Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 40
Filtrar
Más filtros

Tipo del documento
País de afiliación
Intervalo de año de publicación
1.
Biol Res ; 57(1): 30, 2024 May 17.
Artículo en Inglés | MEDLINE | ID: mdl-38760850

RESUMEN

BACKGROUND: Mutations in isocitrate dehydrogenase 1 and 2 (IDH1 and IDH2), are present in most gliomas. IDH1 mutation is an important prognostic marker in glioma. However, its regulatory mechanism in glioma remains incompletely understood. RESULTS: miR-182-5p expression was increased within IDH1-mutant glioma specimens according to TCGA, CGGA, and online dataset GSE119740, as well as collected clinical samples. (R)-2-hydroxyglutarate ((R)-2HG) treatment up-regulated the expression of miR-182-5p, enhanced glioma cell proliferation, and suppressed apoptosis; miR-182-5p inhibition partially eliminated the oncogenic effects of R-2HG upon glioma cells. By direct binding to Cyclin Dependent Kinase Inhibitor 2 C (CDKN2C) 3'UTR, miR-182-5p inhibited CDKN2C expression. Regarding cellular functions, CDKN2C knockdown promoted R-2HG-treated glioma cell viability, suppressed apoptosis, and relieved cell cycle arrest. Furthermore, CDKN2C knockdown partially attenuated the effects of miR-182-5p inhibition on cell phenotypes. Moreover, CDKN2C knockdown exerted opposite effects on cell cycle check point and apoptosis markers to those of miR-182-5p inhibition; also, CDKN2C knockdown partially attenuated the functions of miR-182-5p inhibition in cell cycle check point and apoptosis markers. The engineered CS-NPs (antagomir-182-5p) effectively encapsulated and delivered antagomir-182-5p, enhancing anti-tumor efficacy in vivo, indicating the therapeutic potential of CS-NPs(antagomir-182-5p) in targeting the miR-182-5p/CDKN2C axis against R-2HG-driven oncogenesis in mice models. CONCLUSIONS: These insights highlight the potential of CS-NPs(antagomir-182-5p) to target the miR-182-5p/CDKN2C axis, offering a promising therapeutic avenue against R-2HG's oncogenic influence to glioma.


Asunto(s)
Neoplasias Encefálicas , Glioma , Isocitrato Deshidrogenasa , MicroARNs , Humanos , Isocitrato Deshidrogenasa/genética , Isocitrato Deshidrogenasa/metabolismo , Glioma/genética , Glioma/metabolismo , Animales , Ratones , Xenoinjertos , Trasplante de Neoplasias , Neoplasias Encefálicas/genética , MicroARNs/metabolismo
2.
Cell Mol Neurobiol ; 43(8): 4279-4293, 2023 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-37864627

RESUMEN

To investigate the molecular mechanism of communication network factor 1 (CCN1) regulating pentylenetetrazol (PTZ)-induced epileptogenesis, deepen the understanding of epilepsy seizure pathogenesis, and provide new drug action targets for its clinical prevention and treatment. Differentially expressed genes (DEGs) on microarrays GSE47516 and GSE88992 were analyzed online using GEO2R. Pathway enrichment and protein-protein interaction network (PPI) analysis of DEGs were carried out using Metascape. Brain tissue samples of severe traumatic brain injury patients (named Healthy group) and refractory epilepsy patients (named Epilepsy group) were obtained and analyzed by qRT-PCR and immunohistochemistry (IHC) staining. A PTZ-induced epilepsy mouse model was established and verified. Morphological changes of neurons in mouse brain tissue were detected using hematoxylin and eosin (HE) staining. qRT-PCR was conducted to detect the mRNA expressions of apoptosis-associated proteins Bax, Caspase-3 and bcl2. TUNEL staining was performed to detect brain neuron apoptosis. The levels of myocardial enzymology, GSH, MDA and ROS in blood of mouse were detected by biochemical assay. CCN1 expression was increased in epilepsy brain tissue samples. CCN1 decreasing effectively prolongs seizure incubation period and decreases seizure duration. Silencing of CCN1 also reduces neuronal damage and apoptosis, decreases mRNA and protein expression of proapoptotic proteins Bax and Caspase-3, increases mRNA expression of antiapoptotic protein Bcl2. Moreover, decrease of CCN1 decreases myocardial enzymatic indexes CK and CK-MB levels, reduces myocardial tissue hemorrhage, and relieves oxidative stress response in hippocampal and myocardial tissue. CCN1 expression is increased in epileptic samples. CCN1 decreasing protects brain tissue by attenuating oxidative stress and inhibiting neuronal apoptosis triggered by PTZ injection, which probably by regulating Nrf2/HO-1 pathway.


Asunto(s)
Epilepsia , Pentilenotetrazol , Humanos , Ratones , Animales , Pentilenotetrazol/efectos adversos , Caspasa 3/metabolismo , Proteína X Asociada a bcl-2/metabolismo , Epilepsia/inducido químicamente , Epilepsia/genética , Epilepsia/tratamiento farmacológico , Convulsiones/inducido químicamente , Estrés Oxidativo , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , ARN Mensajero/metabolismo
3.
Addict Biol ; 28(9): e13326, 2023 09.
Artículo en Inglés | MEDLINE | ID: mdl-37644892

RESUMEN

The persistence of maladaptive heroin-associated memory, which is triggered by drug-related stimuli that remind the individual of the drug's pleasurable and rewarding effects, can impede abstinence efforts. Cyclin-dependent kinase 5 (Cdk5), a neuronal serine/threonine protein kinase that plays a role in multiple neuronal functions, has been demonstrated to be involved in drug addiction and learning and memory. Here, we aimed to investigate the role of cdk5 activity in the basolateral amygdala (BLA) in relapse to heroin seeking, using a self-administration rat model. Male rats underwent 10 days of heroin self-administration training, during which an active nose poke resulted in an intravenous infusion of heroin that was accompanied by a cue. The rats then underwent nose poke extinction for 10 days, followed by subsequent tests of heroin-seeking behaviour. We found that intra-BLA infusion of ß-butyrolactone (100 ng/side), a Cdk5 inhibitor, administered 5 min after reactivation, led to a subsequent decrease in heroin-seeking behaviour. Further experiments demonstrated that the effects of ß-butyrolactone are dependent on reactivated memories, temporal-specific and long-lasting on relapse of heroin-associated memory. Results provide suggestive evidence that the activity of Cdk5 in BLA is critical for heroin-associated memory and that the specific inhibitor, ß-butyrolactone, may hold potential as a substance for the treatment of heroin abuse.


Asunto(s)
Complejo Nuclear Basolateral , Heroína , Masculino , Animales , Ratas , Heroína/farmacología , Quinasa 5 Dependiente de la Ciclina , Recurrencia
4.
Addict Biol ; 28(10): e13330, 2023 10.
Artículo en Inglés | MEDLINE | ID: mdl-37753572

RESUMEN

The susceptibility to drug cravings evoked by stimuli poses a formidable hurdle in the treatment of addiction and the prevention of relapse. Pharmacological interventions targeting drug-associated memories hold promise for curbing relapse by impeding the process of memory reconsolidation, predominantly governed by cAMP signalling. Exchange Protein Activated by cAMP (Epac) serves as a distinctive mediator of cAMP signalling, which has been implicated in reinforcing the effects of cocaine and facilitating the acquisition. Nonetheless, the role of Epac in heroin-related memory and the subsequent seeking behaviour remains enigmatic. In this study, we explored the impact of Epac activation on the reconsolidation process of drug-related memories associated with heroin self-administration. Over the course of 10 consecutive days, rats underwent training, wherein they acquired the behaviour of nose poking to obtain heroin accompanied by a tone + light cue. This nose-poking behaviour was subsequently extinguished when heroin infusion and cue presentation were discontinued. Subsequently, we administered 8-pCPT-cAMP (8-CPT), an Epac-specific activator, into the basolateral amygdala at various time points, either in the presence or absence of a conditioned stimulus. Our findings demonstrate that administering 8-CPT immediately after memory retrieval effectively reduces cue- and heroin-induced reinstatement, with the observed effects persisting significantly for a minimum of 28 days. However, infusion of 8-CPT for a duration of 6 h following the memory retrieval trial, or without it altogether, had no discernible impact. Thus, these findings strongly suggest that Epac activation can disrupt the reconsolidation of heroin-associated memory, thereby diminishing the reinstatement of heroin-seeking behaviour.


Asunto(s)
Cocaína , Heroína , Ratas , Animales , Amígdala del Cerebelo , Ratas Sprague-Dawley , Cocaína/farmacología , Recurrencia , Señales (Psicología) , Extinción Psicológica , Autoadministración
5.
Acta Biochim Biophys Sin (Shanghai) ; 55(1): 103-116, 2023 Jan 25.
Artículo en Inglés | MEDLINE | ID: mdl-36718644

RESUMEN

Glioblastoma multiforme (GBM) is one of the most malignant types of central nervous system (CNS) tumors. N6-methyladenine (m6A) RNA modification is a main type of RNA modification in eukaryotic cells. In this study, we find that the m6A RNA methylation eraser FTO is dramatically downregulated in glioma samples and cell lines, particularly in intermediate and core regions and hypoxia-challenged glioma cells. In vitro, FTO overexpression inhibits the hypoxia-induced capacities of glioma cells to proliferate, migrate and invade, and decreases the percentage of cells with m6A RNA methylation. In vivo, FTO overexpression inhibits tumor growth in the xenograft model and decreases the protein levels of migration markers, including Vimentin and Twist. miR-27a-3p is upregulated within glioma intermediate and core regions and hypoxia-challenged glioma cells. miR-27a-3p inhibits the expression of FTO via direct binding to FTO. miR-27a-3p overexpression promotes hypoxia-challenged glioma cell aggressiveness, whereas FTO overexpression partially diminishes the oncogenic effects of miR-27a-3p overexpression. FTO overexpression promotes the nuclear translocation of FOXO3a and upregulates the expression levels of the FOXO3a downstream targets BIM, BNIP3, BCL-6, and PUMA, possibly by interacting with FOXO3a. Conclusively, FTO serves as a tumor suppressor in glioma by suppressing hypoxia-induced malignant behaviors of glioma cells, possibly by promoting the nuclear translocation of FOXO3a and upregulating FOXO3a downstream targets. miR-27a-3p is a major contributor to FTO downregulation in glioma under hypoxia.


Asunto(s)
Glioma , MicroARNs , Humanos , MicroARNs/metabolismo , Glioma/genética , Línea Celular , Regulación hacia Abajo , Hipoxia/genética , Línea Celular Tumoral , Proliferación Celular/genética , Dioxigenasa FTO Dependiente de Alfa-Cetoglutarato/genética , Dioxigenasa FTO Dependiente de Alfa-Cetoglutarato/metabolismo
6.
Exp Cell Res ; 402(1): 112547, 2021 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-33722639

RESUMEN

The high aggressiveness of gliomas remains a huge challenge to clinical therapies, and the hypoxic microenvironment in the core region is a critical contributor to glioma aggressiveness. In this study, it was found that miR-485-5p was low expressed within glioma tissue samples and cells. GO enrichment annotation indicated that the predicted downstream targets miR-485-5p were enriched in hypoxia response and decreased oxygen level. In glioma cells, miR-485-5p overexpression suppressed cell viability, migratory ability, and invasive ability under both normoxic and hypoxic conditions. Through direct binding, miR-485-5p suppressed SRPK1 expression. Under hypoxia, SRPK1 overexpression enhanced hypoxia-induced glioma cell aggressiveness and significantly reversed the effects of miR-485-5p overexpression. Moreover, HIF1A could target the miR-485-5p promoter region to inhibit the transcription. HIF1A, miR-485-5p, and SRPK1 form a regulatory axis, which modulates glioma cell aggressiveness under hypoxia. In conclusion, we identify a HIF1A/miR-485-5p/SRPK1 axis that modulates the aggressiveness of glioma cells under hypoxia. The axis could potentially provide new research avenues in the treatment of gliomas considering the hypoxic environment in its core.


Asunto(s)
Glioma/genética , Subunidad alfa del Factor 1 Inducible por Hipoxia/genética , MicroARNs/genética , Proteínas Serina-Treonina Quinasas/genética , Línea Celular Tumoral , Movimiento Celular/genética , Proliferación Celular/genética , Supervivencia Celular/genética , Femenino , Regulación Neoplásica de la Expresión Génica/genética , Glioma/patología , Humanos , Masculino , Persona de Mediana Edad , Invasividad Neoplásica/genética , Invasividad Neoplásica/patología , Estadificación de Neoplasias , Transducción de Señal/genética , Hipoxia Tumoral/genética , Microambiente Tumoral/genética
7.
Zhong Nan Da Xue Xue Bao Yi Xue Ban ; 47(7): 829-838, 2022 Jul 28.
Artículo en Inglés, Zh | MEDLINE | ID: mdl-36039578

RESUMEN

OBJECTIVES: This study aims to investigate the role of hypoxia-induced long non-coding small nucleolar RNA host gene 14 (lncRNA SNHG14) in glioma temozolomide (TMZ) resistance and underlying mechanisms. METHODS: According to different treatments, the experiment was divided into a normoxia group and a hypoxia group, a control group and a TMZ group. The lncRNA SNHG14 and O6-methylguanine DNA methyltransferase (MGMT) levels in glioma SNB19 and U251 cell line were detected by real-time PCR and Western blotting, respectively, and the association of lncRNA SNHG14 level with hypoxia and TMZ treatment was analyzed. siRNA was used to knockdown the lncRNA SNHG14 expression in glioma cells, and the transfected glioma cells were divided into a negative control group (si-NC group) and a si-SNHG14 group. The interference efficiency was examined by real-time PCR, the key factor MGMT of lncRNA SNHG14 sensitivity regulation was detected by Western blotting, and the cell apoptosis was detected by cytometry. In addition, MTT method was used to detect the cell viability of gliomas in the different groups under the different TMZ concentrations, and the effect of lncRNA SNHG14 on TMZ sensitivity of gliomas was analyzed. Online tools were used to predict miRNAs that could specifically bind to lncRNAs SNHG14 and MGMT. A si-NC group, a si-SNHG14 group, a normoxia group and a hypoxia group were set up, and the changes of miR-143 abundance in different environments were observed by real-time PCR. miR-143 mimics and inhibitor were used to change the level of miR-143 in glioma cells. A NC inhibitor group, a miR-143 inhibitor group, a NC mimics group and a miR-143 mimics group were set up, the interference efficiency was detected by real-time PCR, the expression level of MGMT was detected by Western blotting, and the effect of miR-143 on the level of MGMT were analyzed. The NC inhibitor group, the miR-143 inhibitor group, the NC mimics group and the miR-143 mimics group were treated with different interventions, and the dual luciferase reporter assay was used to observe the changes of lncRNA SNHG14 and MGMT luciferase activities, and to verify the relationship among lncRNA SNHG14, miR-143 and MGMT. Finally, a NC group and a lncRNA SNHG14 overexpression group were set up, and the changes in the abundance of miR-143 and MGMT in each group were detected by RNA-binding protein immunoprecipitation experiments, and the competitive binding relationship among lncRNA SNHG14, miR-143 and MGMT was analyzed. RESULTS: Compared with the normoxia group, the hypoxia group could promote the expression of lncRNA SNHG14 in glioma cells. Compared with the control group, the expression of lncRNA SNHG14 could be significantly inhibited in the TMZ group (P<0.05). Compared with the si-NC group, the expression of lncRNA SNHG14 in the si-SNHG14 group could be effectively inhibited, and the expression level of MGMT was significantly decreased, and the apoptosis rate was significantly increased (all P<0.05). With the increase of TMZ concentrations, the glioma cell viability in the si-SNHG14 group was significantly lower than that in the si-NC group, and the cell viability in the hypoxia group was significantly higher than that in the normoxia group (both P<0.05). Online tool prediction found that miR-143 had binding sites with lncRNA SNHG14 and MGMT. The abundance of miR-143 in the hypoxia group was significantly lower than that in the normoxic group, and the abundance of miR-143 in the si-SNHG14 group was significantly higher than that in the si-NC group (both P<0.05). The miR-143 mimics group or the miR-143 inhibitor group could significantly over-express or under-express miR-143 (both P<0.05). But there was no significant difference between the NC mimics group (or the NC inhibitor group) and the control group (both P>0.05). The level of MGMT protein could significantly up-regulate in the miR-143 inhibitor group, and on the contrary which could significantly down-regulate in the miR-143 mimics group (both P<0.01). The dual luciferase reporter assay showed that there was no significant difference between the NC mimics group (or the NC inhibitor group) and the control group (both P>0.05). The wild-type SNHG14 and MGMT luciferase activities were significantly down-regulated in the miR-143 mimics group, which were significantly up-regulated in the miR-143 inhibitor group (P<0.01 and P<0.05, respectively), but there was no significant change in the luciferase activities of mutant SNHG14 and MGMT (both P>0.05). The results of the RNA-binding protein immunoprecipitation experiment showed that: compared with the NC group, more lncRNA SNHG14 was bound to the precipitated argonaute 2 protein in the cells in the lncRNA SNHG14 overexpression group, but the abundance of MGMT mRNA was decreased significantly, and there were significant differences (both P<0.01). There was a targeting regulatory relationship among lncRNA SNHG14, miR-143 and MGMT. CONCLUSIONS: The up-regulated lncRNA SNHG14 can target miR-143, relieve the inhibition of miR-143 on MGMT, and promote the TMZ resistance in the hypoxia-induced glioma cells.


Asunto(s)
Glioma , MicroARNs , ARN Largo no Codificante , Apoptosis , Línea Celular Tumoral , Proliferación Celular , Resistencia a Antineoplásicos/genética , Glioma/genética , Humanos , Hipoxia , MicroARNs/metabolismo , ARN Largo no Codificante/genética , ARN Nucleolar Pequeño/farmacología , Proteínas de Unión al ARN , Temozolomida/farmacología
8.
Cancer Cell Int ; 21(1): 307, 2021 Jun 10.
Artículo en Inglés | MEDLINE | ID: mdl-34112159

RESUMEN

BACKGROUND: An increasing number of studies have shown that circular RNAs (circRNAs) play important roles in the regulation of tumor progression. Therefore, we explored the expression characteristics, function, and related mechanism of the newly identified circNALCN in glioma. METHODS: RNA sequencing was used to analyze the expression profiles of circRNAs in brain tissue from five glioma cases and four normal controls. Quantitative real-time polymerase chain reaction was implemented to examine the levels of circNALCN, miR-493-3p, and phosphatase and tensin homolog (PTEN). Cell counting kit 8 assays were performed to analyze cell proliferation, and cell migration was assessed by the wound healing test and Transwell assay. Dual-luciferase reporter, fluorescence in situ hybridization, and RNA pulldown assays were performed to confirm the role of circNALCN as an miR-493-3p sponge, weakening the inhibitory effect of miR-493-3p on target PTEN expression. RESULTS: The downregulated expression of circNALCN was observed in both glioma tissues and cell lines. CircNALCN expression was negatively correlated with World Health Organization grade and overall survival in patients with glioma. Functionally, the overexpression of circNALCN significantly inhibited the proliferation and migration of glioma cells, whereas miR-493-3p mimics counteracted these effects. The mechanistic analysis demonstrated that circNALCN acted as a competing endogenous RNA for miR-493-3p to relieve the repressive effects of miR-493-3p on its target, PTEN, suppressing glioma tumorigenesis. CONCLUSIONS: CircNALCN inhibits the progression of glioma through the miR-493-3p/PTEN axis, providing a developable biomarker and therapeutic target for glioma patients.

9.
J Pharmacol Exp Ther ; 368(1): 100-105, 2019 01.
Artículo en Inglés | MEDLINE | ID: mdl-30389723

RESUMEN

Central α4ßδ receptors are the most abundant isoform of δ subunit-containing extrasynaptic GABAA receptors that mediate tonic inhibition. Although the amplitude of GABA-activated currents through α4ßδ receptors is modulated by multiple general anesthetics, the effects of general anesthetics on desensitization and deactivation of α4ßδ receptors remain unknown. In the current study, we investigated the effect of etomidate, a potent general anesthetic, on the kinetics and the pseudo steady-state current amplitude of α4ß3δ receptors inducibly expressed in human embryonic kidney 293 TetR cells. Etomidate directly activates α4ß3δ receptors in a concentration-dependent manner. Etomidate at a clinically relevant concentration (3.2 µM) enhances maximal response without altering the EC50 of GABA concentration response. Etomidate also increases the extent of desensitization and prolongs the deactivation of α4ß3δ receptors in the presence of maximally activating concentrations of GABA (1 mM). To mimic the modulatory effect of etomidate on tonic currents, long pulses (30-60 seconds) of a low GABA concentration (1 µM) were applied to activate α4ß3δ receptors in the absence and presence of etomidate. Although etomidate increases the desensitization of α4ß3δ receptors, the pseudo steady-state current amplitude at 1 µM GABA is augmented by etomidate. Our data demonstrate that etomidate enhances the pseudo steady-state current of α4ß3δ receptors evoked by a GABA concentration comparable to an ambient GABA level, suggesting that α4ß3δ receptors may mediate etomidate's anesthetic effect in the brain.


Asunto(s)
Etomidato/farmacología , Agonistas del GABA/farmacología , Hipnóticos y Sedantes/farmacología , Receptores de GABA-A/biosíntesis , Relación Dosis-Respuesta a Droga , Expresión Génica , Células HEK293 , Humanos , Receptores de GABA-A/genética , Ácido gamma-Aminobutírico/farmacología
10.
J Cell Biochem ; 118(7): 1889-1899, 2017 07.
Artículo en Inglés | MEDLINE | ID: mdl-28121023

RESUMEN

Temozolomide (TMZ)-based chemotherapy is a standard strategy for glioma, while chemoresistance remains a major therapeutic challenge. Recent evidence highlights the crucial regulatory roles of long non-coding RNAs (lncRNA) in tumor biology. However, the roles and regulatory mechanisms of lncRNA cancer susceptibility candidate 2 (CASC2), in glioma tumorigenesis and chemoresistance are poorly understood. In this study, CASC2 expression was down-regulated in glioma tissues and cell lines, and was related to a clinicopathologic features and shorter survival time. Exogenous CACS2 alone was sufficient to inhibit glioma cells' proliferation and amplified TMZ-induced repression of cell proliferation, while CACS2 knockdown could reverse this process. CACS2 overexpression could sensitize TMZ-resistant glioma cells to TMZ, while CACS2 knockdown exerted the opposite function. Moreover, CASC2 could inhibit the miR-181a expression by direct targeting in TMZ-resistant glioma cells. CASC2 up-regulated PTEN protein and down-regulated p-AKT protein through regulating miR-181a, and the effect of CASC2 on PTEN and p-AKT could be partially restored by miR-181a. With TMZ-resistant glioma tissues, miR-181a was up-regulated while PTEN was down-regulated. Taken together, these observations suggest CASC2 up-regulates PTEN through direct inhibiting miR-181a and plays an important role in glioma sensitivity to TMZ and may serve as a potential target for cancer diagnosis and treatment. J. Cell. Biochem. 118: 1889-1899, 2017. © 2017 Wiley Periodicals, Inc.


Asunto(s)
Dacarbazina/análogos & derivados , Glioma/tratamiento farmacológico , Glioma/metabolismo , MicroARNs/metabolismo , Fosfohidrolasa PTEN/metabolismo , ARN Largo no Codificante/metabolismo , Proteínas Supresoras de Tumor/metabolismo , Adulto , Western Blotting , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Proliferación Celular/genética , Dacarbazina/farmacología , Dacarbazina/uso terapéutico , Femenino , Humanos , Técnicas In Vitro , Masculino , MicroARNs/genética , Persona de Mediana Edad , Análisis Multivariante , Fosfohidrolasa PTEN/genética , Modelos de Riesgos Proporcionales , Unión Proteica , ARN Largo no Codificante/genética , Reacción en Cadena en Tiempo Real de la Polimerasa , Temozolomida , Proteínas Supresoras de Tumor/genética
11.
Cell Physiol Biochem ; 42(2): 743-752, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28624824

RESUMEN

AIM: The present study aimed to examine the effect of tumor necrosis factor-α (TNF-α) inhibition on bone marrow-derived mesenchymal stem cells (BMSCs) in neurological function recovery after spinal cord injury (SCI) via the Wnt signaling pathway in a rat model. METHODS: The rat model of SCI was established using Allen's method. Seventy-two adult male Sprague Dawley (SD) rats were randomly assigned into 4 groups (18 rats in each group): the sham control group, saline control group, BMSCs group (injection with BMSCs at the injured site) and BMSCs + TNF-α group (injection with BMSCs under TNF-α treatment at the injured site). Immunochemistry was performed to characterize the culture media after TNF-α-induced differentiation. qRT-PCR and Western blotting analyses were performed to detect the mRNA and protein expression of ß-catenin, Wnt3a, GSK-3ß and Axin. The Basso Beattie Bresnahan (BBB) locomotor score, neurological deficit score (NDS), and balance beam test (BBT) score were used to assess neurological functional recovery of SCI rats. RESULTS: In the BMSC group, numerous spherical cell clusters grew in suspension, and the cells were nestin-, NF200- and GFAP-positive. Compared with the sham control and BMSC groups, the ß-catenin and Wnt3a mRNA and protein expression was increased, but the GSK-3ß and Axin mRNA and protein expression was decreased in the BMSCs + TNF-α group. The SCI rats in the BMSCs + TNF-α group exhibited lower BBB scores, and higher NDSs and BBT scores compared to the BMSCs group. CONCLUSION: Our study provides evidence that TNF-α inhibition may weaken the ability of BMSCs in neurological functional recovery after SCI by activating the Wnt signaling pathway.


Asunto(s)
Trasplante de Células Madre Mesenquimatosas , Células Madre Mesenquimatosas/metabolismo , Traumatismos de la Médula Espinal/terapia , Factor de Necrosis Tumoral alfa/genética , Animales , Médula Ósea/metabolismo , Diferenciación Celular/genética , Humanos , Ratas , Recuperación de la Función/genética , Traumatismos de la Médula Espinal/genética , Traumatismos de la Médula Espinal/patología , Factor de Necrosis Tumoral alfa/antagonistas & inhibidores , Vía de Señalización Wnt/genética , beta Catenina/genética
12.
Cell Physiol Biochem ; 39(6): 2173-2185, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27802437

RESUMEN

BACKGROUND: Valproic acid (VPA), an established antiepileptic drug, was assessed for antitumor activity, including its effects on glioblastoma, but its role has not been determined. METHODS: In the present study, we investigated VPA-induced apoptosis effects on human U87 cells by cell viability, lactate dehydrogenase (LDH) release, TUNEL/Hoechst staining and flow cytometric in vitro, then we further explored the underlying molecular mechanisms using the selective antagonists PD98059, LY294002 and SB216763. RESULTS: The data showed that VPA dose-dependent induction of glioma U87 cells to undergo apoptosis through the mitochondria-dependent pathway in vitro. VPA activated the ERK/Akt pathways by increasing their protein phosphorylation and in turn inhibited GKS3ß activation by the induction of GKS3ß phosphorylation. However, the MAPK inhibitor PD98059 and/or PI3K inhibitor LY294002 were able to antagonize the effects of VPA by abolishing ERK/Akt activations and cancelling GSK3ß suppression, thus it impaired VPA apoptosis-inducing effects on glioma cells. Furthermore, the GSK3ß inhibitor SB216763 caused a strong suppression of GSK3ß activity, which showed similar effects of VPA on regulation of protein expression and apoptosis. CONCLUSION: These findings suggest that GSK3ß may be the central hub for VPA-induced apoptosis and VPA can be further evaluated as a novel agent for glioma therapy.


Asunto(s)
Apoptosis/efectos de los fármacos , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Glioma/enzimología , Glioma/patología , Glucógeno Sintasa Quinasa 3 beta/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Ácido Valproico/farmacología , Neoplasias Encefálicas/enzimología , Neoplasias Encefálicas/patología , Línea Celular Tumoral , Activación Enzimática/efectos de los fármacos , Citometría de Flujo , Glucógeno Sintasa Quinasa 3 beta/antagonistas & inhibidores , Humanos , Mitocondrias/efectos de los fármacos , Mitocondrias/metabolismo , Modelos Biológicos , Inhibidores de Proteínas Quinasas/farmacología , Transducción de Señal/efectos de los fármacos
13.
Arch Biochem Biophys ; 610: 1-7, 2016 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-27693036

RESUMEN

H19, one of the first found cancer-associated long non-coding RNAs (lncRNAs), is involved in the development and progression of many types of tumors. An aberrant expression of H19 was observed in hepatocellular carcinoma, cervical cancer, breast cancer, ovarian cancer, and colorectal cancer. However, the exact effects and molecular mechanisms of H19 in glioma progression are still unknown up to now. In this study, we investigated the role of H19 in human glioma cell lines and clinical tumor samples in order to determine the function of this molecule. In our research, lncRNA-H19 was specifically upregulated in glioma cell lines and promoted glioma cell growth through targeting miR-140. Knockdown of H19 inhibited the proliferation and invasion of human glioma cell and suppressed its metastasis in vitro and in vivo. In addition, miR-140 dependent inhibitor of apoptosis-stimulating protein of p53 (iASPP) regulation was required in H19 induced glioma cell growth. These findings indicated that H19 might regulate the tumor growth and metastasis via miR-140 dependent iASPP regulation. Taken together, our data indicated that H19 might be an oncogenic lncRNA that promoted proliferation and metastasis of glioma and could be regarded as a therapeutic target in human glioma.


Asunto(s)
Neoplasias Encefálicas/metabolismo , Glioma/metabolismo , Péptidos y Proteínas de Señalización Intracelular/metabolismo , MicroARNs/genética , ARN Largo no Codificante/genética , Proteínas Represoras/metabolismo , Neoplasias Encefálicas/genética , Línea Celular Tumoral , Proliferación Celular , Regulación Neoplásica de la Expresión Génica , Glioma/genética , Humanos , Metástasis de la Neoplasia
14.
Cell Physiol Biochem ; 35(2): 419-32, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25613036

RESUMEN

BACKGROUND: Metabotropic glutamate receptors (mGluRs) are G-protein-coupled receptors that mediate neuronal excitability and synaptic plasticity in the central nervous system, and emerging evidence suggests a role of mGluRs in the biology of cancer. Previous studies showed that mGluR1 was a potential therapeutic target for the treatment of breast cancer and melanoma, but its role in human glioma has not been determined. METHODS: In the present study, we investigated the effects of mGluR1 inhibition in human glioma U87 cells using specific targeted small interfering RNA (siRNA) or selective antagonists Riluzole and BAY36-7620. The anti-cancer effects of mGluR1 inhibition were measured by cell viability, lactate dehydrogenase (LDH) release, TUNEL staining, cell cycle assay, cell invasion and migration assays in vitro, and also examined in a U87 xenograft model in vivo. RESULTS: Inhibition of mGluR1 significantly decreased the cell viability but increased the LDH release in a dose-dependent fashion in U87 cells. These effects were accompanied with the induction of caspase-dependent apoptosis and G0/G1 cell cycle arrest. In addition, the results of Matrigel invasion and cell tracking assays showed that inhibition of mGluR1 apparently attenuated cell invasion and migration in U87 cells. All these anti-cancer effects were ablated by the mGluR1 agonist L-quisqualic acid. The results of western blot analysis showed that mGluR1 inhibition overtly decreased the phosphorylation of PI3K, Akt, mTOR and P70S6K, indicating the mitigated activation of PI3K/Akt/mTOR pathway. Moreover, the anti-tumor activity of mGluR1 inhibition in vivo was also demonstrated in a U87 xenograft glioma model in athymic nude mice. CONCLUSION: The remarkable efficiency of mGluR1 inhibition to induce cell death in U87 cells may find therapeutic application for the treatment of glioma patients.


Asunto(s)
Antineoplásicos/administración & dosificación , Neoplasias Encefálicas/tratamiento farmacológico , Glioma/tratamiento farmacológico , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Receptores de Glutamato Metabotrópico/antagonistas & inhibidores , Animales , Antineoplásicos/farmacología , Neoplasias Encefálicas/metabolismo , Ciclo Celular/efectos de los fármacos , Línea Celular Tumoral , Movimiento Celular/efectos de los fármacos , Glioma/metabolismo , Humanos , Ratones , Ratones Desnudos , Terapia Molecular Dirigida , Naftalenos/administración & dosificación , Naftalenos/farmacología , Ácido Quiscuálico/farmacología , ARN Interferente Pequeño/administración & dosificación , ARN Interferente Pequeño/farmacología , Receptores de Glutamato Metabotrópico/metabolismo , Riluzol/administración & dosificación , Riluzol/farmacología , Ensayos Antitumor por Modelo de Xenoinjerto
15.
Biochem Biophys Res Commun ; 457(2): 171-6, 2015 Feb 06.
Artículo en Inglés | MEDLINE | ID: mdl-25542152

RESUMEN

Glioblastoma (GBM) is the most lethal brain tumor due to the resistance to conventional therapies, such as radiotherapy and chemotherapy. TAZ, an important mediator of the Hippo pathway, was found to be up-regulated in diverse cancers, including in GBM, and plays important roles in tumor initiation and progression. However, little is known about the regulation of TAZ expression in tumors. In this study, we found that miR-125a-5p is an important regulator of TAZ in glioma cells by directly targeting the TAZ 3' UTR. MiR-125a-5p levels are inversely correlated with that of TAZ in normal astrocytes and a panel of glioma cell lines. MiR-125a-5p represses the expression of TAZ target genes, including CTGF and survivin, and inhibits cell proliferation and induces the differentiation of GBM cells; whereas over-expression of TAZ rescues the effects of miR-125a-5p. This study revealed a mechanism for TAZ deregulation in glioma cells, and also demonstrated a tumor suppressor role of miR-125a-5p in glioblastoma cells.


Asunto(s)
Neoplasias Encefálicas/genética , Neoplasias Encefálicas/patología , Diferenciación Celular , Glioblastoma/genética , Glioblastoma/patología , Péptidos y Proteínas de Señalización Intracelular/genética , MicroARNs/metabolismo , Regiones no Traducidas 3'/genética , Secuencia de Bases , Diferenciación Celular/genética , Línea Celular Tumoral , Proliferación Celular , Regulación Neoplásica de la Expresión Génica , Genes Relacionados con las Neoplasias , Humanos , Péptidos y Proteínas de Señalización Intracelular/metabolismo , MicroARNs/genética , Datos de Secuencia Molecular , ARN Mensajero/genética , ARN Mensajero/metabolismo , Transactivadores , Factores de Transcripción , Proteínas Coactivadoras Transcripcionales con Motivo de Unión a PDZ
16.
Mol Cell Biochem ; 409(1-2): 103-11, 2015 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-26209061

RESUMEN

Glioblastoma multiforme (GBM) is the most malignant and common brain tumor; it is aggressive growth pattern means that GBM patients face a poor prognosis even when receiving the best available treatment modalities. In recent years, an increasing number of reports suggest that the discovery of microRNAs (miRNAs) might provide a novel therapeutic target for human cancers, including GBM. One miRNA in particular, microRNA-25 (miR-25), is overexpressed in several cancers, wherein accumulating evidence indicates that it functions as an oncogene. However, the function of miR-25 in GBM has not been totally elucidated. In this study, we demonstrated that miR-25 was significantly up-regulated in astrocytoma tissues and glioblastoma cell lines. In vitro studies further demonstrated that overexpressed miR-25 was able to promote, while its antisense oligos inhibited cell proliferation and invasion in U251 cells. Moreover, we identified neurofilament light polypeptide (NEFL) as a novel target molecule of miR-25. Also of note was the fact that NEFL was down-regulated with increased levels of miR-25 expression in human astrocytoma clinical specimens. In addition, via the mTOR signaling pathway, NEFL-siRNA could significantly attenuate the inhibitory effects of knockdown miR-25 on the proliferation and invasion of U251 cells. Overall, our results showed an important role for miR-25 in regulating NEFL expression in GBM, and suggest that miR-25 could be a potential target for GBM treatment.


Asunto(s)
Neoplasias Encefálicas/genética , Glioblastoma/genética , MicroARNs/genética , Proteínas de Neurofilamentos/metabolismo , Serina-Treonina Quinasas TOR/metabolismo , Línea Celular Tumoral , Proliferación Celular/genética , Regulación Neoplásica de la Expresión Génica , Células HEK293 , Humanos , MicroARNs/biosíntesis , Invasividad Neoplásica/genética , Proteínas de Neurofilamentos/genética , Interferencia de ARN , ARN Interferente Pequeño/genética
17.
Biochem Biophys Res Commun ; 443(1): 138-43, 2014 Jan 03.
Artículo en Inglés | MEDLINE | ID: mdl-24284040

RESUMEN

Glutamate-mediated excitotoxicity is involved in many acute and chronic brain diseases. Dynamin related protein 1 (Drp-1), one of the GTPase family of proteins that regulate mitochondrial fission and fusion balance, is associated with apoptotic cell death in cancer and neurodegenerative diseases. Here we investigated the effect of downregulating Drp-1 on glutamate excitotoxicity-induced neuronal injury in HT22 cells. We found that downregulation of Drp-1 with specific small interfering RNA (siRNA) increased cell viability and inhibited lactate dehydrogenase (LDH) release after glutamate treatment. Downregulation of Drp-1 also inhibited an increase in the Bax/Bcl-2 ratio and cleavage of caspase-9 and caspase-3. Drp-1 siRNA transfection preserved the mitochondrial membrane potential (MMP), reduced cytochrome c release, enhanced ATP production, and partly prevented mitochondrial swelling. In addition, Drp-1 knockdown attenuated glutamate-induced increases of cytoplasmic and mitochondrial Ca(2+), and preserved the mitochondrial Ca(2+) buffering capacity after excitotoxicity. Taken together, these results suggest that downregulation of Drp-1 protects HT22 cells against glutamate-induced excitatory damage, and this neuroprotection may be dependent at least in part on the preservation of mitochondrial function through regulating intracellular calcium homeostasis.


Asunto(s)
Apoptosis/fisiología , Calcio/metabolismo , Dinaminas/metabolismo , Ácido Glutámico/metabolismo , Mitocondrias/fisiología , Animales , Apoptosis/efectos de los fármacos , Línea Celular , Regulación hacia Abajo , Dinaminas/genética , Ácido Glutámico/farmacología , Ratones , Mitocondrias/efectos de los fármacos , Mitocondrias/metabolismo , ARN Interferente Pequeño/genética
18.
Zhonghua Wai Ke Za Zhi ; 52(7): 508-13, 2014 Jul.
Artículo en Zh | MEDLINE | ID: mdl-25262607

RESUMEN

OBJECTIVE: To identify factors that predictive of quality of life after microsurgical removal of petroclival meningiomas. METHODS: A consecutive series of 71 cases of petroclival meningiomas received microsurgical removal between July 1991 and April 2010 were analyzed retrospectively. Quality of life was measured using Karnofsky performance scale (KPS). Complete pre-operative, post-operative and follow-up data were obtained from all 71 patients including 18 male and 53 female patients with the mean age of (47 ± 11) years (aging from 15 to 68 years). The duration between onset of symptoms and diagnosis ranged from 1 week to 180 months with the mean duration of (32 ± 30) months. And the tumor size was 15-72 mm with the average of (44 ± 11) mm. Main presentations included headache, unsteady gait, hemiparesis, dysphagia, hoarseness, facial numbness or pain, Bell's palsy, hearing impairment etc. The preoperative KPS was 40-100 with the average of 69 ± 11. The retrosigmoid (-transtentorial) approach was performed in most cases (91.5%). Intergroup χ² test and logistic regression analysis were conducted for prognostic factor characterization. RESULTS: The gross total resection (all were Simpson gradeII) reached in 48 cases (67.6%) and 1 case died postoperatively. The main new neurological dysfunctions were cranial nerve paralysis and hemiplegia with the postoperative KPS of 20-100 with the average of 73 ± 16.Sixty-four cases were followed for 4-132 months with the average of (61 ± 48) months. Seven patients died during follow-up, tumor recurrence and progression were identified in 6 and 8 cases, respectively. The KPS at the last visit ranged from 50 to 100 with the average of 83 ± 13. The extent of tumor resection (OR = 0.280, 95% CI: 0.081-0.967, P = 0.044), preoperative brainstem edema (OR = 0.100, 95% CI: 0.027-0.372, P = 0.001), relationships between tumor and neurovascular structures (OR = 0.288, 95% CI: 0.084-0.985, P = 0.047) and depth of invasion into cavernous sinus (OR = 0.254, 95% CI: 0.061-1.057, P = 0.048) had significant correlations with the prognostic quality of life. CONCLUSIONS: With regard of the choice of surgical approaches, the extent of tumor resection, the protection of neurovascular structures surrounding the tumor and the management of perioperative period, the therapeutic strategies for each patient should be customized to achieve better prognosis.


Asunto(s)
Neoplasias Meníngeas/cirugía , Microcirugia , Adolescente , Adulto , Anciano , Femenino , Estudios de Seguimiento , Humanos , Masculino , Neoplasias Meníngeas/diagnóstico , Persona de Mediana Edad , Pronóstico , Calidad de Vida , Estudios Retrospectivos , Resultado del Tratamiento , Adulto Joven
19.
Zhong Nan Da Xue Xue Bao Yi Xue Ban ; 39(2): 136-41, 2014 Feb.
Artículo en Zh | MEDLINE | ID: mdl-24608386

RESUMEN

OBJECTIVE: To improve the surgical outcome of pituitary adenomas by identifying and preserving the pituitary stalk and the gland during surgery. METHODS: From October 2010 to September 2012, the author from the Department of Neurosurgery of Xiangya Hospital, Central South University operated on 51 patients with pituitary adenoma. During the operations, we carefully identified the normal adenohypophysis, pituitary stalk, neurohypophysis and the abnormal tissues either by direct observation or by medical images, aiming to excise the tumor thoroughly, protect the pituitary function and reduce the postoperative complications. RESULTS: Totally 37 patients (72.5%, 37/51) had total resection of the tumor, 12 (23.5%, 12/51) had subtotal tumor resection and the other 2 had major removal. The gland and the pituitary stalk were well identified and reserved. Detection of hormone content proved that the operation had little effect on the free triiodothyronine (FT3) and adrenocorticotropic hormone (ACTH), while for free tetraiodothyronine (FT4) and thyroid stimulating hormone (TSH) and postoperative followup significant alleviation was found. There was no significant fluctuation for the testosterone in the men preoperatively and postoperatively (all the above results were obtained without hormone replacement therapy). The main postoperative complications were as follows: temporary diabetes insipidus in 5 patients (9.8%, 5/51); electrolyte disorder (the appearance of hyponatremia) in 17 (33.3%, 17/51); and cerebrospinal fluid rhinorrhea and postoperative intracranial infection in 1 (2%, 1/51). No one died during the perioperation period. CONCLUSION: Microscopic transsphenoidal surgery is effective for pituitary adenomas including tumors violating the cavernous sinus. Accurate identification of the pituitary stalk, the gland and the abnormal tissue during the microscopic transsphenoidal operation plays a critical role in preserving the pituitary function and promoting postoperative rehabilitation.


Asunto(s)
Adenoma/cirugía , Microcirugia , Procedimientos Neuroquirúrgicos/métodos , Hipófisis/cirugía , Neoplasias Hipofisarias/cirugía , Humanos , Masculino , Hormonas Hipofisarias/sangre , Complicaciones Posoperatorias , Resultado del Tratamiento
20.
Genes Dis ; 11(3): 100977, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38292193

RESUMEN

Glioma is a common tumor originating in the brain that has a high mortality rate. Temozolomide (TMZ) is the first-line treatment for high-grade gliomas. However, a large proportion of gliomas are resistant to TMZ, posing a great challenge to their treatment. In the study, the specific functions and mechanism(s) by which cortistatin (CORT) regulates TMZ resistance and glioma progression were evaluated. The decreased expression of CORT was detected in glioma tissues, and highly expressed CORT was associated with a better survival rate in patients with glioma. CORT overexpression notably decreased the capacity of glioma cells to proliferate and migrate in vitro and to form tumors in vivo. CORT overexpression also markedly suppressed the viability and enhanced the apoptosis of TMZ-resistant U251 cells by regulating MGMT, p21, and Puma expression. Importantly, CORT overexpression reduced the resistance of gliomas to TMZ in vivo. CORT expression was negatively correlated with MGMT expression in both glioma tissues and cells, and it was found that CORT inhibited NF-κB pathway activation in glioma cells, thereby inhibiting MGMT expression. In conclusion, CORT regulates glioma cell growth, migration, apoptosis, and TMZ resistance by weakening the activity of NF-κB/p65 and thereby regulating MGMT expression. The CORT/NF-κB/MGMT axis might be regarded as a molecular mechanism contributing to the resistance of glioma to TMZ. Our data also suggest that CORT regulates the viability and metastatic potential of glioma cells, independent of its effects on TMZ resistance, providing evidence of novel therapeutic targets for glioma that should be evaluated in further studies.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA