Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 62
Filtrar
Más filtros

Banco de datos
Tipo del documento
País de afiliación
Intervalo de año de publicación
1.
J Cell Mol Med ; 27(18): 2730-2743, 2023 09.
Artículo en Inglés | MEDLINE | ID: mdl-37480215

RESUMEN

PKN1 (protein kinase N1), a serine/threonine protein kinase family member, is associated with various cancers. However, the role of PKN1 in gliomas has rarely been studied. We suggest that PKN1 expression in glioma specimens is considerably upregulated and positively correlates with the histopathological grading of gliomas. Knocking down PKN1 expression in glioblastoma (GBM) cells inhibits GBM cell proliferation, invasion and migration and promotes apoptosis. In addition, yes-associated protein (YAP) expression, an essential effector of the Hippo pathway contributing to the oncogenic role of gliomagenesis, was also downregulated. In contrast, PKN1 upregulation enhances the malignant characteristics of GBM cells and simultaneously upregulates YAP expression. Therefore, PKN1 is a promising therapeutic target for gliomas. Raloxifene (Ralo), a commonly used selective oestrogen-receptor modulator to treat osteoporosis in postmenopausal women, was predicted to target PKN1 according to the bioinformatics team from the School of Mathematics, Tianjin Nankai University. We showed that Ralo effectively targets PKN1, inhibits GBM cells proliferation and migration and sensitizes GBM cells to the major chemotherapeutic drug, Temozolomide. Ralo also reverses the effect of PKN1 on YAP activation. Thus, we confirm that PKN1 contributes to the pathogenesis of gliomas and may be a potential target for Ralo adjuvant glioma therapy.


Asunto(s)
Glioblastoma , Glioma , Femenino , Humanos , Clorhidrato de Raloxifeno/farmacología , Glioma/tratamiento farmacológico , Glioma/genética , Apoptosis , Proliferación Celular
2.
Pharmacol Res ; 187: 106606, 2023 01.
Artículo en Inglés | MEDLINE | ID: mdl-36516884

RESUMEN

Epidermal growth factor receptor variant III (EGFRvIII) is a mutant isoform of EGFR with a deletion of exons 2-7 making it insensitive to EGF stimulation and downstream signal constitutive activation. However, the mechanism underlying the stability of EGFRvIII remains unclear. Based on CRISPR-Cas9 library screening, we found that mucin1 (MUC1) is essential for EGFRvIII glioma cell survival and temozolomide (TMZ) resistance. We revealed that MUC1-C was upregulated in EGFRvIII-positive cells, where it enhanced the stability of EGFRvIII. Knockdown of MUC1-C increased the colocalization of EGFRvIII and lysosomes. Upregulation of MUC1 occurred in an NF-κB dependent manner, and inhibition of the NF-κB pathway could interrupt the EGFRvIII-MUC1 feedback loop by inhibiting MUC1-C. In a previous report, we identified AC1Q3QWB (AQB), a small molecule that could inhibit the phosphorylation of NF-κB. By screening the structural analogs of AQB, we obtained EPIC-1027, which could inhibit the NF-κB pathway more effectively. EPIC-1027 disrupted the EGFRvIII-MUC1-C positive feedback loop in vitro and in vivo, inhibited glioma progression, and promoted sensitization to TMZ. In conclusion, we revealed the pivotal role of MUC1-C in stabilizing EGFRvIII in glioblastoma (GBM) and identified a small molecule, EPIC-1027, with great potential in GBM treatment.


Asunto(s)
Neoplasias Encefálicas , Glioblastoma , Glioma , Humanos , Temozolomida/farmacología , Glioblastoma/tratamiento farmacológico , Glioblastoma/genética , Glioblastoma/metabolismo , FN-kappa B/metabolismo , Neoplasias Encefálicas/tratamiento farmacológico , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/metabolismo , Línea Celular Tumoral , Mucina-1/genética
3.
J Cell Mol Med ; 22(10): 4611-4616, 2018 10.
Artículo en Inglés | MEDLINE | ID: mdl-30073755

RESUMEN

Glioma has been regarded as the most common, highly proliferative and invasive brain tumour. Advances in research of miRNAs in glioma are toward further understanding of the pathogenesis of glioma. MiR-19, a member of miR-17~92 cluster, was reported to play an oncogenic role in tumourigenesis. Here we review the identified data about the effect of miR-19 on proliferation, apoptosis, migration and invasion of glioma cells, the target genes regulated by miR-19, and correlation of miR-19 with the sensitivity of glioma cells to chemotherapy and radiotherapy. It is concluded that miR-19 plays an important role in the pathogenesis of glioma and can be a potential target for gene therapy of glioma.


Asunto(s)
Neoplasias Encefálicas/genética , Carcinogénesis/genética , Regulación Neoplásica de la Expresión Génica , Glioma/genética , MicroARNs/genética , Proteínas de Neoplasias/genética , Antineoplásicos/uso terapéutico , Apoptosis , Neoplasias Encefálicas/metabolismo , Neoplasias Encefálicas/patología , Neoplasias Encefálicas/terapia , Carcinogénesis/metabolismo , Carcinogénesis/patología , Movimiento Celular , Proliferación Celular , Rayos gamma/uso terapéutico , Terapia Genética/métodos , Glioma/metabolismo , Glioma/patología , Glioma/terapia , Humanos , MicroARNs/antagonistas & inhibidores , MicroARNs/metabolismo , Invasividad Neoplásica , Proteínas de Neoplasias/metabolismo , Tolerancia a Radiación , Transducción de Señal
4.
J Neurooncol ; 140(1): 15-26, 2018 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-29916101

RESUMEN

INTRODUCTION: Runt-related transcription factor 3 (RUNX3) exerts a tumor suppressor gene associated with gastric and other cancers, including glioma. However, how its anti-tumor mechanism in functions glioma is unclear. METHODS: We assayed expression of RUNX3 with a tissue microarray (TMA), frozen cancer tissues and malignant glioma cell lines using immunohistochemistry, qRT-PCR and Western bolt analysis. Cell proliferation, invasion, cell cycle distribution and apoptosis were also examined to confirm the effect of RUNX3 medicated malignant phenotype. TOP/FOP experiment was used to detect the ß-catenin/Tcf-4 transcription activity by RUNX3. RESULTS: Enforced RUNX3 expression inhibited proliferation and invasion, induced cell cycle arrest and promoted apoptosis in vitro and in vivo, Bim siRNA partically reversed the effect of RUNX3-induced apoptosis in LN229 and U87 cells, suggesting a dependent role of Bim-caspase pathway. Moreover, Mechanism investigations revealed that restoration of RUNX3 suppressed ß-catenin/Tcf-4 transcription activity. CONCLUSIONS: RUNX3 plays a pivotal role in glioma initiation and progression as a tumor suppressor via attenuation of Wnt signaling, highlighting it as a potential therapeutic target for glioma.


Asunto(s)
Neoplasias Encefálicas , Subunidad alfa 3 del Factor de Unión al Sitio Principal/metabolismo , Glioma , Transducción de Señal/fisiología , Neoplasias Encefálicas/metabolismo , Neoplasias Encefálicas/patología , Neoplasias Encefálicas/fisiopatología , Regulación Neoplásica de la Expresión Génica , Glioma/metabolismo , Glioma/patología , Glioma/fisiopatología , Humanos , Invasividad Neoplásica/fisiopatología , Análisis de Matrices Tisulares , Factor de Transcripción 4/metabolismo , beta Catenina/metabolismo
5.
Biochem Biophys Res Commun ; 458(2): 307-12, 2015 Mar 06.
Artículo en Inglés | MEDLINE | ID: mdl-25656572

RESUMEN

We demonstrated that IKBKE is overexpressed in human gliomas and that the downregulation of IKBKE markedly inhibits the proliferative and invasive abilities of glioma cells, which is consistent with the results reported by several different research groups. Therefore, IKBKE represents a promising therapeutic target for the treatment of glioma. In the present study, we verified that the microRNAs let-7b and let-7i target IKBKE through luciferase assays and found that let-7b/i mimics can knock down IKBKE and upregulate E-cadherin through western blot analysis. Moreover, the expression levels of let-7b/i were significantly lower in glioma cell lines than that in normal brain tissues, as determined by quantitative real-time PCR. Furthermore, let-7b/i inhibit the invasion and migration of glioma cells, as determined through wound healing and Transwell assays. The above-mentioned data suggest that let-7b/i inhibit the invasive ability of glioma cells by directly downregulating IKBKE and indirectly upregulating E-cadherin.


Asunto(s)
Movimiento Celular/genética , Marcación de Gen/métodos , Glioblastoma/genética , Glioblastoma/patología , Quinasa I-kappa B/genética , MicroARNs/genética , Línea Celular Tumoral , Silenciador del Gen , Humanos , Invasividad Neoplásica/genética , Invasividad Neoplásica/patología
6.
Int J Oncol ; 64(2)2024 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-38063241

RESUMEN

Following the publication of the above article, a concerned reader drew to the Editor's attention that, regarding the western blots featured in Fig. 3B on p. 670, the bands featured in the U251 and U251­MC lanes for the miR­21 and U6 experiments appeared to be duplicates of each other. Moreover, certain of these data were strikingly similar to data that appeared in another article published at around the same time featuring some of the same authors (again, with apparent duplications of bands within the same gel slices, as they were presented). After having conducted an internal investigation of this matter, the Editor of International Journal of Oncology has judged that the apparently anomalous grouping of the data could not have been attributed to pure coincidence. Therefore, the Editor has decided that this article should be retracted from the publication on the grounds of an overall lack of confidence in the data. The authors were asked for an explanation to account for these concerns, but the Editorial Office did not receive a reply. The Editor sincerely apologizes to the readership for any incovenience caused, and we thank the reader for bringing this matter to our attention. [International Journal of Oncology 36: 665­672, 2010; DOI: 10.3892/ijo_00000542].

7.
J Neurooncol ; 112(2): 179-89, 2013 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-23377830

RESUMEN

Recently, many studies have found that the miR-106b ~25 cluster plays an oncogenic role in tumor progression. However, the precise role of each microRNAs (miRNAs) in the cluster is not yet clear. In the present study, we examined the expression of miR-106b in glioma samples and a tissue microarray by real-time PCR and in situ hybridization (ISH), respectively, finding that miR-106b is overexpressed in the majority of gliomas. Meanwhile, the expression of miR-106b was positively correlated with tumor grade (p < 0.05). The transfection of a miR-106b anti-sense oligonucleotide (ASON) into three human glioma cell lines (U251, LN229 and TJ905) suppressed the proliferation of these cells. Moreover, the growth of xenograft tumors in nude mice treated with miR-106b ASON was significantly impaired. A bioinformatics analysis predicted that RBL2 may be the target of miR-106b, and dual-luciferase reporter assays identified RBL2, but not RB1 or RBL1, as a target of miR-106b. These results suggest that miR-106b facilitates glioma cell growth by promoting cell cycle progression through the negative regulation of RBL2.


Asunto(s)
Neoplasias Encefálicas/prevención & control , Proliferación Celular , Regulación Neoplásica de la Expresión Génica , Glioma/prevención & control , MicroARNs/genética , Animales , Apoptosis , Western Blotting , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/patología , Femenino , Citometría de Flujo , Glioma/genética , Glioma/patología , Humanos , Hibridación in Situ , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Clasificación del Tumor , Oligonucleótidos Antisentido/farmacología , Pronóstico , ARN Mensajero/genética , Reacción en Cadena en Tiempo Real de la Polimerasa , Proteína de Retinoblastoma/genética , Proteína de Retinoblastoma/metabolismo , Proteína p107 Similar a la del Retinoblastoma/genética , Proteína p107 Similar a la del Retinoblastoma/metabolismo , Proteína p130 Similar a la del Retinoblastoma/genética , Proteína p130 Similar a la del Retinoblastoma/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Análisis de Matrices Tisulares , Ensayos Antitumor por Modelo de Xenoinjerto
8.
Cancer Biol Med ; 20(5)2023 06 05.
Artículo en Inglés | MEDLINE | ID: mdl-37283490

RESUMEN

OBJECTIVE: Epidermal growth factor receptor variant III (EGFRvIII) is a constitutively-activated mutation of EGFR that contributes to the malignant progression of glioblastoma multiforme (GBM). Temozolomide (TMZ) is a standard chemotherapeutic for GBM, but TMZ treatment benefits are compromised by chemoresistance. This study aimed to elucidate the crucial mechanisms leading to EGFRvIII and TMZ resistance. METHODS: CRISPR-Cas13a single-cell RNA-seq was performed to thoroughly mine EGFRvIII function in GBM. Western blot, real-time PCR, flow cytometry, and immunofluorescence were used to determine the chemoresistance role of E2F1 and RAD51-associated protein 1 (RAD51AP1). RESULTS: Bioinformatic analysis identified E2F1 as the key transcription factor in EGFRvIII-positive living cells. Bulk RNA-seq analysis revealed that E2F1 is a crucial transcription factor under TMZ treatment. Western blot suggested enhanced expression of E2F1 in EGFRvIII-positive and TMZ-treated glioma cells. Knockdown of E2F1 increased sensitivity to TMZ. Venn diagram profiling showed that RAD51AP1 is positively correlated with E2F1, mediates TMZ resistance, and has a potential E2F1 binding site on the promoter. Knockdown of RAD51AP1 enhanced the sensitivity of TMZ; however, overexpression of RAD51AP1 was not sufficient to cause chemotherapy resistance in glioma cells. Furthermore, RAD51AP1 did not impact TMZ sensitivity in GBM cells with high O6-methylguanine-DNA methyltransferase (MGMT) expression. The level of RAD51AP1 expression correlated with the survival rate in MGMT-methylated, but not MGMT-unmethylated TMZ-treated GBM patients. CONCLUSIONS: Our results suggest that E2F1 is a key transcription factor in EGFRvIII-positive glioma cells and quickly responds to TMZ treatment. RAD51AP1 was shown to be upregulated by E2F1 for DNA double strand break repair. Targeting RAD51AP1 could facilitate achieving an ideal therapeutic effect in MGMT-methylated GBM cells.


Asunto(s)
Neoplasias Encefálicas , Glioblastoma , Glioma , Humanos , Antineoplásicos Alquilantes/farmacología , Antineoplásicos Alquilantes/uso terapéutico , Neoplasias Encefálicas/tratamiento farmacológico , Neoplasias Encefálicas/genética , Factor de Transcripción E2F1/genética , Factor de Transcripción E2F1/metabolismo , Factor de Transcripción E2F1/uso terapéutico , Glioblastoma/tratamiento farmacológico , Glioblastoma/genética , Glioma/tratamiento farmacológico , O(6)-Metilguanina-ADN Metiltransferasa/genética , O(6)-Metilguanina-ADN Metiltransferasa/metabolismo , O(6)-Metilguanina-ADN Metiltransferasa/uso terapéutico , Temozolomida/farmacología , Temozolomida/uso terapéutico , Factores de Transcripción/metabolismo
9.
Mol Ther Oncolytics ; 20: 290-305, 2021 Mar 26.
Artículo en Inglés | MEDLINE | ID: mdl-33614912

RESUMEN

miR-19a/b belong to the miR-17-92 family. We have demonstrated previously that miR-19a/b are overexpressed in glioma and glioma cell lines. However, the role of miR-19a/b in glioma remains unclear. In the present study, we aim to identify the biological function and molecular mechanism of miR-19a/b in glioma cell proliferation and epithelial-mesenchymal transition (EMT). Knocking down miR-19a/b in LN308 glioblastoma (GBM) cells with higher expression of miR-19a/b inhibits cell proliferation and invasion, induces apoptosis, and suppresses EMT by downregulating the expression of Akt, phosphorylated p-Akt, nuclear factor κB (NF-κB), Snail, N-cadherin, and Vimentin and upregulating E-cadherin in vitro and in vivo. Enhanced proliferation and EMT are also observed when miR-19a/b are transfected into SNB19 GBM cells, with lowered expression of miR-19a/b. miR-19a is more effective than miR-19b in the regulation of biological behavior of glioma cells. miR-19a/b modulate molecular events for the promotion of EMT via the Akt-NF-κB pathway. SEPT7 has been confirmed as the target gene of miR-19a/b. The effect of miR-19a/b on proliferation and EMT of glioma cells and the Akt-NF-κB pathway could be reversed by transfection with SEPT7. Our study strongly suggests that miR-19a/b play a significant role in glioma progression and EMT through regulating target gene-SEPT7 and the SEPT7-Akt-NF-κB pathway.

10.
Lab Invest ; 90(2): 144-55, 2010 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-20048743

RESUMEN

MicroRNAs (miRNAs) are a class of endogenous small noncoding RNAs that regulate gene expression after transcription. Aberrant expression of miRNAs has been shown to be involved in tumorigenesis. We showed that miR-21 was one of the most frequently overexpressed miRNA in human glioblastoma (GBM) cell lines. To explore whether miR-21 can serve as a therapeutic target for glioblastoma, we downregulated miR-21 with a specific antisense oligonucleotide and found that apoptosis was induced and cell-cycle progression was inhibited in vitro in U251 (PTEN mutant) and LN229 (PTEN wild-type) GBM cells; xenograft tumors from antisense-treated U251 cells were suppressed in vivo. Antisense-miR-21-treated cells showed a decreased expression of EGFR, activated Akt, cyclin D, and Bcl-2. Although miR-21 is known to regulate PTEN and downregulation of miR-21 led to increased PTEN expression both endogenously and in a reporter gene assay, the GBM suppressor effect of antisense-miR-21 is most likely independent of PTEN regulation because U251 has mutant PTEN. Microarray analysis showed that the knockdown of miR-21 significantly altered expression of 169 genes involved in nine cell-cycle and signaling pathways. Taken together, our studies provide evidence that miR-21 may serve as a novel therapeutic target for malignant gliomas independent of PTEN status.


Asunto(s)
Regulación hacia Abajo , Receptores ErbB/metabolismo , Glioblastoma/metabolismo , MicroARNs/metabolismo , Fosfohidrolasa PTEN/fisiología , Animales , Apoptosis , Línea Celular Tumoral , Ciclina D/metabolismo , Femenino , Perfilación de la Expresión Génica , Humanos , Redes y Vías Metabólicas , Ratones , Ratones Desnudos , Trasplante de Neoplasias , Análisis de Secuencia por Matrices de Oligonucleótidos , Oligonucleótidos Antisentido , Proteínas Proto-Oncogénicas c-akt/metabolismo , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , Transducción de Señal , Trasplante Heterólogo
11.
Mol Cancer ; 9: 229, 2010 Sep 02.
Artículo en Inglés | MEDLINE | ID: mdl-20813046

RESUMEN

BACKGROUND: MiR-221 and miR-222 (miR-221/222) are frequently up-regulated in various types of human malignancy including glioblastoma. Recent studies have reported that miR-221/222 regulate cell growth and cell cycle progression by targeting p27 and p57. However the underlying mechanism involved in cell survival modulation of miR-221/222 remains elusive. RESULTS: Here we showed that miR-221/222 inhibited cell apoptosis by targeting pro-apoptotic gene PUMA in human glioma cells. Enforced expression of miR-22/222 induced cell survival whereas knockdown of miR-221/222 rendered cells to apoptosis. Further, miR-221/222 reduced PUMA protein levels by targeting PUMA-3'UTR. Introducing PUMA cDNA without 3'UTR abrogated miR-221/222-induced cell survival. Notably, knockdown of miR-221/222 induces PUMA expression and cell apoptosis and considerably decreases tumor growth in xenograft model. Finally, there was an inverse relationship between PUMA and miR-221/222 expression in glioma tissues. CONCLUSION: To our knowledge, these data indicate for the first time that miR-221/222 directly regulate apoptosis by targeting PUMA in glioblastoma and that miR-221/222 could be potential therapeutic targets for glioblastoma intervention.


Asunto(s)
Proteínas Reguladoras de la Apoptosis/metabolismo , Glioblastoma/metabolismo , Glioblastoma/terapia , MicroARNs/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , Animales , Apoptosis/genética , Proteínas Reguladoras de la Apoptosis/genética , Northern Blotting , Western Blotting , Línea Celular Tumoral , Supervivencia Celular/fisiología , Femenino , Glioblastoma/genética , Humanos , Inmunohistoquímica , Hibridación in Situ , Técnicas In Vitro , Ratones , Ratones Desnudos , MicroARNs/genética , Células 3T3 NIH , Proteínas Proto-Oncogénicas/genética , Ensayos Antitumor por Modelo de Xenoinjerto
12.
Cancer Invest ; 28(3): 248-58, 2010 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-19916744

RESUMEN

OBJECTIVES: To explore the role of SEPT7 in glioma cell invasion. METHODS: SEPT7 was transfected into human glioma cell lines U251 and TJ899, the invasive abilities were evaluated by transwell assay, scratch assay, and 3-D/2-D Matrigel growth. The expression of MMP2/9, MT1-MMP, integrin alpha(v)beta(3), and TIMP1/2 was detected by immunohistochemistry, immunofluorescence, and Western blot analyses. Distribution of alpha-tubulin was examined by laser scanning confocal analysis. RESULT: After SEPT7 trasfection, cell invasion was inhibited, expression of MMP2/9, MT1-MMP, and integrin alpha(v)beta(3) was decreased, while TIMP1/2 was increased, and alpha-tubulin was redistributed. CONCLUSION: These results suggest that SEPT7 plays an important role in the glioma cell invasion.


Asunto(s)
Proteínas del Citoesqueleto/fisiología , Proteínas de Unión al GTP/fisiología , Glioma/patología , Animales , Proteínas de Ciclo Celular , Línea Celular Tumoral , Proteínas del Citoesqueleto/análisis , Proteínas del Citoesqueleto/genética , Femenino , Proteínas de Unión al GTP/análisis , Proteínas de Unión al GTP/genética , Glioblastoma/patología , Humanos , Metaloproteinasa 14 de la Matriz/análisis , Metaloproteinasa 2 de la Matriz/análisis , Metaloproteinasa 9 de la Matriz/análisis , Ratones , Ratones Endogámicos BALB C , Invasividad Neoplásica , Septinas , Inhibidor Tisular de Metaloproteinasa-1/análisis , Transfección , Tubulina (Proteína)/análisis , Regulación hacia Arriba , Ensayos Antitumor por Modelo de Xenoinjerto
13.
BMC Cancer ; 10: 27, 2010 Jan 31.
Artículo en Inglés | MEDLINE | ID: mdl-20113523

RESUMEN

BACKGROUND: Substantial data indicate that the oncogene microRNA 21 (miR-21) is significantly elevated in glioblastoma multiforme (GBM) and regulates multiple genes associated with cancer cell proliferation, apoptosis, and invasiveness. Thus, miR-21 can theoretically become a target to enhance the chemotherapeutic effect in cancer therapy. So far, the effect of downregulating miR-21 to enhance the chemotherapeutic effect to taxol has not been studied in human GBM. METHODS: Human glioblastoma U251 (PTEN-mutant) and LN229 (PTEN wild-type) cells were treated with taxol and the miR-21 inhibitor (in a poly (amidoamine) (PAMAM) dendrimer), alone or in combination. The 50% inhibitory concentration and cell viability were determined by the MTT assay. The mechanism between the miR-21 inhibitor and the anticancer drug taxol was analyzed using the Zheng-Jun Jin method. Annexin V/PI staining was performed, and apoptosis and the cell cycle were evaluated by flow cytometry analysis. Expression of miR-21 was investigated by RT-PCR, and western blotting was performed to evaluate malignancy related protein alteration. RESULTS: IC(50) values were dramatically decreased in cells treated with miR-21 inhibitor combine with taxol, to a greater extent than those treated with taxol alone. Furthermore, the miR-21 inhibitor significantly enhanced apoptosis in both U251 cells and LN229 cells, and cell invasiveness was obviously weakened. Interestingly, the above data suggested that in both the PTEN mutant and the wild-type GBM cells, miR-21 blockage increased the chemosensitivity to taxol. It is worth noting that the miR-21 inhibitor additively interacted with taxol on U251cells and synergistically on LN229 cells. Thus, the miR-21 inhibitor might interrupt the activity of EGFR pathways, independently of PTEN status. Meanwhile, the expression of STAT3 and p-STAT3 decreased to relatively low levels after miR-21 inhibitor and taxol treatment. The data strongly suggested that a regulatory loop between miR-21 and STAT3 might provide an insight into the mechanism of modulating EGFR/STAT3 signaling. CONCLUSIONS: Taken together, the miR-21 inhibitor could enhance the chemo-sensitivity of human glioblastoma cells to taxol. A combination of miR-21 inhibitor and taxol could be an effective therapeutic strategy for controlling the growth of GBM by inhibiting STAT3 expression and phosphorylation.


Asunto(s)
Antineoplásicos Fitogénicos/farmacología , Neoplasias Encefálicas/tratamiento farmacológico , Glioblastoma/tratamiento farmacológico , MicroARNs/genética , Fosfohidrolasa PTEN/genética , Paclitaxel/farmacología , Línea Celular Tumoral , Receptores ErbB/metabolismo , Regulación Neoplásica de la Expresión Génica , Humanos , Concentración 50 Inhibidora , Modelos Biológicos , Fosforilación , Factor de Transcripción STAT3/metabolismo , Sales de Tetrazolio/farmacología , Tiazoles/farmacología
14.
J Neurooncol ; 97(1): 41-51, 2010 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-19771395

RESUMEN

Notch receptors play an essential role in cellular processes during embryonic and postnatal development, including maintenance of stem cell self-renewal, proliferation, and determination of cell fate and apoptosis. Deregulation of Notch signaling has been implicated in some genetic diseases and tumorigenesis. The function of Notch signaling in a variety of tumors can be either oncogenic or tumor-suppressive, depending on the cellular context. In this study, Notch1 overexpression was observed in the majority of 45 astrocytic gliomas with different grades and in U251MG glioma cells. Transfection of siRNA targeting Notch1 into U251 cells in vitro downregulated Notch1 expression, associated with inhibition of cell growth, arrest of cell cycle, reduction of cell invasiveness, and induction of cell apoptosis. Meanwhile, tumor growth was delayed in established subcutaneous gliomas in nude mice treated with Notch1 siRNA in vivo. These results suggest that Notch1 plays an important oncogenic role in the development and progression of astrocytic gliomas. Furthermore, knockdown of Notch1 expression by siRNA simultaneously downregulated the expression of EGFR and the important components of its downstream pathways, including PI3K, p-AKT, K-Ras, cyclin D1 and MMP9, indicating the crosstalk and interaction of Notch and EGFR signaling pathways.


Asunto(s)
Astrocitoma/genética , Regulación Neoplásica de la Expresión Génica/fisiología , Receptor Notch1/genética , Animales , Anexina A5/metabolismo , Apoptosis/efectos de los fármacos , Apoptosis/fisiología , Astrocitoma/tratamiento farmacológico , Astrocitoma/fisiopatología , Ciclo Celular/efectos de los fármacos , Ciclo Celular/fisiología , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Ciclina D1/metabolismo , Modelos Animales de Enfermedad , Receptores ErbB/metabolismo , Citometría de Flujo/métodos , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Etiquetado Corte-Fin in Situ/métodos , Metaloproteinasa 9 de la Matriz/metabolismo , Ratones , Proteína Oncogénica v-akt/metabolismo , Fosfatidilinositol 3-Quinasas/metabolismo , Antígeno Nuclear de Célula en Proliferación/metabolismo , Proteínas Proto-Oncogénicas p21(ras)/metabolismo , ARN Interferente Pequeño/farmacología , ARN Interferente Pequeño/uso terapéutico , Receptor Notch1/metabolismo , Transducción de Señal/efectos de los fármacos , Transducción de Señal/genética , Transfección/métodos
15.
J Neurooncol ; 98(3): 329-40, 2010 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-20035367

RESUMEN

Our previous study demonstrated that SEPT7 was downregulated at mRNA level in human gliomas. This study is to further examine the expression of SEPT7 in glioma samples and characterizes its role on cell cycle progression and growth of glioma cells. mRNA and protein expression of SEPT7 were detected by RT-PCR, immunohistochemical staining, and western blot analysis in human glioma specimens and normal brain tissues. A pcDNA3-SEPT7 expression plasmid was constructed and transfected into human glioblastoma cell line U251, and cell proliferation and apoptosis were examined. The growth of established U251 and TJ905 subcutaneous xenograft gliomas was measured in nude mice treated with pcDNA3-SEPT7 and U251 xenograft tumors treated with SEPT7 siRNA. SEPT7 expression is negatively correlated with the increase of glioma grade. Overexpression of SEPT7 is able to inhibit cell proliferation and arrest cell cycle progression in the G0/G1 phase both in vitro and in vivo. Knocking down further the already low endogenous expression of SEPT7 in U251 xenograft tumors with siRNA leads to faster tumor growth compared with control tumors. This study demonstrates that SEPT7 is involved in gliomagenesis and suppresses glioma cell growth.


Asunto(s)
Neoplasias Encefálicas/metabolismo , Proteínas del Citoesqueleto/metabolismo , Proteínas de Unión al GTP/metabolismo , Regulación Neoplásica de la Expresión Génica/fisiología , Glioma/metabolismo , Análisis de Varianza , Animales , Apoptosis/fisiología , Encéfalo/metabolismo , Neoplasias Encefálicas/patología , Caspasa 3/metabolismo , Ciclo Celular/fisiología , Proteínas de Ciclo Celular , Línea Celular Tumoral , Proliferación Celular , Proteínas del Citoesqueleto/genética , Modelos Animales de Enfermedad , Citometría de Flujo/métodos , Proteínas de Unión al GTP/genética , Proteína Ácida Fibrilar de la Glía/metabolismo , Glioma/patología , Humanos , Etiquetado Corte-Fin in Situ/métodos , Ratones , Ratones Desnudos , Trasplante de Neoplasias/mortalidad , Trasplante de Neoplasias/patología , Antígeno Nuclear de Célula en Proliferación/metabolismo , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , ARN Mensajero/metabolismo , ARN Interferente Pequeño/farmacología , Septinas , Factores de Tiempo , Análisis de Matrices Tisulares/métodos , Transfección/métodos
16.
Int J Oncol ; 35(3): 583-91, 2009 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-19639178

RESUMEN

Cyclooxygenase-2 (COX-2) and phosphatidylinositol 3-kinase (PI3K)/Akt play a critical role in the formation of many malignant tumors, and have been shown to be important therapeutic targets. In the present study, small hairpin RNA (shRNA) expression constructs that target sequences of human COX-2, Akt1 and PIK3R1 were used to examine the proliferation and invasion inhibition effects on SGC7901 gastric adenocarcinoma cells and U251 glioma cells. Cell growth was inhibited by over 70%, as indicated by a MTT assay, and was accompanied by G1/G0 phase arrest in the shRNA treated group, indicating poor cell growth activities. The number of cells invading through the matrigel in the shRNA treated group were significantly decreased (26.4+/-4.6) compared with that of the control group (105+/-4.0) and the nonsense sequence group (102.5+/-6.4). In addition, the tumor volumes in the SGC7901 subcutaneous nude mouse model treated with shRNA was significantly smaller than those of the control group and nonsense sequence group. When COX-2, Akt1 and PIK3R1 were dramatically downregulated, proliferating cell nuclear antigen (PCNA), CyclinD1 and matrix metalloproteinases (MMP-2, MMP-9) were downregulated, while tissue-inhibitor of metalloproteinase-2 (TIMP-2) and P53 were upregulated. Our results demonstrated that shRNA targeting COX-2, Akt1 and PIK3R1 downregulates their expression significantly in a sequence-specific manner, exerting proliferation and invasion inhibition effects on SGC7901 and U251 cells. In conclusion, our data suggest a novel mechanism for the regulation of malignant tumor cell growth and provide evidence for new combinatory gene therapy for malignant tumors.


Asunto(s)
Ciclooxigenasa 2/genética , Neoplasias Experimentales/genética , Fosfatidilinositol 3-Quinasas/genética , Proteínas Proto-Oncogénicas c-akt/genética , ARN Interferente Pequeño , Adenoviridae/genética , Animales , Western Blotting , Línea Celular Tumoral , Proliferación Celular , Ciclooxigenasa 2/metabolismo , Regulación hacia Abajo , Citometría de Flujo , Vectores Genéticos , Humanos , Inmunohistoquímica , Ratones , Ratones Desnudos , Invasividad Neoplásica/genética , Invasividad Neoplásica/patología , Neoplasias Experimentales/metabolismo , Neoplasias Experimentales/patología , Fosfatidilinositol 3-Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transfección , Ensayos Antitumor por Modelo de Xenoinjerto
17.
Int J Oncol ; 34(6): 1653-60, 2009 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-19424584

RESUMEN

MicroRNAs are short regulatory RNAs that negatively modulate protein expression at a post-transcriptional level. Emerging evidence suggests that altered regulation of miRNA may be involved in the pathogenesis of several types of cancers. In the current study, an inverse relationship between the expression of miR-221/miR-222 and the cell cycle inhibitor p27Kip1 was identified in U251 glioma cells. Co-suppression of miR-221/222 directly resulted in the up-regulation of p27Kip1 in the tested cells, consequently, affects their growth potential by reducing a G1 to S shift in the cell cycle. Consistently, miR-221/222 knocked-down through antisense 2'-OME-oligonucleotides increased p27Kip1 in U251 glioma subcutaneous mice and strongly reduced tumor growth in vivo through up regulation of p27Kip1. Our results suggest that miR-221/222 is a regulator of the tumor suppressor gene p27Kip1, and co-suppression of miR-221/222 expression in advanced gliomas may inhibit glioma cell proliferation by a mechanism involving the up-regulation of p27Kip1 in vitro and in vivo.


Asunto(s)
Inhibidor p27 de las Quinasas Dependientes de la Ciclina/antagonistas & inhibidores , Regulación Neoplásica de la Expresión Génica , Glioblastoma/prevención & control , MicroARNs/antagonistas & inhibidores , Animales , Northern Blotting , Western Blotting , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/patología , Neoplasias Encefálicas/prevención & control , Ciclo Celular , Proliferación Celular , Inhibidor p27 de las Quinasas Dependientes de la Ciclina/genética , Inhibidor p27 de las Quinasas Dependientes de la Ciclina/metabolismo , Femenino , Glioblastoma/genética , Glioblastoma/patología , Humanos , Técnicas para Inmunoenzimas , Hibridación Fluorescente in Situ , Técnicas In Vitro , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , MicroARNs/fisiología , Interferencia de ARN , ARN Mensajero/genética , ARN Mensajero/metabolismo , Tasa de Supervivencia
18.
Zhonghua Zhong Liu Za Zhi ; 31(10): 721-6, 2009 Oct.
Artículo en Zh | MEDLINE | ID: mdl-20021821

RESUMEN

OBJECTIVE: To study the inhibitory effect of knocking down microRNA(miR)-221 and miR-222 on human glioma cell growth and its possible mechanism. METHODS: miRNA-221/222 antisense oligonucleotides (antisense miR221/222) were transfected into human glioma U251 cells by lipofectamine. Northern blot analysis was conducted to detect the mRNA expression of miR-221/222 in the control and transfected cell groups. The proliferation activity of cells was determined by MTT assay. Cell invasion ability was examined by transwell assay, and cell cycle kinetics and apoptosis were detected with flow cytometry. The expression of relevant proteins was analyzed by Western blotting. The therapeutic efficacy of antisense miR221/222 on the growth of xenograft tumors in nude mice were also observed. RESULTS: In the antisense miR-221/222-transfected cells, the expression of miR-221/222 was significantly reduced; the cell invasion ability was suppressed, cell cycle was blocked at G(0)/G(1) phase, and apoptotic cells were increased. The growth of xenograft tumors treated with antisense miR-221/222 was also inhibited. In antisense miR-221/222 treated tumor cells, the expression of bcl-2 was down-regulated while connexin43, p27, PUMA, caspase-3, PTEN, TIMP3 and Bax up-regulated, and p53 expression not changed. CONCLUSION: There is a significant inhibitory effect of antisense miR-221/222 on the growth of human glioma U251 cells. miR-221/222 may be considered as a candidate target for gene therapy of human gliomas.


Asunto(s)
Proliferación Celular , Glioma/patología , MicroARNs/biosíntesis , Animales , Apoptosis , Secuencia de Bases , Caspasa 3/metabolismo , Ciclo Celular , Línea Celular Tumoral , Regulación hacia Abajo , Regulación Neoplásica de la Expresión Génica , Técnicas de Silenciamiento del Gen , Terapia Genética , Glioma/metabolismo , Humanos , Antígeno Ki-67/metabolismo , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , MicroARNs/genética , Datos de Secuencia Molecular , Trasplante de Neoplasias , Oligonucleótidos Antisentido/farmacología , Fosfohidrolasa PTEN/metabolismo , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , ARN Mensajero/metabolismo , Inhibidor Tisular de Metaloproteinasa-3/metabolismo , Transfección
19.
Zhonghua Yi Xue Yi Chuan Xue Za Zhi ; 26(6): 634-8, 2009 Dec.
Artículo en Zh | MEDLINE | ID: mdl-19953484

RESUMEN

OBJECTIVE: To study the radiation-sensitizing effect of up-regulating p27(kip1) expression by knocking down miR-221/222 in the U251 human glioblastoma cell line. METHODS: By bioinformatic analysis, we searched the miRNA-221/222 sequence and found the relationship between p27(kip1) and miRNA-221/222. miRNA-221/222 antisense oligonucleotides were transfected into U251 human glioblastoma cells. Northern blot analysis was conducted to detect the expression of miR-221/222 in control, scrambled oligonucleotide (ODN) transfected and anti-mi-221/222ODNs transfected cell groups. The cell cycle kinetics was detected by flow cytometry. Clonogenic assay was used to measure the mitotic cell death and p27(kip1) expression was examined by Western blot analysis. RESULTS: Based on bioinformatic analysis, we found that the seed sequences of miR-221 and miR-222 coincide with each other, and p27(kip1) is a target for miRNA-221/222. The expression level of miR-221/222 was significantly knocked down in cells transfected with antimiR-221/222 as compared to the parental cells or cells transfected with scrambled ODN. Cell cycle was arrested in G0 or G1 phase in the anti-miR-221/222 group. When combined with irradiation, S phase fraction in the anti-miR-221/222 cell group is lower than that in the other two control groups. Anti-miR-221/222 combined with irradiation could synergistically enhance mitotic cell death. The expression of p27(kip1) was up regulated in the anti-miR-221/222 group revealed by Western blot analysis. CONCLUSION: Anti-miR-221/222 may enhance the radiosensitivity of U251 human glioblastoma through upregulation of p27(kip1).


Asunto(s)
Inhibidor p27 de las Quinasas Dependientes de la Ciclina/genética , Glioblastoma/genética , MicroARNs/genética , Regulación hacia Arriba , Secuencia de Bases , Ciclo Celular/efectos de la radiación , Línea Celular Tumoral , Inhibidor p27 de las Quinasas Dependientes de la Ciclina/metabolismo , Glioblastoma/metabolismo , Humanos , MicroARNs/metabolismo , Datos de Secuencia Molecular , Oligonucleótidos Antisentido/genética , Oligonucleótidos Antisentido/metabolismo , Tolerancia a Radiación , Alineación de Secuencia , Regulación hacia Arriba/efectos de la radiación , Rayos X
20.
Zhonghua Yi Xue Yi Chuan Xue Za Zhi ; 26(5): 521-4, 2009 Oct.
Artículo en Zh | MEDLINE | ID: mdl-19806572

RESUMEN

OBJECTIVE: To study the effect of silencing Dicer by small interference RNA (siRNA) to suppress the global microRNA (miRNAs) expression on the biological characteristics of TJ905 glioblastoma cells. METHODS: The silencing effect of RNA interference on Dicer expression was evaluated by reverse transcription-polymerase chain reaction (RT-PCR), Western blot analysis and immunofluorescence staining. The cell proliferation rate and cell cycle kinetics were detected by MTT assay and flow cytometry respectively, and the cell invasive ability was evaluated by transwell assay. RESULTS: The siRNA targeting Dicer suppressed the expression of Dicer in TJ905 cells. Meanwhile, the proliferation activity and invasive ability were significantly enhanced in cells transfected with Dicer siRNA compared to those cells transfected with scrambled siRNA and the control cells. CONCLUSION: Suppression of Dicer expression renders the glioma cells harboring more aggressive phenotype. This preliminary finding suggests that global lower expression of miRNAs may play an oncogenic role.


Asunto(s)
ARN Helicasas DEAD-box/genética , Silenciador del Gen , Glioblastoma/genética , ARN Interferente Pequeño/genética , Ribonucleasa III/genética , Ciclo Celular , Línea Celular Tumoral , Proliferación Celular , ARN Helicasas DEAD-box/metabolismo , Regulación Neoplásica de la Expresión Génica , Glioblastoma/metabolismo , Glioblastoma/fisiopatología , Humanos , ARN Interferente Pequeño/metabolismo , Ribonucleasa III/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA