Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Hereditas ; 160(1): 29, 2023 Jun 22.
Artículo en Inglés | MEDLINE | ID: mdl-37349788

RESUMEN

BACKGROUND: Glioma stem cells (GSCs) are responsible for glioma recurrence and drug resistance, yet the mechanisms underlying their maintenance remains unclear. This study aimed to identify enhancer-controlled genes involved in GSCs maintenance and elucidate the mechanisms underlying their regulation. METHODS: We analyzed RNA-seq data and H3K27ac ChIP-seq data from GSE119776 to identify differentially expressed genes and enhancers, respectively. Gene Ontology analysis was performed for functional enrichment. Transcription factors were predicted using the Toolkit for Cistrome Data Browser. Prognostic analysis and gene expression correlation was conducted using the Chinese Glioma Genome Atlas (CGGA) data. Two GSC cell lines, GSC-A172 and GSC-U138MG, were isolated from A172 and U138MG cell lines. qRT-PCR was used to detect gene transcription levels. ChIP-qPCR was used to detect H3K27ac of enhancers, and binding of E2F4 to target gene enhancers. Western blot was used to analyze protein levels of p-ATR and γH2AX. Sphere formation, limiting dilution and cell growth assays were used to analyze GSCs growth and self-renewal. RESULTS: We found that upregulated genes in GSCs were associated with ataxia-telangiectasia-mutated-and-Rad3-related kinase (ATR) pathway activation, and that seven enhancer-controlled genes related to ATR pathway activation (LIN9, MCM8, CEP72, POLA1, DBF4, NDE1, and CDKN2C) were identified. Expression of these genes corresponded to poor prognosis in glioma patients. E2F4 was identified as a transcription factor that regulates enhancer-controlled genes related to the ATR pathway activation, with MCM8 having the highest hazard ratio among genes positively correlated with E2F4 expression. E2F4 bound to MCM8 enhancers to promote its transcription. Overexpression of MCM8 partially restored the inhibition of GSCs self-renewal, cell growth, and the ATR pathway activation caused by E2F4 knockdown. CONCLUSION: Our study demonstrated that E2F4-mediated enhancer activation of MCM8 promotes the ATR pathway activation and GSCs characteristics. These findings offer promising targets for the development of new therapies for gliomas.


Asunto(s)
Glioma , Humanos , Glioma/genética , Glioma/metabolismo , Factores de Transcripción/metabolismo , Proliferación Celular/genética , Células Madre Neoplásicas/metabolismo , Proteínas de Mantenimiento de Minicromosoma/metabolismo , Factor de Transcripción E2F4/metabolismo , Proteínas Asociadas a Microtúbulos , Proteínas de la Ataxia Telangiectasia Mutada/metabolismo
2.
CNS Neurosci Ther ; 28(12): 2319-2330, 2022 12.
Artículo en Inglés | MEDLINE | ID: mdl-36184801

RESUMEN

AIMS: We aimed to investigate the role of receptor-interacting protein 2 (RIP2) in regulation of stemness of glioma cells and chemotherapy resistance. METHODS: Plasmid transfection was used to overexpress RIP2. Chemical inhibitors were used to inhibit RIP2 or NF-κB activity. Cancer stemness of glioma cells was investigated by sphere formation assays, clone formation assays, and xenograft tumor formation assays. The expression of RIP2, p-NF-κB, IκBα, CD133, or SOX-2 was detected by Western blotting and immunofluorescence. Apoptosis was detected by flow cytometry. Immunohistochemical staining was used to detect the expression of RIP2, CD133, and SOX-2 in xenograft tumor tissue. The effect of the RIP2/NF-κB pathway on temozolomide (TMZ) resistance was evaluated by xenograft tumor assay. RESULTS: Transfection with RIP2 plasmid enhanced the sphere formation capability of U251 cells, clone formation capability, and xenograft tumor formation capability. RIP2 could mediate TMZ resistance by upregulating the expression of CD133 and SOX-2 by activating the NF-κB pathway. Both RIP2 inhibitor GSK583 and the NF-κB inhibitor SC75741 could reverse the resistance of U251 cells to TMZ. CONCLUSION: RIP2 mediates TMZ resistance by regulating the maintenance of stemness in glioma cells through NF-κB. Interventions targeting the RIP2/NF-κB pathway may be a new strategy for TMZ-resistant gliomas.


Asunto(s)
Neoplasias Encefálicas , Glioma , Células Madre Neoplásicas , Proteína Serina-Treonina Quinasa 2 de Interacción con Receptor , Humanos , Neoplasias Encefálicas/metabolismo , Línea Celular Tumoral , Resistencia a Antineoplásicos , Glioma/metabolismo , FN-kappa B/metabolismo , Temozolomida/uso terapéutico , Animales , Proteína Serina-Treonina Quinasa 2 de Interacción con Receptor/genética
3.
Exp Ther Med ; 22(3): 924, 2021 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-34306193

RESUMEN

Proinflammatory polarization of microglia aggravates brain injury in cerebral infarction. The present study focused on the role of long non-coding (lnc)RNA X-inactive specific transcript (XIST) in the phenotype modulation of microglia. It was revealed that lncRNA XIST was significantly upregulated in both a mouse cerebral infarction model induced by middle cerebral artery occlusion (MCAO) and an activated microglial model induced by oxygen/glucose deprivation (OGD). The overexpression of XIST enhanced the expression and release of pro-inflammatory mediators [such as tumor necrosis factor (TNF)-α, IL-6, and iNOS] in microglia. Culture supernatant from lncRNA XIST-overexpressed microglial cells induced the apoptosis of primary neurons, while TNF-α antibody counteracted this neurotoxic effect. LncRNA XIST served as a sponge for miR-96-5p, counteracting its inhibitory effect on IKKß/NF-κB signaling and TNF-α production. Notably, TNF-α was positively regulated by XIST and in turn enhanced XIST expression in microglia. The lncRNA XIST-TNF-α feedback promoted the proinflammatory polarization of microglia, thereby exacerbating cerebral neuron apoptosis.

4.
CNS Neurol Disord Drug Targets ; 18(1): 78-87, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-30394221

RESUMEN

BACKGROUND AND OBJECTIVE: Exosomes communicate inter-cellularly and miRNAs play critical roles in this scenario. MiR-214-5p was implicated in multiple tumors with diverse functions uncovered. However, whether miR-214-5p is mechanistically involved in glioblastoma, especially via exosomal pathway, is still elusive. Here we sought to comprehensively address the critical role of exosomal miR-214-5p in glioblastoma (GBM) microenvironment. METHODS: The relative expression of miR-214-5p was determined by real-time PCR. Cell viability and migration were measured by MTT and transwell chamber assays, respectively. The secretory cytokines were measured with ELISA kits. The regulatory effect of miR-214-5p on CXCR5 expression was interrogated by luciferase reporter assay. Protein level was analyzed by Western blot. RESULTS: We demonstrated that miR-214-5p was aberrantly overexpressed in GBM and associated with poorer clinical prognosis. High level of miR-214-5p significantly contributed to cell proliferation and migration. GBM-derived exosomal miR-214-5p promoted inflammatory response in primary microglia upon lipopolysaccharide challenge. We further identified CXCR5 as the direct target of miR-214- 5p in this setting. CONCLUSION: Overexpression of miR-214-5p in GBM modulated the inflammatory response in microglia via exosomal transfer.


Asunto(s)
Glioblastoma/metabolismo , Inflamación/metabolismo , MicroARNs/metabolismo , Microglía/metabolismo , Receptores CXCR5/metabolismo , Línea Celular Tumoral , Movimiento Celular/fisiología , Supervivencia Celular/fisiología , Células Cultivadas , Exosomas/metabolismo , Glioblastoma/fisiopatología , Humanos , Inflamación/inducido químicamente , Interleucina-6/metabolismo , Interleucina-8/metabolismo , Lipopolisacáridos , Cultivo Primario de Células , Factor de Necrosis Tumoral alfa/metabolismo
5.
Oncol Lett ; 16(2): 2525-2532, 2018 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-30013647

RESUMEN

Radiation therapy is important for the comprehensive treatment of intracranial tumors. However, the molecular mechanisms underlying the pathogenesis of delayed cognitive dysfunction are not well-defined and effective treatments or prevention measures remain insufficient. In the present study, 60 adult male Wistar rats were randomly divided into three groups, which included a control, whole brain radiotherapy (WBRT) (single dose of 30 Gy of WBRT) and nimodipine (single dose of 30 Gy of WBRT followed by nimodipine injection intraperitoneally) groups. The rats were sacrificed 7 days or 3 months following irradiation. At 3 months, the Morris water maze test was used to assess spatial learning and memory function in rats. The results demonstrated that the WBRT group demonstrated a significantly impaired cognitive performance, decreased numbers of hippocampal Cornu Ammonis (CA)1 neurons and upregulated expression of caspase-3 in the dentate gyrus compared with those in the control and nimodipine groups. Reverse transcription-quantitative polymerase chain reaction analysis demonstrated that the WBRT group exhibited increased ratio of B-cell lymphoma 2 (Bcl-2)-associated X protein (Bax)/Bcl-2 compared with that in control and nimodipine groups on day 7 following irradiation. However, the WBRT group exhibited decreased levels of brain-derived neurotrophic factor (BDNF) compared with that in control and nimodipine groups at 3 months following brain irradiation. The levels of growth-associated protein 43 and amyloid precursor protein between the nimodipine group and WBRT group were not statistically significant. The present study demonstrated that neuron apoptosis may lead to delayed cognitive deficits in the hippocampus, in response to radiotherapy. The cognitive impairment may be alleviated in response to a calcium antagonist nimodipine. The molecular mechanisms involved in nimodipine-mediated protection against cognitive decline may involve the regulation of Bax/Bcl-2 and BDNF in the hippocampus.

6.
Biochem Biophys Res Commun ; 494(3-4): 674-680, 2017 12 16.
Artículo en Inglés | MEDLINE | ID: mdl-29066350

RESUMEN

ZMYND11 (zinc finger MYND-type containing 11) has been widely regarded to be involved in a variety of cancers as a potential suppressor. However, the biological role and mechanism of ZMYND11 in glioblastoma multiform (GBM) remain unknown. In this study, we found that ZMYND11 expression was remarkably decreased in GBM tissues from 20 cases and cell line (U87) compared to normal brain tissue from 10 cases (P < 0.001). Furthermore, we explored that ZMYND11 upregulation significantly suppressed U87 cells proliferation and invasion, induced cell cycle arrest and apoptosis in vitro. Subsequently, we identified increased ZMYND11 inhibited the tumor growth using tumor cells xenograft experiment on rude mice. Moreover, we explored that ZMYND11 was a new direct and functional target of miR-196a-5p in U87 via luciferase reporter assay. In addition, we confirmed the negative correlation between miR-196a-5p and ZMYND11 in GBM tissue and U87 cells by changing the expression level of miR-196a-5p with lentivirus and plasmid vector. Furthermore, we demonstrated that decreased ZMYND11 could reverse suppressive effect of downregulated miR-196a-5p on U87 by rescue experiment. Taken together, ZMYND11 was demonstrated to be a potential and extremely promising suppressor of GBM, while miRNA-196a-5p was quite an important target of treatment of GBM.


Asunto(s)
Neoplasias Encefálicas/metabolismo , Neoplasias Encefálicas/patología , Proteínas Portadoras/metabolismo , Glioblastoma/metabolismo , Glioblastoma/patología , MicroARNs/metabolismo , Adulto , Anciano , Animales , Proteínas de Ciclo Celular , Línea Celular Tumoral , Proliferación Celular , Proteínas Co-Represoras , Proteínas de Unión al ADN , Regulación hacia Abajo , Femenino , Humanos , Masculino , Ratones , Ratones Desnudos , Persona de Mediana Edad , Invasividad Neoplásica
7.
Ther Adv Med Oncol ; 9(12): 741-754, 2017 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-29449895

RESUMEN

BACKGROUND: Glioblastoma multiforme (GBM) is the most aggressive and deadly primary brain cancer that arises from astrocytes and classified as grade IV. Recently, exosomes have been reported as an essential mediator in diverse cancer carcinogenesis and metastasis. However, their role in GBM is still unclear. In this study, we aimed to investigate whether blood exosomes can be potential clinical diagnostic markers for GBM. METHODS: We used a xenograft orthotopic mouse model to detect the differentially expressed genes in the brain and blood exosomes of original/recurrent GBM. RESULTS: We found that recurrent GBM had stronger growth capacity and lethality than original GBM in the mouse model. A gene microarray of original tumors and blood exosomes from GBM orthotopic xenografts results showed that DNM3, p65 and CD117 expressions increased, whereas PTEN and p53 expressions decreased in both original tumors and blood exosomes. In the recurrent GBM tumor model, DNM3 and p65 showed increased expressions, whereas ST14 and p53 showed decreased expressions in tumor and blood exosomes of the recurrent GBM mouse model. CONCLUSION: In summary, we found that DNM3, p65 and p53 had a similar trend in brain and blood exosomes both for original and recurrent GBM, and could serve as potential clinical diagnostic markers for GBM.

8.
J Neurooncol ; 131(2): 255-265, 2017 01.
Artículo en Inglés | MEDLINE | ID: mdl-27837435

RESUMEN

MicroRNA is an important regulator of glioblastoma. This study aims at validating microRNA-221 (miR-221) as a biomarker for glioblastoma, and understanding how miR-221 regulates glioblastoma progression. Using clinical samples, miR-221 expression was analyzed by quantitative reverse-transcriptase PCR (qPCR). SHG-44 cells were treated with anti-miR-221 or U87MG-derived exosomes followed by monitoring changes in cell viability, migration and temozolomide (TMZ) resistance. Bioinformatics approach was used to identify targets of miR-221. The interaction between miR-221 and its target, DNM3 gene, was studied with dual-luciferase reporter assay, Spearman's correlation analysis, and western blotting. To verify that RELA regulates miR-221 expression, RELA-expressing vector or shRNA was introduced into SHG-44 cells and its effect on miR-221 expression was monitored. Both tissue-level and exosomal miR-221 expression increased with glioma grades. In SHG-44 cells, downregulating miR-221 expression inhibited cell proliferation, migration, and TMZ resistance, whereas incubation with U87MG-derived exosomes exerted tumor-promoting effects. DNM3 gene is a target of miR-221. RELA induced miR-221 expression. In glioma, elevated miR-221 expression is a biomarker for glioma. DNM3 is a target of miR-221 and RELA regulates miR-221 expression. The RELA/miR-221 axis is a target for glioma diagnosis and therapy.


Asunto(s)
Antineoplásicos Alquilantes/uso terapéutico , Neoplasias Encefálicas/metabolismo , Dacarbazina/análogos & derivados , Dinamina III/metabolismo , Glioma/metabolismo , MicroARNs/metabolismo , Factor de Transcripción ReIA/metabolismo , Apoptosis , Neoplasias Encefálicas/tratamiento farmacológico , Línea Celular Tumoral , Proliferación Celular , Supervivencia Celular , Dacarbazina/uso terapéutico , Progresión de la Enfermedad , Resistencia a Antineoplásicos , Exosomas/metabolismo , Regulación Neoplásica de la Expresión Génica , Glioma/tratamiento farmacológico , Humanos , Temozolomida
9.
Int J Oncol ; 47(3): 1025-33, 2015 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-26165303

RESUMEN

Gliomas are the most common and aggressive types of tumors in human brain, of which the prognosis remains dismal because of their biological behavior. The involvement of miRNAs in tumorigenesis of various kinds of cancers drives us to explore new miRNAs related to gliomas. We measured expression level of miR­95­3p by qRT-PCR in human glioma and non-neoplasm brain tissues and found that higher level of miR­95­3p in glioma tissues of higher grade. Biological functions of miR­95­3p on glioma cells were investigated by MTT assay, flow cytometry and transwell assay. We discovered the cell lines transfected with miR­95­3p ASO (antisense oligonucleotide) had retarded proliferation and invasion but enhanced apoptosis ability. We searched on-line tool Targetscan and selected CELF (CUGBP- and ETR-3-like family 2) as a putative target. Luciferase reporter was employed to confirm the binding sites in 3'UTR region of CELF2 for miR­95­3p. The correlation between expression of CELF2 and miR­95­3p was determined by western blotting and qRT-PCR both in cell lines and human samples. Results showed CELF2 was a direct target of miR­95­3p and expression levels of CELF2 and miR­95­3p were negatively correlated. Finally, CELF2 largely abrogated the effects of miR­95­3p on proliferation, invasion and apoptosis of glioma cells in rescue experiments, which verified the role of CELF2 in miR­95­3p regulating glioma biological behavior. In conclusion, our data suggest the expression level of miR­95­3p is positively related to glioma grade and downregulation of miR­95­3p affects proliferation, invasion and apoptosis of glioma cells by targeting CELF2. We identified miR­95­3p as a putative therapeutic target and CELF2 as a potential tumor suppressor.


Asunto(s)
Neoplasias Encefálicas/genética , Neoplasias Encefálicas/patología , Proteínas CELF/genética , Glioma/genética , Glioma/patología , MicroARNs/genética , Proteínas del Tejido Nervioso/genética , Adulto , Sitios de Unión , Neoplasias Encefálicas/metabolismo , Proteínas CELF/química , Proteínas CELF/metabolismo , Línea Celular Tumoral , Proliferación Celular , Regulación hacia Abajo , Femenino , Regulación Neoplásica de la Expresión Génica , Glioma/metabolismo , Humanos , Masculino , MicroARNs/metabolismo , Persona de Mediana Edad , Invasividad Neoplásica , Proteínas del Tejido Nervioso/química , Proteínas del Tejido Nervioso/metabolismo , Regiones Promotoras Genéticas
10.
Chin Med J (Engl) ; 125(21): 3831-5, 2012 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-23106883

RESUMEN

BACKGROUND: In response to the injury of the central nervous system (CNS), the astrocytes upregulate the expression of glial fibrillary acidic protein (GFAP), which largely contributes to the reactive gliosis after brain injury. The regulatory mechanism of this process is still not clear. In this study, we aimed to compare the ephrin-B2 deficient mice with the wild type ones with regard to gliosis after traumatic brain injury. METHODS: We generated ephrin-B2 knockout mice specifically in CNS astrocytes. Twelve mice from this gene-knockout strain were randomly selected along with twelve mice from the wild type littermates. In both groups, a modified controlled cortical impact injury model was applied to create a closed traumatic brain injury. Twenty-eight days after the injury, Nissl staining and GFAP immunofluorescence staining were used to compare the brain atrophy and GFAP immunoreactivity between the two groups. All the data were analyzed by t-test for between-group comparison. RESULTS: We successfully set up the conditional ephrin-B2 knockout mice strain, which was confirmed by genotyping and ephrin-B2/GFAP double staining. These mice developed normally without apparent abnormality in general appearance. Twenty-eight days following brain injury, histopathology revealed by immunohistochemistry showed different degrees of cerebral injuries in both groups. Compared with wild-type group, the ephrin-B2 knockout group exhibited less brain atrophy ratio for the injured hemispheres (P = 0.005) and hippocampus (P = 0.027). Also the wild-type group demonstrated greater GFAP immunoreactivity increment within hippocampal regions (P = 0.008). CONCLUSIONS: The establishment of conditional ephrin-B2 knockout mice provides us with a new way to explore the role of ephrin-B2 in astrocytes. Our findings revealed less atrophy and GFAP immunoreactivity in the knockout mice strain after traumatic brain injury, which implied ephrin-B2 could be one of the promoters to upregulate gliosis following brain injury.


Asunto(s)
Lesiones Encefálicas/complicaciones , Efrina-B2/fisiología , Gliosis/etiología , Animales , Atrofia , Encéfalo/patología , Lesiones Encefálicas/patología , Efrina-B2/deficiencia , Proteína Ácida Fibrilar de la Glía , Inmunohistoquímica , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Proteínas del Tejido Nervioso/análisis
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA