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











Base de datos
Intervalo de año de publicación
1.
World Neurosurg ; 161: e90-e100, 2022 05.
Artículo en Inglés | MEDLINE | ID: mdl-35077893

RESUMEN

OBJECTIVE: Patients who experience vertebral compression fractures are vulnerable to subsequent vertebral compression fractures (SVCFs). The purpose of this nationwide population-based study was to determine the age-specific cumulative incidence and factors associated with SVCFs in South Korea. METHODS: Diagnostic codes, medical costs, and comorbid diseases in patients who had a vertebral compression fracture in 2011 and 2012 were collected from the National Health Insurance Service database of South Korea from 2007 to 2018. Demographic data, mortality rate, medical cost, and frequency of vertebroplasty or kyphoplasty were compared between patients with an initial fracture (IF) and those with a subsequent fracture (SF). RESULTS: The cumulative incidence of SVCFs over 4 years was 24.4% and increased rapidly within a few months after the IF. In 2011, SVCFs occurred in 17,004 patients, and the incidence rate per 100,000 people was 113.6 (84.9 in men vs. 138.5 in women). The odds ratio (OR) of SVCFs in units of 10 years was the highest in women in their 60s, at 2.89. However, in men in their 70s, the OR was the highest, at 2.51. The rates of vertebroplasty or kyphoplasty, medical expenses, and mortality rate were significantly higher in the SF group than in the IF group (P < 0.01). CONCLUSIONS: The age-specific cumulative incidence of SVCFs per 100,000 people was 113.6. SVCFs were more frequent among women, the elderly, and patients who underwent vertebroplasty or kyphoplasty. Women in their 60s or above and men in their 70s or above were at highest risk.


Asunto(s)
Fracturas por Compresión , Cifoplastia , Fracturas Osteoporóticas , Fracturas de la Columna Vertebral , Vertebroplastia , Anciano , Femenino , Fracturas por Compresión/epidemiología , Fracturas por Compresión/etiología , Fracturas por Compresión/cirugía , Humanos , Incidencia , Cifoplastia/efectos adversos , Masculino , Fracturas Osteoporóticas/complicaciones , Fracturas de la Columna Vertebral/epidemiología , Fracturas de la Columna Vertebral/etiología , Fracturas de la Columna Vertebral/cirugía , Resultado del Tratamiento , Vertebroplastia/efectos adversos
2.
J Neurosci ; 40(2): 447-458, 2020 01 08.
Artículo en Inglés | MEDLINE | ID: mdl-31719166

RESUMEN

Cognitive impairments are key features in multiple sclerosis (MS), a progressive disorder characterized by neuroinflammation-induced demyelination in the central nervous system. To understand the neural substrates that link demyelination to cognitive deficits in MS, we investigated hippocampal neurogenesis and synaptic connectivity of adult-born neurons, which play an essential role in cognitive function. The administration and withdrawal of the combination of cuprizone and rapamycin (Cup/Rap) in C57BL/6J male mice efficiently demyelinated and remyelinated the hippocampus, respectively. In the demyelinated hippocampus, neurogenesis was nearly absent in the dentate gyrus, which was due to inhibited proliferation of neural stem cells (NSCs). Specifically, radial glia-like type 1 NSCs were shifted from a proliferative state to a mitotically-quiescent state in the demyelinated hippocampus. In addition, dendritic spine densities of adult-born neurons were significantly decreased, indicating a reduction in synaptic connections between hippocampal newborn neurons and excitatory input neurons. Concomitant with hippocampal remyelination induced by withdrawal of Cup/Rap, proliferation of type 1 NSCs and dendritic spine densities of adult-born neurons reverted to normal in the hippocampus. Our study shows that proliferation of hippocampal NSCs and synaptic connectivity of adult-born neurons are inversely correlated with the level of demyelination, providing critical insight into hippocampal neurogenesis as a potential therapeutic target to treat cognitive deficits associated with MS.SIGNIFICANCE STATEMENT To identify the neural substrates that mediate cognitive dysfunctions associated with a majority of MS patients, we investigated hippocampal neurogenesis and structural development of adult-born neurons using a Cup/Rap model, which recapitulates the hippocampal demyelination that occurs in MS patients. A shift of NSCs from a proliferatively-active state to mitotically-quiescent state dramatically decreased neurogenesis in the demyelinated hippocampus. Formation of dendritic spines on newborn neurons was also impaired following demyelination. Interestingly, the altered neurogenesis and synaptic connectivity of newborn neurons were reversed to normal levels during remyelination. Thus, our study revealed reversible genesis and synaptic connectivity of adult-born neurons between the demyelinated and remyelinated hippocampus, suggesting hippocampal neurogenesis as a potential target to normalize cognitive impairments in MS patients.


Asunto(s)
Disfunción Cognitiva/patología , Hipocampo/patología , Esclerosis Múltiple , Vías Nerviosas/patología , Neurogénesis/fisiología , Animales , Proliferación Celular/fisiología , Disfunción Cognitiva/etiología , Cuprizona/toxicidad , Modelos Animales de Enfermedad , Masculino , Ratones , Ratones Endogámicos C57BL , Inhibidores de la Monoaminooxidasa/toxicidad , Esclerosis Múltiple/inducido químicamente , Esclerosis Múltiple/complicaciones , Esclerosis Múltiple/patología , Células-Madre Neurales/patología , Neuronas/patología
3.
PLoS One ; 10(10): e0137703, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26444992

RESUMEN

Glioblastoma (GBM) is the most aggressive and most lethal brain tumor. As current standard therapy consisting of surgery and chemo-irradiation provides limited benefit for GBM patients, novel therapeutic options are urgently required. Forkhead box M1 (FoxM1) transcription factor is an oncogenic regulator that promotes the proliferation, survival, and treatment resistance of various human cancers. The roles of FoxM1 in GBM remain incompletely understood, due in part to pleotropic nature of the FoxM1 pathway. Here, we show the roles of FoxM1 in GBM stem cell maintenance and radioresistance. ShRNA-mediated FoxM1 inhibition significantly impeded clonogenic growth and survival of patient-derived primary GBM cells with marked downregulation of Sox2, a master regulator of stem cell phenotype. Ectopic expression of Sox2 partially rescued FoxM1 inhibition-mediated effects. Conversely, FoxM1 overexpression upregulated Sox2 expression and promoted clonogenic growth of GBM cells. These data, with a direct binding of FoxM1 in the Sox2 promoter region in GBM cells, suggest that FoxM1 regulates stemness of primary GBM cells via Sox2. We also found significant increases in FoxM1 and Sox2 expression in GBM cells after irradiation both in vitro and in vivo orthotopic tumor models. Notably, genetic or a small-molecule FoxM1 inhibitor-mediated FoxM1 targeting significantly sensitized GBM cells to irradiation, accompanying with Sox2 downregulation. Finally, FoxM1 inhibition combined with irradiation in a patient GBM-derived orthotopic model significantly impeded tumor growth and prolonged the survival of tumor bearing mice. Taken together, these results indicate that the FoxM1-Sox2 signaling axis promotes clonogenic growth and radiation resistance of GBM, and suggest that FoxM1 targeting combined with irradiation is a potentially effective therapeutic approach for GBM.


Asunto(s)
Neoplasias Encefálicas/patología , Resistencia a Antineoplásicos/genética , Factores de Transcripción Forkhead/genética , Glioblastoma/patología , Factores de Transcripción SOXB1/genética , Animales , Neoplasias Encefálicas/mortalidad , Neoplasias Encefálicas/terapia , Proteína Forkhead Box M1 , Factores de Transcripción Forkhead/antagonistas & inhibidores , Regulación Neoplásica de la Expresión Génica , Glioblastoma/mortalidad , Glioblastoma/terapia , Xenoinjertos , Humanos , Ratones , Ratones Endogámicos BALB C , Trasplante de Neoplasias , Células Madre Neoplásicas/citología , Regiones Promotoras Genéticas/genética , Interferencia de ARN , ARN Interferente Pequeño , Tolerancia a Radiación/genética , Factores de Transcripción SOXB1/biosíntesis , Transducción de Señal/genética
4.
Oncotarget ; 6(17): 15077-94, 2015 Jun 20.
Artículo en Inglés | MEDLINE | ID: mdl-25987130

RESUMEN

The most abundant populations of non-neoplastic cells in the glioblastoma (GBM) microenvironment are resident microglia, macrophages and infiltrating monocytes from the blood circulation. The mechanisms by which monocytes infiltrate into GBM, their fate following infiltration, and their role in GBM growth are not known. Here we tested the hypothesis that loss of the fractalkine receptor CX3CR1 in microglia and monocytes would affect gliomagenesis. Deletion of Cx3cr1 from the microenvironment resulted in increased tumor incidence and shorter survival times in glioma-bearing mice. Loss of Cx3cr1 did not affect accumulation of microglia/macrophages in peri-tumoral areas, but instead indirectly promoted the trafficking of CD11b+CD45hiCX3CR1lowLy-6ChiLy-6G-F4/80-/low circulating inflammatory monocytes into the CNS, resulting in their increased accumulation in the perivascular area. Cx3cr1-deficient microglia/macrophages and monocytes demonstrated upregulation of IL1ß expression that was inversely proportional to Cx3cr1 gene dosage. The Proneural subgroup of the TCGA GBM patient dataset with high IL1ß expression showed shorter survival compared to patients with low IL1ß. IL1ß promoted tumor growth and increased the cancer stem cell phenotype in murine and human Proneural glioma stem cells (GSCs). IL1ß activated the p38 MAPK signaling pathway and expression of monocyte chemoattractant protein (MCP-1/CCL2) by tumor cells. Loss of Cx3cr1 in microglia in a monocyte-free environment had no impact on tumor growth and did not alter microglial migration. These data suggest that enhancing signaling to CX3CR1 or inhibiting IL1ß signaling in intra-tumoral macrophages can be considered as potential strategies to decrease the tumor-promoting effects of monocytes in Proneural GBM.


Asunto(s)
Neoplasias Encefálicas/genética , Glioblastoma/genética , Microglía/metabolismo , Monocitos/metabolismo , Receptores de Quimiocina/genética , Animales , Neoplasias Encefálicas/metabolismo , Neoplasias Encefálicas/patología , Receptor 1 de Quimiocinas CX3C , Línea Celular , Femenino , Regulación Neoplásica de la Expresión Génica , Glioblastoma/metabolismo , Glioblastoma/patología , Humanos , Immunoblotting , Interleucina-1beta/genética , Interleucina-1beta/metabolismo , Interleucina-1beta/farmacología , Macrófagos/metabolismo , Macrófagos/patología , Masculino , Ratones Noqueados , Ratones Transgénicos , Microglía/patología , Microscopía Confocal , Monocitos/patología , Células Madre Neoplásicas/efectos de los fármacos , Células Madre Neoplásicas/metabolismo , Receptores de Quimiocina/metabolismo , Receptores Tipo I de Interleucina-1/genética , Receptores Tipo I de Interleucina-1/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Análisis de Supervivencia , Células Tumorales Cultivadas , Microambiente Tumoral/genética , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
5.
Oncotarget ; 6(24): 20145-59, 2015 Aug 21.
Artículo en Inglés | MEDLINE | ID: mdl-26023737

RESUMEN

Glioblastoma (GBM) is the most lethal brain cancer with profound genomic alterations. While the bona fide tumor suppressor genes such as PTEN, NF1, and TP53 have high frequency of inactivating mutations, there may be the genes with GBM-suppressive roles for which genomic mutation is not a primary cause for inactivation. To identify such genes, we employed in vivo RNAi screening approach using the patient-derived GBM xenograft models. We found that Nemo-Like Kinase (NLK) negatively regulates mesenchymal activities, a characteristic of aggressive GBM, in part via inhibition of WNT/ß-catenin signaling. Consistent with this, we found that NLK expression is especially low in a subset of GBMs that harbors high WNT/mesenchymal activities. Restoration of NLK inhibited WNT and mesenchymal activities, decreased clonogenic growth and survival, and impeded tumor growth in vivo. These data unravel a tumor suppressive role of NLK and support the feasibility of combining oncogenomics with in vivo RNAi screen.


Asunto(s)
Neoplasias Encefálicas/genética , Glioblastoma/genética , Proteínas Quinasas Activadas por Mitógenos/genética , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Animales , Neoplasias Encefálicas/enzimología , Neoplasias Encefálicas/patología , Proliferación Celular/genética , Femenino , Glioblastoma/enzimología , Glioblastoma/patología , Humanos , Péptidos y Proteínas de Señalización Intracelular/genética , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Proteínas Serina-Treonina Quinasas/genética , Proteínas Serina-Treonina Quinasas/metabolismo , Interferencia de ARN
6.
Thromb Haemost ; 111(5): 873-82, 2014 May 05.
Artículo en Inglés | MEDLINE | ID: mdl-24429998

RESUMEN

Cyclophilin A (CyPA) is an important mediator in cardiovascular diseases. It possesses peptidyl-prolyl cis-trans isomerase activity (PPIase) and chaperone functions, which regulate protein folding, intracellular trafficking and reactive oxygen species (ROS) production. Platelet glycoprotein receptor αIIbß3 integrin activation is the common pathway for platelet activation. It was our objective to understand the mechanism by which CyPA-regulates αIIbß3 activation in platelets. Mice deficient for CyPA (CyPA-/-) had prolonged tail bleeding time compared to wild-type (WT) controls despite equivalent platelet numbers. In vitro studies revealed that CyPA-/- platelets exhibited dramatically decreased thrombin-induced platelet aggregation. In vivo, formation of occlusive thrombi following FeCl3 injury was also significantly impaired in CyPA-/- mice compared with WT-controls. Furthermore, CyPA deficiency inhibited flow-induced thrombus formation in vitro. Flow cytometry demonstrated that thrombin-induced ROS production and αIIbß3 activation were reduced in CyPA-/- platelets. Coimmunoprecipitation studies showed ROS-dependent increased association of CyPA and αIIbß3. This association was dependent upon the PPIase activity of CyPA. Significantly, fibrinogen-platelet binding, platelet spreading and cytoskeleton reorganisation were also altered in CyPA-/- platelets. Moreover, CyPA deficiency prevented thrombin-induced αIIbß3 and cytoskeleton association. In conclusion, CyPA is an important mediator in platelet function by regulation of αIIbß3 bidirectionalsignalling through increased ROS production and facilitating interaction between αIIbß3 and the cell cytoskeleton.


Asunto(s)
Plaquetas/fisiología , Ciclofilina A/metabolismo , Arterias Mesentéricas/patología , Complejo GPIIb-IIIa de Glicoproteína Plaquetaria/metabolismo , Trombosis/sangre , Animales , Adhesión Celular/genética , Células Cultivadas , Cloruros/administración & dosificación , Ciclofilina A/genética , Citoesqueleto/genética , Compuestos Férricos/administración & dosificación , Fibrinógeno/metabolismo , Hemorragia/genética , Arterias Mesentéricas/efectos de los fármacos , Ratones Endogámicos C57BL , Ratones Noqueados , Agregación Plaquetaria , Especies Reactivas de Oxígeno/metabolismo , Transducción de Señal , Trombina/metabolismo , Trombosis/inducido químicamente
7.
Cardiovasc Res ; 101(3): 444-53, 2014 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-24293519

RESUMEN

AIMS: Cyclophilin A (CyPA) is a pro-inflammatory mediator involved in oxidative stress-related cardiovascular diseases. It is secreted from vascular smooth muscle cell (VSMC) in response to reactive oxygen species (ROS) in a highly regulated manner. Extracellular CyPA activates VSMCs and endothelial cells (ECs) promoting inflammation, cell growth, and cell death. Recently, it was shown that acetylated CyPA (AcK-CyPA) affects its function. We investigated the role of acetylation of CyPA for its secretion and signalling in vascular cells. METHODS AND RESULTS: We used angiotensin II (Ang II) to create sustained ROS and found significantly increased AcK-CyPA in VSMC. Site-directed mutagenesis showed that lysines K82 and K125 were the predominant CyPA residues acetylated in response to Ang II. Importantly, acetylation of K82 and K125 were required for Ang II-mediated CyPA secretion. ROS inhibitors, Tiron, and N-acetylcysteine inhibited Ang II-induced intracellular CyPA acetylation and also AcK-CyPA secretion. Using secreted CyPA from wild type and K82/125R mutants expressed in transduced VSMC or in vitro acetylated recombinant CyPA, we showed that extracellular AcK-CyPA significantly increased pERK1/2, matrix metalloproteinase-2 activation, and ROS production in VSMC compared with non-acetylated CyPA. Moreover, extracellular AcK-CyPA increased adhesion molecule expression (VCAM-1 and ICAM-1) in EC, which promoted monocyte adhesion. CONCLUSIONS: ROS-dependent acetylation of CyPA is required for the generation of extracellular CyPA. Acetylated extracellular CyPA regulates VSMC and EC activation, suggesting that inhibition of acetylation of CyPA may prevent the pathogenesis of oxidative stress-related cardiovascular diseases.


Asunto(s)
Ciclofilina A/farmacología , Músculo Liso Vascular/efectos de los fármacos , Acetilación , Angiotensina II/farmacología , Animales , Células Cultivadas , Ciclofilina A/química , Células Endoteliales/efectos de los fármacos , Células Endoteliales/metabolismo , Inflamación/tratamiento farmacológico , Inflamación/metabolismo , Molécula 1 de Adhesión Intercelular/metabolismo , Ratones , Músculo Liso Vascular/metabolismo , Miocitos del Músculo Liso/metabolismo , Ratas , Especies Reactivas de Oxígeno/metabolismo , Molécula 1 de Adhesión Celular Vascular/metabolismo
8.
Arterioscler Thromb Vasc Biol ; 33(9): 2147-53, 2013 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-23846495

RESUMEN

OBJECTIVE: Angiotensin II (AngII) signal transduction in vascular smooth muscle cells (VSMC) is mediated by reactive oxygen species (ROS). Cyclophilin A (CyPA) is a ubiquitously expressed cytosolic protein that possesses peptidyl-prolyl cis-trans isomerase activity, scaffold function, and significantly enhances AngII-induced ROS production in VSMC. We hypothesized that CyPA regulates AngII-induced ROS generation by promoting translocation of NADPH oxidase cytosolic subunit p47phox to caveolae of the plasma membrane. APPROACH AND RESULTS: Overexpression of CyPA in CyPA-deficient VSMC (CyPA(-/-)VSMC) significantly increased AngII-stimulated ROS production. Nicotinamide adenine dinucleotide phosphate (NADPH) oxidase inhibitors (VAS2870 or diphenylene iodonium) significantly attenuated AngII-induced ROS production in CyPA and p47phox-overexpressing CyPA(-/-)VSMC. Cell fractionation and sucrose gradient analyses showed that AngII-induced p47phox plasma membrane translocation, specifically to the caveolae, was reduced in CyPA(-/-)VSMC compared with wild-type-VSMC. Immunofluorescence studies demonstrated that AngII increased p47phox and CyPA colocalization and translocation to the plasma membrane. In addition, immunoprecipitation of CyPA followed by immunoblotting of p47phox and actin showed that AngII increased CyPA and p47phox interaction. AngII-induced p47phox and actin cell cytoskeleton association was attenuated in CyPA(-/-)VSMC. Mechanistically, inhibition of p47phox phosphorylation and phox homology domain deletion attenuated CyPA and p47phox interaction. Finally, cyclosporine A and CyPA-peptidyl-prolyl cis-trans isomerase mutant, R55A, inhibited AngII-stimulated CyPA and p47phox association in VSMC, suggesting that peptidyl-prolyl cis-trans isomerase activity was required for their interaction. CONCLUSIONS: These findings provide the mechanism by which CyPA is an important regulator for AngII-induced ROS generation in VSMC through interaction with p47phox and cell cytoskeleton, which enhances the translocation of p47phox to caveolae.


Asunto(s)
Angiotensina II/farmacología , Caveolas/efectos de los fármacos , Ciclofilina A/metabolismo , Músculo Liso Vascular/efectos de los fármacos , Miocitos del Músculo Liso/efectos de los fármacos , NADPH Oxidasas/metabolismo , Citoesqueleto de Actina/efectos de los fármacos , Citoesqueleto de Actina/enzimología , Animales , Western Blotting , Caveolas/enzimología , Ciclofilina A/genética , Relación Dosis-Respuesta a Droga , Inhibidores Enzimáticos/farmacología , Técnica del Anticuerpo Fluorescente , Células HeLa , Humanos , Inmunoprecipitación , Ratones , Ratones Noqueados , Ratones Transgénicos , Microscopía Fluorescente , Músculo Liso Vascular/enzimología , Mutación , Miocitos del Músculo Liso/enzimología , NADPH Oxidasas/antagonistas & inhibidores , NADPH Oxidasas/genética , Oligopéptidos , Péptidos/genética , Péptidos/metabolismo , Fosforilación , Unión Proteica , Dominios y Motivos de Interacción de Proteínas , Mapeo de Interacción de Proteínas , Transporte de Proteínas , Ratas , Especies Reactivas de Oxígeno/metabolismo , Receptor de Angiotensina Tipo 1/efectos de los fármacos , Receptor de Angiotensina Tipo 1/genética , Receptor de Angiotensina Tipo 1/metabolismo , Proteínas Recombinantes de Fusión/metabolismo , Factores de Tiempo
9.
Cancer Res ; 72(16): 4214-24, 2012 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-22719067

RESUMEN

Arsenic trioxide (As2O3) is used, in current practice, as an effective chemotherapeutic agent for acute promyelocytic leukemia (APL). However, the side effects and relatively low efficacy of As2O3 in treating other leukemias have limited its wider use in therapeutic applications. In the present study, we found that the expression of carbonyl reductase 1 (CBR1) affects the resistance to As2O3 in leukemias, including APL; As2O3 upregulated CBR1 expression at the transcriptional level by stimulating the activity of the transcription factor activator protein-1. Moreover, CBR1 overexpression was sufficient to protect cells against As2O3 through modulation of the generation of reactive oxygen species, whereas the attenuation of CBR1 was sufficient to sensitize cells to As2O3. A combination treatment with the specific CBR1 inhibitor hydroxy-PP-Me remarkably increased As2O3-induced apoptotic cell death compared with As2O3 alone, both in vitro and in vivo. These results were confirmed in primary cultured human acute and chronic myeloid leukemia cells, with no significant cell death observed in normal leukocytes. Taken together, our findings indicate that CBR1 contributes to the low efficacy of As2O3 and, therefore, is a rational target for the development of combination chemotherapy with As2O3 in diverse leukemias including APL.


Asunto(s)
Oxidorreductasas de Alcohol/antagonistas & inhibidores , Oxidorreductasas de Alcohol/metabolismo , Protocolos de Quimioterapia Combinada Antineoplásica/farmacología , Arsenicales/farmacología , Inhibidores Enzimáticos/farmacología , Leucemia Mieloide/tratamiento farmacológico , Leucemia Mieloide/enzimología , Óxidos/farmacología , Oxidorreductasas de Alcohol/biosíntesis , Oxidorreductasas de Alcohol/genética , Animales , Apoptosis/efectos de los fármacos , Trióxido de Arsénico , Arsenicales/administración & dosificación , Sinergismo Farmacológico , Activación Enzimática/efectos de los fármacos , Inhibidores Enzimáticos/administración & dosificación , Femenino , Técnicas de Silenciamiento del Gen , Células HL-60 , Humanos , Células K562 , Leucemia Mieloide/genética , Leucemia Mieloide/patología , Ratones , Ratones Endogámicos BALB C , NADPH Oxidasas/metabolismo , Óxidos/administración & dosificación , Regiones Promotoras Genéticas , Especies Reactivas de Oxígeno/metabolismo , Factor de Transcripción AP-1/metabolismo , Células U937 , Regulación hacia Arriba/efectos de los fármacos , Ensayos Antitumor por Modelo de Xenoinjerto
10.
Antioxid Redox Signal ; 16(3): 245-61, 2012 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-21902453

RESUMEN

AIM: Many studies have reported that the generation of reactive oxygen species (ROS) increases during the differentiation of muscle-derived C2C12 cells. Peroxiredoxin-2 (Prx-2) is an abundant mammalian enzyme that protects against oxidative stress. However, the role of Prx-2 in muscle differentiation has not been investigated. RESULTS: In this study, we demonstrated that Prx-2 expression increases during muscle differentiation and regeneration in response to exogenous H(2)O(2). This increase occurs only in myoblast cell lines because no increase in Prx-2 expression was observed in the NIH3T3, MEF, Chang, or HEK293 cell lines. The antioxidants, N-acetyl L-cysteine (NAC) and 4,5-dihydroxy-1,3-benzenedisulfonic acid (Tiron), both suppressed myogenesis and Prx-2 expression. Moreover, Prx-2 was upregulated at the transcriptional level by NF-κB during the differentiation of muscle-derived C2C12 cells. We also found that inhibition of phosphatidylinositol 3-kinase (PI3K) blocks NF-κB activation and suppresses Prx-2 expression. Interestingly, Prx-2 knockdown increased the expression levels of other antioxidant enzymes, including all of the other Prx family member, thioredoxin-1 (Trx-1) and catalase, but also enhanced the accumulation of endogenous ROS during muscle differentiation. INNOVATION: In this study, we demonstrated for the first time that Prx-2 is unregulated during the muscle differentiation and regeneration. CONCLUSION: Prx-2 is upregulated via the PI3K/NF-κB pathway and attenuates oxidative stress during muscle differentiation and regeneration.


Asunto(s)
Diferenciación Celular , Proteínas de Homeodominio/metabolismo , Músculo Esquelético/fisiología , FN-kappa B/metabolismo , Estrés Oxidativo , Regulación hacia Arriba , Acetilcisteína/farmacología , Animales , Antioxidantes/farmacología , Línea Celular , Femenino , Proteínas de Homeodominio/genética , Ratones , Ratones Endogámicos C57BL , Desarrollo de Músculos/efectos de los fármacos , Músculo Esquelético/citología , Fosfatidilinositol 3-Quinasas/metabolismo , Regiones Promotoras Genéticas , Unión Proteica , Especies Reactivas de Oxígeno/metabolismo , Regeneración , Células Satélite del Músculo Esquelético/metabolismo , Células Satélite del Músculo Esquelético/fisiología , Transcripción Genética
11.
Cell Physiol Biochem ; 28(5): 873-86, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-22178940

RESUMEN

Mitochondrial oxidative damage is thought to play a key role in pancreatic ß-cell failure in the pathogenesis of type 2 diabetes. Despite this, the potential of mitochondria-targeted antioxidants to protect pancreatic ß-cells against oxidative stress has not yet been studied. Therefore, we investigated if mitochondria-targeted antioxidants protect pancreatic ß-cells such as RINm5F and HIT-T15 cells against oxidative stress under glucotoxic and glucolipotoxic conditions. When ß-cells were incubated under these conditions, the expression levels of mitochondrial electron transport chain complex subunits, mitochondrial antioxidant enzymes (such as MnSOD and Prx3), ß-cell apoptosis, lipogenic enzymes (such as ACC, FAS and ABCA1), intracellular lipid accumulation, oxidative stress, ER stress, mitochondrial membrane depolarization, nuclear NF- κB and sterol regulatory element binding protein 1c (SREBP1c) were all increased, in parallel with decreases in intracellular ATP content, citrate synthase enzymatic activity and glucose-stimulated insulin secretion. These changes were consistent with elevated mitochondrial oxidative stress, and incubation with the mitochondria-targeted antioxidants, MitoTempol or Mitoquinone (MitoQ), prevented these effects. In conclusion, mitochondria-targeted antioxidants protect pancreatic ß-cells against oxidative stress, promote their survival, and increase insulin secretion in cell models of the glucotoxicity and glucolipotoxicity associated with Type 2 diabetes.


Asunto(s)
Antioxidantes/farmacología , Glucosa/toxicidad , Células Secretoras de Insulina/efectos de los fármacos , Insulina/metabolismo , Mitocondrias/efectos de los fármacos , Estrés Oxidativo/efectos de los fármacos , Animales , Línea Celular , Cricetinae , Proteínas de Homeodominio/metabolismo , Secreción de Insulina , Mitocondrias/metabolismo , Membranas Mitocondriales/efectos de los fármacos , FN-kappa B/metabolismo , Compuestos Organofosforados/farmacología , Ratas , Proteína 1 de Unión a los Elementos Reguladores de Esteroles/metabolismo , Superóxido Dismutasa/metabolismo , Ubiquinona/farmacología
12.
Hepatology ; 54(5): 1661-78, 2011 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-21748762

RESUMEN

UNLABELLED: Cyclophilin B (CypB) performs diverse roles in living cells, but its role in hepatocellular carcinoma (HCC) is largely unclear. To reveal its role in HCC, we investigated the induction of CypB under hypoxia and its functions in tumor cells in vitro and in vivo. Here, we demonstrated that hypoxia-inducible factor 1α (HIF-1α) induces CypB under hypoxia. Interestingly, CypB protected tumor cells, even p53-defective HCC cells, against hypoxia- and cisplatin-induced apoptosis. Furthermore, it regulated the effects of HIF-1α, including those in angiogenesis and glucose metabolism, via a positive feedback loop with HIF-1α. The tumorigenic and chemoresistant effects of CypB were confirmed in vivo using a xenograft model. Finally, we showed that CypB is overexpressed in 78% and 91% of the human HCC and colon cancer tissues, respectively, and its overexpression in these cancers reduced patient survival. CONCLUSIONS: These results indicate that CypB induced by hypoxia stimulates the survival of HCC via a positive feedback loop with HIF-1α, indicating that CypB is a novel candidate target for developing chemotherapeutic agents against HCC and colon cancer.


Asunto(s)
Carcinoma Hepatocelular/tratamiento farmacológico , Carcinoma Hepatocelular/patología , Cisplatino/farmacología , Ciclofilinas/genética , Neoplasias Hepáticas/tratamiento farmacológico , Neoplasias Hepáticas/patología , Animales , Antineoplásicos/farmacología , Apoptosis/efectos de los fármacos , Carcinoma Hepatocelular/metabolismo , Neoplasias del Colon/metabolismo , Neoplasias del Colon/patología , Ciclofilinas/metabolismo , Resistencia a Antineoplásicos/fisiología , Femenino , Células Hep G2 , Humanos , Peróxido de Hidrógeno/farmacología , Hipoxia/patología , Subunidad alfa del Factor 1 Inducible por Hipoxia/genética , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Neoplasias Hepáticas/metabolismo , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Neovascularización Patológica/metabolismo , Neovascularización Patológica/patología , Oxidantes/farmacología , Regiones Promotoras Genéticas/fisiología , Factor de Transcripción STAT3/metabolismo , Factor A de Crecimiento Endotelial Vascular/metabolismo , Ensayos Antitumor por Modelo de Xenoinjerto
13.
Br J Nutr ; 106(10): 1544-51, 2011 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-21676280

RESUMEN

It has been reported that high-fat, high-carbohydrate (HFHC) meals increase oxidative stress and inflammation. We examined whether repeated intake of excess energy in the form of HFHC meals alters reactive oxygen species (ROS) generation and the expression levels of antioxidant enzymes and mitochondrial proteins in mononuclear cells, and to determine whether this is associated with insulin resistance. We recruited healthy lean individuals (n 10). The individuals were divided into two groups: one group (n 5) ingested 10878·4 kJ/d (2600 kcal/d; 55-70 % carbohydrate, 9·5-16 % fat, 7-20 % protein) recommended by the Dietary Reference Intake for Koreans for 4 d and the other group (n 5) ingested a HFHC meal containing 14 644 kJ/d (3500 kcal/d). Then, measurements of blood insulin and glucose levels, together with suppressor of cytokine signalling-3 (SOCS-3) expression levels, were performed in both groups. Also, cellular and mitochondrial ROS levels as well as malondialdehyde (MDA) levels were measured. Expression levels of cytosolic and mitochondrial antioxidant enzymes, and mitochondrial complex proteins were analysed. Repeated intake of HFHC meals induced an increase in homeostasis model of assessment-insulin resistance (HOMA-IR), together with an increase in SOCS-3 expression levels. While a single intake of the HFHC meal increased cytosolic and mitochondrial ROS, repeated intake of HFHC meals reduced them and increased the levels of MDA, cytosolic and mitochondrial antioxidant enzymes, and several mitochondrial complex proteins. Repeated intake of HFHC meals induced cellular antioxidant mechanisms, which in turn increased lipid peroxidation (MDA) and SOCS-3 expression levels, induced hyperinsulinaemia and increased HOMA-IR, an index of insulin resistance. In conclusion, excess energy added to a diet can generate detrimental effects in a short period.


Asunto(s)
Antioxidantes/metabolismo , Carbohidratos de la Dieta/administración & dosificación , Grasas de la Dieta/administración & dosificación , Estrés Oxidativo , Adulto , Secuencia de Bases , Western Blotting , Composición Corporal , Cartilla de ADN , Prueba de Tolerancia a la Glucosa , Humanos , Masculino , Malondialdehído/metabolismo , Reacción en Cadena de la Polimerasa , Especies Reactivas de Oxígeno/metabolismo , Proteína 3 Supresora de la Señalización de Citocinas , Proteínas Supresoras de la Señalización de Citocinas/metabolismo
14.
Free Radic Biol Med ; 51(4): 905-20, 2011 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-21683784

RESUMEN

Accumulated amyloid-ß (Aß) is a well-known cause of neuronal apoptosis in Alzheimer disease and functions in part by generating oxidative stress. Our previous work suggested that cyclophilin B (CypB) protects against endoplasmic reticulum (ER) stress. Therefore, in this study we examined the ability of CypB to protect against Aß toxicity. CypB is present in the neurons of rat and mouse brains, and treating neural cells with Aß(25-35) mediates apoptotic cell death. Aß(25-35)-induced neuronal toxicity was inhibited by the overexpression of CypB as measured by cell viability, apoptotic morphology, sub-G1 cell population, intracellular reactive oxygen species accumulation, activated caspase-3, PARP cleavage, Bcl-2 proteins, mitogen-activated protein kinase (MAPK) activation, and phosphoinositide 3-kinase (PI-3-K) activation. CypB/R95A PPIase mutants did not reduce Aß(25-35) toxicity. We showed that Aß(25-35)-induced apoptosis is more severe in a CypB knockdown model, confirming that CypB protects against Aß(25-35)-induced toxicity. Consequently, these findings suggest that CypB may protect against Aß toxicity by its antioxidant properties, by regulating MAPK and PI-3-K signaling, and through the ER stress pathway.


Asunto(s)
Enfermedad de Alzheimer/metabolismo , Ciclofilinas/metabolismo , Neuronas/efectos de los fármacos , Enfermedad de Alzheimer/tratamiento farmacológico , Enfermedad de Alzheimer/patología , Péptidos beta-Amiloides/farmacología , Animales , Apoptosis/efectos de los fármacos , Apoptosis/genética , Caspasa 3/metabolismo , Línea Celular Tumoral , Ciclofilinas/genética , Citoprotección/genética , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Humanos , Mutación/genética , Neuronas/metabolismo , Neuronas/patología , Fosfatidilinositol 3-Quinasas/metabolismo , Poli(ADP-Ribosa) Polimerasa-1 , Poli(ADP-Ribosa) Polimerasas/metabolismo , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , Ratas , Especies Reactivas de Oxígeno/metabolismo , Transgenes/genética
15.
J Cell Sci ; 121(Pt 21): 3636-48, 2008 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-18946027

RESUMEN

Prolonged accumulation of misfolded proteins in the endoplasmic reticulum (ER) results in ER stress-mediated apoptosis. Cyclophilins are protein chaperones that accelerate the rate of protein folding through their peptidyl-prolyl cis-trans isomerase (PPIase) activity. In this study, we demonstrated that ER stress activates the expression of the ER-localized cyclophilin B (CypB) gene through a novel ER stress response element. Overexpression of wild-type CypB attenuated ER stress-induced cell death, whereas overexpression of an isomerase activity-defective mutant, CypB/R62A, not only increased Ca(2+) leakage from the ER and ROS generation, but also decreased mitochondrial membrane potential, resulting in cell death following exposure to ER stress-inducing agents. siRNA-mediated inhibition of CypB expression rendered cells more vulnerable to ER stress. Finally, CypB interacted with the ER stress-related chaperones, Bip and Grp94. Taken together, we concluded that CypB performs a crucial function in protecting cells against ER stress via its PPIase activity.


Asunto(s)
Apoptosis , Calcio/metabolismo , Ciclofilinas/fisiología , Retículo Endoplásmico/metabolismo , Especies Reactivas de Oxígeno , Transcripción Genética , Animales , Secuencia de Bases , Ciclofilinas/biosíntesis , Ciclofilinas/metabolismo , Regulación de la Expresión Génica , Homeostasis , Potenciales de la Membrana , Mitocondrias/metabolismo , Modelos Biológicos , Datos de Secuencia Molecular , Ratas
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA