyl Violet staining revealed substantial loss of cellular connections, distorted architecture and increased pyknosis in hippocampal CA1 and CA3 regions of MG132 mice. Immunohistochemical analysis of MG132 mice showed increased accumulation of intracellular amyloid-ß in hippocampal cells when compared to control. Moreover, double immunostaining revealed increased expression of amyloid precursor protein C-terminal fragments (APP-CTFß) without affecting ß-secretase expression in MG132 mice. Real-Time PCR analyses showed significant increase in hippocampal expression of α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA) receptor subunit glutamate A1 (GluA1), but no change in the brain-derived neurotrophic factor (Bdnf) expression in MG132 mice. Western blot analyses showed decreased levels of pThr286-CaMKIIα:CaMKIIα and pSer133-CREB:CREB ratio but increased pro:mature BDNF ratio in the hippocampus of MG132 mice. Taken together, postnatal proteasome inhibition could lead to accumulation of intracellular amyloid-ß protein aggregates, which mediate hippocampus-dependent spatial memory impairments in adult mice.


Asunto(s)
Péptidos beta-Amiloides/metabolismo , Hipocampo/metabolismo , Discapacidades para el Aprendizaje/patología , Complejo de la Endopetidasa Proteasomal/metabolismo , Aprendizaje Espacial/fisiología , Secretasas de la Proteína Precursora del Amiloide/metabolismo , Precursor de Proteína beta-Amiloide/metabolismo , Animales , Animales Recién Nacidos , Ácido Aspártico Endopeptidasas/metabolismo , Modelos Animales de Enfermedad , Factor 2 Eucariótico de Iniciación/genética , Factor 2 Eucariótico de Iniciación/metabolismo , Femenino , Hipocampo/efectos de los fármacos , Hipocampo/crecimiento & desarrollo , Discapacidades para el Aprendizaje/inducido químicamente , Leupeptinas/toxicidad , Masculino , Aprendizaje por Laberinto/efectos de los fármacos , Ratones , Ratones Endogámicos BALB C , Inhibidores de Proteasoma/toxicidad , Agregación Patológica de Proteínas/inducido químicamente , Receptores AMPA/genética , Receptores AMPA/metabolismo , Aprendizaje Espacial/efectos de los fármacos
5.
Mol Pharmacol ; 92(5): 564-575, 2017 11.
Artículo en Inglés | MEDLINE | ID: mdl-28830914

RESUMEN

N-acetyl-l-cysteine (NAC) exhibits protective properties in brain injury models and has undergone a number of clinical trials. Most studies of NAC have focused on neurons. However, neuroprotection may be complemented by the protection of astrocytes because healthier astrocytes can better support the viability of neurons. Here, we show that NAC can protect astrocytes against protein misfolding stress (proteotoxicity), the hallmark of neurodegenerative disorders. Although NAC is thought to be a glutathione precursor, NAC protected primary astrocytes from the toxicity of the proteasome inhibitor MG132 without eliciting any increase in glutathione. Furthermore, glutathione depletion failed to attenuate the protective effects of NAC. MG132 elicited a robust increase in the folding chaperone heat shock protein 70 (Hsp70), and NAC mitigated this effect. Nevertheless, three independent inhibitors of Hsp70 function ablated the protective effects of NAC, suggesting that NAC may help preserve Hsp70 chaperone activity and improve protein quality control without need for Hsp70 induction. Consistent with this view, NAC abolished an increase in ubiquitinated proteins in MG132-treated astrocytes. However, NAC did not affect the loss of proteasome activity in response to MG132, demonstrating that it boosted protein homeostasis and cell viability without directly interfering with the efficacy of this proteasome inhibitor. The thiol-containing molecules l-cysteine and d-cysteine both mimicked the protective effects of NAC, whereas the thiol-lacking molecule N-acetyl-S-methyl-l-cysteine failed to exert protection or blunt the rise in ubiquitinated proteins. Collectively, these findings suggest that the thiol group in NAC is required for its effects on glial viability and protein quality control.


Asunto(s)
Acetilcisteína/farmacología , Astrocitos/efectos de los fármacos , Citoprotección/efectos de los fármacos , Glutatión , Pliegue de Proteína/efectos de los fármacos , Animales , Astrocitos/fisiología , Supervivencia Celular/efectos de los fármacos , Supervivencia Celular/fisiología , Citoprotección/fisiología , Relación Dosis-Respuesta a Droga , Femenino , Proteínas HSP70 de Choque Térmico/antagonistas & inhibidores , Proteínas HSP70 de Choque Térmico/fisiología , Leupeptinas/toxicidad , Masculino , Ratas , Ratas Sprague-Dawley
6.
Neurotox Res ; 30(3): 392-406, 2016 10.
Artículo en Inglés | MEDLINE | ID: mdl-27194525

RESUMEN

N-acetyl-leucyl-leucyl-norleucinal (ALLN), an inhibitor of proteasomes and calpain, is widely used to reduce proteasomes or calpain-mediated cell death in rodents. However, ALLN is toxic to retinal neurons to some extent. At the concentration of 10 µM, ALLN is non-toxic to cortical neurons, but induces cell death of retinal neurons in vitro. The tolerance concentration of ALLN for retinal neurons is unclear, and the precise mechanism of cell death induced by ALLN remains elusive. In this study, we investigated the toxic effect of ALLN on primary retinal neurons. The 3-(4,5-dimethylthiazole-2-yl)-2,5-diphenyltetrazolium bromide (MTT) assay showed no significant changes of cell viability at 1 µM but decreased cell viability after treatment of ALLN at 2.5, 5, and 7.5 µM. Lactate dehydrogenase (LDH) release was highly elevated and propidium iodide (PI)-positive cells were significantly increased at 2.5, 5, and 7.5 µM after all treatment times. Moreover, the protein levels of caspase-3 were up-regulated at 5 and 7.5 µM after 12 and 24 h of ALLN treatment. The ratio of Bax/Bcl-2 was raised and Annexin V-positive cells were increased at 5 and 7.5 µM after 12 and 24 h of ALLN treatment. However, there were no significant changes in either the ratio of microtubule-associated protein 1 light chain 3 (LC3) II/LC3 I or monodansylcadaverine (MDC) staining. Our data clearly show that at the concentrations equal to and higher than 2.5 µM, ALLN may induce cell death of primary retinal neurons by necrosis and apoptosis, but not autophagy. These suggest that primary retinal neurons are more susceptible to ALLN treatment and provide a possible mechanism for the cell death of ALLN-sensitive cells in ALLN injury.


Asunto(s)
Leupeptinas/toxicidad , Neuronas Retinianas/efectos de los fármacos , Animales , Apoptosis/efectos de los fármacos , Apoptosis/fisiología , Autofagia/efectos de los fármacos , Autofagia/fisiología , Western Blotting , Caspasa 3/metabolismo , Supervivencia Celular/efectos de los fármacos , Supervivencia Celular/fisiología , Células Cultivadas , Relación Dosis-Respuesta a Droga , Evaluación Preclínica de Medicamentos , Técnica del Anticuerpo Fluorescente , L-Lactato Deshidrogenasa/metabolismo , Proteínas Asociadas a Microtúbulos/metabolismo , Mitocondrias/efectos de los fármacos , Mitocondrias/metabolismo , Necrosis/inducido químicamente , Necrosis/metabolismo , Necrosis/patología , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , Ratas Sprague-Dawley , Neuronas Retinianas/metabolismo , Neuronas Retinianas/patología , Factores de Tiempo , Proteína X Asociada a bcl-2/metabolismo
7.
Hippocampus ; 26(8): 980-94, 2016 08.
Artículo en Inglés | MEDLINE | ID: mdl-26934478

RESUMEN

The dual-hit hypothesis of neurodegeneration states that severe stress sensitizes vulnerable cells to subsequent challenges so that the two hits are synergistic in their toxic effects. Although the hippocampus is vulnerable to a number of neurodegenerative disorders, there are no models of synergistic cell death in hippocampal neurons in response to combined proteotoxic and oxidative stressors, the two major characteristics of these diseases. Therefore, a relatively high-throughput dual-hit model of stress synergy was developed in primary hippocampal neurons. In order to increase the rigor of the study and strengthen the interpretations, three independent, unbiased viability assays were employed at multiple timepoints. Stress synergy was elicited when hippocampal neurons were treated with the proteasome inhibitor MG132 followed by exposure to the oxidative toxicant paraquat, but only after 48 h. MG132 and paraquat only elicited additive effects 24 h after the final hit and even loss of heat shock protein 70 activity and glutathione did not promote stress synergy at this early timepoint. Dual hits of MG132 elicited modest glutathione loss and slightly synergistic toxic effects 48 h after the second hit, but only at some concentrations and only according to two viability assays (metabolic fitness and cytoskeletal integrity). The thiol N-acetyl cysteine protected hippocampal neurons against dual MG132/MG132 hits but not dual MG132/paraquat hits. These findings support the view that proteotoxic and oxidative stress propel and propagate each other in hippocampal neurons, leading to synergistically toxic effects, but not as the default response and only after a delay. The neuronal stress synergy observed here lies in contrast to astrocytic responses to dual hits, because astrocytes that survive severe proteotoxic stress resist additional cell loss following second hits. In conclusion, a new model of hippocampal vulnerability was developed for the testing of therapies, because neuroprotective treatments that are effective against severe, synergistic stress are more likely to succeed in the clinic. © 2016 Wiley Periodicals, Inc.


Asunto(s)
Hipocampo/fisiopatología , Neuronas/fisiología , Estrés Fisiológico/fisiología , Acetilcisteína/farmacología , Animales , Supervivencia Celular/efectos de los fármacos , Células Cultivadas , Glutatión/metabolismo , Proteínas HSP70 de Choque Térmico/metabolismo , Hipocampo/efectos de los fármacos , Hipocampo/patología , Leupeptinas/toxicidad , Proteínas Asociadas a Microtúbulos/metabolismo , Modelos Neurológicos , Enfermedades Neurodegenerativas/tratamiento farmacológico , Enfermedades Neurodegenerativas/patología , Enfermedades Neurodegenerativas/fisiopatología , Neuronas/efectos de los fármacos , Neuronas/patología , Fármacos Neuroprotectores/farmacología , Estrés Oxidativo/fisiología , Paraquat/toxicidad , Complejo de la Endopetidasa Proteasomal/metabolismo , Ratas Sprague-Dawley , Estrés Fisiológico/efectos de los fármacos
8.
Toxicol In Vitro ; 29(8): 1965-76, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-26165741

RESUMEN

E-cadherin/ß-catenin complex plays an important role in maintaining the homeostasis of tissues and regulating cell proliferation, survival and apoptosis. To address the relationships between the change of E-cadherin/ß-catenin complex and cell apoptosis, human oral squamous carcinoma SCC-25 cells were used to investigate whether the dissociation of the E-cadherin/ß-catenin complex was the main reason of MG132- or bortezomib-induced apoptosis. We found that MG132 or bortezomib alone induced remarkable loss of cell integrity and contact, inhibited cell growth, survival, migration and caused cell cycle arrest, intracellular ROS production. Further experiments showed that colony formations were significantly decreased by MG132 and bortezomib alone or plus cis-diaminedichloroplatinum (CDDP). Immunofluorescence staining showed that SCC-25 cells exhibited remarkable accumulations of ß-catenin in cytoplasm and few E-cadherin in cell membranes after MG132 or bortezomib treatment. Western blot results showed that MG132 or bortezomib induced high accumulation of ubiquitinated proteins and activation of apoptosis related protein caspase-3. Meanwhile, the combinational use of MG132 or bortezomib with CDDP led to synergistic effects on SCC-25 cells. However, knockdown of ß-catenin could decrease MG132 or bortezomib induced cell death. Taken together, our data suggest that the regulation of E-cadherin/ß-catenin complex could be a promising therapeutic target to overcome the multidrug resistance of oral cancer.


Asunto(s)
Bortezomib/toxicidad , Cadherinas/metabolismo , Muerte Celular/efectos de los fármacos , Cisplatino/toxicidad , Leupeptinas/toxicidad , beta Catenina/metabolismo , Cadherinas/química , Adhesión Celular/efectos de los fármacos , Ciclo Celular/efectos de los fármacos , Línea Celular Tumoral , Movimiento Celular , Roturas del ADN/efectos de los fármacos , Resistencia a Antineoplásicos , Humanos , Neoplasias de la Boca , beta Catenina/química
9.
Neurobiol Aging ; 36(5): 1924-37, 2015 May.
Artículo en Inglés | MEDLINE | ID: mdl-25771395

RESUMEN

The telencephalic allocortex develops protein inclusions before the neocortex in many age-related proteinopathies. One major defense mechanism against proteinopathic stress is the heat shock protein (Hsp) network. We therefore contrasted Hsp defenses in stressed primary neocortical and allocortical cells. Neocortical neurons were more resistant to the proteasome inhibitor MG132 than neurons from 3 allocortical subregions: entorhinal cortex, piriform cortex, and hippocampus. However, allocortical neurons exhibited higher MG132-induced increases in Hsp70 and heat shock cognate 70 (Hsc70). MG132-treated allocortical neurons also exhibited greater levels of protein ubiquitination. Inhibition of Hsp70/Hsc70 activity synergistically exacerbated MG132 toxicity in allocortical neurons more than neocortical neurons, suggesting that the allocortex is more reliant on these Hsp defenses. In contrast, astrocytes harvested from the neocortex or allocortex did not differ in their response to Hsp70/Hsc70 inhibition. Consistent with the idea that chaperones are maximally engaged in allocortical neurons, an increase in Hsp70/Hsc70 activity was protective only in neocortical neurons. Finally, the levels of select Hsps were altered in the neocortex and allocortex in vivo with aging.


Asunto(s)
Corteza Cerebral/metabolismo , Proteínas del Choque Térmico HSC70/metabolismo , Proteínas HSP70 de Choque Térmico/metabolismo , Neocórtex/metabolismo , Neuronas/metabolismo , Telencéfalo , Envejecimiento/metabolismo , Animales , Células Cultivadas , Corteza Cerebral/citología , Femenino , Proteínas del Choque Térmico HSC70/antagonistas & inhibidores , Proteínas HSP70 de Choque Térmico/antagonistas & inhibidores , Leupeptinas/toxicidad , Chaperonas Moleculares , Neocórtex/citología , Inhibidores de Proteasoma/toxicidad , Ratas Sprague-Dawley , Ubiquitinación/efectos de los fármacos
10.
Cell Biol Int ; 38(10): 1221-6, 2014 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-24805111

RESUMEN

Glioblastoma multiforme (GBM) is the most common and lethal type of brain cancer. Proteasome inhibitors are emerging as a new class of anti-glioma agents; however, the mechanisms of their killing malignant cells are still unclear. We treated U87MG cells with the proteasome inhibitor MG132 and found that cell death correlated with caspase-8 activation and autophagy protein p62/SQSTM1.To explore the role of autophagy and p62/SQSTM1 in MG132-induced cancer cell death, we measured the alteration of MG132's cytotoxicity by autophagy inhibition, autophagy induction or variation of p62/SQSTM1 gene expression. Autophagy was activated upon MG132 treatment for short periods, while inhibition of autophagy aggravated MG132-induced cell death followed by high levels of p62/SQSTM1 and active caspase-8 (p18). Moreover, U87MG cell death was dependent on p62/SQSTM1, and its function required its C-terminus UBA domain to attenuate the MG132-induced cell death. The results suggest that p62/SQSTM1 is a potential contributor in determining the fate of U87MG cells deficient in proteolytic activity.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Apoptosis/efectos de los fármacos , Caspasa 8/metabolismo , Leupeptinas/toxicidad , Inhibidores de Proteasas/toxicidad , Proteínas Adaptadoras Transductoras de Señales/antagonistas & inhibidores , Proteínas Adaptadoras Transductoras de Señales/genética , Línea Celular Tumoral , Humanos , Interferencia de ARN , ARN Interferente Pequeño/metabolismo , Proteína Sequestosoma-1
11.
Folia Neuropathol ; 52(1): 41-55, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24729342

RESUMEN

The proteins' ubiquitination and their further degradation by proteasomes are crucial for cell cycle regulation, transcription and DNA replication, inflammatory response, and apoptosis. Proteasome inhibitors have recently become considered as a promising method in cancer and inflammatory disease therapy. In this study, utilizing the rat model, we try to establish the influence of proteasome inhibitor MG-132: (1) on the basis of spontaneous and evoked locomotor activity and (2) on the condition of nigrostriatal projections eight weeks after MG-132 intraperitoneal administration. We also discuss the current status of knowledge about intraperitoneal administration of MG-132, a laboratory method that is being used more and more. Our results revealed a lack of motor abnormalities, but significant loss (20%) of substantia nigra pars compacta dopaminergic neurons after systemic MG-132 administration. This loss was accompanied by a corresponding decrease (8%) of density of dopaminergic terminals in dorsolateral striatum. Moreover, evidence of very limited but ongoing fibre degeneration within the dorsal striatum suggests that MG-132 severely disturbed the nigrostriatal pathway. In summary, intraperitoneal application of proteasome inhibitor MG-132, despite the encouraging results of experimental treatment and prevention of many pathological processes, should be used with caution because of the potential adverse effects on the structure of the central nervous system, especially elements of the nigrostriatal pathway.


Asunto(s)
Leupeptinas/toxicidad , Degeneración Nerviosa/inducido químicamente , Inhibidores de Proteasoma/toxicidad , Sustancia Negra/efectos de los fármacos , Sustancia Negra/patología , Animales , Inmunohistoquímica , Inyecciones Intraperitoneales , Leupeptinas/administración & dosificación , Masculino , Actividad Motora/efectos de los fármacos , Inhibidores de Proteasoma/administración & dosificación , Ratas , Ratas Wistar
12.
J Periodontal Res ; 49(6): 801-9, 2014 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-24494651

RESUMEN

BACKGROUND: The domesticated legume, Canavalia gladiata (commonly called the sword bean), is known to contain canavanine. The fruit is used in Chinese and Japanese herbal medicine for treating the discharge of pus, but its pharmacological mechanisms are still unclear. OBJECTIVES: This study examined the effect of sword bean extract (SBE) on (i) oral bacteria and human oral epithelial cells in vitro, and (ii) the initiation and progression of experimental Porphyromonas gingivalis-induced alveolar bone resorption in rats. MATERIAL AND METHODS: A high-performance liquid chromatography/ultraviolet method was applied to quantitate canavanine in SBE. By assessing oral bacterial growth, we estimated the minimum inhibitory concentration and minimum bactericidal concentration of SBE, canavanine, chlorhexidine gluconate (CHX) solution. The cytotoxicity of SBE, canavanine, CHX, leupeptin and cystatin for KB cells was determined using a trypan blue assay. The effects of SBE, canavanine, leupeptin and cystatin on Arg-gingipain (Rgp) and Lys-gingipain (Kgp) were evaluated by colorimetric assay using synthetic substrates. To examine its effects on P. gingivalis-associated periodontal tissue breakdown, SBE was orally administered to P. gingivalis-infected rats. RESULT: Sword bean extract contained 6.4% canavanine. SBE and canavanine inhibited the growth of P. gingivalis and Fusobacterium nucleatum. The cytotoxicity of SBE, canavanine and cystatin on KB cells was significantly lower than that of CHX. Inhibition of Rgp with SBE was comparable to that with leupeptin, a known Rgp inhibitor, and inhibition of Kgp with SBE was significantly higher than that with leupeptin at 500 µg/mL ( p < 0.05). P. gingivalis-induced alveolar bone resorption was significantly suppressed by administration of SBE, with bone levels remaining comparable to non-infected animals ( p < 0.05). CONCLUSION: The present study suggests that SBE might be effective against P. gingivalis-associated alveolar bone resorption.


Asunto(s)
Pérdida de Hueso Alveolar/prevención & control , Infecciones por Bacteroidaceae/microbiología , Canavalia , Fitoterapia/métodos , Extractos Vegetales/uso terapéutico , Porphyromonas gingivalis/efectos de los fármacos , Adhesinas Bacterianas/efectos de los fármacos , Pérdida de Hueso Alveolar/microbiología , Animales , Canavalia/química , Canavanina/análisis , Canavanina/farmacología , Canavanina/toxicidad , Clorhexidina/análogos & derivados , Clorhexidina/farmacología , Clorhexidina/toxicidad , Cromatografía Líquida de Alta Presión , Cistatinas/farmacología , Cistatinas/toxicidad , Cisteína Endopeptidasas/efectos de los fármacos , Progresión de la Enfermedad , Células Epiteliales/efectos de los fármacos , Cisteína-Endopeptidasas Gingipaínas , Bacterias Gramnegativas/efectos de los fármacos , Bacterias Grampositivas/efectos de los fármacos , Humanos , Células KB , Leupeptinas/farmacología , Leupeptinas/toxicidad , Masculino , Pruebas de Sensibilidad Microbiana , Mucosa Bucal/citología , Mucosa Bucal/efectos de los fármacos , Extractos Vegetales/análisis , Ratas , Ratas Wistar , Organismos Libres de Patógenos Específicos
13.
Neuroscience ; 255: 19-32, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-24096134

RESUMEN

N-Acetyl cysteine, a glutathione precursor, has been shown to benefit patients with Alzheimer's disease and reduce the symptoms of traumatic brain injury in soldiers. Parkinson's and Alzheimer's disease are both characterized by stress from protein misfolding, or proteotoxicity. We have developed a high-throughput model of proteotoxicity by treating neuroblastoma N2a cells with the proteasome inhibitor MG132 and performing three independent assays for viability. Our previous study showed that N-acetyl cysteine protects N2a cells against two sequential treatments of MG132 and raises glutathione levels in a two-hit model of synergistic neurodegeneration. In the present study, however, N-acetyl cysteine was found to reduce the toxicity of a single hit of MG132 independent of its effect on glutathione. All three viability assays confirmed this protection. We measured heat shock protein 70 (Hsp70) levels because Hsp70 is a protective chaperone that helps refold proteins or guides ubiquitinated proteins toward degradation by the proteasome. Hsp70 levels were higher in MG132-treated cells when N-acetyl cysteine was applied. No parallel change in heat shock cognate 70 (Hsc70) was elicited. Inhibition of Hsp70/Hsc70 activity with VER 155008 attenuated the protection afforded by N-acetyl cysteine in a dose-responsive manner. MG132 induced a large rise in ubiquitinated proteins and N-acetyl cysteine reduced this effect. Consistent with the chaperone functions of Hsp70, VER 155008 also prevented the reduction in ubiquitin-conjugated proteins by N-acetyl cysteine. These data reveal a new role for N-acetyl cysteine: this compound may reduce misfolded protein levels and ameliorate proteotoxicity through heat shock proteins. These findings broaden the potential mechanisms of action for this dietary supplement in neurodegenerative proteinopathies.


Asunto(s)
Acetilcisteína/farmacología , Proteínas HSP70 de Choque Térmico/metabolismo , Neuronas/efectos de los fármacos , Fármacos Neuroprotectores/farmacología , Pliegue de Proteína/efectos de los fármacos , Animales , Western Blotting , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Inhibidores de Cisteína Proteinasa/toxicidad , Depuradores de Radicales Libres/farmacología , Ensayos Analíticos de Alto Rendimiento , Leupeptinas/toxicidad , Ratones , Neuronas/metabolismo , Neuronas/patología , Complejo de la Endopetidasa Proteasomal/efectos de los fármacos
14.
PLoS One ; 8(3): e58596, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23536801

RESUMEN

In Parkinson's and Alzheimer's diseases, the allocortex accumulates aggregated proteins such as synuclein and tau well before neocortex. We present a new high-throughput model of this topographic difference by microdissecting neocortex and allocortex from the postnatal rat and treating them in parallel fashion with toxins. Allocortical cultures were more vulnerable to low concentrations of the proteasome inhibitors MG132 and PSI but not the oxidative poison H2O2. The proteasome appeared to be more impaired in allocortex because MG132 raised ubiquitin-conjugated proteins and lowered proteasome activity in allocortex more than neocortex. Allocortex cultures were more vulnerable to MG132 despite greater MG132-induced rises in heat shock protein 70, heme oxygenase 1, and catalase. Proteasome subunits PA700 and PA28 were also higher in allocortex cultures, suggesting compensatory adaptations to greater proteasome impairment. Glutathione and ceruloplasmin were not robustly MG132-responsive and were basally higher in neocortical cultures. Notably, neocortex cultures became as vulnerable to MG132 as allocortex when glutathione synthesis or autophagic defenses were inhibited. Conversely, the glutathione precursor N-acetyl cysteine rendered allocortex resilient to MG132. Glutathione and ceruloplasmin levels were then examined in vivo as a function of age because aging is a natural model of proteasome inhibition and oxidative stress. Allocortical glutathione levels rose linearly with age but were similar to neocortex in whole tissue lysates. In contrast, ceruloplasmin levels were strikingly higher in neocortex at all ages and rose linearly until middle age. PA28 levels rose with age and were higher in allocortex in vivo, also paralleling in vitro data. These neo- and allocortical differences have implications for the many studies that treat the telencephalic mantle as a single unit. Our observations suggest that the topographic progression of protein aggregations through the cerebrum may reflect differential responses to low level protein-misfolding stress but also reveal impressive compensatory adaptations in allocortex.


Asunto(s)
Senescencia Celular , Corteza Cerebral/metabolismo , Neocórtex/metabolismo , Estrés Fisiológico , Animales , Autofagia , Supervivencia Celular/efectos de los fármacos , Corteza Cerebral/efectos de los fármacos , Femenino , Glutatión/metabolismo , Peróxido de Hidrógeno/toxicidad , Leupeptinas/toxicidad , Proteínas Asociadas a Microtúbulos/metabolismo , Neocórtex/efectos de los fármacos , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Complejo de la Endopetidasa Proteasomal/metabolismo , Ratas , Ubiquitina/metabolismo
15.
J Clin Invest ; 122(10): 3742-54, 2012 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-22945628

RESUMEN

Intrinsic stress response pathways are frequently mobilized within tumor cells. The mediators of these adaptive mechanisms and how they contribute to carcinogenesis remain poorly understood. A striking example is heat shock factor 1 (HSF1), master transcriptional regulator of the heat shock response. Surprisingly, we found that loss of the tumor suppressor gene neurofibromatosis type 1 (Nf1) increased HSF1 levels and triggered its activation in mouse embryonic fibroblasts. As a consequence, Nf1-/- cells acquired tolerance to proteotoxic stress. This activation of HSF1 depended on dysregulated MAPK signaling. HSF1, in turn, supported MAPK signaling. In mice, Hsf1 deficiency impeded NF1-associated carcinogenesis by attenuating oncogenic RAS/MAPK signaling. In cell lines from human malignant peripheral nerve sheath tumors (MPNSTs) driven by NF1 loss, HSF1 was overexpressed and activated, which was required for tumor cell viability. In surgical resections of human MPNSTs, HSF1 was overexpressed, translocated to the nucleus, and phosphorylated. These findings reveal a surprising biological consequence of NF1 deficiency: activation of HSF1 and ensuing addiction to this master regulator of the heat shock response. The loss of NF1 function engages an evolutionarily conserved cellular survival mechanism that ultimately impairs survival of the whole organism by facilitating carcinogenesis.


Asunto(s)
Transformación Celular Neoplásica/genética , Proteínas de Unión al ADN/fisiología , Genes de Neurofibromatosis 1 , Proteínas de Neoplasias/fisiología , Neurofibromina 1/deficiencia , Factores de Transcripción/fisiología , Transporte Activo de Núcleo Celular , Animales , Línea Celular Tumoral/metabolismo , Proteínas de Unión al ADN/biosíntesis , Proteínas de Unión al ADN/deficiencia , Proteínas de Unión al ADN/genética , Regulación Neoplásica de la Expresión Génica , Técnicas de Silenciamiento del Gen , Factores de Transcripción del Choque Térmico , Calor , Humanos , Leupeptinas/toxicidad , Sistema de Señalización de MAP Quinasas , Macrólidos/toxicidad , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Células 3T3 NIH , Proteínas de Neoplasias/biosíntesis , Proteínas de Neoplasias/deficiencia , Proteínas de Neoplasias/genética , Neoplasias de la Vaina del Nervio/patología , Neurofibromina 1/fisiología , Fosforilación , Procesamiento Proteico-Postraduccional , ARN Mensajero/biosíntesis , ARN Mensajero/genética , ARN Neoplásico/biosíntesis , ARN Neoplásico/genética , ARN Interferente Pequeño/farmacología , Factores de Transcripción/biosíntesis , Factores de Transcripción/deficiencia , Factores de Transcripción/genética , Witanólidos/toxicidad
16.
Brain Res Bull ; 88(6): 609-16, 2012 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-22664331

RESUMEN

Parkinson disease (PD) is the second most common neurodegenerative disease, and it cannot be completely cured by current medications. In this study, DJ-1 protein was administrated into medial forebrain bundle of PD model rats those had been microinjected with 6-hydroxydopamine (6-OHDA) or MG-132. We found that DJ-1 protein could reduce apomorphine-induced rotations, inhibit reduction of dopamine contents and tyrosine hydroxylase levels in the striatum, and decrease dopaminergic neuron death in the substantia nigra. In 6-OHDA lesioned rats, uncoupling protein-4, uncoupling protein-5 and superoxide dismutase-2 (SOD2) mRNA and SOD2 protein were increased when DJ-1 protein was co-injected. Simultaneously, administration of DJ-1 protein reduced α-synuclein and hypoxia-inducible factor 1α mRNA and α-synuclein protein in MG-132 lesioned rats. Therefore, DJ-1 protein protected dopaminergic neurons in two PD model rats by increasing antioxidant capacity and inhibiting α-synuclein expression.


Asunto(s)
Antiparkinsonianos/uso terapéutico , Neuronas Dopaminérgicas/efectos de los fármacos , Péptidos y Proteínas de Señalización Intracelular/uso terapéutico , Leupeptinas/toxicidad , Fármacos Neuroprotectores/uso terapéutico , Proteínas Oncogénicas/uso terapéutico , Oxidopamina/toxicidad , Trastornos Parkinsonianos/prevención & control , Animales , Antiparkinsonianos/administración & dosificación , Apomorfina/antagonistas & inhibidores , Cuerpo Estriado/efectos de los fármacos , Cuerpo Estriado/metabolismo , Cuerpo Estriado/patología , Dopamina/análisis , Neuronas Dopaminérgicas/enzimología , Evaluación Preclínica de Medicamentos , Humanos , Subunidad alfa del Factor 1 Inducible por Hipoxia/biosíntesis , Subunidad alfa del Factor 1 Inducible por Hipoxia/genética , Péptidos y Proteínas de Señalización Intracelular/administración & dosificación , Canales Iónicos/biosíntesis , Canales Iónicos/genética , Masculino , Microinyecciones , Proteínas de Transporte de Membrana Mitocondrial/biosíntesis , Proteínas de Transporte de Membrana Mitocondrial/genética , Proteínas Mitocondriales/biosíntesis , Proteínas Mitocondriales/genética , Proteínas Desacopladoras Mitocondriales , Actividad Motora/efectos de los fármacos , Proteínas del Tejido Nervioso/análisis , Proteínas del Tejido Nervioso/biosíntesis , Proteínas del Tejido Nervioso/genética , Fármacos Neuroprotectores/administración & dosificación , Proteínas Oncogénicas/administración & dosificación , Estrés Oxidativo/efectos de los fármacos , Trastornos Parkinsonianos/inducido químicamente , Trastornos Parkinsonianos/tratamiento farmacológico , Proteína Desglicasa DJ-1 , Ratas , Ratas Sprague-Dawley , Sustancia Negra/efectos de los fármacos , Sustancia Negra/metabolismo , Sustancia Negra/patología , Superóxido Dismutasa/biosíntesis , Superóxido Dismutasa/genética , Tirosina 3-Monooxigenasa/análisis , alfa-Sinucleína/biosíntesis , alfa-Sinucleína/genética
17.
Brain Res ; 1404: 21-30, 2011 Aug 02.
Artículo en Inglés | MEDLINE | ID: mdl-21737065

RESUMEN

Nitric oxide is linked with neurodegeneration in Parkinson's disease (PD) through the involvement of both inducible (iNOS) and neuronal nitric oxide synthase (nNOS). While non-selective NOS inhibitors are neuroprotective, the role of nNOS has not been determined using selective NOS inhibitors. The present study investigated the neuroprotective effect of selective iNOS and nNOS inhibitors on MPP(+)- and MG-132-induced cell death in cell lines with differing levels of nNOS expression. Inhibition of endogenously expressed nNOS by 7-NI and ARR17477 enhanced the toxicity of MPP(+) and MG-132 in N1E-115 cells, whereas in transfected SH-SY5Y cells overexpressing nNOS, ARR17477 and 7-NI protected against MPP(+)- and MG-132-induced cell death. In contrast, inhibition of iNOS by 1400W was ineffective in preventing MPP(+) and MG-132 toxicity in these cell lines. These results suggest a dual role for NOS in dopaminergic cell viability. nNOS is protective against toxic insult when produced endogenously. When nNOS is overexpressed, it becomes neurotoxic to cells suggesting that inhibition of nNOS may be a promising strategy to prevent cell death in PD.


Asunto(s)
Amidinas/farmacología , Dopamina/metabolismo , Inhibidores Enzimáticos/farmacología , Indazoles/farmacología , Óxido Nítrico Sintasa de Tipo I/metabolismo , Tiofenos/farmacología , 1-Metil-4-fenil-1,2,3,6-Tetrahidropiridina/farmacología , Animales , Asparagina/metabolismo , Caspasa 3/metabolismo , Muerte Celular/efectos de los fármacos , Línea Celular Tumoral , Citrulina/metabolismo , Proteínas de Transporte de Dopamina a través de la Membrana Plasmática/metabolismo , Relación Dosis-Respuesta a Droga , Humanos , L-Lactato Deshidrogenasa/metabolismo , Leupeptinas/toxicidad , Ratones , Neuroblastoma/patología , Neurotoxinas/farmacología , Óxido Nítrico Sintasa de Tipo I/genética , Transfección/métodos , Tritio/metabolismo , Tirosina 3-Monooxigenasa/metabolismo , Proteínas de Transporte Vesicular de Monoaminas/metabolismo
18.
Hum Exp Toxicol ; 30(12): 1945-54, 2011 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-21421692

RESUMEN

Carbobenzoxy-Leu-Leu-leucinal (MG132) as a proteasome inhibitor can induce growth inhibition and death in lung cancer or normal cells. However, little is known about relationship between proteasome inhibition and mitogen-activated protein kinase (MAPK) signaling in normal lung cells. Thus, in the present study, we investigated the effects of MAPK inhibitors on MG132-treated human pulmonary fibroblast (HPF) cells in relation to cell growth inhibition, cell death, reactive oxygen species (ROS) and glutathione (GSH). Treatment with 15 µM MG132 increased ROS levels including mitochondrial O(2•)(-) and GSH depleted cell numbers in HPF cells at 24 hours. MAP kinase or ERK kinase (MEK) inhibitor did not significantly affect cell growth inhibition, cell death, the loss of mitochondrial membrane potential (MMP; ΔΨ(m)), ROS level and GSH depletion in MG132-treated HPF cells. c-Jun N-terminal kinase (JNK) inhibitor attenuated the growth inhibition and death by MG132. This inhibitor also significantly decreased O(2•)(-) level in MG132-treated HPF cells. Although p38 inhibitor slightly enhanced HPF cell growth inhibition by MG132, this inhibitor and siRNA prevented HPF cell death induced by MG132. p38 inhibitor also attenuated d O(2•)(-) level and GSH depletion. Moreover, extracellular signal regulated kinase (ERK), JNK or p38 siRNA did not strongly affect ROS levels in MG132-treated HPF cells. ERK and JNK siRNAs decreased anonymous ubiquitinated protein levels in MG132-treated HPF cells. In conclusion, MAPK inhibitors differently affected the growth inhibition and death of MG132-treated HPF cells. Especially, p38 inhibitor attenuated HPF cell death by MG132, which was in part related to changes in ROS and GSH levels.


Asunto(s)
Inhibidores de Cisteína Proteinasa/toxicidad , Fibroblastos/efectos de los fármacos , Leupeptinas/toxicidad , Proteínas Quinasas Activadas por Mitógenos/antagonistas & inhibidores , Inhibidores de Proteasoma , Apoptosis/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Células Cultivadas , Antagonismo de Drogas , Fibroblastos/enzimología , Fibroblastos/patología , Glutatión/metabolismo , Humanos , Pulmón/citología , Potencial de la Membrana Mitocondrial/efectos de los fármacos , Mitocondrias/efectos de los fármacos , Mitocondrias/metabolismo , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Estrés Oxidativo , Oxígeno/metabolismo , Complejo de la Endopetidasa Proteasomal/metabolismo , Especies Reactivas de Oxígeno/metabolismo
19.
J Biomol Screen ; 16(3): 313-22, 2011 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-21310850

RESUMEN

Strict quality control of cells is required for the standardization and interpretation of results in all areas of cell-based research, especially in drug discovery. Real-time cellular analysis using electrical impedance as a readout offers a rapid and highly reproducible method for quality control as it provides a quantitative measure of overall cell morphology and growth. In a case study, the authors demonstrate that samples of a single cell line obtained from several different labs show clear differences in their impedance profiles when compared with the corresponding standard cell line. A number of kinetic parameters were derived from the impedance profiles and used to quantify the differences among these cell lines. Our findings indicate that this methodology can detect cell line differences including mix-ups or contaminations, genetic alterations, and potential epigenetic changes occurring during passaging, all of which can occur in the time scale of a screening campaign. Finally, we provide evidence that these impedance profile differences can be predictive of different outcomes in cell-based functional assays for the effects of small molecules on otherwise seemingly identical cell lines.


Asunto(s)
Bioensayo/métodos , Técnicas Citológicas/métodos , Antineoplásicos/toxicidad , Línea Celular Tumoral , Células/citología , Células/efectos de los fármacos , Células HeLa , Humanos , Leupeptinas/toxicidad , Repeticiones de Microsatélite/genética , Control de Calidad , Receptores Acoplados a Proteínas G/metabolismo , Transducción de Señal/efectos de los fármacos
20.
Biochem Pharmacol ; 81(3): 412-24, 2011 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-21040711

RESUMEN

Identification of novel target pathways in glioblastoma (GBM) remains critical due to poor prognosis, inefficient therapies and recurrence associated with these tumors. In this work, we evaluated the role of nuclear-factor-kappa-B (NFκB) in the growth of GBM cells, and the potential of NFκB inhibitors as antiglioma agents. NFκB pathway was found overstimulated in GBM cell lines and in tumor specimens compared to normal astrocytes and healthy brain tissues, respectively. Treatment of a panel of established GBM cell lines (U138MG, U87, U373 and C6) with pharmacological NFκB inhibitors (BAY117082, parthenolide, MG132, curcumin and arsenic trioxide) and NFκB-p65 siRNA markedly decreased the viability of GBMs as compared to inhibitors of other signaling pathways such as MAPKs (ERK, JNK and p38), PKC, EGFR and PI3K/Akt. In addition, NFκB inhibitors presented a low toxicity to normal astrocytes, indicating selectivity to cancerous cells. In GBMs, mitochondrial dysfunction (membrane depolarization, bcl-xL downregulation and cytochrome c release) and arrest in the G2/M phase were observed at the early steps of NFκB inhibitors treatment. These events preceded sub-G1 detection, apoptotic body formation and caspase-3 activation. Also, NFκB was found overstimulated in cisplatin-resistant C6 cells, and treatment of GBMs with NFκB inhibitors overcame cisplatin resistance besides potentiating the effects of the chemotherapeutics, cisplatin and doxorubicin. These findings support NFκB as a potential target to cell death induction in GBMs, and that the NFκB inhibitors may be considered for in vivo testing on animal models and possibly on GBM therapy.


Asunto(s)
Antineoplásicos/toxicidad , Neoplasias Encefálicas/tratamiento farmacológico , Glioblastoma/tratamiento farmacológico , FN-kappa B/antagonistas & inhibidores , Animales , Antineoplásicos/farmacología , Apoptosis/efectos de los fármacos , Trióxido de Arsénico , Arsenicales , Astrocitos/efectos de los fármacos , Astrocitos/metabolismo , Neoplasias Encefálicas/metabolismo , Neoplasias Encefálicas/patología , Ciclo Celular/efectos de los fármacos , Muerte Celular/efectos de los fármacos , Línea Celular Tumoral , Cisplatino/toxicidad , Curcumina/toxicidad , Doxorrubicina/toxicidad , Sinergismo Farmacológico , Glioblastoma/metabolismo , Glioblastoma/patología , Humanos , Leupeptinas/toxicidad , Terapia Molecular Dirigida , FN-kappa B/genética , FN-kappa B/metabolismo , Nitrilos/toxicidad , Óxidos/toxicidad , Ratas , Sesquiterpenos/toxicidad , Transducción de Señal/efectos de los fármacos , Sulfonas/toxicidad
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA