Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
1.
J Neurooncol ; 164(1): 157-170, 2023 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-37523046

RESUMEN

OBJECTIVE: Recent studies have increasingly shown that glioma stem cells (GSCs) are extremely important for developing and treating glioblastoma multiforme (GBM). The Broad-complex, Tram-track, and Bric-a-brac protein family is functionally related to a variety of tumor stem cells, and the role of SPOPL as a member of this family in GSCs deserves to be investigated. METHODS: To investigate the expression of SPOPL in GSCs and its impact on the prognosis of GBM patients by using clinical specimens, patient-derived primary GSCs and public databases. In vivo and in vitro, the effect of SPOPL on the proliferation, self-renewal, and differentiation ability of GSCs was explored. Probing the mechanism by which SPOPL affects the biological function of GSCs using RNA sequencing (RNA-seq) and rescue experiments. RESULTS: The expression of SPOPL was significantly upregulated in GSCs and GBM, and patients with high SPOPL expression had a poorer prognosis. SPOPL enhanced the proliferation and self-renewal ability of GSCs and enhanced the tumorigenicity of GSCs. The Notch signaling pathway was significantly inhibited in SPOPL knockdown GSCs. Activation or inhibition of the Notch signaling pathway rescued changes in the biological function of GSCs caused by altered SPOPL expression. CONCLUSION: SPOPL can be used as a potential prognostic biomarker for GBM in clinical work and promotes the proliferation and stemness of GSCs by activating the Notch signaling pathway, which may be a potential molecule for targeting GSCs to treat GBM.


Asunto(s)
Neoplasias Encefálicas , Glioblastoma , Glioma , Humanos , Neoplasias Encefálicas/patología , Línea Celular Tumoral , Proliferación Celular , Glioblastoma/patología , Glioma/patología , Células Madre Neoplásicas/patología , Transducción de Señal/genética
2.
Neuro Oncol ; 23(5): 743-756, 2021 05 05.
Artículo en Inglés | MEDLINE | ID: mdl-33325513

RESUMEN

BACKGROUND: Aberrant epidermal growth factor receptor (EGFR) activation is observed in over 50% of cases of adult glioblastoma (GBM). Nevertheless, EGFR antibodies are ineffective in clinical GBM treatment, suggesting the existence of redundant EGFR activation mechanisms. Whether circular RNA (circRNA) encodes a protein involved in EGFR-driven GBM remains unclear. We reported an unexpected mechanism in which circular EGFR RNA (circ-EGFR) encodes a novel EGFR variant to sustained EGFR activation. METHOD: We used RNA-seq, Northern blot, and Sanger sequencing to confirm the existence of circ-EGFR. Antibodies and a liquid chromatograph tandem mass spectrometer were used to identify circ-EGFR protein products. Lentivirus-transfected stable cell lines were used to assess the biological functions of the novel protein in vitro and in vivo. Clinical implications of circ-EGFR were assessed using 97 pathologically diagnosed GBM patient samples. RESULTS: The infinite open reading frame (iORF) in circ-EGFR translated repeating amino acid sequences via rolling translation and programmed -1 ribosomal frameshifting (-1PRF) induced out-of-frame stop codon (OSC), forming a polymetric novel protein-complex, which we termed rolling-translated EGFR (rtEGFR). rtEGFR directly interacted with EGFR, maintained EGFR membrane localization and attenuated EGFR endocytosis and degradation. Importantly, circ-EGFR levels correlated with the EGFR signature and predicted the poor prognosis of GBM patients. Deprivation of rtEGFR in brain tumor-initiating cells (BTICs) attenuated tumorigenicity and enhanced the anti-GBM effect. CONCLUSION: Our findings identified the endogenous rolling-translated protein and provided strong clinical evidence that targeting rtEGFR could improve the efficiency of EGFR-targeting therapies in GBM.


Asunto(s)
Neoplasias Encefálicas , Receptores ErbB/genética , Glioblastoma , Transducción de Señal , Neoplasias Encefálicas/genética , Línea Celular Tumoral , Proliferación Celular , Receptores ErbB/metabolismo , Glioblastoma/genética , Humanos , ARN Circular
3.
World J Clin Cases ; 9(7): 1631-1638, 2021 Mar 06.
Artículo en Inglés | MEDLINE | ID: mdl-33728306

RESUMEN

BACKGROUND: Most small intestinal lipomas are treated surgically, and some require repeated surgeries for multiple lipomas. However, application of endoscopic submucosal dissection (ESD) technology in the deep small intestine is rarely reported owing to the special anatomical structure of the small intestine, medical equipment limitations, and the lack of relevant experience among endoscopists. CASE SUMMARY: Two patients with small intestinal lipomas treated at the Air Force Medical Center from November 2015 to September 2019 were selected to undergo balloon-assisted ESD to treat the lipomas and explore the technical feasibility and safety of ESD for treating small intestinal lipomas. The two patients successfully underwent balloon-assisted ESD to treat four small intestinal lipomas, with a complete resection rate of 100% (4/4), without intraoperative or postoperative bleeding, perforation, or other complications. After 3-6 mo of postoperative follow-up, the clinical symptoms caused by the lipomas were significantly relieved or disappeared after treatment. CONCLUSION: Balloon-assisted ESD is a safe and reliable new method for treating deep intestinal lipomas and shows good clinical feasibility.

4.
AJNR Am J Neuroradiol ; 41(8): 1423-1429, 2020 08.
Artículo en Inglés | MEDLINE | ID: mdl-32675337

RESUMEN

BACKGROUND AND PURPOSE: Previous studies have suggested that increased mortality and disability in patients with brain tumor are associated with peritumoral brain edema. However, the mechanism of peritumoral brain edema in brain tumors is unknown. This study aimed to investigate the effect of Piezo1 overexpression on peritumoral brain edema in glioblastomas. MATERIALS AND METHODS: The Piezo1 expression in cell lines and paired samples was detected by quantitative reverse transcription polymerase chain reaction, Western blot, and immunohistochemistry. Sixty-four patients with glioblastomas were analyzed retrospectively. The Piezo1 expression of tumor tissue was detected by immunohistochemistry. The diameters of tumor and edema were measured by preoperative MR imaging, and the edema index value was calculated. RESULTS: Western blot and quantitative reverse transcription polymerase chain reaction showed that Piezo1 expression was higher in 6 glioma cell lines than in the normal astrocyte cell line. Compared with peritumoral tissues, Piezo1 was up-regulated in tumor tissues. Sixty-four patients with glioblastomas were enrolled in further study. Piezo1 was higher in the moderate edema group than in the mild edema group (P < .001), higher in the severe edema group than in the moderate edema group (P < .001), and correlated with the edema index (r = 0.73; P < .001). Receiver operating characteristic curve analysis showed that the edema index yielded an area under the curve of 0.867 (95% CI, 0.76-0.97; P < .001), with a sensitivity of 100% and a specificity of 70%. CONCLUSIONS: Piezo1 overexpression is positively correlated with the degree of peritumoral brain edema in glioblastomas. Predicting high Piezo1 expression in tumor tissues based on the edema extent shows good sensitivity and specificity.


Asunto(s)
Edema Encefálico/metabolismo , Neoplasias Encefálicas/patología , Glioblastoma/patología , Canales Iónicos/biosíntesis , Adulto , Anciano , Edema Encefálico/etiología , Neoplasias Encefálicas/complicaciones , Femenino , Glioblastoma/complicaciones , Humanos , Imagen por Resonancia Magnética/métodos , Masculino , Persona de Mediana Edad , Curva ROC , Estudios Retrospectivos , Sensibilidad y Especificidad
5.
Cell Cycle ; 16(18): 1705-1718, 2017 Sep 17.
Artículo en Inglés | MEDLINE | ID: mdl-28767320

RESUMEN

Ubiquitin-conjugating enzyme E2C (UBE2C) is characterized as a crucial molecule in cancer cell growth that plays an essential role in the development of gliomas, but the detailed mechanisms have not been fully elucidated. In this study, we found that Forkhead box transcription factor M1 (FoxM1) overexpression increased UBE2C expression, whereas FoxM1 suppression inhibited UBE2C expression in glioma cells. In addition, high FoxM1/UBE2C expression was significantly correlated with poor prognosis in glioma. We subsequently demonstrated that UBE2C was a direct transcriptional target of FoxM1, and site-directed mutations markedly down-regulated UBE2C promoter activity. Moreover, UBE2C siRNA (si-UBE2C) significantly induced glioma cell autophagy and increased both mCherry-LC3 punctate fluorescence and LC3B-II/LC3-I expression. Notably, the si-UBE2C-induced decrease in cell viability was markedly inhibited by the autophagy inhibitor bafilomycin A1. The silencing of UBE2C resulted in a distinct inhibition of the PI3K-Akt-mTOR pathway, which functions in the negative modulation of autophagy. Collectively, our findings provide clinical and molecular evidence that FoxM1 promotes glioma progression by enhancing UBE2C transcription and that the inhibition of UBE2C partially induces autophagic glioma cell death. Thus, targeting the FoxM1-UBE2C axis has therapeutic potential in the treatment of gliomas.


Asunto(s)
Autofagia , Neoplasias Encefálicas/metabolismo , Neoplasias Encefálicas/patología , Proteína Forkhead Box M1/metabolismo , Glioma/metabolismo , Glioma/patología , Neuroprotección , Enzimas Ubiquitina-Conjugadoras/metabolismo , Adolescente , Adulto , Apoptosis/efectos de los fármacos , Autofagia/genética , Línea Celular Tumoral , Supervivencia Celular/genética , Biología Computacional , Femenino , Técnicas de Silenciamiento del Gen , Silenciador del Gen , Humanos , Estimación de Kaplan-Meier , Macrólidos/farmacología , Masculino , Regiones Promotoras Genéticas , Unión Proteica , Proteínas Proto-Oncogénicas c-akt/metabolismo , Transducción de Señal/efectos de los fármacos , Serina-Treonina Quinasas TOR/metabolismo
6.
Neuro Oncol ; 19(3): 352-363, 2017 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-27923917

RESUMEN

Background: Forkhead box M1 (FOXM1) is overexpressed and activates numerous oncoproteins in tumors. However, the mechanism by which the FOXM1 protein aberrantly accumulates in human cancer remains uncertain. This study was designed to clarify the upstream signaling pathway(s) that regulate FOXM1 protein stability and transcriptional activity. Methods: Mass spectrometry and immunoprecipitation were performed to identify the FOXM-metadherin (MTDH) interaction. In vivo and in vitro ubiquitination assays were conducted to test the effect of MTDH on FOXM1 stability. Chromatin immunoprecipitation assays were used to determine the involvement of MTDH in FOXM1 transcriptional activity. Cell invasion assays, tube formation assays, and in vivo tumor formation assays were performed to evaluate the cooperative activities of FOXM1 and MTDH during tumorigenesis. Results: MTDH directly interacts with FOXM1 via the N-terminal inhibitory domain of MTDH, and this interaction disrupted the binding of cadherin-1 to FOXM1, thus protecting FOXM1 from subsequent proteasomal degradation. Deleting the MTDH-binding sites of FOXM1 abolished the MTDH overexpression-mediated stabilization of FOXM1. MTDH also bound to FOXM1 target gene promoters and enhanced FOXM1 transcriptional activity. MTDH knockdown destabilized FOXM1 and attenuated its transcriptional activity, consequently inhibiting cell cycle progression, angiogenesis, and cancer cell invasion in vitro and in vivo; these effects were abolished via forced overexpression of a stabilized mutant form of FOXM1. Thus, MTDH stabilized FOXM1 and supported the sustained activation of FOXM1 target genes. Conclusion: These findings highlight a novel MTDH-regulated mechanism of FOXM1 stabilization and provide profound insight into the tumorigenic events simultaneously mediated by FOXM1 and MTDH.


Asunto(s)
Astrocitos/patología , Moléculas de Adhesión Celular/metabolismo , Transformación Celular Neoplásica/patología , Proteína Forkhead Box M1/genética , Regulación Neoplásica de la Expresión Génica , Glioblastoma/patología , Animales , Apoptosis , Astrocitos/metabolismo , Cadherinas/genética , Cadherinas/metabolismo , Moléculas de Adhesión Celular/genética , Proliferación Celular , Transformación Celular Neoplásica/genética , Transformación Celular Neoplásica/metabolismo , Proteína Forkhead Box M1/química , Proteína Forkhead Box M1/metabolismo , Glioblastoma/genética , Glioblastoma/metabolismo , Humanos , Proteínas de la Membrana , Ratones , Estabilidad Proteica , Proteínas de Unión al ARN , Transducción de Señal , Células Tumorales Cultivadas , Ubiquitinación , Ensayos Antitumor por Modelo de Xenoinjerto
7.
Int J Oncol ; 28(5): 1225-32, 2006 May.
Artículo en Inglés | MEDLINE | ID: mdl-16596239

RESUMEN

Telomerase is implicated in the development of cellular immortality and oncogenesis. It has been shown that telomerase activity is considerably higher in the tissue of many different cancers than in normal tissue, and that the inhibition or downregulation of telomerase activity can prevent the malignant proliferation of tumor cells. Antisense oligonucleotides have been widely used in suppressing the expression of genes and, therefore, in the present research, we evaluated the effect of antisense human telomerase RNA (hTR) on glioma cell growth in vitro and in vivo. We showed that antisense hTR cDNA significantly inhibited TJ905 human glioma cell proliferation in vitro and tumor growth in vivo, as determined by MTT assay and by measuring the volume of glioma in nude mice. Consistent with these results, we found that telomerase activity and the mRNA levels of hTR and hTERT (human telomerase reverse transcriptase) expression were markedly decreased in tumor cells treated with antisense hTR cDNA, as assessed by TRAP (telomeric repeat amplification protocol) assay and RT-PCR (reverse transcription-polymerase chain reaction) analysis. Our study conclusively demonstrates that antisense hTR effectively inhibits the growth of human glioma cells in vitro and in vivo and, thus, may be potentially used for gene therapy of malignant gliomas and other cancers.


Asunto(s)
Neoplasias Encefálicas/patología , Glioma/patología , ARN sin Sentido/farmacología , ARN sin Sentido/uso terapéutico , Telomerasa/genética , Actinas/genética , Secuencia de Bases , División Celular/efectos de los fármacos , Línea Celular Tumoral , Cartilla de ADN , Humanos , Cinética , ARN Mensajero/genética
8.
Mol Biol Cell ; 26(3): 430-9, 2015 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-25428991

RESUMEN

Midkine (MDK) expression is associated with the proliferation of many cancers, including glioma. However, the upstream signaling that leads to MDK accumulation remains elusive. This study investigates the molecular mechanism that induces MDK overexpression in human glioma. The Repository for Molecular Brain Neoplasia Data was analyzed to identify potential MDK regulators. Expression of MDK and specificity protein 1 (SP1) was compared in glioma specimens. Chromatin immunoprecipitation assay was used to confirm the transcriptional regulation. MDK-force-expressed, SP1-silenced glioma cells were used to test rescue effects in vitro and in vivo. MDK and SP1 expression in gliomas was significantly higher than in adjacent tissues and was positively correlated in glioma clinical samples and cell lines. The promoter of the human MDK gene has a putative SP1 binding site. SP1 binds to the promoter of the MDK gene and directly regulates MDK expression. MDK or SP1 gene silencing inhibited the proliferation of glioma cells and reduced the tumor volume in nude mice. Overexpression of MDK in SP1-silenced cells could partially rescue the SP1 inhibition effects in vivo and in vitro. SP1 directly up-regulated the expression of MDK, and the SP1-MDK axis cooperated in glioma tumorigenesis.


Asunto(s)
Regulación Neoplásica de la Expresión Génica , Glioma/genética , Factores de Crecimiento Nervioso/genética , Factor de Transcripción Sp1/metabolismo , Activación Transcripcional , Animales , Línea Celular Tumoral , Proliferación Celular , Glioma/metabolismo , Humanos , Ratones , Ratones Desnudos , Midkina , Regiones Promotoras Genéticas , Regulación hacia Arriba
9.
Clin Neurol Neurosurg ; 107(1): 49-54, 2004 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-15567553

RESUMEN

OBJECTIVE: To evaluate the Cx43 expression of astrocytic tumors and correlate their expression with degrees of malignancy and proliferation activity of tumors. METHODS: Cx43 expression in eight normal brain tissues, 44 freshly resected astrocytic tumor specimens and four malignant glioma cell lines were examined by Northern blot analysis and immunohistochemical staining. The proliferation activity of tumors was measured by Ki67 labeling index (Ki67LI) with immunostaining. Scrape loading and dye transfer assay was used for examination of gap junction intercellular communication (GJIC) in glioma cell lines. RESULTS: Twenty-three out of 44 astrocytic tumors (52%) expressed both Cx43 mRNA and Protein. Cx43 expression was decreased with the ascending of tumor grade and negatively correlated with Ki67LI. GJIC was interrupted in glioma cell lines deficient in Cx43 expression. CONCLUSIONS: Cx43 expression level is inversely correlated with the tumor grade and proliferation activity of tumor, suggesting the potential role of Cx43 in the malignant progression of astrocytic tumors.


Asunto(s)
Astrocitoma/metabolismo , Neoplasias Encefálicas/metabolismo , Conexina 43/metabolismo , Animales , Astrocitoma/patología , Neoplasias Encefálicas/patología , Estudios de Casos y Controles , Línea Celular Tumoral , Proliferación Celular , Conexina 43/genética , Humanos , ARN Mensajero/metabolismo , Ratas
10.
Zhonghua Wai Ke Za Zhi ; 40(2): 90-3, 2002 Feb.
Artículo en Zh | MEDLINE | ID: mdl-11955387

RESUMEN

OBJECTIVE: To investigate telomerase activity and expression of hTR and hTERT in human neuroepithelial tumors for exploring new strategy for clinical diagnosis and treatment. METHODS: Telomerase activity was detected by modified TRAP method and the expression of hTR and hTERT was measured by RT-PCR method in 65 human neuroepithelial tumors, respectively. RESULTS: The positive rates of telomerase and hTERT were 61.54% and 70.77% respectively in human neuroepithelial tumors, and the positive rate and their level of expression were correlated with the degree of malignancy of tumors positively. CONCLUSIONS: Telomerase activity and hTERT are significantly correlated with the degree of malignancyin human neuroepithelial tumors. hTERT may play a key role in the regulation of telomerase activity.


Asunto(s)
Regulación Enzimológica de la Expresión Génica , Regulación Neoplásica de la Expresión Génica , Neoplasias Neuroepiteliales/enzimología , Telomerasa/metabolismo , Proteínas de Unión al ADN , Humanos , Neoplasias Neuroepiteliales/genética , Telomerasa/biosíntesis , Telomerasa/genética
11.
Mol Cancer Res ; 11(9): 1101-11, 2013 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-23761169

RESUMEN

UNLABELLED: Glioma-initiating cells (GIC), which are characterized by their self-renewal capacity and tumorigenicity, were recently identified as a highly tumorigenic subpopulation of glioblastoma multiforme and are considered responsible for glioblastoma recurrence and chemo/radiation resistance. Previously, it was revealed that Wnt signaling activation is critical to the self-renewal of GICs. However, the molecular mechanism underlying the high expression of ß-catenin, the key transcription factor of the Wnt signaling pathway, remains elusive. In this investigation, it was determined that aurora kinase A (AURKA) regulates the self-renewal and tumorigenicity of GICs by stabilizing ß-catenin. In GICs, AURKA directly interacts with AXIN and disrupts the AXIN/GSK3ß/ß-catenin destruction complex and stabilizes ß-catenin, thereby activating Wnt signaling to promote self-renewal. Stable knockdown of AURKA destabilizes ß-catenin by increasing phosphorylated ß-catenin bound to AXIN and suppresses Wnt signaling, which inhibits the ability of GICs to self-renew. This effect is rescued by expression of an AURKA kinase dead mutant, D274A, which lacks the ability to phosphorylate GSK3ß, indicating that stabilization of ß-catenin by AURKA in GICs is independent from phosphorylation of GSK3ß. Functional experiments confirm that inhibition of AUKRA in GICs could suppress their "stemness," self-renewal ability, and tumorigenicity both in vitro and in vivo, and these effects could be rescued by stabilized ß-catenin mutant. These findings indicate that AURKA competes away the binding of AXIN from ß-catenin, induces ß-catenin stabilization, and activates Wnt signaling in GICs. IMPLICATIONS: AURKA kinase inhibition could effectively attenuate Wnt signaling, thereby inhibiting the self-renewal and tumorigenicity of GICs, and may be a novel target for glioblastoma treatment strategies.


Asunto(s)
Aurora Quinasa A/metabolismo , Glioma/metabolismo , Glioma/patología , Células Madre Neoplásicas/fisiología , Proteínas Wnt/metabolismo , Vía de Señalización Wnt , beta Catenina/metabolismo , Aurora Quinasa A/genética , Proteína Axina/metabolismo , Línea Celular Tumoral , Transformación Celular Neoplásica , Resistencia a Antineoplásicos , Silenciador del Gen , Glioblastoma/metabolismo , Humanos , Recurrencia Local de Neoplasia , Fosforilación
12.
Pathol Oncol Res ; 16(4): 509-14, 2010 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-20084481

RESUMEN

Suicide gene therapy using herpes simplex virus-thymidine kinase (HSV-TK)/ganciclovir (GCV), has been extensively tested for the treatment of glioma. Our previous study showed that exogenous wild type p53 (wt-p53) enhanced the anti-tumor effect of HSV-TK/GCV therapy. However, the use of GCV is hindered by its low penetration to the brain and its toxicity when used at higher dose. In the present study, we used another pro-drug, acyclovir (ACV), and examined the therapeutic efficacy of HSV-TK/ACV combining with wt-p53 in C6 glioma cells. We observed that wt-p53 combined with HSV-TK/ACV resulted in the super-additive anti-tumor effect in vitro. Exogenous wt-p53 significantly enhanced the sensitivity of TK positive C6 cells to ACV in vitro. Our in vivo experiment demonstrated that the effect of wt-p53 and HSV-TK/ACV combination therapy was better than that of HSV-TK/ACV alone. The survival time of tumor-bearing rats treated with wt-p53 in combination with HSV-TK/ACV was also significantly prolonged than those treated with HSV-TK/ACV alone. These results suggest that wt-p53 can enhance the therapeutic efficacy of HSV-TK/ACV both in vitro and in vivo. These findings are considerably valuable with the respect of using less toxic ACV as prodrug. This novel strategy could provide benefit to HSV-TK/prodrug gene therapy.


Asunto(s)
Aciclovir/farmacología , Antineoplásicos/farmacología , Genes p53 , Terapia Genética/métodos , Glioma/genética , Glioma/terapia , Simplexvirus/enzimología , Timidina Quinasa/metabolismo , Aciclovir/farmacocinética , Animales , Antineoplásicos/farmacocinética , Apoptosis , Línea Celular , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Terapia Combinada , Sinergismo Farmacológico , Ganciclovir/farmacocinética , Ganciclovir/farmacología , Glioma/enzimología , Glioma/patología , Humanos , Etiquetado Corte-Fin in Situ , Imagen por Resonancia Magnética , Masculino , Ratas , Ratas Sprague-Dawley , Simplexvirus/genética , Timidina Quinasa/biosíntesis , Timidina Quinasa/genética
13.
Oncol Rep ; 23(3): 725-32, 2010 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-20127012

RESUMEN

Angiogenesis is a key pathologic feature of glioblastoma, which is the most common and most lethal primary brain tumor in adults. The degree of angiogenesis has been shown to be inversely related to patient survival. However, the molecular changes leading to angiogenesis in glioblastoma remain poorly understood. In the present study, we found a direct correlation between nuclear factor (NF)-kappaB activation and angiogenesis in glioblastomas. Blockade of NF-kappaB signaling significantly inhibited glioblastoma growth and angiogenesis in nude mice. These effects were consistent with significant inhibition of the expression of multiple angiogenic molecules, including vascular endothelial growth factor, and interleukin-8, in vitro and in vivo. Furthermore, blockade of NF-kappaB signaling also significantly inhibited the angiogenic potential of glioblastoma cells in vitro and angiogenesis of brain tumors in mouse xenograft models. Collectively, these results suggest that NF-kappaB activation plays a critical role in the growth and progression of glioblastoma and is a potential target for therapy for human glioblastoma.


Asunto(s)
Regulación Neoplásica de la Expresión Génica , Glioblastoma/irrigación sanguínea , Interleucina-8/genética , FN-kappa B/fisiología , Neovascularización Patológica/etiología , Factor A de Crecimiento Endotelial Vascular/genética , Animales , Línea Celular Tumoral , Femenino , Humanos , Interleucina-8/análisis , Ratones , Ratones Endogámicos BALB C , FN-kappa B/antagonistas & inhibidores , Neovascularización Patológica/prevención & control , ARN Mensajero/análisis , Factor A de Crecimiento Endotelial Vascular/análisis
14.
Clin Neurol Neurosurg ; 112(5): 413-9, 2010 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-20236756

RESUMEN

OBJECTIVE: To investigate the expression of astrocyte elevated gene-1 (AEG-1) in human oligodendrogliomas and the association between AEG-1 expression and progression of oligodendrogliomas. METHODS: The expression of AEG-1 in normal human oligodendroglial cells, oligodendroglioma cell line, and four pairs of matched oligodendroglioma tissues and their adjacent normal brain tissues was detected by quantitative RT-PCR and western blotting. In addition, AEG-1 protein expression was examined in 75 cases of histologically characterized oligodendrogliomas by immunohistochemistry. Statistical analyses were applied to test for prognostic and diagnostic associations. RESULTS: Western blotting and RT-PCR showed that AEG-1 mRNA and protein were elevated in the oligodendroglioma cell line and significantly upregulated in primary oligodendrogliomas compared with the adjacent non-cancerous brain tissues. Immunohistochemical analysis showed that 51 of 75 (68.0%) paraffin-embedded archival oligodendroglioma samples exhibited high expression of AEG-1. Statistical analysis suggested that upregulation of AEG-1 was significantly correlated with the histological grade of oligodendroglioma (p=0.000) and that patients with high AEG-1 level exhibited shorter survival time (p=0.000). Multivariate analysis revealed that AEG-1 upregulation might be an independent prognostic indicator for the survival of patients with oligodendroglioma. CONCLUSIONS: AEG-1 might represent a novel, useful diagnostic and prognostic marker for oligodendroglioma and play a role during the development and progression of the disease.


Asunto(s)
Neoplasias Encefálicas/genética , Neoplasias Encefálicas/patología , Moléculas de Adhesión Celular/genética , Oligodendroglioma/genética , Oligodendroglioma/patología , Adulto , Western Blotting , Neoplasias Encefálicas/metabolismo , Cartilla de ADN/genética , Femenino , Humanos , Inmunohistoquímica , Antígeno Ki-67/metabolismo , Masculino , Proteínas de la Membrana , Persona de Mediana Edad , Oligodendroglioma/metabolismo , ARN Mensajero/genética , Proteínas de Unión al ARN , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
15.
J Neurooncol ; 82(3): 239-48, 2007 May.
Artículo en Inglés | MEDLINE | ID: mdl-17102907

RESUMEN

OBJECTIVE: To study on the antitumor effect of combining wt-p53 gene with suicide gene therapy (HSV-tk+GCV) for malignant gliomas. METHODS: AdCMV-p53 was transfected into C6 glioma cells at MOI of (Multiplicity of infection) 0(G100), 10(TPG1), 100(TPG2), then AdCMV-tk was transducted to C6 glioma cells of G100, TPG1 and TPG2, respectively, at MOI of 100. The C6 glioma cells tranfected with both AdCMV-p53 and AdCMV-tk were exposed to various concentration of GCV. The cell survival rate was measured by MTT assay in vitro. Rat glioma model was established by injecting 5 x 10(5) C6 glioma cells into right caudate nucleus of SD rats. AdCMV-p53 and AdCMV-tk were injected into glioma on day 5 and 6, respectively. On day 7, ganciclovir (GCV) was administrated intraperitoneally at 15 mg/kg/day for 14 days. The survival time of all rats was observed. The growth of intracerebral tumors was monitored dynamically by enhanced MRI. Cell apoptosis was evaluated by TUNEL method. Expression of HSV-tk gene was identified by in situ hybridization and expression of exogenous p53 gene was detected with Western blotting. RESULTS: In vitro, wt-p53 significantly enhanced antitumor effect of HSV-tk/GCV. The concentration of GCV for ID50 of TPG2 cells (0.001 microg/ml GCV) was 10 times lower than that for the cells of tk-GCV group (MOI = 100), while the concentration of GCV for ID100 of TPG2 (0.01 microg/ml GCV) and TPG1(0.1 microg/ml GCV) was 100 and 10 times lower than that for the cells of tk-GCV group (MOI = 100), respectively. Apoptosis of C6 glioma cells also could be induced by transfection with wt-p53 gene slightly. For in vivo study, the survival time of tumor-bearing rats treated with HSV-TK/GCV or wt-p53 combined with HSV-TK/GCV was significantly prolonged and the intracerebral tumors were regressed and disappeared earlier in the combined gene therapy group than those in the HSV-TK/GCV therapy group as shown in enhanced MRI. However, only half dose of GCV for the rats treated with both wt-p53 and HSV-TK/GCV was needed to obtain the same efficacy as those rats treated with HSV-TK/GCV alone. These results indicate that the transfection of wt-p53 potentiates the effect of HSV-TK/GCV therapy. CONCLUSIONS: The combination of HSV-tk/GCV system with wt-p53 gene transduction is optimal for clinical therapeutic trials of suicide gene therapy for malignant gliomas.


Asunto(s)
Genes Transgénicos Suicidas , Terapia Genética/métodos , Glioma/terapia , Neoplasias Experimentales/terapia , Timidina Quinasa/genética , Proteína p53 Supresora de Tumor/genética , Adenoviridae/genética , Animales , Antivirales/metabolismo , Antivirales/farmacología , Southern Blotting , Western Blotting , Línea Celular Tumoral , Ganciclovir/metabolismo , Ganciclovir/farmacología , Vectores Genéticos , Humanos , Hibridación in Situ , Etiquetado Corte-Fin in Situ , Ratas , Ratas Sprague-Dawley , Timidina Quinasa/efectos de los fármacos , Timidina Quinasa/metabolismo , Transfección
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA