Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 35
Filtrar
Más filtros

Tipo del documento
Intervalo de año de publicación
1.
Anticancer Drugs ; 31(9): 950-958, 2020 10.
Artículo en Inglés | MEDLINE | ID: mdl-32590393

RESUMEN

In the present study, to delve into the effect of granulocyte-macrophage colony-stimulating factor (GM-CSF) combined with temozolomide (TMZ) on high-grade glioma cells and related mechanism, six cases of high-grade glioma cells from patient's tumor tissues were cultured. 3-(4,5)-dimethylthiahiazo (-z-y1)-3,5-di- phenytetrazoliumromide 3-(4,5)-dimethylthiahiazo (-z-y1)-3,5-di- phenytetrazoliumromide (MTT) assay was performed to detect cell proliferation and toxicity. Flow cytometry was performed to ascertain cell cycle and apoptosis rate. To detect the expressions of O6-methylguanine-DNA methyltransferase (MGMT) methylation status and MGMT protein, respectively, specific PCR and immunofluorescence were performed. According to the results of MTT assay, compared with the results of control group, GM-CSF group exhibited enhanced cell viability in varying degrees. In three cases of cells (MGMT gene methylation), the combination group [(67.67 ± 1.16), (68.13 ± 1.06), (68.42 ± 1.73)] had noticeably lower cell viability than the corresponding TMZ group [(90.00 ± 1.73), (82.33 ± 1.53), (82.67 ± 2.11)] (P < 0.01). Nevertheless, the two groups showed no significant difference in another three cases (MGMT gene unmethylated) (P > 0.05). In combination group, the apoptosis rate of the MGMT methylation cells was higher than that in the corresponding TMZ group (P < 0.01), which is consistent with MTT assay results. In all six cases of primary glioma cells, the fraction of cells in G1 phase of GM-CSF-treated group was noticeably down-regulated and was up-regulated in S phase (P < 0.01). GM-CSF could induce high-grade glioma cells to rapidly enter the cell cycle, thereby enhancing the lethal effect of TMZ on glioma cells with MGMT gene promoter methylation. However, this effect is not ideal on glioma cells with MGMT unmethylation.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/farmacología , Astrocitoma/tratamiento farmacológico , Neoplasias del Sistema Nervioso Central/tratamiento farmacológico , Glioblastoma/tratamiento farmacológico , Factor Estimulante de Colonias de Granulocitos y Macrófagos/farmacología , Temozolomida/farmacología , Astrocitoma/enzimología , Astrocitoma/genética , Astrocitoma/patología , Ciclo Celular/efectos de los fármacos , Neoplasias del Sistema Nervioso Central/enzimología , Neoplasias del Sistema Nervioso Central/genética , Neoplasias del Sistema Nervioso Central/patología , Metilación de ADN , Metilasas de Modificación del ADN/biosíntesis , Metilasas de Modificación del ADN/genética , Enzimas Reparadoras del ADN/biosíntesis , Enzimas Reparadoras del ADN/genética , Sinergismo Farmacológico , Glioblastoma/enzimología , Glioblastoma/genética , Glioblastoma/patología , Factor Estimulante de Colonias de Granulocitos y Macrófagos/administración & dosificación , Humanos , Inmunohistoquímica , Clasificación del Tumor , Regiones Promotoras Genéticas , Temozolomida/administración & dosificación , Células Tumorales Cultivadas , Proteínas Supresoras de Tumor/biosíntesis , Proteínas Supresoras de Tumor/genética
2.
J Cell Physiol ; 234(5): 6783-6800, 2019 05.
Artículo en Inglés | MEDLINE | ID: mdl-30317578

RESUMEN

Malignant gliomas are aggressive primary neoplasms that originate in the glial cells of the brain or the spine with notable resistance to standard treatment options. We carried out the study with the aim to shed light on the sensitization of resveratrol to temozolomide (TMZ) against glioma through the Wnt signaling pathway. Initially, glioma cell lines with strong resistance to TMZ were selected by 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) assay. Then, the glioma cells were subjected to resveratrol, TMZ, Wnt signaling pathway inhibitors, and activators. Cell survival rate and inhibitory concentration at half maximum value were detected by MTT, apoptosis by flow cytometry, and terminal deoxynucleotidyl transferase-mediated dUTP nick-end labeling staining, in vitro proliferation by hanging drop method and ß-catenin translocation into nuclei by TOP/FOP-FLASH assay. The expressions of the Wnt signaling pathway-related and apoptosis-related factors were determined by western blot analysis. Nude mice with glioma xenograft were established to detect tumorigenic ability. Glioma cell lines T98G and U138 which were highly resistant to TMZ were selected for subsequent experiments. Resveratrol increased the efficacy of TMZ by restraining cell proliferation, tumor growth, and promoting cell apoptosis in glioma cells. Resveratrol inhibited Wnt2 and ß-catenin expressions yet elevated GSK-3ß expression. Moreover, the Wnt signaling pathway participates in the sensitivity enhancing of resveratrol to TMZ via regulating O 6 -methylguanine-DNA methyltransferase (MGMT) expression. Resveratrol sensitized TMZ-induced glioma cell apoptosis by repressing the activation of the Wnt signaling pathway and downregulating MGMT expression, which may confer new thoughts to the chemotherapy of glioma.


Asunto(s)
Neoplasias Encefálicas/tratamiento farmacológico , Glioma/tratamiento farmacológico , Resveratrol/farmacología , Temozolomida/farmacología , Vía de Señalización Wnt/efectos de los fármacos , Animales , Apoptosis/efectos de los fármacos , Neoplasias Encefálicas/metabolismo , Línea Celular , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Regulación hacia Abajo/efectos de los fármacos , Resistencia a Antineoplásicos/efectos de los fármacos , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Glioma/metabolismo , Glucógeno Sintasa Quinasa 3 beta/metabolismo , Humanos , Ratones , Ratones Desnudos , O(6)-Metilguanina-ADN Metiltransferasa/metabolismo
3.
Clin Lab ; 65(3)2019 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-30868865

RESUMEN

BACKGROUND: Long noncoding RNAs (lncRNAs) are a novel type of endogenous noncoding RNA that are involved in the regulation of gene expression and play important roles in several types of cancer. LncRNA001089 is an intergenic lncRNA associated with cancer progression and tumor occurrence; however, the role and function of LncRNA001089 in glioma remains unclear. METHODS: In this study, we determined the expression levels of LncRNA001089 in tissues of glioma patients and explored its effect on glioma cell metastasis and proliferation using U251 glioma cell lines. Quantitative real-time PCR (qRT-PCR) technology was used to verify the expression levels of LncRNA001089 in the 127 tissues of glioma patients. Functional studies of LncRNA001089 were performed in glioma cells, including a colony formation assay, evaluation of proliferation by CCK-8, evaluation of apoptosis by flow cytometry, and evaluation of migration and invasion in vitro by Transwell assays. Tumorigenesis was evaluated in vivo in nude mice. RESULTS: LncRNA001089 was downregulated in glioma tissues, evaluated by qRT-PCR. Kaplan-Meier analysis indicated that the downregulation of LncRNA001089 expression was associated with poor prognoses in glioma patients. Multivariate analysis demonstrated that LncRNA001089 downregulation and WHO high-grade glioma were independent factors that both predicted poor outcomes for glioma patients. Cells with LncRNA001089 stable overexpression exhibited decreased capacities for proliferation, migration, and invasion in vitro, and restrained nude mouse tumorigenesis in vivo, but LncRNA001089 overexpression enhanced apoptosis. CONCLUSIONS: Our data suggest that LncRNA001089 plays a critical role in the development of glioma and may function as a potential novel biomarker and/or a therapeutic target for treatment of glioma.


Asunto(s)
Glioma/metabolismo , ARN Largo no Codificante/metabolismo , Animales , Biomarcadores de Tumor/metabolismo , Proliferación Celular , Regulación hacia Abajo , Femenino , Glioma/diagnóstico , Humanos , Masculino , Ratones Endogámicos BALB C , Ratones Desnudos , Persona de Mediana Edad , Trasplante de Neoplasias , Pronóstico
4.
J Cell Biochem ; 119(2): 2278-2287, 2018 02.
Artículo en Inglés | MEDLINE | ID: mdl-28857242

RESUMEN

This study aims to explore the role of long noncoding RNA (lncRNA)-HOX transcript antisense intergenic RNA (HOTAIR) in the occurrence and progression of glioma. Fresh glioma and normal brain tissues were classified into a glioma group (n = 67) and a normal group (n = 64) respectively. U87 cells were assigned into the blank, sh-NC, and sh-HOTAIR groups. Quantitative real-time polymerase chain reaction (qRT-PCR) was utilized to determine HOTAIR expression. Cell proliferation, cell cycle and cell apoptosis rates were detected by cell counting kit-8 (CCK-8) and flow cytometry (FCM). Scratch test and transwell assay were conducted for cell migration and invasion. Orthotopic glioma tumor model in nude mice was established by inoculating tumor cell suspension. Hematoxylin-Eosin (HE) staining was used to observe the growth and invasion of orthotopic glioma tumors. The expression of HOTAIR and cell viability was found to be lowest in the sh-HOTAIR group among the three groups. The sh-HOTAIR group exhibited a higher apoptotic rate and lower number of cell migration compared with the blank and sh-NC groups. Additionally, the speed of wound healing was slower, the migration distance decreased and the survival time of nude mice was extended in the sh-HOTAIR compared to the other groups. Moreover, the sh-HOTAIR group demonstrated reduced lesion sizes and inflammation, no convulsions or hemiplegia and lesser number of satellite metastases. Our findings support that down-regulation of HOTAIR could inhibit cell proliferation, promote cell apoptosis as well as suppress cell invasion and migration in the progression of glioma.


Asunto(s)
Neoplasias Encefálicas/genética , Regulación hacia Abajo , Glioma/genética , ARN Largo no Codificante/genética , Animales , Apoptosis , Línea Celular Tumoral , Movimiento Celular , Progresión de la Enfermedad , Regulación Neoplásica de la Expresión Génica , Humanos , Ratones , Trasplante de Neoplasias
5.
Clin Transl Oncol ; 25(5): 1378-1388, 2023 May.
Artículo en Inglés | MEDLINE | ID: mdl-36484954

RESUMEN

PURPOSE: Patients diagnosed with cancer often suffer from emotional stressors, such as anxiety, depression, and fear of death. However, whether fear stress could influence the glioma progression is still unclear. METHODS: Xenograft glioma animal models were established in nude mice. Tumor-bearing mice were subjected to fear stress by living closely with cats and then their depressive behaviors were measured using an open field test. Hematoxylin and eosin staining, the TUNEL staining and immunochemical staining were used to detect the histopathological changes of tumor tissues. Gene expression profiling was used to screen the aberrant gene expression. Methylated RNA immunoprecipitation was used to identify the RNA m6A level. Gene expression was measured by western blot and real-time PCR, respectively. RESULTS: We found that fear stress promoted glioma tumor progression in mice. Fear stress-induced upregulation of METTL3 and FSP1, increased m6A level of glioma tumor tissues, and inhibited ferroptosis in glioma progression, which were reversed by knockdown of METTL3 and FSP1 in vivo. In addition, we found that when iFSP1 (a ferroptosis inducer by targeting inhibition of FSP1) was introduced to glioma cells, the cells viability of glioma significantly was decreased and ferroptosis was enhanced in glioma cells. CONCLUSIONS: Fear stress-induced upregulation of METTL3 stabilized FSP1 mRNA by m6A modification, leading to tumor progression through inhibition of ferroptosis. Our study provides a new understanding of psychological effects on glioma development, and new insights for glioma therapy.


Asunto(s)
Miedo , Ferroptosis , Glioma , Estrés Psicológico , Animales , Humanos , Ratones , Línea Celular Tumoral , Depresión/etiología , Depresión/genética , Depresión/psicología , Modelos Animales de Enfermedad , Miedo/fisiología , Miedo/psicología , Ferroptosis/genética , Ferroptosis/fisiología , Expresión Génica , Glioma/genética , Glioma/psicología , Metiltransferasas/genética , Ratones Desnudos , ARN Mensajero , Estrés Psicológico/etiología , Estrés Psicológico/genética , Estrés Psicológico/psicología , Regulación hacia Arriba/genética
6.
Front Oncol ; 13: 1238607, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37920153

RESUMEN

Objective: Tumor in situ fluid (TISF) refers to the fluid within surgical cavities of glioma. Several studies preliminarily proved the value of cell-free tumor DNA (cf-tDNA) from TISF in the dynamic characterization of the glioma genome. Here, we assessed the potential utility of TISF cf-tDNA in broad aspects of tumor evolution under therapeutic pressure. Methods: This study was conducted under an Institutional Review Board-approved protocol at Henan Provincial People's Hospital (China). Cf-tDNA samples were sequenced with a designed 68-gene panel. A total of 205 cf-tDNA samples from 107 patients were studied. The clinical relevance of serial cf-tDNA profiling during the postoperative course was analyzed. Results: At least one tumor mutations were detected in 179/205 (87.3%) TISF cf-tDNA samples. Serial cf-tDNA was complementary to molecular residual disease and to initial tumors. Serial cf-tDNA revealed the selection of pre-existing mismatch repair-deficient cells by temozolomide as a resistant mechanism. Cf-tDNA parameters during treatment were predictive of recurrence, and serial cf-tDNA monitoring had diagnostic value for early recurrence. A total of 223 potentially actionable genomic alterations were assessed in cf-tDNA samples, wherein 78% were not found in any tumor tissue. Conclusions: In conclusion, serial TISF cf-tDNA profiling is valuable in tracking the tumor evolution of glioma during treatment and may be a feasible non-invasive option for monitoring glioma in future prospective studies and clinical practice.

7.
Cancer Med ; 12(16): 17171-17183, 2023 08.
Artículo en Inglés | MEDLINE | ID: mdl-37533228

RESUMEN

BACKGROUND: Oligodendroglioma is known for its relatively better prognosis and responsiveness to radiotherapy and chemotherapy. However, little is known about the evolution of genetic changes as oligodendroglioma progresses. METHODS: In this study, we evaluated gene evolution invivo during tumor progression based on deep whole-genome sequencing data (ctDNA). We analyzed longitudinal genomic data from six patients during tumor evolution, of which five patients developed distant recurrence. RESULTS: Whole-exome sequencing demonstrated that the rate of shared mutations between the primary and recurrent samples was relatively low. In two cases, even well-known major driver mutations in CIC and FUBP1 that were detected in primary tumors were not detected in the relapse samples. Among these cases, two patients had a conversion from the IDH mutation in the originating state to the IDH1 wild state during the process of gene evolution under chemotherapy treatment, indicating that the cell phenotype and genetic characteristics of oligodendroglioma may change during tumor evolution. Two patients received long-term temozolomide (TMZ) treatment before the operation, and we found that recurrence tumors harbored mutations in the PI3K/AKT and Sonic hedgehog (SHh) signaling pathways. Hypermutation occurred with mutations in MMR genes in one patient, contributing to the rapid progression of the tumor. CONCLUSION: Oligodendroglioma displayed great spatial and temporal heterogeneity during tumor evolution. The PI3K/AKT and SHh signaling pathways may play an important role in promoting treatment resistance and distant relapse during oligodendroglioma evolution. In addition, there was a tendency to increase the degree of tumor malignancy during evolution. Distant recurrence may be a later event duringoligodendroglioma progression. CLINICALTRIALS: gov, Identifier: NCT05512325.


Asunto(s)
Neoplasias Encefálicas , Oligodendroglioma , Humanos , Oligodendroglioma/genética , Oligodendroglioma/terapia , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/terapia , Neoplasias Encefálicas/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Fosfatidilinositol 3-Quinasas/metabolismo , Proteínas Hedgehog/metabolismo , Recurrencia Local de Neoplasia/genética , Recurrencia Local de Neoplasia/patología , Mutación , Genómica , Isocitrato Deshidrogenasa/genética , Isocitrato Deshidrogenasa/metabolismo , Proteínas de Unión al ADN/genética , Proteínas de Unión al ARN/genética
8.
iScience ; 26(9): 107528, 2023 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-37649695

RESUMEN

The evolutionary trajectories of genomic alterations underlying distant recurrence in glioma remain largely unknown. To elucidate glioma evolution, we analyzed the evolutionary trajectories of matched pairs of primary tumors and relapse tumors or tumor in situ fluid (TISF) based on deep whole-genome sequencing data (ctDNA). We found that MMR gene mutations occurred in the late stage in IDH-mutant glioma during gene evolution, which activates multiple signaling pathways and significantly increases distant recurrence potential. The proneural subtype characterized by PDGFRA amplification was likely prone to hypermutation and distant recurrence following treatment. The classical and mesenchymal subtypes tended to progress locally through subclonal reconstruction, trunk genes transformation, and convergence evolution. EGFR and NOTCH signaling pathways and CDNK2A mutation play an important role in promoting tumor local progression. Glioma subtypes displayed distinct preferred evolutionary patterns. ClinicalTrials.gov, NCT05512325.

9.
Technol Health Care ; 31(2): 635-645, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-36314174

RESUMEN

BACKGROUND: Glioblastoma is the most common and most aggressive type of primary brain tumor. OBJECTIVE: The aim of this study was to investigate the efficacy and safety of intranasal granulocyte-macrophage colony stimulating factor (GM-CSF) administration combined with chemoradiotherapy in patients with glioblastoma who underwent surgery. METHODS: Ninety-two patients were randomly divided into two groups: a control group (n= 46), who received radiotherapy with adjuvant local delivery of nimustine hydrochloride (ACNU) and systemic administration of temozolomide, and an intervention group (n= 46), who received intranasal GM-CSF prior to each cycle of adjuvant chemotherapy in addition to the treatment of the control group. Karnofsky performance status (KPS) scores, progression-free survival (PFS), overall survival (OS), and adverse effects were calculated and compared between the two groups. RESULTS: Compared with the control group, the intervention group had longer PFS (7.8 vs. 6.9 months, P= 0.016) and OS (19.2 vs. 17.1 months, P= 0.045, without adjustment for interim analyses). The KPS scores were also higher in the intervention group than in the control group after 6 months (84.35 ± 8.86 vs. 80.65 ± 7.72; t= 4.552, P= 0.036). Furthermore, the patients in the intervention group had lower incidence of neutropenia and thrombocytopenia (8.7% vs. 29.5%, P= 0.012; 8.7% vs. 18.2%, P= 0.186). Other adverse events were similar in both groups, and most adverse events were grade I/II and resolved spontaneously. CONCLUSION: Intranasal GM-CSF enhances the efficacy of the local ACNU administration combined with oral temozolomide chemotherapy. The survival and performance status were significantly improved in patients with glioblastoma after surgery. Additionally, the GM-CSF therapy was able to reduce the occurrence of chemotherapy-related neutropenia and thrombocytopenia.


Asunto(s)
Neoplasias Encefálicas , Glioblastoma , Neutropenia , Trombocitopenia , Humanos , Temozolomida/uso terapéutico , Glioblastoma/tratamiento farmacológico , Glioblastoma/radioterapia , Nimustina/uso terapéutico , Factor Estimulante de Colonias de Macrófagos/uso terapéutico , Factor Estimulante de Colonias de Granulocitos y Macrófagos/uso terapéutico , Neutropenia/tratamiento farmacológico , Trombocitopenia/tratamiento farmacológico , Granulocitos , Neoplasias Encefálicas/tratamiento farmacológico , Neoplasias Encefálicas/radioterapia
10.
Int J Neurosci ; 122(12): 723-33, 2012 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-22862301

RESUMEN

BACKGROUND: Recent studies have shown that autologous adult stem cells may be a protocol of treatment for spinal cord injury (SCI) in humans. The purpose of this study was to compare the effect of an injection of autologous bone marrow mononuclear cells (BMMCs) and bone-marrow cell mobilization induced by granulocyte colony stimulating factor (G-CSF) in rats with SCI. METHODS: Adult rats were assigned into three groups: control group (SCI only), SCI + BMMCs, and SCI + G - CSF. Neurological scores and electrophysiological testing were done in all rats before SCI, and at 1, 7, 14, 28, 56 days post-SCI. Simultaneously, immunohistochemical labeling and TUNEL assay were performed at the given time. RESULTS: From 1 week post-SCI onward, animals treated with BMMCs or G-CSF had higher BBB scores than control group. Motor and somatosensory evoked potentials (MEPs and SEPs, respectively) of the treated group were significantly better than those in control group at 2 weeks. After sacrifice, compared with the control, animals treated with BMMCs and G-CSF significantly increased expressions of Brdu, NSE, GFAP, Factor VIII, BDNF, and reduced expression of apoptosis cells around the lesion site. Our results indicate that administration of BMMCs and G-CSF in a SCI model achieves similarly positive effect on functional and histologic recovery. CONCLUSIONS: The use of G-CSF may be a viable alternative to BMMCs for autograft in patients with SCI.


Asunto(s)
Trasplante de Médula Ósea/métodos , Factor Estimulante de Colonias de Granulocitos/uso terapéutico , Traumatismos de la Médula Espinal/metabolismo , Análisis de Varianza , Animales , Antígenos CD/metabolismo , Apoptosis/efectos de los fármacos , Factor Neurotrófico Derivado del Encéfalo/metabolismo , Bromodesoxiuridina/metabolismo , Modelos Animales de Enfermedad , Electroencefalografía , Potenciales Evocados Motores/fisiología , Potenciales Evocados Somatosensoriales/fisiología , Proteína Ácida Fibrilar de la Glía/metabolismo , Masculino , Examen Neurológico , Fosfopiruvato Hidratasa/metabolismo , Ratas , Ratas Sprague-Dawley , Traumatismos de la Médula Espinal/tratamiento farmacológico , Traumatismos de la Médula Espinal/cirugía , Trasplante Autólogo/métodos
11.
J Immunol Res ; 2022: 8972730, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35647198

RESUMEN

Background: Glioma is the most common primary brain tumor with high mortality and poor outcomes. As a hallmark of cancers, inflammatory responses are crucial for their progression. The present study is aimed at exploring the prognostic value of inflammatory response-related genes (IRRGs) and constructing a prognostic IRRG signature for gliomas. Materials and Methods: We investigated the relationship between IRRGs and gliomas by integrating the transcriptomic data for gliomas from public databases. Differentially expressed IRRGs (DE-IRRGs) were identified in the GSE4290 cohort. Further, univariate, least absolute shrinkage and selection operator, and multivariate Cox regression analyses were conducted to construct an IRRG signature using The Cancer Genome Atlas (TCGA) cohort. Gliomas from the Chinese Glioma Genome Atlas (CGGA) cohort were employed for independent validation. The performance of gene signature was assessed by survival and receiver operating characteristic curve analyses. The differences in clinical correlations, immune infiltrate types, immunotherapeutic response predictions, and pathway enrichment among subgroups were investigated via bioinformatic algorithms. Results: In total, 37 DE-IRRGs were determined, of which 31 were found to be associated with survival. Ultimately, eight genes were retained to construct an IRRG signature that further classified glioma patients into two groups; the high-risk group suffered a poorer outcome as compared to the low-risk group. Furthermore, the high-risk group was significantly correlated with several risk factors, including older age, higher tumor grade, IDH wild type, 1p19q noncodel, and MGMT unmethylation. The nomogram was constructed by integrating the risk scores and other independent clinical characteristics. Moreover, the high-risk group had a greater immune infiltration and was most likely to benefit from immunotherapy. Gene set enrichment analysis suggested that immune and oncogenic pathways were enriched in high-risk glioma patients. Conclusion: We constructed a signature composed of eight IRRGs for gliomas, which could effectively predict survival and guide decision-making for treatment.


Asunto(s)
Neoplasias Encefálicas , Glioma , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/patología , Glioma/metabolismo , Humanos , Nomogramas , Pronóstico , Transcriptoma
12.
J Mol Neurosci ; 72(10): 2136-2149, 2022 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-36040678

RESUMEN

An increasing number of evidences have shown that the carcinogenic effect of DRAXIN plays an important role in the malignant process of tumors, but the mechanism of its involvement in glioma has not yet been revealed. The main aim of this study is to explore the relationship between DRAXIN and the prognosis and pathogenesis of glioma through a large quality of data analysis. Firstly, thousands of tissue samples with clinical information were collected based on various public databases. Then, a series of bioinformatics analyses were performed to mine data from information of glioma samples extracted from several reputable databases to reveal the key role of DRAXIN in glioma development and progression, with the confirmation of basic experiments. Our results showed that high expression of the oncogene DRAXIN in tumor tissue and cells could be used as an independent risk factor for poor prognosis in glioma patients and was strongly associated with clinical risk features. The reverse transcription-quantitative PCR technique was then utilized to validate the DRAXIN expression results we obtained. In addition, co-expression analysis identified, respectively, top 10 genes that were closely associated with DRAXIN positively or negatively. Finally, in vitro experiments demonstrated that knockdown of DRAXIN significantly inhibited proliferation and invasion of glioma cell. To sum up, this is the first report of DRAXIN being highly expressed in gliomas and leading to poor prognosis of glioma patients. DRAXIN may not only benefit to explore the pathogenesis of gliomas, but also serve as a novel biological target for the treatment of glioma.


Asunto(s)
Neoplasias Encefálicas , Glioma , Humanos , Neoplasias Encefálicas/diagnóstico , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/metabolismo , Línea Celular Tumoral , Proliferación Celular , Glioma/diagnóstico , Glioma/genética , Glioma/metabolismo
13.
Brain Res Bull ; 176: 121-129, 2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-34274429

RESUMEN

Glioma, featured with high incidence and low survival rate, is the most common type of primary brain tumor, severely affecting human life worldwide. LINC02381 is an interesting lncRNA functioning as oncogenic lncRNA in some cancers but as tumor-suppressor in others, but no report demonstrates its association with and function in glioma. Intriguingly, we found in a bioinformatics website LncRNADisease that LINC02381 was closely related to malignant glioma, so this study aimed to figure out the expression and function of LINC02381 in glioma. By RT-qPCR, we confirmed LINC02381 upregulation in glioma cells. Functional experiments demonstrated that LINC02381 knockdown repressed glioma cell proliferation and induced apoptosis. Boinformatics tools and RT-qPCR revealed the positive correlation between LINC02381 and CBX5 in glioma cells. More importantly, we confirmed that LINC02381 could interact and work synergistically with CEBPß to bind to CBX5 promoter and activate CBX5 transcriptionally. Additionally, rescue experiments indicated that CBX5 up-regulation reversed the decline in cell proliferation and the augment in cell apoptosis caused by LINC02381 knockdown. To conclude, LINC02381 could facilitate CBX5 transcription via interaction with CEBPß, thus exerting its oncogenic role in glioma cells, which could contribute to better understanding of glioma.


Asunto(s)
Apoptosis/genética , Neoplasias Encefálicas/genética , Proliferación Celular/genética , Homólogo de la Proteína Chromobox 5/genética , Glioma/genética , ARN Largo no Codificante/genética , Astrocitos/metabolismo , Neoplasias Encefálicas/metabolismo , Neoplasias Encefálicas/patología , Homólogo de la Proteína Chromobox 5/metabolismo , Regulación Neoplásica de la Expresión Génica , Glioma/metabolismo , Glioma/patología , Humanos , ARN Largo no Codificante/metabolismo , Transcripción Genética
14.
Front Surg ; 8: 646465, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34395506

RESUMEN

Introduction: Tractography has demonstrated utility for surgical resection in the setting of primary brain tumors involving eloquent white matter (WM) pathways. Methods: Twelve patients with glioma in or near eloquent motor areas were analyzed. The motor status was recorded before and after surgery. Two different tractography approaches were used to generate the motor corticospinal tract (CST): Constrained spherical deconvolution probabilistic tractography (CSD-Prob) and single tensor deterministic tractography (Tens-DET). To define the degree of disruption of the CST after surgical resection of the tumor, we calculated the percentage of the CST affected by surgical resection, which was then correlated with the postoperative motor status. Moreover, the fractional anisotropy (FA), mean diffusivity (MD), axial diffusivity (AD), and radial diffusivity (RD) of the CST generated by the CSD-Prob and the Tens-DET was measured and compared between the ipsilesional and contralesional side. Results: The CST was identified in all patients and its trajectory was displaced by the tumor. Only the CSD-Prob approach showed the CST with the characteristic fan-like projections from the precentral gyrus to the brainstem. Disruption of the CST was identified in 6/6 with postoperative motor deficit by CSD-Prob approach and in 5/6 in the Tens-DET. The degree of disruption was significantly associated with the motor deficit with the CSD-Prob approach (rho = -0.88, p = 0.021). However, with the Tens-DET approach the CST disruption did not show significant association with the motor function (rho = -0.27, p = 0.6). There was a significant decrease in FA (p = 0.006) and a significant increase in MD (p = 0.0004) and RD (p = 0.005) on the ipsilesional CST compared with the contralesional CST only with the CSD-Prob approach. Conclusion: CSD-Prob accurately represented the known anatomy of the CST and provided a meaningful estimate of microstructural changes of the CST affected by the tumor and its macrostructural damage after surgery. Newer surgical planning stations should include advanced models and algorithms of tractography in order to obtain more meaningful reconstructions of the WM pathways during glioma surgery.

15.
Front Oncol ; 11: 584988, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33868989

RESUMEN

Tumor in situ fluid (TISF) refers to the fluid at the local surgical cavity. We evaluated the feasibility of TISF-derived circulating tumor DNA (ctDNA) characterizing the genomic landscape for glioma. This retrospective study included TISF and tumor samples from 10 patients with glioma, we extracted cell-free DNA (cfDNA) from the TISF and then performed deep sequencing on that. And we compared genomic alterations between TISF and tumor tissue. Results showed that the concentration of cfDNA fragments from the patients for TISF ranged from 7.2 to 1,397 ng/ml. At least one tumor-specific mutation was identified in all 10 patients (100%). Further analysis of TISF ctDNA revealed a broad spectrum of genetic mutations, which have been reported to have clinical relevance. The analysis of concordance between TISF and tumor tissue reflected the spatiotemporal heterogeneity of glioma. Collectively, TISF ctDNA was a powerfully potential source for characterizing the genomic landscape of glioma, which provided new possibilities for precision medicine in patients with glioma.

16.
J Exp Clin Cancer Res ; 40(1): 164, 2021 May 11.
Artículo en Inglés | MEDLINE | ID: mdl-33975615

RESUMEN

BACKGROUND: Temozolomide (TMZ) resistance limits its application in glioma. Exosome can carry circular RNAs (circRNAs) to regulate drug resistance via sponging microRNAs (miRNAs). miRNAs can control mRNA expression by regulate the interaction with 3'UTR and methylation. Nanog homeobox (NANOG) is an important biomarker for TMZ resistance. Hitherto, it is unknown about the role of exosomal hsa_circ_0072083 (circ_0072083) in TMZ resistance in glioma, and whether it is associated with NANOG via regulating miRNA sponge and methylation. METHODS: TMZ-resistant (n = 36) and sensitive (n = 33) patients were recruited. The sensitive cells and constructed resistant cells were cultured and exposed to TMZ. circ_0072083, miR-1252-5p, AlkB homolog H5 (ALKBH5) and NANOG levels were examined via quantitative reverse transcription polymerase chain reaction and western blot. The half maximal inhibitory concentration (IC50) of TMZ, cell proliferation, apoptosis, migration and invasion were analyzed via Cell Counting Kit-8, colony formation, flow cytometry, wound healing and transwell assays. The in vivo function was assessed using xenograft model. The N6-methyladenosine (m6A) level was analyzed via methylated RNA immunoprecipitation (MeRIP). Target relationship was investigated via dual-luciferase reporter assay and RNA immunoprecipitation. Warburg effect was investigated via lactate production, glucose uptake and key enzymes expression. Exosome was isolated and confirmed via transmission electron microscopy and specific protein expression. RESULTS: circ_0072083 expression was increased in TMZ-resistant glioma tissues and cells. circ_0072083 knockdown restrained the resistance of resistant cells via decreasing IC50 of TMZ, proliferation, migration, invasion and xenograft tumor growth and increasing apoptosis. circ_0072083 silence reduced NANOG expression via blocking ALKBH5-mediated demethylation. circ_0072083 could regulate NANOG and ALKBH5 via targeting miR-1252-5p to control TMZ resistance. Warburg effect promoted the release of exosomal circ_0072083 in resistant cells. Exosomal circ_0072083 from resistant cells increased the resistance of sensitive cells to TMZ in vitro and xenograft model. Exosomal circ_0072083 level was enhanced in resistant patients, and it had a diagnostic value and indicated a lower overall survival in glioma. CONCLUSION: Exosomal circ_0072083 promoted TMZ resistance via increasing NANOG via regulating miR-1252-5p-mediated degradation and demethylation in glioma.


Asunto(s)
Neoplasias Encefálicas/tratamiento farmacológico , Glioma/tratamiento farmacológico , Proteína Homeótica Nanog/biosíntesis , ARN Circular/metabolismo , Temozolomida/farmacología , Desmetilasa de ARN, Homólogo 5 de AlkB/biosíntesis , Desmetilasa de ARN, Homólogo 5 de AlkB/genética , Desmetilasa de ARN, Homólogo 5 de AlkB/metabolismo , Antineoplásicos Alquilantes/farmacología , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/metabolismo , Resistencia a Antineoplásicos , Exosomas/genética , Exosomas/metabolismo , Glioma/metabolismo , Glioma/patología , Humanos , Persona de Mediana Edad , Proteína Homeótica Nanog/genética , Proteína Homeótica Nanog/metabolismo , ARN Circular/genética , Transducción de Señal , Regulación hacia Arriba , Efecto Warburg en Oncología
17.
Front Oncol ; 11: 742037, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34712610

RESUMEN

The recurrence of glioma is a difficult problem in clinical treatment. The molecular markers of primary tumors after resection cannot fully represent the characteristics of recurrent tumors. Here, abundant tumor DNA was detected in tumor in situ fluid (TISF). We report that TISF-derived tumor DNA (TISF-DNA) can detect genomic changes in recurrent tumors and facilitate recurrence risk analysis, providing valuable information for diagnosis and prognosis. The tumor DNA in TISF is more representative and sensitive than that in cerebrospinal fluid. It reveals the mutational landscape of minimal residual disease after glioma surgery and the risk of early recurrence, contributing to the clinical management and clinical research of glioma patients.

18.
Cancer Lett ; 479: 1-12, 2020 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-32194140

RESUMEN

Development of chemotherapy resistance remains a major obstacle for glioma management. Exosome-mediated transfer of circular RNAs (circRNAs) are being found to have relevance to many human cancers, including glioma. The purpose of this study is to explore the effect and underlying mechanism of exosomal circRNA nuclear factor I X (CircNFIX) on temozolomide (TMZ) chemoresistance in glioma. Our results indicated that exosomal CircNFIX was up-regulated in the serum of TMZ-resistant patients and predicted poor prognosis. Exosomal CircNFIX from TMZ-resistant cells conferred TMZ resistance to recipient sensitive cells through the enhancement of cell migration and invasion and the repression of cell apoptosis under TMZ exposure. CircNFIX directly interacted with miR-132 by binding to miR-132. CircNFIX knockdown enhanced TMZ sensitivity in resistant glioma cells by up-regulating miR-132. Additionally, exosomal CircNFIX promoted tumor growth and its depletion enhanced TMZ sensitivity in glioma cells in vivo. Taken together, our study suggests that exosome-mediated transfer of CircNFIX enhances TMZ resistance in glioma at least partially through sponging miR-132, highlighting a potentially prognostic biomarker and therapeutic target for improving the clinical benefits of TMZ treatment in patients with glioma.


Asunto(s)
Neoplasias Encefálicas/genética , Resistencia a Antineoplásicos , Exosomas/genética , Glioma/genética , ARN Circular/genética , Temozolomida/farmacología , Animales , Neoplasias Encefálicas/tratamiento farmacológico , Línea Celular Tumoral , Movimiento Celular , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Femenino , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Glioma/tratamiento farmacológico , Humanos , Masculino , Ratones , Trasplante de Neoplasias , Pronóstico , Regulación hacia Arriba
19.
Front Oncol ; 10: 537763, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33251130

RESUMEN

Meningioma (MEN) is a common central nervous system disease. Accumulating evidence indicated that long non-coding RNA maternally expressed gene 3 (MEG3) participated in the progression of MEN. However, the potential mechanisms of MEG3 in altering the aggressive phenotypes of MEN need further exploration. Levels of MEG3, microRNA (miR)-29c, and A-kinase anchor protein 12 (AKAP12) were determined using quantitative real-time Polymerase Chain Reaction (qRT-PCR) assay. Dual-luciferase reporter and RNA immunoprecipitation (RIP) assays were performed to verify the relationship between miR-29c and MEG3 or AKAP12. The protein level of AKAP12 was detected by western blot. Moreover, cell-cycle arrest, migration, invasion, and proliferation were assessed by flow cytometry, wound healing, transwell assays, and CCK-8 assay, respectively. Levels of MEG3 and AKAP12 were downregulated, while miR-29c was effectively increased in MEN tissues and cell line. Mechanically, MEG3 was a sponge of miR-29c to regulate the expression of AKAP12. Functionally, increase of MEG3 diminished cell-cycle, migration, invasion, and proliferation in MEN cells, and reintroduction of miR-29c could eliminate these effects. In addition, AKAP12 depletion overturned the inhibitory effects of miR-29c absence on cell-cycle, migration, invasion, and proliferation in vitro. Also, AKAP12 was co-regulated by MEG3/miR-29c axis. MEG3 mediated the aggressive behaviors of MEN cells via miR-29c/AKAP12 axis, supporting that MEG3 served as a promising biomarker for the diagnosis and treatment of human MEN.

20.
Oncol Lett ; 15(6): 9877-9883, 2018 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-29805689

RESUMEN

To investigate the effect of valproic acid (VPA) on the susceptibility of glioma stem cells to temozolomide (TMZ) and nimustine (ACNU), the O6-methylguanine-DNA methyltransferase (MGMT) promoter methylation and its expression of MGMT were examined. A total of 3 glioma cell populations were isolated from human glioma tissues, and immunocytochemistry was used to detect the expression of MGMT. VPA inhibition on the growth of the 3 glioma cell populations exposed to various concentrations of TMZ and ACNU was evaluated. Flow cytometry was applied to detect the apoptosis of glioma cells, and a methylation-specific polymerase chain reaction was used to identify methylation of MGMT promoter. Immunocytochemistry results indicated that MGMT was negatively expressed in the G1 population, but positively expressed in the G2 and G3 populations. Cell growth inhibition assays demonstrated that the survival rate in the VPA + TMZ or ACNU groups was decreased compared with that of the TMZ or ACNU alone groups (P<0.05). As for the apoptotic rate, those in the VPA alone group were increased compared with the control group (P<0.05), and the rates in the VPA + TMZ or ACNU groups were increased compared with TMZ or ACNU alone groups (P<0.05). The expression of MGMT remained negative in the G1 population following treatment with VPA, but MGMT expression became negative in the 2 MGMT-positive cell populations (G2 and G3) following VPA treatment. The MGMT promoter in the G1 population was partially methylated in the control group, but was fully methylated following VPA treatment, while the promoters of G2, G3 were unmethylated in the control group and became partially methylated in the VPA treatment group. Taken together, TMZ and ACNU may suppress the growth of glioma stem cells in vitro in a dose-dependent manner. VPA may enhance the inhibitory effects of various concentrations of TMZ and ACNU on the growth of MGMT-negative/positive cells, particularly on MGMT-positive cell populations. VPA itself may induce the apoptosis of glioma cells, and VPA combined with TMZ or ACNU may enhance TMZ/ACNU-induced apoptosis of glioma stem cells. Furthermore, VPA may also promote the methylation of the MGMT promoter to silence MGMT expression in glioma cells, which may be an important mechanism through which VPA enhances the efficacy of TMZ and ACNU in targeting glioma stem cells.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA