Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 133
Filtrar
Mais filtros

Base de dados
País/Região como assunto
Tipo de documento
Intervalo de ano de publicação
1.
Mol Biol Rep ; 50(10): 8015-8023, 2023 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-37541997

RESUMO

BACKGROUND: The tumor microenvironment contains chemokines that play a crucial role in various processes, such as tumorigenesis, inflammation, and therapy resistance, in different types of cancer. CXCL5 is a significant chemokine that has been shown to promote tumor proliferation, invasion, angiogenesis, and therapy resistance when overexpressed in various types of cancer. This research aims to investigate the impact of CXCL5 on the biological functions of glioblastoma (GBM). METHODS: The TCGA GBM and GEO databases were utilized to perform transcriptome microarray analysis and oncogenic signaling pathway analysis of CXCL5 in GBM. Validation of CXCL5 expression was performed using RT-qPCR and Western Blot. The impact of CXCL5 on cell proliferation, tumorigenesis, and angiogenesis in GBM was assessed through various methods, including cell proliferation assay, cloning assay, intracranial xenograft tumor models, and tube formation assay. Clinical prognosis was evaluated in 59 samples of gliomas with varying degrees of malignancy (grades 2, 3, and 4) and the TCGA GBM database, based on CXCL5 expression levels. The activities of the JAK-STAT and NF-κB signaling pathways were detected using Western Blot. RESULTS: The expression of CXCL5 was highly enriched in GBM. Moreover, the inhibition of CXCL5 showed a significant efficacy in suppressing cellular proliferation and angiogenesis, resulting in extended survival rates in xenograft mouse models in comparison to the control group. Notably, pretreatment with dapsone exhibited a reversal of the impact of CXCL5 on the formation of colonies and tubes in GBM cells. Elevated expression of CXCL5 was correlated with poor outcomes in GBM patients. Furthermore, the overexpression of CXCL5 has been associated with the activation of JAK-STAT and NF-κB signaling pathways. CONCLUSIONS: CXCL5 plays an important role in tumorigenesis and angiogenesis, indicating the potential for novel therapies targeting CXCL5 in GBM.


Assuntos
Neoplasias Encefálicas , Glioblastoma , Humanos , Animais , Camundongos , NF-kappa B/metabolismo , Glioblastoma/metabolismo , Transdução de Sinais , Carcinogênese/genética , Transformação Celular Neoplásica , Neoplasias Encefálicas/metabolismo , Linhagem Celular Tumoral , Proliferação de Células , Regulação Neoplásica da Expressão Gênica , Microambiente Tumoral , Quimiocina CXCL5/genética , Quimiocina CXCL5/metabolismo
2.
Exp Cell Res ; 414(1): 113077, 2022 05 01.
Artigo em Inglês | MEDLINE | ID: mdl-35219647

RESUMO

Glioblastoma (GBM) is one of the most lethal types of primary brain tumors in adults with a median survival of less than 15 months. Although comprehensive clinical treatment strategies including surgical resection followed by radiotherapy and chemotherapy are widely applied, the prognosis for GBM patients remains dismal. The Nuclear Factor-κB (NF-κB) signaling pathway is a complex network linking extracellular stimuli to cell survival and proliferation, and aberrant activation of NF-κB signaling has been implicated in the propagation of a wide range of cancers. However, the underlying mechanism of NF-κB activation still requires further investigation. Here, we report that crumbs homolog 2 (CRB2) is markedly up-regulated in human GBM relative to non-tumor tissues or normal astrocytes. Clinically, enriched CRB2 could be observed in high grade glioma with IDH IDH wild-type and 1p19q co-deletion and implied poor outcome in GBM. Consistent with this, malignant characteristics of GBM cells including proliferation, migration, invasion and tumorigenesis were significantly suppressed by lentivirus knock-down of CRB2. Furthermore, exogenous overexpression of CRB2 enhanced the malignant biological signatures of GBM cells as well as therapy resistance to temozolomide (TMZ). To further investigate the molecular mechanisms responsible, bioinformatics analysis was performed using 3 public databases, with the result that CRB2 was found to correlate closely with tumor necrosis factor α (TNFα)-NF-κB signaling. Mechanistically, elevated CRB2 increased the phosphorylation of IκB-kinase α (IKKα), thus activating NF-κB via reduction of Ikß protein. Taken together, these data suggest that CRB2 might be a reliable prognostic biomarker and potential therapeutic target for GBM.


Assuntos
Neoplasias Encefálicas , Proteínas de Transporte , Glioblastoma , Glioma , Proteínas de Membrana , Neoplasias Encefálicas/patologia , Proteínas de Transporte/metabolismo , Linhagem Celular Tumoral , Regulação Neoplásica da Expressão Gênica , Glioblastoma/patologia , Glioma/patologia , Humanos , Proteínas de Membrana/genética , Proteínas de Membrana/metabolismo , NF-kappa B/genética , NF-kappa B/metabolismo , Temozolomida/uso terapêutico
3.
J Cell Mol Med ; 26(13): 3736-3750, 2022 07.
Artigo em Inglês | MEDLINE | ID: mdl-35615996

RESUMO

Gliomas, as the most lethal and malignant brain tumours in adults, remain a major challenge worldwide. DNA damage and repair-related genes (DDRRGs) appear to play a significant role in gliomas, but the studies of DDRRGs are still insufficient. Herein, we systematically explored and analysed 1547 DDRRGs in 938 glioma samples from TCGA and CGGA datasets. Using least absolute shrinkage and selection operator (LASSO) Cox regression analysis, we identified a 16-DDRRG signature, characterized by high-risk and low-risk patterns. This risk model harbours robust predictive capability for overall survival of glioma patients. We found the high-risk score is strongly associated with well-known malignant features of gliomas, such as the mesenchymal subtype, IDH-wildtype, 1p/19q non-codeletion and MGMT promoter unmethylated status. In addition, we found that the high-risk score is also linked with multiple oncogenic pathways and therapeutic resistance. Significantly, we found the high-risk group has higher enrichment of immunosuppressive cells (M2-type macrophages, Tregs and MDSCs) and immune inhibition biomarkers (PD-1, PD-L1 and CTLA-4). Lastly, we proved that SMC4, which has the highest positive regression coefficient in our risk model, is strongly linked with malignant progression and TMZ resistance of gliomas in a E2F1-dependent manner.


Assuntos
Neoplasias Encefálicas , Glioma , Adulto , Biomarcadores , Neoplasias Encefálicas/patologia , Aberrações Cromossômicas , Dano ao DNA/genética , Glioma/patologia , Humanos , Isocitrato Desidrogenase/genética , Mutação
4.
Cancer Sci ; 113(2): 517-528, 2022 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-34866280

RESUMO

Nonglioblastomatous diffuse glioma (non-GDG) is a heterogeneous neuroepithelial tumor that exhibits a varied survival range from 4 to 13 years based on the diverse subtypes. Recent studies demonstrated novel molecular markers can predict prognosis for non-GDG patients; however, these findings as well as pathological classification strategies show obvious limitations on malignant transition due to the heterogeneity among non-GDGs. Therefore, developing reliable prognostic biomarkers and therapeutic targets have become an urgent need for precisely distinguishing non-GDG subtypes, illuminating the underlying mechanism. Nuclear factor κß (NF-κB) has been proved to be a significant nuclear transcriptional regulator with specific DNA-binding sequences to participate in multiple pathophysiological processes. However, the underlying mechanism of NF-κB activation still needs to be further investigated. Herein, our results indicated retinol-binding protein 1 (RBP1) was significantly upregulated in the IDHWT and 1p19qNon co-del non-GDG subtypes and enriched RBP1 expression was markedly correlated with more severe outcomes. Additionally, malignant signatures of the non-GDG cells including proliferation, migration, invasion, and self-renewal were significantly suppressed by lentiviral knockdown of RBP1. To further explore the underlying molecular mechanism, bioinformatics analysis was performed using databases, and the results demonstrated RBP1 was strongly correlated with tumor necrosis factor α (TNFα)-NF-κB signaling. Moreover, exogenous silencing of RBP1 reduced phosphorylation of IkB-kinase α (IKKα) and thus decreased NF-κB expression via decreasing the degradation of the IκBα protein. Altogether, these data suggested RBP1-dependent activation of NF-κB signaling promoted malignancy of non-GDG, indicating that RBP1 could be a reliable prognostic biomarker and potential therapeutic target for non-GDG.


Assuntos
Glioma/patologia , NF-kappa B/metabolismo , Proteínas Celulares de Ligação ao Retinol/metabolismo , Biomarcadores Tumorais/metabolismo , Movimento Celular , Proliferação de Células , Transformação Celular Neoplásica , Transição Epitelial-Mesenquimal , Glioma/genética , Glioma/metabolismo , Humanos , Quinase I-kappa B/metabolismo , Isocitrato Desidrogenase/metabolismo , Fosforilação , Prognóstico , Proteínas Celulares de Ligação ao Retinol/genética , Transdução de Sinais , Fator de Necrose Tumoral alfa/metabolismo
5.
Anticancer Drugs ; 33(10): 1058-1068, 2022 11 01.
Artigo em Inglês | MEDLINE | ID: mdl-36206098

RESUMO

Long noncoding RNAs (lncRNAs) have been demonstrated to participate in various biological processes and play key roles in tumorigenesis and metastasis. Pituitary adenoma (PA) is one of the most common malignancies in central nervous system. Recently, multiple lncRNAs have been identified to regulate PA initiation, progression and metastasis. we aimed to elucidate the expression pattern and function of lncRNA MYMLR in PA development. The expression of lncRNA MYMLR in PA tissues and cells was examined by real-time quantitative PCR. Knockdown of MYMLR expression was achieved by using shRNA. The function of MYMLR and regulatory network were analyzed using CCK-8 assay, wound-healing assay, migration assay and Annexin V/PI staining. Xenograft tumor model was used to explore the function of MYMLR in vivo . Bioinformatics analysis and luciferase reporter assay were conducted to investigate the interaction between MYMLR and its regulatory network. LncRNA MYMLR was highly expressed in PA tissues compared with that in normal tissues. Knockdown of MYMLR suppressed cell proliferation, migration and invasion, while promoting PA cell apoptosis. Mechanistically, MYMLR functioned as a competing endogenous RNA (ceRNA) sponging microRNA miR-197-3p. Furthermore, miR-197-3p exerted its tumor inhibitory role via negatively regulating carbonyl reductase 1 (CBR1). Overexpression of CBR1 antagonized the inhibitory effect of lncRNA MYMLR knockdown or miR-197-3p overexpression. In addition, xenograft tumor model revealed that knockdown of lncRNA MYMLR suppressed PA tumor development in vivo via regulating CBR1. Our findings suggest a regulatory network of lncRNA MYMLR/miR-197-3p/CBR1, which benefits the understanding of PA development and provides a promising lncRNA-direct therapeutic strategy against PA.


Assuntos
Carbonil Redutase (NADPH) , MicroRNAs , Neoplasias Hipofisárias , RNA Longo não Codificante , Humanos , Anexina A5/genética , Anexina A5/metabolismo , Carbonil Redutase (NADPH)/genética , Carbonil Redutase (NADPH)/metabolismo , Linhagem Celular Tumoral , Movimento Celular/genética , Proliferação de Células/fisiologia , Regulação Neoplásica da Expressão Gênica , Luciferases/genética , Luciferases/metabolismo , MicroRNAs/genética , MicroRNAs/metabolismo , Neoplasias Hipofisárias/genética , RNA Longo não Codificante/genética , RNA Longo não Codificante/metabolismo , RNA Interferente Pequeno , Animais
6.
J Biochem Mol Toxicol ; 36(2): e22949, 2022 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-34850494

RESUMO

Parkinson's disease (PD) ranks as the second most neurodegenerative disease characterized by loss of neurons, bradykinesia, anosmia, sleep disorder, and motor deficiency with increased global prevalence. Here, we have analyzed daidzein's neuroprotective functions in in vitro and in vivo models of PD. BV2 microglial cells induced with lipopolysaccharide (LPS) and C57BL6 mice induced with MPTP (1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine) were used in this study to investigate neuroprotective functions of daidzein. BV2 cells induced with LPS do not exert and significant (p < 0.05) reduction in cell viability up to concentration range (5-100 µM/ml). Furthermore, LPS exposed BV2 microglia exhibited significantly (p < 0.05) increased NO production, pro-inflammatory mediators PGE2, interleukin-6 (IL6), and interleukin-1ß (IL-1ß) levels. Treatment with daidzein (10, 25, and 50 µM/ml) to LPS-induced BV2 microglia exhibited significantly (p < 0.05) decreased NO, pro-inflammatory mediators PGE2, IL6, and IlL-1ß. Similar to the in vitro results, C57BL6 mice induced with MPTP showed defects in motor functions as observed from altered forelimb and hindlimb footprint analyses, grip strength, and perturbed motor coordination observed via rotarod tests. Additionally, levels of dopamine were significantly reduced, and pro-inflammatory mediators tumor necrosis factor alpha (TNF-α), IL-1ß, IL6 were found to be increased in MPTP-induced C57BL6 PD mice. Administering daidzein significantly restored the functional levels of dopamine and pro-inflammatory mediators TNF-α, IL-1ß, IL6 to near normal physiology as seen in healthy C57BL6 mice controls. Similarly, daidzein treatment to PD mice also restored the histological architecture to near normal levels as in control mice. Together, our results collectively endorse the neuroprotective functions of daidzein as observed from our initial studies, and further studies aimed at investigating daidzein's ability in regulating the catecholamine synthesis pathway to protect substantia nigra pars compacta (SNpc) neurons are in focus.


Assuntos
Isoflavonas/farmacologia , Lipopolissacarídeos/toxicidade , Microglia/metabolismo , Fármacos Neuroprotetores/farmacologia , Transtornos Parkinsonianos , Animais , Masculino , Camundongos , Transtornos Parkinsonianos/induzido quimicamente , Transtornos Parkinsonianos/tratamento farmacológico , Transtornos Parkinsonianos/metabolismo
7.
Metab Brain Dis ; 37(5): 1351-1363, 2022 06.
Artigo em Inglês | MEDLINE | ID: mdl-35486208

RESUMO

Cerebral ischemia is a common cerebrovascular disease with high mortality and disability rate. Exploring its mechanism is essential for developing effective treatment for cerebral ischemia. Therefore, this study aims to explore the regulatory effect and mechanism of retinoid X receptor γ (RXRγ) on cerebral ischemia-reperfusion (I/R) injury. A mouse intraluminal middle cerebral artery occlusion model was established, and PC12 cells were exposed to anaerobic/reoxygenation (A/R) as an in vitro model in this study. Cerebral I/R surgery or A/R treatment induced ferroptosis, downregulated RXRγ and GPX4 (glutathione peroxidase 4) levels, upregulated cyclooxygenase-2 (COX-2) level and increased ROS (reactive oxygen species) level in A/R induced cells or I/R brain tissues in vivo or PC12 cells in vitro. Knockdown of RXRγ downregulated GPX4 and increased COX-2 and ROS levels in A/R induced cells. RXRγ overexpression has the opposite effect. GPX4 knockdown reversed the improvement of RXRγ overexpression on COX-2 downregulation, GPX4 upregulation and ferroptosis in PC12 cells. Furthermore, chromatin immunoprecipitation (ChIP) and luciferase reporter gene assays revealed that RXRγ bound to GPX4 promoter region and activated its transcription. Overexpression of RXRγ or GPX4 alleviated brain damage and inhibited ferroptosis in I/R mice. In conclusion, RXRγ-mediated transcriptional activation of GPX4 might inhibit ferroptosis during I/R-induced brain injury.


Assuntos
Isquemia Encefálica , Ferroptose , Traumatismo por Reperfusão , Receptor X Retinoide gama/metabolismo , Animais , Isquemia Encefálica/metabolismo , Ciclo-Oxigenase 2/metabolismo , Modelos Animais de Doenças , Camundongos , Neurônios/metabolismo , Fosfolipídeo Hidroperóxido Glutationa Peroxidase , Ratos , Espécies Reativas de Oxigênio/metabolismo , Reperfusão , Traumatismo por Reperfusão/metabolismo
8.
J Cell Mol Med ; 25(16): 7720-7733, 2021 08.
Artigo em Inglês | MEDLINE | ID: mdl-34173716

RESUMO

Glioma is a common malignant tumour of the brain. In this study, we aimed to investigate diagnostic biomarkers and its role in glioma. Weighted gene co-expression network analysis (WGCNA) and Cytoscape software were used to screen the marker genes in glioma. RT-qPCR and Western blotting methods were performed to determine the expression of PAICS, ERCC1 and XPA genes in glioma tissues. Expression level of PAICS in different grades of glioma was examined by immunohistochemistry. CCK8 and Colony formation assays were used to detect cell proliferation. Cell adhesion assay was used to detect adhesion ability. Wound healing and transwell tests were used to detect cell migration ability. Flow cytometry was used to detect cell cycle and apoptosis. According to the predicted co-expression network, we identified the hub gene PAICS. Furthermore, we observed that PAICS expression level was up-regulated in glioma tissues compared with normal tissues, and the expression level was correlated with the grade of glioma. Moreover, we found PAICS can promote glioma cells proliferation and migration in vitro. Flow cytometry results showed that si-PAICS cells were stalled at the G1 phase compared with the si-NC cells and knocking down PAICS expression can increase apoptotic rate. PAICS can regulate the mRNA and protein levels of nucleotide excision repair pathway core genes ERCC1 and XPA. l-aspartic acid can affect the expression of PAICS and then inhibit glioma cell proliferation. Our results indicated that PAICS can promote glioma proliferation and migration. PAICS may act as a potential diagnostic marker and a therapeutic target for glioma.


Assuntos
Biomarcadores Tumorais/genética , Neoplasias Encefálicas/patologia , Regulação Neoplásica da Expressão Gênica , Glioma/patologia , Peptídeo Sintases/genética , Biomarcadores Tumorais/metabolismo , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/metabolismo , Ciclo Celular , Linhagem Celular Tumoral , Movimento Celular , Proliferação de Células , Biologia Computacional/métodos , Bases de Dados Genéticas , Glioma/genética , Glioma/metabolismo , Humanos , Invasividade Neoplásica , Peptídeo Sintases/metabolismo , Transdução de Sinais
9.
J Cell Mol Med ; 25(9): 4487-4500, 2021 05.
Artigo em Inglês | MEDLINE | ID: mdl-33837625

RESUMO

Low-grade gliomas (LGGs) are grade III gliomas based on the WHO classification with significant genetic heterogeneity and clinical properties. Traditional histological classification of gliomas has been challenged by the improvement of molecular stratification; however, the reproducibility and diagnostic accuracy of LGGs classification still remain poor. Herein, we identified fatty acid binding protein 5 (FABP5) as one of the most enriched genes in malignant LGGs and elevated FABP5 revealed severe outcomes in LGGs. Functionally, lentiviral suppression of FABP5 reduced malignant characters including proliferation, cloning formation, immigration, invasion and TMZ resistance, contrarily, the malignancies of LGGs were enhanced by exogenous overexpression of FABP5. Mechanistically, epithelial-mesenchymal transition (EMT) was correlated to FABP5 expression in LGGs and tumour necrosis factor α (TNFα)-dependent NF-κB signalling was involved in this process. Furthermore, FABP5 induced phosphorylation of inhibitor of nuclear factor kappa-B kinase α (IKKα) thus activated nuclear factor kappa-B (NF-κB) signalling. Taken together, our study indicated that FABP5 enhances malignancies of LGGs through canonical activation of NF-κB signalling, which could be used as individualized prognostic biomarker and potential therapeutic target of LGGs.


Assuntos
Biomarcadores Tumorais/metabolismo , Neoplasias Encefálicas/patologia , Proteínas de Ligação a Ácido Graxo/metabolismo , Regulação Neoplásica da Expressão Gênica , Glioma/patologia , NF-kappa B/metabolismo , Apoptose , Biomarcadores Tumorais/genética , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/metabolismo , Proliferação de Células , Proteínas de Ligação a Ácido Graxo/genética , Glioma/genética , Glioma/metabolismo , Humanos , NF-kappa B/genética , Invasividade Neoplásica , Prognóstico , Transdução de Sinais , Taxa de Sobrevida , Células Tumorais Cultivadas , Cicatrização
10.
J Bioenerg Biomembr ; 53(5): 621-632, 2021 10.
Artigo em Inglês | MEDLINE | ID: mdl-34251584

RESUMO

Dysregulation of protein O-fucosyl transferase 1 (POFUT1) contributes to the occurrence and progression of multiple cancers. However, whether POFUT1 has a relationship with the pathogenesis of glioblastoma (GBM) is unknown. This work was aimed at evaluating the detailed relevance of POFUT1 in GBM. Here, we demonstrated high levels of POFUT1 in GBM tissue and elucidated that GBM patients with high levels of POFUT1 had a shorter survival rate than those with low levels of POFUT1. POFUT1 knockdown in GBM cells markedly downregulated the ability to proliferate and invade, while overexpression of POFUT1 potentiated the proliferative and invasive ability of GBM cells. Further mechanistic studies indicated that silencing POFUT1 prohibited the activation of Notch signaling, leading to a reduction in the expression of HES1 and HEY1. On the contrary, overexpression of POFUT1 enhanced the activation of Notch signaling. Notably, inhibition of Notch signaling markedly reversed POFUT1-overexpression-induced tumor promotion effects in GBM cells. In addition, POFUT1 silencing markedly repressed the potential of GBM cells to form tumors in vivo. In conclusion, the data of this work indicates that POFUT1 serves a tumor promotion role in GBM by enhancing the activation of Notch signaling. This study underlines the potential role of the POFUT1/Notch axis in GBM progression and proposes POFUT1 as a promising anticancer target for GBM.


Assuntos
Neoplasias Encefálicas/metabolismo , Fucosiltransferases/metabolismo , Glioblastoma/metabolismo , Receptores Notch/metabolismo , Animais , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/patologia , Linhagem Celular Tumoral , Proliferação de Células/fisiologia , Fucosiltransferases/biossíntese , Fucosiltransferases/genética , Glioblastoma/genética , Glioblastoma/patologia , Xenoenxertos , Humanos , Camundongos , Camundongos Nus , Invasividade Neoplásica , Transdução de Sinais , Regulação para Cima
11.
Eur Radiol ; 31(7): 5253-5262, 2021 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-33758954

RESUMO

OBJECTIVE: The study aimed to evaluate the predictive validity of the neural network (NN) method for presurgical mapping of motor areas using resting-state functional MRI (rs-fMRI) data of patients with brain tumor located in the perirolandic cortex (PRC). METHODS: A total of 109 patients with brain tumors occupying PRC underwent rs-fMRI and hand movement task-based fMRI (tb-fMRI) scans. Using a NN model trained on fMRI data of 47 healthy controls, individual task activation maps were predicted from their rs-fMRI data. NN-predicted maps were compared with task activation and independent component analysis (ICA)-derived maps. Spatial Pearson's correlation coefficients (CC) matrices and Dice coefficients (DC) between task activation and predicted activation using NN (DCNN_Act) and ICA (DCICA_Act) were calculated and compared using non-parametric tests. The effects of tumor types and head motion on predicted maps were demonstrated. RESULTS: The CC matrix of NN-predicted maps showed higher diagonal values compared with ICA-derived maps (p < 0.001). DCNN_Act were higher than DCICA_Act (p < 0.001) for patients with or without motor deficits. Lower DCs were found in subjects with head motion greater than one voxel. DCs were higher on the nontumor side than on the tumor side (p < 0.001), especially in the glioma group compared with meningioma and metastatic groups. CONCLUSIONS: This study indicated that the NN approach could predict individual motor activation using rs-fMRI data and could have promising clinical applications in brain tumor patients with anatomical and functional reorganizations. KEY POINTS: • The neural network machine learning approach successfully predicted hand motor activation in patients with a tumor in the perirolandic cortex, despite space-occupying effects and possible functional reorganization. • Compared to the conventional independent component analysis, the neural network approach utilizing resting-state fMRI data yielded a higher correlation to the active task hand activation data. • The Dice coefficient of machine learning-predicted activation vs. task fMRI activation was different between tumor and nontumor side, also between tumor types, which might indicate different effects of possible neurovascular uncoupling on resting-state and task fMRI.


Assuntos
Neoplasias Encefálicas , Imageamento por Ressonância Magnética , Encéfalo , Mapeamento Encefálico , Neoplasias Encefálicas/diagnóstico por imagem , Humanos , Aprendizado de Máquina , Descanso
12.
BMC Neurol ; 21(1): 195, 2021 May 12.
Artigo em Inglês | MEDLINE | ID: mdl-33980169

RESUMO

BACKGROUND: The effects of BRAFnon-V600E and BRAFV600E on the outcomes and the molecular characteristics of adult glioma patients are unknown and need to be explored, although BRAFV600E has been extensively studied in pediatric glioma. METHODS: Co-occurring mutations and copy number alterations of associated genes in the MAPK and p53 pathways were investigated using data from The Cancer Genome Atlas (TCGA) public database retrieved by cBioPortal. The prognosis of available adult glioma cohorts with BRAFV600E and BRAFnon-V600E mutations were also investigated. RESULTS: Ninety patients with BRAFV600E or BRAFnon-V600E were enrolled in this study, and data from 52 nonredundant patients were investigated. Glioblastoma multiform was the most common cancer type, with BRAF non-V600E and BRAFV600E. TP53 (56.00% vs. 7.41%), IDH1/2 (36.00% vs. 3.70%), and ATRX (32.00% vs. 7.41%) exhibited more mutations in BRAFnon-V600E than in BRAFV600E, and TP53 was an independent risk factor (56.00% vs. 7.41%). Both BRAFnon-V600E and BRAFV600E frequently overlapped with CDKN2A/2B homozygous deletions (HDs), but there was no significant difference. Survival analysis showed no difference between the BRAF non-V600E and BRAFV600E cohorts, even after excluding the survival benefit of IDH1/2 mutations and considering the BRAFnon-V600E mutations in the glycine-rich loop (G-loop) and in the activation segment. The estimated mean survival of patients with BRAFnon-V600E & IDH1/2WT with mutations in the G-loop groups was the shortest. CONCLUSIONS: BRAFnon-V600E exhibited a stronger association with IDH1/2 mutations than BRAFV600E, but no survival advantage was found.


Assuntos
Neoplasias Encefálicas/genética , Glioma/genética , Proteínas Proto-Oncogênicas B-raf/genética , Adulto , Idoso , Idoso de 80 Anos ou mais , Feminino , Glioblastoma/genética , Homozigoto , Humanos , Isocitrato Desidrogenase/genética , Masculino , Pessoa de Meia-Idade , Mutação , Prognóstico , Análise de Sobrevida , Adulto Jovem
13.
J Cell Biochem ; 121(3): 2406-2415, 2020 03.
Artigo em Inglês | MEDLINE | ID: mdl-31692079

RESUMO

The long noncoding RNAs (lncRNAs) are associated with tumorigenesis and progression of cancer. While DNA methylation is a common epigenetic regulator of gene expression, the methylation of lncRNAs was rarely studied. To address this gap, we integrated DNA methylation and RNA-seq data to characterize the landscape of lncRNA methylation in colon adenocarcinoma (COAD). We collected and analyzed the lncRNA expression and methylation data from The Cancer Genome Atlas and Cancer Cell Line Encyclopedia to identify the epigenetically regulated lncRNAs. We further investigated the biological and clinical relevance of the identified lncRNAs via bioinformatics analysis. We identified 20 epigenetically upregulated lncRNAs in COAD, including several well-studied lncRNAs whose methylation regulation were poorly investigated, such as PVT1 and UCA1. We also revealed several novel tumor-associated lncRNAs in COAD, including GATA2-As1 and CYTOR. Next, we explored their biology function using gene set enrichment analysis and competitive endogenous RNA analysis. We characterized the methylation landscape of lncRNA in COAD and identified 20 epigenetically upregulated lncRNAs. Our findings will shed new light on the epigenetic regulation of lncRNA expression by DNA methylation.


Assuntos
Adenocarcinoma/patologia , Biomarcadores Tumorais/genética , Neoplasias do Colo/patologia , Metilação de DNA , Epigênese Genética , Regulação Neoplásica da Expressão Gênica , RNA Longo não Codificante/genética , Adenocarcinoma/genética , Estudos de Casos e Controles , Neoplasias do Colo/genética , Biologia Computacional , Perfilação da Expressão Gênica , Redes Reguladoras de Genes , Humanos , Prognóstico , Taxa de Sobrevida
14.
Mol Med ; 26(1): 29, 2020 04 08.
Artigo em Inglês | MEDLINE | ID: mdl-32268875

RESUMO

BACKGROUND: Glioma has the characteristics of high incidence and mortality, and is a common malignant tumor of the central nervous system. Circular RNAs (circRNAs) have been reported to play vital roles in progression of cancer including glioma, and circKIF4A is up-regulated in glioma tissues. However, its role and mechanisms in gliomas are unclear. METHODS: circKIF4A and miR-139-3p were determined by qRT-PCR. Transwell assay, wound-healing assay, cell colony formation and flow cytometry were performed to measure cell invasion, migration, proliferation and apoptosis. Western blotting was used to evaluate Wnt/ß-catenin pathway-related protein. Luciferase reporter assays confirmed the relationship among circKIF4A, miR-139-3p and Wnt5a. Sphere formation was performed to measure the ability of glioma-initiating cells (GICs) spheroid formation. A nude mouse xenograft model was established and immunohistochemical staining was used to detect Ki-67 and Wnt5a levels. RESULTS: circKIF4A and Wnt5a were up-regulated and miR-139-3p was down-regulated in both glioma cells and tissues. circKIF4A promoted Wnt5a expression by sponging miR-139-3p. Knockdown of circKIF4A inhibited the colony formation ability, migration and invasion, and promoted the apoptosis of glioma cells by regulating miR-139-3p. Knockdown of circKIF4A inhibited Wnt/ß-catenin signaling pathway and proliferation-related signal via miR-139-3p. Furthermore, knockdown of circKIF4A or overexpression of miR-139 suppressed the ability of sphere formation of GICs and inhibitd Wnt/ß-catenin signaling pathway and proliferation-related signal in GICs. Additionally, depletion of circKIF4A decreased the expression level of Wnt5a and Ki-67, inhibited tumorigenesis in xenograft modes. CONCLUSION: circKIF4A was overexpressed in glioma, and knockdown of circKIF4A suppressed glioma progression via miR-139-3p/Wnt5a axis. The results indicated that circKIF4A may be a potential target for clinical treatment of glioma.


Assuntos
Neoplasias Encefálicas/patologia , Glioma/patologia , MicroRNAs/genética , RNA Circular/genética , Proteína Wnt-5a/genética , Animais , Apoptose , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/metabolismo , Linhagem Celular Tumoral , Movimento Celular , Proliferação de Células , Progressão da Doença , Regulação Neoplásica da Expressão Gênica , Glioma/genética , Glioma/metabolismo , Humanos , Masculino , Camundongos , Transplante de Neoplasias , Via de Sinalização Wnt , Proteína Wnt-5a/metabolismo
15.
Acta Biochim Biophys Sin (Shanghai) ; 52(12): 1394-1403, 2020 Dec 29.
Artigo em Inglês | MEDLINE | ID: mdl-33200790

RESUMO

Human glioma is the most common primary brain tumor and is associated with high morbidity and mortality. Aberrant expressions of microRNAs (miRNAs) are involved in glioma progression. In the present study, we aimed to elucidate the roles of miR-4530 in the pathogenesis of gliomas. miR-4530 expression was examined in human glioma clinical tissues and cell lines including U251 and T98G. The target gene of miR-4530, RTEL1, was predicted with online tools and validated by luciferase reporter assay. Lentivirus infection, transfection of plasmids, and miRNA mimics were used to manipulate gene expression. Cell proliferation was determined using the CCK-8 method, and migration and invasion assays were determined with transwell experiments. Colony formation was measured by crystal violet staining, while apoptosis was determined by Annexin V/PI staining. The anti-tumor effects of miR-4530 were evaluated in nude mice xenografted using U251 cells. Our results showed that miR-4530 was significantly down-regulated in human glioma tissues and cell lines. miR-4530 over-expression inhibited the malignant behaviors of U251 and T98G cells, including reduced proliferation, diminished colony formation, migration and invasion, and increased apoptosis. Further mechanistic investigations revealed that RTEL1 is a direct functional target of miR-4530 in gliomas, and its over-expression remarkably reverses the effects of miR-4530 mimics on inhibiting these malignant behaviors. In addition, miR-4530 over-expression inhibited the growth of xenografted U251 glioma in nude mice. Therefore, miR-4530 acts as a tumor-suppressor gene and inhibits the malignant biological behaviors of human glioma cells, which is associated with directly targeting RTEL1. The miR-4530/RTEL1 axis is a potential therapeutic target for gliomas.


Assuntos
Neoplasias Encefálicas/genética , DNA Helicases/metabolismo , Glioma/genética , MicroRNAs/genética , MicroRNAs/metabolismo , Animais , Apoptose/genética , Neoplasias Encefálicas/metabolismo , Neoplasias Encefálicas/patologia , Linhagem Celular Tumoral , Movimento Celular/genética , Proliferação de Células/genética , Regulação para Baixo , Regulação Neoplásica da Expressão Gênica , Genes Supressores de Tumor , Glioma/metabolismo , Glioma/patologia , Humanos , Camundongos Nus , Ensaio Tumoral de Célula-Tronco , Ensaios Antitumorais Modelo de Xenoenxerto
16.
Biochem Biophys Res Commun ; 516(3): 999-1006, 2019 08 27.
Artigo em Inglês | MEDLINE | ID: mdl-31277942

RESUMO

Regulator of chromosome condensation 2 (RCC2) is a regulator of cell-cycle progression linked in multiple cancers to pro-tumorigenic phenomena including promotion of tumor growth, tumor metastases and poorer patient prognoses. However, the role of RCC2 in GBM remains under-investigated. Here, we sought to determine the relevance of RCC2 in GBM, as well as its roles in GBM development, progression and prognosis. Initial clinical evaluation determined significant RCC2 enrichment in GBM when compared to normal brain tissue, and elevated expression was closely associated with a poorer prognosis in glioma patients. Via shRNA inhibition, we determined that RCC2 is essential to tumor proliferation and tumorigenicity in vitro and in vivo. Additionally, RCC2 was determined to promote radioresistance of GBM tumor cells. Investigation of the underlying mechanisms implicated DNA mismatch repair, JAK-STAT pathway and activated transcription of DNA methyltransferase 1 (DNMT1). For validation, pharmacologic inhibition via administration of a DNMT1 inhibitor demonstrated attenuated GBM tumor growth both in vitro and in vivo. Collectively, this study determined a novel therapeutic target for GBM in the form of RCC2, which plays a pivotal role in GBM proliferation and radio-resistance via regulation of DNMT1 expression in a p-STAT3 dependent manner.


Assuntos
Neoplasias Encefálicas/genética , Proteínas Cromossômicas não Histona/genética , DNA (Citosina-5-)-Metiltransferase 1/genética , Regulação Neoplásica da Expressão Gênica , Glioblastoma/genética , Fatores de Troca do Nucleotídeo Guanina/genética , Tolerância a Radiação/genética , Animais , Neoplasias Encefálicas/mortalidade , Neoplasias Encefálicas/patologia , Neoplasias Encefálicas/terapia , Carcinogênese/genética , Carcinogênese/metabolismo , Carcinogênese/patologia , Linhagem Celular Tumoral , Proliferação de Células/efeitos da radiação , Proteínas Cromossômicas não Histona/antagonistas & inibidores , Proteínas Cromossômicas não Histona/metabolismo , DNA (Citosina-5-)-Metiltransferase 1/antagonistas & inibidores , DNA (Citosina-5-)-Metiltransferase 1/metabolismo , Decitabina/farmacologia , Progressão da Doença , Inibidores Enzimáticos/farmacologia , Glioblastoma/mortalidade , Glioblastoma/patologia , Glioblastoma/terapia , Fatores de Troca do Nucleotídeo Guanina/antagonistas & inibidores , Fatores de Troca do Nucleotídeo Guanina/metabolismo , Xenoenxertos , Humanos , Janus Quinase 1/genética , Janus Quinase 1/metabolismo , Camundongos , Camundongos SCID , Gradação de Tumores , Neuroglia/metabolismo , Neuroglia/patologia , Neuroglia/efeitos da radiação , Prognóstico , RNA Interferente Pequeno/genética , RNA Interferente Pequeno/metabolismo , Fatores de Transcrição STAT/genética , Fatores de Transcrição STAT/metabolismo , Transdução de Sinais , Análise de Sobrevida , Transcrição Gênica
17.
Artigo em Inglês | MEDLINE | ID: mdl-31668814

RESUMO

This article has been withdrawn at the request of the author(s) and/or editor. The Publisher apologizes for any inconvenience this may cause. The full Elsevier Policy on Article Withdrawal can be found at https://www.elsevier.com/about/our-business/policies/article-withdrawal.

18.
Epilepsia ; 60(9): 1942-1948, 2019 09.
Artigo em Inglês | MEDLINE | ID: mdl-31329275

RESUMO

OBJECTIVE: Various forms of vascular imaging are performed to identify vessels that should be avoided during stereoelectroencephalography (SEEG) planning. Digital subtraction angiography (DSA) is the gold standard for intracranial vascular imaging. DSA is an invasive investigation, and a balance is necessary to identify all clinically relevant vessels and not to visualize irrelevant vessels that may unnecessarily restrict electrode placement. We sought to estimate the size of vessels that are clinically significant for SEEG planning. METHODS: Thirty-three consecutive patients who underwent 354 SEEG electrode implantations planned with computer-assisted planning and DSA segmentation between 2016 and 2018 were identified from a prospectively maintained database. Intracranial positions of electrodes were segmented from postimplantation computed tomography scans. Each electrode was manually reviewed using "probe-eye view" with the raw preoperative DSA images for vascular conflicts. The diameter of vessels and the location of conflicts were noted. Vessel conflicts identified on raw DSA images were cross-referenced against other modalities to determine whether the conflict could have been detected. RESULTS: One hundred sixty-six vessel conflicts were identified between electrodes and DSA-identified vessels, with 0-3 conflicts per electrode and a median of four conflicts per patient. The median diameter of conflicting vessels was 1.3 mm (interquartile range [IQR] = 1.0-1.5 mm). The median depth of conflict was 31.0 mm (IQR = 14.3-45.0 mm) from the cortical surface. The addition of sulcal models to DSA, magnetic resonance venography (MRV), and T1 + gadolinium images, as an exclusion zone during computer-assisted planning, would have prevented the majority of vessel conflicts. We were unable to determine whether vessels were displaced or transected by the electrodes. SIGNIFICANCE: Vascular segmentation from DSA images was significantly more sensitive than T1 + gadolinium or MRV images. Electrode conflicts with vessels 1-1.5 mm in size did not result in a radiologically detectable or clinically significant hemorrhage and could potentially be excluded from consideration during SEEG planning.


Assuntos
Vasos Sanguíneos/diagnóstico por imagem , Encéfalo/diagnóstico por imagem , Eletrodos Implantados , Eletroencefalografia/métodos , Procedimentos Neurocirúrgicos/métodos , Angiografia Digital , Encéfalo/cirurgia , Angiografia Cerebral , Feminino , Humanos , Masculino
19.
Cell Mol Biol (Noisy-le-grand) ; 65(5): 32-37, 2019 Jun 30.
Artigo em Inglês | MEDLINE | ID: mdl-31304903

RESUMO

To investigate the effects of emodin on learning and memory and protein kinase C (PKC) signaling pathway in Alzheimer's disease (AD) model mice. 60 APP/PS1 double transgenic AD mice were selected as model mice at the age of 7-8 months, 36 healthy male C57BL/6 mice served as the control group. Morris water maze method and passive avoidance experiment were used to evaluate the memory ability of mice. The thiazole blue (MTT) method and the lactate dehydrogenase (LDH) cytotoxicity test kit were used to evaluate the effect of emodin on the cell viability of hippocampal neurons in HT22 mice treated with ß-amyloid peptide 1-42 (Aß1-42). The effect of emodin on PKC levels was explored using the modified Takai method and Western blotting. Behavioral test results showed that the escape latency of the mice in the model group was longer than that in the control group (P<0.05), and the escape latency was significantly shortened given a emodin prognosis. The MTT and LDH test results showed that emodin to Aß- overexpression induced the protective effect of hippocampus cells in HT22 mice. Western blot analysis showed that the phosphorylation level of PKC in mice increased significantly after emodin administration. Emodin can attenuate oxidative stress and inflammatory response in Alzheimer's model mice by activating PKC pathway, thereby improving cognitive function.


Assuntos
Doença de Alzheimer/metabolismo , Medicamentos de Ervas Chinesas/farmacologia , Emodina/farmacologia , Substâncias Protetoras/farmacologia , Proteína Quinase C/metabolismo , Transdução de Sinais/efeitos dos fármacos , Doença de Alzheimer/tratamento farmacológico , Peptídeos beta-Amiloides/farmacologia , Animais , Comportamento Animal/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Cognição/efeitos dos fármacos , Modelos Animais de Doenças , Medicamentos de Ervas Chinesas/administração & dosagem , Medicamentos de Ervas Chinesas/uso terapêutico , Emodina/administração & dosagem , Emodina/uso terapêutico , Hipocampo/patologia , Inflamação/metabolismo , Masculino , Memória/efeitos dos fármacos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Estresse Oxidativo/efeitos dos fármacos , Fragmentos de Peptídeos/farmacologia , Fosforilação/efeitos dos fármacos , Substâncias Protetoras/administração & dosagem , Substâncias Protetoras/uso terapêutico
20.
Biotechnol Appl Biochem ; 66(2): 247-253, 2019 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-30548933

RESUMO

The present study aimed to investigate the protective effects of salidroside (SAL) on 1-methyl-4-phenylpyridinium (MPP+ )-induced PC12 cell model for Parkinson's disease. PC12 cells were pretreated with SAL in different concentrations and then exposed to MPP+ . To evaluate the effects of SAL on cytotoxicity, the survival rate was tested by the 3-(4,5-dimethylthiazol-2-yl)-2,5-dimethyltetrazolium bromide (MTT) assay and the apoptosis was tested via flow cytometry and Western blot. Reactive oxygen species (ROS), glutathione (GSH), and malondialdehyde (MDA) were detected to analyze the effects of SAL on oxidative stress. The mRNA and protein levels of inflammatory factors TNF-α and IL-1ß were also determined by real-time quantitative polymerase chain reaction and Western blot. Pretreatment with SAL effectively relieved the MPP+ cytotoxic effects and decreased the release of ROS production and inflammatory cytokines. SAL also inhibited apoptosis, suppressed MDA activity, and increased GSH levels in MPP+ -treated PC12 cells. Moreover, the expression levels of caspase-9, caspase-3, and Bax were significantly decreased in the SAL treatment groups compared with the MPP+ group, whereas Bcl-2 expression was significantly increased in the SAL treatment groups. In summary, the overall results suggested that SAL have neuroprotective effects on the MPP+ -induced PC12 cell model by inhibiting inflammation, oxidative stress, and cell apoptosis. SAL may be a potential active product to protect against Parkinson's disease.


Assuntos
Apoptose/efeitos dos fármacos , Glucosídeos/farmacologia , Intoxicação por MPTP/prevenção & controle , Estresse Oxidativo/efeitos dos fármacos , Fenóis/farmacologia , Animais , Caspase 3/metabolismo , Caspase 9/metabolismo , Inflamação/induzido quimicamente , Inflamação/metabolismo , Inflamação/patologia , Inflamação/prevenção & controle , Intoxicação por MPTP/metabolismo , Intoxicação por MPTP/patologia , Células PC12 , Ratos , Proteína X Associada a bcl-2/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA