Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 44
Filtrar
Más filtros

Banco de datos
Tipo del documento
País de afiliación
Intervalo de año de publicación
1.
Molecules ; 25(6)2020 Mar 23.
Artículo en Inglés | MEDLINE | ID: mdl-32210117

RESUMEN

Temozolomide (TMZ)-induced chemoresistance to human glioblastomas is a critical challenge now. Our previous studies showed that honokiol, a major bioactive constituent of Magnolia officinalis (Houpo), can kill human glioblastoma cells and suppresses glioblastoma growth. This study was further aimed to evaluate the effects of honokiol on human drug-resistant glioblastoma cells and the possible mechanisms. The results by data mining in the cancer genome atlas (TCGA) database and immunohistochemistry displayed that expression of caspase-9 mRNA and protein in human glioblastomas was induced. Human TMZ-resistant U87-MG-R9 glioblastoma cells were selected and prepared from human drug-sensitive U87-MG cells. Compared to human drug-sensitive U87-MG cells, TMZ did not affect viability of U87-MG-R9 glioblastoma cells. Interestingly, treatment with honokiol suppressed proliferation and survival of human drug-resistant glioblastoma cells in concentration- and time-dependent manners. Compared to caspase-8 activation, honokiol chiefly increased activity of caspase-9 in U87-MG-R9 cells. Successively, levels of cleaved caspase-3 and activities of caspase-3 and caspase-6 in human TMZ-tolerant glioblastoma cells were augmented following honokiol administration. In parallel, honokiol triggered DNA fragmentation of U87-MG-R9 cells. Accordingly, treatment of human TMZ-resistant glioblastoma cells with honokiol induced cell apoptosis but did not affect cell necrosis. Fascinatingly, suppressing caspase-9 activity using its specific inhibitors repressed honokiol-induced caspase-6 activation, DNA fragmentation, and cell apoptosis. Taken together, this study has shown the major roles of caspase-9 in transducing honokiol-induced mitochondria-dependent apoptosis in human drug-resistant glioblastoma cells. Thus, honokiol may be clinically applied as a drug candidate for treatment of glioblastoma patients with chemoresistance.


Asunto(s)
Apoptosis/efectos de los fármacos , Compuestos de Bifenilo/farmacología , Caspasa 9/metabolismo , Resistencia a Antineoplásicos/efectos de los fármacos , Glioblastoma , Lignanos/farmacología , Proteínas de Neoplasias/metabolismo , Línea Celular Tumoral , Activación Enzimática/efectos de los fármacos , Glioblastoma/tratamiento farmacológico , Glioblastoma/enzimología , Glioblastoma/patología , Humanos , Mitocondrias/metabolismo , Mitocondrias/patología , Temozolomida/farmacología
2.
J Cell Biochem ; 118(9): 2635-2644, 2017 09.
Artículo en Inglés | MEDLINE | ID: mdl-27987330

RESUMEN

Dexmedetomidine, an agonist of alpha2-adrenergic receptors, is used for critically ill patients to induce and maintain sedation and analgesia. Brain ischemia/reperfusion (I/R) usually causes severe neuronal injuries to intensive care unit patients. This study was aimed to evaluate the effects of dexmedetomidine on I/R-induced insults to neuronal cells and the possible mechanisms. Treatment of neuro-2a cells with dexmedetomidine did not affect cell viability but could protect against I/R-induced cell death. Separately, the I/R-triggered cell shrinkage, DNA fragmentation, and apoptosis in neuro-2a cells were alleviated by dexmedetomidine. As to the mechanisms, exposure of neuro-2a cells to dexmedetomidine substantially attenuated I/R-induced translocation of Bax protein from the cytosol to mitochondria and reduction in the mitochondrial membrane potential (MMP). Successively, dexmedetomidine decreased cytochrome c release from mitochondria to the cytoplasm and consequent cascade activations of caspases-9, -3, and -6 in I/R-treated neuro-2a cells. Interestingly, downregulating caspase-6 activity synergistically improved dexmedetomidine-induced defense against I/R-induced apoptosis of neuro-2a cells. The dexmedetomidine-involved neuroprotection was further confirmed in the other NB41A3 neuronal cells by significantly attenuating I/R-induced changes in the MMP, caspase-3 activation, DNA fragmentation, and cell apoptosis. Taken together, this study has shown the neuroprotective effects of dexmedetomidine against I/R-induced apoptotic insults via an intrinsic Bax-mitochondria-cytochrome c-caspase protease pathway. J. Cell. Biochem. 118: 2635-2644, 2017. © 2016 Wiley Periodicals, Inc.


Asunto(s)
Apoptosis/efectos de los fármacos , Dexmedetomidina/farmacología , Mitocondrias/metabolismo , Proteínas Mitocondriales/metabolismo , Neuronas/metabolismo , Daño por Reperfusión/prevención & control , Caspasas/metabolismo , Línea Celular Tumoral , Fragmentación del ADN/efectos de los fármacos , Humanos , Mitocondrias/patología , Neuronas/patología , Daño por Reperfusión/metabolismo , Daño por Reperfusión/patología
3.
Toxicol Appl Pharmacol ; 304: 59-69, 2016 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-27236003

RESUMEN

Honokiol, an active constituent extracted from the bark of Magnolia officinalis, possesses anticancer effects. Apoptosis is classified as type I programmed cell death, while autophagy is type II programmed cell death. We previously proved that honokiol induces cell cycle arrest and apoptosis of U87 MG glioma cells. Subsequently in this study, we evaluated the effect of honokiol on autophagy of glioma cells and examined the molecular mechanisms. Administration of honokiol to mice with an intracranial glioma increased expressions of cleaved caspase 3 and light chain 3 (LC3)-II. Exposure of U87 MG cells to honokiol also induced autophagy in concentration- and time-dependent manners. Results from the addition of 3-methyladenine, an autophagy inhibitor, and rapamycin, an autophagy inducer confirmed that honokiol-induced autophagy contributed to cell death. Honokiol decreased protein levels of PI3K, phosphorylated (p)-Akt, and p-mammalian target of rapamycin (mTOR) in vitro and in vivo. Pretreatment with a p53 inhibitor or transfection with p53 small interfering (si)RNA suppressed honokiol-induced autophagy by reversing downregulation of p-Akt and p-mTOR expressions. In addition, honokiol caused generation of reactive oxygen species (ROS), which was suppressed by the antioxidant, vitamin C. Vitamin C also inhibited honokiol-induced autophagic and apoptotic cell death. Concurrently, honokiol-induced alterations in levels of p-p53, p53, p-Akt, and p-mTOR were attenuated following vitamin C administration. Taken together, our data indicated that honokiol induced ROS-mediated autophagic cell death through regulating the p53/PI3K/Akt/mTOR signaling pathway.


Asunto(s)
Autofagia/efectos de los fármacos , Compuestos de Bifenilo/toxicidad , Glioma/tratamiento farmacológico , Lignanos/toxicidad , Fosfatidilinositol 3-Quinasas/biosíntesis , Proteínas Proto-Oncogénicas c-akt/biosíntesis , Serina-Treonina Quinasas TOR/biosíntesis , Proteína p53 Supresora de Tumor/biosíntesis , Adenina/análogos & derivados , Adenina/farmacología , Animales , Apoptosis/efectos de los fármacos , Ácido Ascórbico/farmacología , Caspasa 3/biosíntesis , Caspasa 3/efectos de los fármacos , Línea Celular Tumoral , Relación Dosis-Respuesta a Droga , Regulación hacia Abajo , Ratones , Proteínas Asociadas a Microtúbulos/biosíntesis , Proteínas Asociadas a Microtúbulos/efectos de los fármacos , Fosfatidilinositol 3-Quinasas/efectos de los fármacos , Proteínas Proto-Oncogénicas c-akt/efectos de los fármacos , ARN Interferente Pequeño/biosíntesis , Especies Reactivas de Oxígeno/metabolismo , Transducción de Señal , Sirolimus/farmacología , Serina-Treonina Quinasas TOR/efectos de los fármacos , Factores de Tiempo , Proteína p53 Supresora de Tumor/efectos de los fármacos
4.
Arch Toxicol ; 90(4): 905-16, 2016 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-25726415

RESUMEN

Nitric oxide (NO) has biphasic effects on regulating osteoblast survival and death. This study was aimed to evaluate the effects of NO pretreatment on hydrogen peroxide (HP)-induced insults of rat osteoblasts and the possible mechanisms. Exposure of osteoblasts prepared from rat calvarias to HP significantly increased intracellular reactive oxygen species levels, decreased alkaline phosphatase activity and cell survival, and ultimately induced cell apoptosis. However, NO pretreatment lowered HP-induced oxidative stress and apoptotic insults. In parallel, HP increased Bax levels and its translocation from the cytoplasm to mitochondria. NO pretreatment caused significant attenuations in HP-induced modulations in Bax synthesis and translocation. In contrast, pretreatment with NO enhanced levels and translocation of antiapoptotic Bcl-XL protein in rat osteoblasts. RNA analyses further revealed that HP inhibited Bcl-XL mRNA expression without affecting Bax mRNA levels. In comparison, NO induced Bcl-XL mRNA production and alleviated HP-caused inhibition of this mRNA expression. As to the mechanism, HP suppressed RNA and protein levels of transcription factor GATA-5 in rat osteoblasts. Pretreatment with NO induced GATA-5 mRNA and protein expressions and simultaneously attenuated HP-induced inhibition of this gene's expression. Consequently, GATA-5 knockdown using RNA interference inhibited Bcl-XL mRNA expression and concurrently lowered NO's protection against HP-induced apoptotic insults. Therefore, this study showed that NO can protect rat osteoblasts from HP-induced apoptotic insults. The protective mechanisms are mediated by GATA-5-mediated transcriptional induction of Bcl-X L gene, and translocational modulation of Bcl-XL and Bax proteins.


Asunto(s)
Factor de Transcripción GATA5/metabolismo , Óxido Nítrico/farmacología , Osteoblastos/efectos de los fármacos , Estrés Oxidativo/fisiología , Proteína bcl-X/genética , Animales , Apoptosis/efectos de los fármacos , Células Cultivadas , Factor de Transcripción GATA5/genética , Expresión Génica/efectos de los fármacos , Peróxido de Hidrógeno/toxicidad , Óxido Nítrico/metabolismo , Osteoblastos/patología , Osteoblastos/fisiología , Estrés Oxidativo/efectos de los fármacos , Transporte de Proteínas/efectos de los fármacos , Ratas Wistar , Proteína X Asociada a bcl-2/genética , Proteína X Asociada a bcl-2/metabolismo , Proteína bcl-X/metabolismo
5.
J Formos Med Assoc ; 112(10): 635-43, 2013 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-24120154

RESUMEN

BACKGROUND/PURPOSE: MicroRNA-208a (miR208a) and mechanical stress play a key role in cardiac hypertrophy. The relationship between miR208a and mechanical stress in cultured cardiomyocytes has not been investigated. The molecular mechanisms underlying miR208a-induced hypertrophy of cardiomyocytes by mechanical stress is poorly understood. This study investigated whether miR208a is a critical regulator in cardiomyocyte hypertrophy under mechanical stretch. METHODS: Neonatal rat cardiomyocytes grown on a flexible membrane base were stretched at 60 cycles/minute. MiR real-time quantitative assays were used to quantify miRs. A quantitative sandwich enzyme immunoassay technique was used to measure transforming growth factor-ß1 (TGF-ß1). A (3)H-proline incorporation assay was used to measure protein synthesis. RESULTS: Mechanical stretch significantly enhanced miR208a expression. Stretch significantly induced cardiomyocyte hypertrophic protein expression such as ß-myosin heavy chain (MHCß), thyroid hormone receptor-associated protein 100, myostatin, connexin 40, GATA4, and brain natriuretic peptide. MHCα was not induced by stretch. Overexpression of miR208a significantly increased MHCß protein expression while pretreatment with antagomir208a significantly attenuated MHCß protein expression induced by stretch and overexpression of miR208a. Mechanical stretch significantly increased the secretion of TGF-ß1 from cultured cardiomyocytes. Exogenous addition of TGF-ß1 recombinant protein significantly increased miR208a expression and pretreatment with TGF-ß1 antibody attenuated miR208a expression induced by stretch. Mechanical stretch and overexpression of miR208a increased protein synthesis while antagomir208a attenuated protein synthesis induced by stretch and overexpression of miR208a. CONCLUSION: Cyclic stretch enhances miR208a expression in cultured rat cardiomyocytes. MiR208a plays a role in stretch-induced cardiac hypertrophy. The stretch-induced miR208a is mediated by TGF-ß1.


Asunto(s)
MicroARNs/metabolismo , Miocitos Cardíacos/metabolismo , Miocitos Cardíacos/patología , Estrés Mecánico , Factor de Crecimiento Transformador beta1/metabolismo , Animales , Células Cultivadas , Conexinas/biosíntesis , Factor de Transcripción GATA4/biosíntesis , Hipertrofia , Complejo Mediador/biosíntesis , MicroARNs/antagonistas & inhibidores , Cadenas Pesadas de Miosina/biosíntesis , Miostatina/biosíntesis , Péptido Natriurético Encefálico/biosíntesis , Biosíntesis de Proteínas/efectos de los fármacos , Ratas , Factor de Crecimiento Transformador beta1/farmacología , Proteína alfa-5 de Unión Comunicante
6.
J Biomed Sci ; 19: 72, 2012 Aug 03.
Artículo en Inglés | MEDLINE | ID: mdl-22862895

RESUMEN

BACKGROUND: PUMA (p53-up-regulated modulator of apoptosis), an apoptosis regulated gene, increased during endoplasmic reticulum stress. However, the expression of PUMA in cardiomyocytes under mechanical stress is little known. We aimed to investigate the regulation mechanism of PUMA expression and apoptosis induced by mechanical stress in cardiomyocytes. METHODS: Aorta-caval (AV) shunt was performed in adult Wistar rats to induce volume overload. Rat neonatal cardiomyocytes were stretched by vacuum to 20% of maximum elongation at 60 cycles/min. RESULTS: PUMA protein and mRNA were up-regulated in the shunt group as compared with sham group. The increased PUMA protein expression and apoptosis induced by shunt was reversed by treatment with atorvastatin at 30 mg/kg/ day orally for 7 days. TUNEL assay showed that treatment with atorvastatin inhibited the apoptosis induced by volume overload. Cyclic stretch significantly enhanced PUMA protein and gene expression. Addition of c-jun N-terminal kinase (JNK) inhibitor SP600125, JNK small interfering RNA (siRNA) and interferon-γ (INF-γ) antibody 30 min before stretch reduced the induction of PUMA protein. Gel shift assay demonstrated that stretch increased the DNA binding activity of interferon regulatory factor-1. Stretch increased, while PUMA-Mut plasmid, SP600125 and INF-γ antibody abolished the PUMA promoter activity induced by stretch. PUMA mediated apoptosis induced by stretch was reversed by PUMA siRNA and atorvastatin. CONCLUSIONS: Mechanical stress enhanced apoptosis and PUMA expression in cardiomyocytes. Treatment with atorvastatin reversed both PUMA expression and apoptosis induced by mechanical stress in cardiomyocytes.


Asunto(s)
Proteínas Reguladoras de la Apoptosis , Apoptosis , Miocitos Cardíacos/metabolismo , Estrés Mecánico , Animales , Aorta/cirugía , Apoptosis/efectos de los fármacos , Apoptosis/genética , Proteínas Reguladoras de la Apoptosis/genética , Proteínas Reguladoras de la Apoptosis/metabolismo , Atorvastatina , Volumen Sanguíneo , Células Cultivadas , Regulación de la Expresión Génica/efectos de los fármacos , Regulación de la Expresión Génica/genética , Ácidos Heptanoicos/administración & dosificación , Masculino , Pirroles/administración & dosificación , ARN Mensajero/genética , ARN Mensajero/metabolismo , Ratas , Ratas Wistar , Transducción de Señal
7.
J Biomed Sci ; 18: 27, 2011 May 04.
Artículo en Inglés | MEDLINE | ID: mdl-21542902

RESUMEN

BACKGROUND: Visfatin, a adipocytokine with insulin-mimetic effect, plays a role in endothelial angiogenesis. Hyperbaric oxygen (HBO) has been used in medical practice. However, the molecular mechanism of beneficial effects of HBO is poorly understood. We sought to investigate the cellular and molecular mechanisms of regulation of visfatin by HBO in human coronary arterial endothelial cells (CAECs). METHODS: Human CAECs were exposed to 2.5 atmosphere absolute (ATA) of oxygen in a hyperbaric chamber. Western blot, real-time polymerase chain reaction, and promoter activity assay were performed. In vitro glucose uptake and tube formation was detected. RESULTS: Visfatin protein (2.55-fold) and mRNA (2.53-fold) expression were significantly increased after exposure to 2.5 ATA HBO for 4 to 6 h. Addition of SP600125 and JNK siRNA 30 min before HBO inhibited the induction of visfatin protein. HBO also significantly increased DNA-protein binding activity of AP-1 and visfatin promoter activity. Addition of SP600125 and TNF-α monoclonal antibody 30 min before HBO abolished the DNA-protein binding activity and visfatin promoter activity induced by HBO. HBO significantly increased secretion of TNF-α from cultured human CAECs. Exogenous addition of TNF-α significantly increased visfatin protein expression while TNF-α antibody and TNF-α receptor antibody blocked the induction of visfatin protein expression induced by HBO. HBO increased glucose uptake in human CAECs as HBO and visfatin siRNA and TNF-α antibody attenuated the glucose uptake induced by HBO. HBO significantly increased the tube formation of human CAECs while visfatin siRNA, TNF-α antibody inhibited the tube formation induced by HBO. CONCLUSIONS: HBO activates visfatin expression in cultured human CAECs. HBO-induced visfatin is mediated by TNF-α and at least in part through JNK pathway.


Asunto(s)
Vasos Coronarios/metabolismo , Células Endoteliales/metabolismo , Oxigenoterapia Hiperbárica , Proteínas Quinasas JNK Activadas por Mitógenos/metabolismo , Nicotinamida Fosforribosiltransferasa/metabolismo , Transducción de Señal , Factor de Necrosis Tumoral alfa/metabolismo , Antracenos/farmacología , Células Cultivadas , Vasos Coronarios/citología , Células Endoteliales/citología , Humanos , Proteínas Quinasas JNK Activadas por Mitógenos/antagonistas & inhibidores , Regiones Promotoras Genéticas , ARN Mensajero/metabolismo , ARN Interferente Pequeño/metabolismo , Factor de Transcripción AP-1/metabolismo , Factor de Necrosis Tumoral alfa/genética
8.
J Agric Food Chem ; 69(44): 13020-13033, 2021 Nov 10.
Artículo en Inglés | MEDLINE | ID: mdl-34723490

RESUMEN

Phytoestrogens are strongly recommended for treating osteoporosis. Our previous study showed that naringin, a citrus flavonoid, can enhance the bone mass in ovariectomized rats. In this study, we further elucidated the mechanisms of naringin-induced osteoblast maturation and bone healing. Treatment of human osteoblasts with naringin increased cell viability and proliferation. In parallel, exposure to naringin enhanced translocation of estrogen receptor alpha (ERα) to nuclei and its transactivation activity. Sequentially, naringin induced alkaline phosphatase (ALP) mRNA and protein expression and its enzyme activity. Pretreatment with methylpiperidinopyrazole (MPP), a specific inhibitor of ERα, attenuated naringin-induced augmentations in ERα transactivation activity, ALP gene expression, and cell mineralization. The beneficial effects of naringin were also confirmed in mouse MC3T3-E1 cells. Moreover, administration of mice with a bone defect with naringin increased levels of ERα and ALP in damaged sites and simultaneously enhanced the healing rate and bone strength. Nevertheless, treatment with MPP weakened naringin-triggered expression of ERα and ALP and improved bone healing and mass. Therefore, naringin could improve osteoblast mineralization and bone healing via regulating ERα-dependent ALP gene expression. Naringin can be clinically applied for treatment of osteoporosis-related bone diseases.


Asunto(s)
Fosfatasa Alcalina , Receptor alfa de Estrógeno , Fosfatasa Alcalina/genética , Animales , Diferenciación Celular , Receptor alfa de Estrógeno/genética , Flavanonas , Expresión Génica , Ratones , Osteoblastos , Ratas
9.
Am J Chin Med ; 49(4): 901-923, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33853499

RESUMEN

Our previous study showed that estrogen can induce mitochondrial adenosine triphosphate (ATP) synthesis-associated gene expressions and osteoblast maturation. Genistein, a phytoestrogenic isoflavone that is widely found in various foods and traditional herb products, is beneficial for osteogenesis by selectively triggering estrogen receptor alpha (ER[Formula: see text] expression. In this study, we further investigated the mechanisms of genistein-induced energy production and osteoblast activation. Exposure of rat calvarial osteoblasts and human U-2 OS cells to genistein triggered osteoblast activation without affecting cell survival. Treatment with genistein time-dependently induced ER[Formula: see text] mRNA and protein expressions in rat calvarial osteoblasts. Analyses by confocal microscopy and immunoblotting showed that genistein stimulated translocation of ER[Formula: see text] from the cytoplasm to mitochondria. Subsequently, expressions of mitochondrial cytochrome c oxidase (COX) I and II mRNAs and proteins in primary rat osteoblasts were induced after exposure to genistein. Knocking-down ER[Formula: see text] concurrently inhibited genistein-induced COX I and II mRNA expressions. In addition, mitochondrial complex enzyme activities, the mitochondrial membrane potential, and cellular ATP levels in rat calvarial osteoblasts were time-dependently augmented by genistein. Suppressing ER[Formula: see text] expression instantaneously lowered genistein-induced enhancements of mitochondrial energy production and osteoblast activation. Effects of genistein on ER[Formula: see text] translocation, COX I and II mRNA expressions, ATP synthesis, and osteoblast activation were further confirmed in human U-2 OS cells. This study showed that genistein can stimulate energy production and consequent osteoblast activation via inducing ER[Formula: see text]-mediated mitochondrial ATP synthesis-linked gene expressions.


Asunto(s)
Metabolismo Energético/efectos de los fármacos , Receptor alfa de Estrógeno/genética , Expresión Génica/efectos de los fármacos , Genisteína/farmacología , ATPasas de Translocación de Protón Mitocondriales/genética , Osteoblastos/efectos de los fármacos , Animales , Línea Celular Tumoral , Modelos Animales de Enfermedad , Receptor alfa de Estrógeno/metabolismo , Femenino , Humanos , ATPasas de Translocación de Protón Mitocondriales/metabolismo , Osteoporosis/tratamiento farmacológico , Ratas , Ratas Wistar
10.
J Agric Food Chem ; 68(39): 10639-10650, 2020 Sep 30.
Artículo en Inglés | MEDLINE | ID: mdl-32897066

RESUMEN

Osteoporosis-associated fractures may cause higher morbidity and mortality. Our previous study showed the effects of genistein, a phytoestrogen, on the induction of estrogen receptor alpha (ERα) gene expression and stimulation of osteoblast mineralization. In this study, rat calvarial osteoblasts and an animal bone defect model were used to investigate the effects of genistein on bone healing. Treatment with genistein caused a time-dependent increase in alkaline phosphatase (ALP) activity in rat osteoblasts. Levels of cytosolic and nuclear ERα significantly augmented following exposure to genistein. Subsequently, genistein elevated levels of ALP mRNA and protein in rat osteoblasts. Moreover, genistein induced other osteogenesis-associated osteocalcin and Runx2 mRNA and protein expressions. Knocking-down ERα using RNA interference concurrently inhibited genistein-induced Runx2, osteocalcin, and ALP mRNA expression. Attractively, administration of ICR mice suffering bone defects with genistein caused significant increases in the callus width, chondrocyte proliferation, and ALP synthesis. Results of microcomputed tomography revealed that administration of genistein increased trabecular bone numbers and improved the bone thickness and volume. This study showed that genistein can improve bone healing via triggering ERα-mediated osteogenesis-associated gene expressions and subsequent osteoblast maturation.


Asunto(s)
Receptor alfa de Estrógeno/metabolismo , Genisteína/administración & dosificación , Osteoblastos/efectos de los fármacos , Osteogénesis/efectos de los fármacos , Osteoporosis/tratamiento farmacológico , Fitoestrógenos/administración & dosificación , Fosfatasa Alcalina/genética , Fosfatasa Alcalina/metabolismo , Animales , Regeneración Ósea/efectos de los fármacos , Huesos/metabolismo , Huesos/fisiopatología , Subunidad alfa 1 del Factor de Unión al Sitio Principal/genética , Subunidad alfa 1 del Factor de Unión al Sitio Principal/metabolismo , Receptor alfa de Estrógeno/genética , Femenino , Humanos , Ratones , Ratones Endogámicos ICR , Osteoblastos/citología , Osteoblastos/metabolismo , Osteocalcina/genética , Osteocalcina/metabolismo , Osteoporosis/genética , Osteoporosis/metabolismo , Osteoporosis/fisiopatología , Ratas
11.
Life Sci ; 258: 118195, 2020 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-32781073

RESUMEN

AIMS: The estrogen-ERα axis participates in osteoblast maturation. This study was designed to further evaluated the roles of the estrogen-ERα axis in bone healing and the possible mechanisms. MAIN METHODS: Female ICR mice were created a metaphyseal bone defect in the left femurs and administered with methylpiperidinopyrazole (MPP), an inhibitor of ERα. Bone healing was evaluated using micro-computed tomography. Colocalization of ERα with alkaline phosphatase (ALP) and ERα translocation to mitochondria were determined. Levels of ERα, ERß, PECAM-1, VEGF, and ß-actin were immunodetected. Expression of chromosomal Runx2, ALP, and osteocalcin mRNAs and mitochondrial cytochrome c oxidase (COX) I and COXII mRNAs were quantified. Angiogenesis was measured with immunohistochemistry. KEY FINDINGS: Following surgery, the bone mass was time-dependently augmented in the bone-defect area. Simultaneously, levels of ERα were specifically upregulated and positively correlated with bone healing. Administration of MPP to mice consistently decreased levels of ERα and bone healing. As to the mechanisms, osteogenesis was enhanced in bone healing, but MPP attenuated osteoblast maturation. In parallel, expressions of osteogenesis-related ALP, Runx2, and osteocalcin mRNAs were induced in the injured zone. Treatment with MPP led to significant inhibition of the alp, runx2, and osteocalcin gene expressions. Remarkably, administration of MPP lessened translocation of ERα to mitochondria and expressions of mitochondrial energy production-related coxI and coxII genes. Furthermore, exposure to MPP decreased levels of PECAM-1 and VEGF in the bone-defect area. SIGNIFICANCE: The present study showed the contributions of the estrogen-ERα axis to bone healing through stimulation of energy production, osteoblast maturation, and angiogenesis.


Asunto(s)
Regeneración Ósea , Diferenciación Celular , Metabolismo Energético , Receptor alfa de Estrógeno/metabolismo , Neovascularización Fisiológica , Osteoblastos/citología , Transducción de Señal , Fosfatasa Alcalina/metabolismo , Animales , Peso Corporal/efectos de los fármacos , Regeneración Ósea/efectos de los fármacos , Callo Óseo/efectos de los fármacos , Callo Óseo/patología , Diferenciación Celular/efectos de los fármacos , Cromosomas de los Mamíferos/metabolismo , Subunidad alfa 1 del Factor de Unión al Sitio Principal/metabolismo , Complejo IV de Transporte de Electrones/genética , Complejo IV de Transporte de Electrones/metabolismo , Metabolismo Energético/efectos de los fármacos , Femenino , Regulación de la Expresión Génica/efectos de los fármacos , Técnicas de Silenciamiento del Gen , Ratones Endogámicos ICR , Mitocondrias/efectos de los fármacos , Mitocondrias/metabolismo , Neovascularización Fisiológica/efectos de los fármacos , Tamaño de los Órganos/efectos de los fármacos , Osteoblastos/efectos de los fármacos , Osteoblastos/metabolismo , Osteocalcina/metabolismo , Osteogénesis/efectos de los fármacos , Transporte de Proteínas/efectos de los fármacos , Pirazoles/administración & dosificación , Pirazoles/farmacología , Regulación hacia Arriba/efectos de los fármacos , Cicatrización de Heridas/efectos de los fármacos
12.
Life Sci ; 213: 279-286, 2018 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-30352244

RESUMEN

AIMS: Our previous study showed that propofol can protect against sepsis-induced insults through suppressing liver nitrosation and inflammation. This study further evaluated the mechanisms of propofol-caused protection from sepsis-induced liver dysfunction. MAIN METHODS: Male Wistar rats were subjected to cecal ligation and puncture (CLP) and then exposed to propofol. Levels of hepatic oxidative stress and lipid peroxidation were consecutively measured. Expressions of tumor necrosis factor (TNF)-α, interleukin (IL)-1ß, and IL-4 messenger (m)RNA or proteins were quantified. Effects of propofol on microsomal pentoxyresorufin O-dealkelase (PROD) and ethoxycoumarin O-deethylase (ECOD) activities were determined. KEY FINDINGS: Administration of propofol to CLP-treated rats significantly attenuated sepsis-induced insults. CLP caused augmented serum aspartate aminotransferase and alanine aminotransferase activities and concurrently triggered liver damage. In contrast, treatment with propofol protected against CLP-induced liver dysfunction. As to the mechanisms, the CLP-induced increases in oxidative stress and lipid peroxidation levels and TNF-α and IL-1ß mRNA and protein expressions were subsequently attenuated by propofol. Furthermore, administration of CLP-treated rats with propofol augmented levels of IL-4 in the liver. Phenobarbital treatment of liver microsomes in CLP-treated rats produced less amplification of PROD and ECOD activities, and a smaller amount of 4-hydroxypropofol was metabolized from propofol by liver microsomes. In contrast, more drug interactions occurred with propofol, which decreased PROD and ECOD activities in liver microsomes of CLP-treated rats. SIGNIFICANCE: Taken together, the present study showed that propofol can protect against sepsis-induced liver dysfunction through suppressing hepatic oxidative stress, lipid peroxidation, inflammation, and drug biotransformation and interactions in the liver.


Asunto(s)
Peroxidación de Lípido/efectos de los fármacos , Hepatopatías/terapia , Propofol/farmacología , Animales , Interacciones Farmacológicas , Inflamación/tratamiento farmacológico , Interleucina-1beta , Interleucina-4 , Hígado/metabolismo , Masculino , Estrés Oxidativo/efectos de los fármacos , Propofol/uso terapéutico , Ratas , Ratas Wistar , Sepsis/complicaciones , Factor de Necrosis Tumoral alfa
13.
Life Sci ; 195: 25-32, 2018 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-29307523

RESUMEN

AIMS: Propofol can be applied as an anesthetic or sedative agent for septic patients. Our previous studies showed that propofol ameliorated inflammation- and nitrosative stress-induced cellular insults. This study further evaluated effects of propofol on cecal ligation and puncture (CLP)-induced septic insults to rats and its possible mechanisms. MAIN METHODS: Wistar rats were administered with CLP and effects of propofol on CLP-induced liver dysfunction and rat death were evaluated. Levels of hepatic or systemic nitrogen oxides (NOx) and interleukin (IL)-6 were quantified. Sequentially, inducible nitric oxide synthase (iNOS) and IL-6 gene expressions, toll-like receptor 4 (TLR4) protein levels, and nuclear factor (NF)-κB translocation were determined. KEY FINDINGS: Subjecting rats to CLP led to body weight loss, liver weight gain, and death. Administration of propofol lessened CLP-induced augmentations of serum and hepatic nitrosative stress and IL-6 levels. Additionally, propofol suppressed CLP-induced enhancements in levels of hepatic iNOS protein. Furthermore, the CLP-induced iNOS and IL-6 mRNA expressions in the liver were inhibited following propofol administration. Sequentially, subjecting rats to CLP enhanced hepatic TLR4 protein levels and NF-κB translocation to nuclei, but propofol inhibited these augmentations. SIGNIFICANCE: Consequently, exposure to propofol protected against CLP-induced liver dysfunction and increased the survival rates of the animals. This study shows that propofol can protect rats against septic insults through suppression of systemic and hepatic nitrosative and inflammatory stress due to inhibition of TLR4/NF-κB-mediated iNOS and IL-6 mRNA and protein expressions.


Asunto(s)
Hepatitis/tratamiento farmacológico , Hipnóticos y Sedantes/uso terapéutico , Interleucina-6/biosíntesis , Hígado/metabolismo , Hígado/patología , FN-kappa B/efectos de los fármacos , Óxido Nítrico Sintasa de Tipo II/biosíntesis , Nitrosación/efectos de los fármacos , Propofol/uso terapéutico , Sepsis/tratamiento farmacológico , Receptor Toll-Like 4/efectos de los fármacos , Actinas/metabolismo , Animales , Enfermedades del Ciego/metabolismo , Enfermedades del Ciego/patología , Regulación hacia Abajo , Regulación de la Expresión Génica/efectos de los fármacos , Hepatitis/metabolismo , Hepatitis/patología , Interleucina-6/genética , Masculino , Óxido Nítrico Sintasa de Tipo II/genética , Ratas , Ratas Wistar , Sepsis/metabolismo , Sepsis/patología , Translocación Genética/efectos de los fármacos
14.
Food Funct ; 9(8): 4500-4507, 2018 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-30083664

RESUMEN

Licochalcone A (LA), an active ingredient of licorice, has multiple biological activities, including antioxidative and anti-inflammatory activities. Although LA exerts antitumor effects in various cancer cells, its role in gliomas remains unclear. Therefore, this study determined whether LA inhibits glioma cell growth in vitro and in vivo. The present data revealed that LA effectively inhibited the growth of U87 glioma cells by inducing cell cycle arrest in the G0/G1 and G2/M phases; cell cycle arrest was attributed to the LA-mediated reduction of mRNA and protein levels of cyclins and cyclin-dependent kinases. Moreover, subcutaneous (flank) and orthotopic (brain) tumor models were used to determine the role of LA in gliomas. LA significantly alleviated tumor growth in both models. These findings indicate that LA exerts antitumor effects in gliomas in vitro and in vivo and that it is a potential agent for treating glioblastoma multiforme.


Asunto(s)
Antineoplásicos/administración & dosificación , Puntos de Control del Ciclo Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Chalconas/administración & dosificación , Glioma/tratamiento farmacológico , Animales , Línea Celular Tumoral , Glioma/fisiopatología , Humanos , Masculino , Ratones , Ratones Desnudos
15.
Anesth Analg ; 104(3): 726-30, 2007 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-17312234

RESUMEN

BACKGROUND: Excitatory amino acids play an important role in morphine tolerance. Recently, we demonstrated that a single morphine challenge induces an increase in spinal cerebrospinal fluid excitatory amino acid concentrations in morphine-tolerant rats, and that dexamethasone inhibits the development of morphine tolerance. We further examined the effect of intrathecal dexamethasone infusion on the development of morphine tolerance and on expression of the intracellular glutamate metabolizing enzymes, glutamate dehydrogenase and glutamine synthetase, in the spinal cord. METHODS: Male Wistar rats, implanted with an intrathecal catheter, were divided into four groups that were infused for 5 days with intrathecal morphine (15 microg/h), saline (1 microL/h), dexamethasone (2 microg/h), or dexamethasone (2 microg/h) plus morphine (15 microg/h). On Day 5, the spinal cords were removed and prepared for Western blot analysis of glutamate dehydrogenase and glutamate synthetase. RESULTS: Glutamate dehydrogenase and glutamate synthetase concentrations were downregulated in the morphine-tolerant rat spinal cords. Concurrent infusion of dexamethasone attenuated morphine tolerance and the associated glutamate dehydrogenase and glutamate synthetase downregulation. CONCLUSION: Intrathecal dexamethasone attenuates long-term morphine infusion-induced glutamate dehydrogenase and glutamate synthetase downregulation and antinociceptive tolerance.


Asunto(s)
Analgésicos Opioides/farmacología , Dexametasona/farmacología , Glutamato Deshidrogenasa/líquido cefalorraquídeo , Glutamato-Amoníaco Ligasa/líquido cefalorraquídeo , Morfina/farmacología , Médula Espinal/metabolismo , Animales , Regulación hacia Abajo , Tolerancia a Medicamentos , Glucocorticoides/farmacología , Masculino , Dependencia de Morfina/patología , Ratas , Ratas Wistar , Factores de Tiempo
16.
J Pain Symptom Manage ; 32(5): 444-52, 2006 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-17085270

RESUMEN

The epidemiology of cancer pain and the outcomes associated with pain treatments were investigated through a retrospective survey of 772 patients with advanced cancer. The cumulative prevalence of pain was 87%, including all types of cancer. Mean duration of pain was 6.9+/-8.1 months. The prevalence of pain was 28%, 46%, 67%, 75%, and 79% at 6 months, 3 months, 1 month, 1 week, and 1 day before the time of death, respectively. The so-called "strong" opioids had been used in 85% of the 669 patients with pain. Seventy-nine percent of patients with pain received nonsurgical antineoplastic treatment for pain control. No more than 11% of patients ultimately experienced substantial pain in the last 6 months of life (defined as pain score 5-10 on a 0-10 numeric rating scale). We conclude that the application of a multidisciplinary approach to pain management offers effective pain control for most patients with advanced cancer.


Asunto(s)
Neoplasias/complicaciones , Manejo del Dolor , Dolor/epidemiología , Cuidado Terminal , Adolescente , Adulto , Anciano , Anciano de 80 o más Años , Niño , Estudios de Cohortes , Femenino , Humanos , Masculino , Persona de Mediana Edad , Dimensión del Dolor , Prevalencia , Estudios Retrospectivos , Resultado del Tratamiento
17.
Life Sci ; 78(16): 1801-7, 2006 Mar 13.
Artículo en Inglés | MEDLINE | ID: mdl-16271370

RESUMEN

Neuropathic pain syndromes respond poorly to opioid treatment. In our previous studies, we found that intrathecal (i.t.) injection of pertussis toxin (PTX) produces thermal hyperalgesia, which is poorly responsive to morphine and is accompanied by an increase in cerebrospinal fluid (CSF) levels of excitatory amino acids (EAAs) and protein kinase C (PKC) activation. In the present study, rats were implanted with an i.t. catheter for drug injection and a microdialysis probe for CSF dialysate collection. On the fourth day after injection of PTX (2 microg, i.t.), there was a significant reduction in the antinociceptive effect of morphine (10 microg, i.t.) which was accompanied by an increase in levels of EAAs. Pretreatment with the PKC inhibitor, chelerythrine (25 microg, i.t.) one hour before morphine injection markedly inhibited both effects. These results suggest that, in PTX-treated rats, PKC plays an important role in inhibiting the morphine-induced spinal EAA release, which might be related to the reduced antinociceptive effect of morphine.


Asunto(s)
Analgésicos Opioides/farmacología , Inhibidores Enzimáticos/farmacología , Aminoácidos Excitadores/líquido cefalorraquídeo , Morfina/farmacología , Toxina del Pertussis/administración & dosificación , Fenantridinas/farmacología , Proteína Quinasa C/antagonistas & inhibidores , Alcaloides , Animales , Benzofenantridinas , Inyecciones Espinales , Masculino , Ratas , Ratas Wistar
18.
Data Brief ; 9: 667-672, 2016 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-27774504

RESUMEN

This article contains raw and processed data related to a research, "Honokiol induces autophagic cell death in malignant glioma through reactive oxygen species-mediated regulation of the p53/PI3K/Akt/mTOR signaling pathway" (C.J. Lin, T.L. Chen, Y.Y. Tseng, G.J. Wu, M.H. Hsieh, Y.W. Lin, R.M. Chen, 2016) [1]. Data were obtained by immunoblotting analyses of light chain 3 (LC3)-II, beclin-1, Akt, and mTOR in human glioma U87 MG cells and mouse glioma tissues treated with honokiol, an active constituent extracted from the bark of Magnolia officinalis, "Honokiol induces autophagy of neuroblastoma cells through activating the PI3K/Akt/mTOR and endoplasmic reticular stress/ERK1/2 signaling pathways and suppressing cell migration" (P.S. Yeh, W. Wang, Y.A. Chang, C.J. Lin, J.J. Wang, R.M. Chen, 2016) [2]. The processed data show the effects of honokiol on induction of autophagy in human glioma U87 MG cells by analyzing levels of LC3-II, p62, and bectin-1, "Honokiol-induced apoptosis and autophagy in glioblastoma multiforme cells" (K.H. Chang, M.D Yan, C.J. Yao, P.C. Lin, G.M. Lai, 2013) [3]. In addition, chloroquine, a lysosomal inhibitor, was administered to the cells to further confirm honokiol-induced cell autophagy. Sequentially, mice with gliomas were created and treated with honokiol. Amounts of phosphorylated and non-phosphorylated Akt and mTOR in glioma tissues were analyzed to determine the possible mechanisms of honokiol-induced autophagy.

19.
Ann N Y Acad Sci ; 1042: 168-76, 2005 May.
Artículo en Inglés | MEDLINE | ID: mdl-15965060

RESUMEN

Propofol is a widely used intravenous anesthetic agent. Our previous study showed that a therapeutic concentration of propofol can modulate macrophage functions. Mitochondria play critical roles in the maintenance of macrophage activities. This study attempted to evaluate further the effects of mitochondria on the propofol-induced suppression of macrophage functions using mouse macrophage-like Raw 264.7 cells as the experimental model. Macrophages were exposed to a clinically relevant concentration of propofol for 1, 6, and 24 h. Analysis by the Trypan blue exclusion method revealed that propofol was not cytotoxic to macrophages. Exposure of macrophages to propofol did not affect mitochondrial NADH dehydrogenase activity of complex I. However, analysis of flow cytometry showed that propofol significantly decreased the mitochondrial membrane potential of macrophages. Cellular levels of ATP in macrophages were significantly reduced after propofol administration. In parallel with the dysfunction of mitochondria, the chemotactic analysis showed that exposure to propofol significantly inhibited the migration of macrophages. This study shows that a therapeutic concentration of propofol can specifically reduce the mitochondrial membrane potential, but there is no such effect on complex I NADH dehydrogenase activity. Modulation of the mitochondrial membrane potential may decrease the biosynthesis of cellular ATP and thus reduce the chemotactic activity of macrophages. This study provides in vitro data to validate mitochondrial dysfunction as a possible critical cause for propofol-induced immunosuppression of macrophage functions.


Asunto(s)
Adenosina Trifosfato/biosíntesis , Quimiotaxis/efectos de los fármacos , Complejo I de Transporte de Electrón/metabolismo , Macrófagos/efectos de los fármacos , Membranas Mitocondriales/efectos de los fármacos , Membranas Mitocondriales/metabolismo , Propofol/farmacología , Animales , Línea Celular , Supervivencia Celular , Macrófagos/citología , Macrófagos/metabolismo , Potenciales de la Membrana/efectos de los fármacos , Ratones
20.
Ann N Y Acad Sci ; 1042: 448-59, 2005 May.
Artículo en Inglés | MEDLINE | ID: mdl-15965091

RESUMEN

2,6-Diisopropylphenol is an intravenous anesthetic agent used for induction and maintenance of anesthesia. Since it is similar to alpha-tocopherol, 2,6-diisopropylphenol may have antioxidant effects. Osteoblasts play important roles in bone remodeling. In this study, we attempted to evaluate the protective effects of 2,6-diisopropylphenol on oxidative stress-induced osteoblast insults and their possible mechanisms, using neonatal rat calvarial osteoblasts as the experimental model. Clinically relevant concentrations of 2,6-diisopropylphenol (3 and 30 microM) had no effect on osteoblast viability. However, 2,6-diisopropylphenol at 300 microM time-dependently caused osteoblast death. Exposure to sodium nitroprusside (SNP), a nitric oxide donor, increased amounts of nitrite in osteoblasts. 2,6-Diisopropylphenol did not scavenge basal or SNP-releasing nitric oxide. Hydrogen peroxide (HP) enhanced levels of intracellular reactive oxygen species in osteoblasts. 2,6-Diisopropylphenol significantly reduced HP-induced oxidative stress. Exposure of osteoblasts to SNP and HP decreased cell viability time-dependently. 2,6-Diisopropylphenol protected osteoblasts from SNP- and HP-induced cell damage. Analysis by a flow cytometric method revealed that SNP and HP induced osteoblast apoptosis. 2,6-Diisopropylphenol significantly blocked SNP- and HP-induced osteoblast apoptosis. Administration of SNP and HP increased caspase-3 activities. However, 2,6-diisopropylphenol significantly decreased SNP- and HP-enhanced caspase-3 activities. This study shows that a therapeutic concentration of 2,6-diisopropylphenol can protect osteoblasts from SNP- and HP-induced cell insults, possibly via suppression of caspase-3 activities.


Asunto(s)
Apoptosis/efectos de los fármacos , Caspasas/metabolismo , Osteoblastos/citología , Osteoblastos/efectos de los fármacos , Estrés Oxidativo , Propofol/farmacología , Animales , Caspasa 3 , Células Cultivadas , Activación Enzimática/efectos de los fármacos , Peróxido de Hidrógeno/farmacología , Nitritos/metabolismo , Nitroprusiato/farmacología , Osteoblastos/enzimología , Ratas , Ratas Wistar , Especies Reactivas de Oxígeno/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA