Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 132
Filtrar
Mais filtros

Bases de dados
País/Região como assunto
Tipo de documento
Intervalo de ano de publicação
1.
FASEB J ; 37(10): e23160, 2023 10.
Artigo em Inglês | MEDLINE | ID: mdl-37750502

RESUMO

Glioblastoma multiforme (GBM) is the most malignant brain tumor with rapid angiogenesis. How to inhibit GBM angiogenesis is a key problem to be solved. To explore the targets of inhibiting GBM angiogenesis, this study confirmed that the expression of circMTA1 (hsa_circ_0033614) was significantly upregulated in human brain microvascular endothelial cells exposed to glioma cell-conditioned medium (GECs). The expression of circMTA1 in the cytoplasm was significantly higher than that in the nucleus. Upregulated circMTA1 in GECs can promote cell proliferation, migration, and tube formation. Further exploration of the circularization mechanism of circMTA1 confirmed that KHDRBS1 protein can bind to the upstream and downstream flanking sequences of circMTA1 and promote circMTA1 biogenesis by coordinating Alu element pairing. KHDRBS1 upregulated the proliferation, migration, and tube formation of GECs by promoting the biogenesis of circMTA1. CircMTA1 can encode the protein MTA1-134aa by internal ribosome entry site sequence-mediated translation mechanism, and promote the proliferation, migration, and tube formation of GECs through the encoded MTA1-134aa. This study provides a new target for inhibiting angiogenesis in brain GBM and a new strategy for improving the therapeutic efficacy of GBM.


Assuntos
Neoplasias Encefálicas , Glioblastoma , Glioma , Humanos , Glioblastoma/genética , Células Endoteliais , Elementos Alu , Neoplasias Encefálicas/genética , Proteínas de Ciclo Celular , Proteínas de Ligação a DNA , Proteínas de Ligação a RNA , Proteínas Adaptadoras de Transdução de Sinal
2.
BMC Cancer ; 23(1): 1015, 2023 Oct 20.
Artigo em Inglês | MEDLINE | ID: mdl-37864150

RESUMO

BACKGROUND: Glioblastoma (GBM) is the most aggressive malignant primary brain tumor. The transfer RNA-derived fragments (tRFs) are a new group of small noncoding RNAs, which are dysregulated in many cancers. Until now, the expression and function of tRFs in glioma remain unknown. METHODS: The expression profiles of tRF subtypes were analyzed using the Cancer Genome Atlas (TCGA)-low-grade gliomas (LGG)/GBM dataset. The target genes of tRFs were subjected to Gene Ontology, Kyoto Encyclopedia and Gene set enrichment analysis of Genes and Genomes pathway enrichment analysis. The protein-protein interaction enrichment analysis was performed by STRING. QRT-PCR was performed to detect the expressions of tRFs in human glioma cell lines U87, U373, U251, and human astrocyte cell line SVG p12. Western blot assay was used to detect to the expression of S100A11. The interaction between tRF-19-R118LOJX and S100A11 mRNA 3'UTR was detected by dual-luciferase reporter assay. The effects of tRF-19-R118LOJX, tRF-19-6SM83OJX and S100A11 on the glioma cell proliferation, migration and in vitro vasculogenic mimicry formation ability were examined by CCK-8 proliferation assay, EdU assay, HoloMonitor cell migration assay and tube formation assay, respectively. RESULTS: tRF-19-R118LOJX and tRF-19-6SM83OJX are the most differentially expressed tRFs between LGG and GBM groups. The functional enrichment analysis showed that the target genes of tRF-19-R118LOJX and tRF-19-6SM83OJX are enriched in regulating blood vessel development. The upregulated target genes are linked to adverse survival outcomes in glioma patients. tRF-19-R118LOJX and tRF-19-6SM83OJX were identified to suppress glioma cell proliferation, migration, and in vitro vasculogenic mimicry formation. The mechanism of tRF-19-R118LOJX might be related to its function as an RNA silencer by targeting the S100A11 mRNA 3'UTR. CONCLUSION: tRFs would become novel diagnostic biomarkers and therapeutic targets of glioma, and the mechanism might be related to its post-transcriptionally regulation of gene expression by targeting mRNA 3'UTR.


Assuntos
Glioma , RNA de Transferência , Humanos , Regiões 3' não Traduzidas , RNA de Transferência/genética , RNA de Transferência/metabolismo , Linhagem Celular , Diferenciação Celular , Glioma/genética
3.
BMC Cancer ; 23(1): 237, 2023 Mar 13.
Artigo em Inglês | MEDLINE | ID: mdl-36915038

RESUMO

Glioma is primary brain tumour with a poor prognosis. Metabolic reprogramming is a hallmark of glioma, and is critical in the development of antiglioma agents and glioma therapy. Cuproptosis is a novel form of cell death mediated by protein lipidation and highly associated with mitochondrial metabolism. However, the clinical impact of cuproptosis-related genes (CRGs) in glioma remains largely unknown. The purpose of this study is to create a new CRGs signature that can be used to predict survival and immunotherapy in glioma patients. LASSO regression analysis was applied to establish prognostic gene signatures. Furthermore, a CRGs signature-based nomogram was developed and demonstrated good predictive potential. We also analyzed the relationship of CRGs and immune infiltration and the correlation with the pathological grade of glioma. Finally, we explored the miRNA that may regulate cuproptosis-related gene FDX1. We found that miR-606 was markedly downregulated in GBM, overexpression of miR-606 can significantly inhibit aerobic glycolysis and proliferation of GBM cells. FDX1 was upregulated in GBM, knockdown of FDX1 significantly inhibit aerobic glycolysis and proliferation of GBM cells. And luciferase assay was used to verified that miR-606 binds to and regulates FDX1 mRNA. These results provide a basis for further exploring the biological mechanisms of cuproptosis. This study may provide new potential therapeutic perspectives for patients with glioma.


Assuntos
Apoptose , Glioma , MicroRNAs , Humanos , Morte Celular , Cobre , Glioma/genética , MicroRNAs/genética , Nomogramas , Prognóstico
4.
Cell Biol Toxicol ; 39(6): 2841-2860, 2023 12.
Artigo em Inglês | MEDLINE | ID: mdl-37058271

RESUMO

Glioblastoma (GBM) is a primary tumor in the intracranial compartment. Vasculogenic mimicry (VM) is a process in which a pipeline of tumor cells that provide blood support to carcinogenic cells is formed, and studying VM could provide a new strategy for clinical targeted treatment of GBM. In the present study, we found that SNORD17 and ZNF384 were significantly upregulated and promoted VM in GBM, whereas KAT6B was downregulated and inhibited VM in GBM. RTL-P assays were performed to verify the 2'-O-methylation of KAT6B by SNORD17; IP assays were used to detect the acetylation of ZNF384 by KAT6B. In addition, the binding of ZNF384 to the promoter regions of VEGFR2 and VE-cadherin promoted transcription, as validated by chromatin immunoprecipitation and luciferase reporter assays. And finally, knockdown of SNORD17 and ZNF384 combined with KAT6B overexpression effectively reduced the xenograft tumor size, prolonged the survival time of nude mice and reduced the number of VM channels. This study reveals a novel mechanism of the SNORD17/KAT6B/ZNF384 axis in modulating VM development in GBM that may provide a new goal for the comprehensive treatment of GBM.


Assuntos
Glioblastoma , Animais , Camundongos , Humanos , Glioblastoma/genética , Glioblastoma/tratamento farmacológico , Camundongos Nus , Metilação , Linhagem Celular Tumoral , RNA Mensageiro , Histona Acetiltransferases/uso terapêutico
5.
Cell Biol Toxicol ; 39(6): 2881-2898, 2023 12.
Artigo em Inglês | MEDLINE | ID: mdl-37097350

RESUMO

RNA-binding proteins (RBPs), long non-coding RNAs (lncRNAs), and small nucleolar RNAs (snoRNAs) were found to play crucial regulatory roles in ischemic injury. Based on GEO databases and our experimental results, we selected Dcp2, lncRNA-RNCR3, Dkc1, and Snora62 and Foxh1 as research candidates. We found that expression levels of Dcp2, RNCR3, Dkc1, Snora62, and Foxh1 were upregulated in oxygen glucose deprivation-treated HT22 cells and hippocampal tissues subject to chronic cerebral ischemia (CCI). Silencing of Dcp2, RNCR3, Dkc1, Snora62, and Foxh1 all inhibited apoptosis of oxygen glucose deprivation-treated HT22 cells. Moreover, Dcp2 promoted RNCR3 expression by increasing its stability. Importantly, RNCR3 may act as a molecular skeleton to bind to Dkc1 and recruit Dck1 to promote snoRNP assembly. Snora62 was responsible for pseudouridylation at 28S rRNA U3507 and U3509 sites. Pseudouridylation levels of 28S rRNA were reduced after knockdown of Snora62. Decreased pseudouridylation levels inhibited the translational activity of its downstream target, Foxh1. Our study further confirmed that Foxh1 transcriptionally promoted the expression of Bax and Fam162a. Notably, experiments in vivo showed that Dcp2 knockdown combined with RNCR3 knockdown and Snora62 knockdown resulted in an anti-apoptosis effect. In conclusion, this study suggests that the axis Dcp2/RNCR3/Dkc1/Snora621 is important for the regulation of neuronal apoptosis induced by CCI.


Assuntos
Isquemia Encefálica , MicroRNAs , RNA Longo não Codificante , Humanos , MicroRNAs/genética , RNA Ribossômico 28S , Isquemia Encefálica/genética , Isquemia Encefálica/metabolismo , Apoptose/genética , Glucose , Oxigênio , RNA Longo não Codificante/genética , RNA Longo não Codificante/metabolismo
6.
Cell Biol Toxicol ; 39(6): 3323-3340, 2023 12.
Artigo em Inglês | MEDLINE | ID: mdl-37906341

RESUMO

Human malignant gliomas are the most common and aggressive primary malignant tumors of the human central nervous system. Vasculogenic mimicry (VM), which refers to the formation of a tumor blood supply system independently of endothelial cells, contributes to the malignant progression of glioma. Therefore, VM is considered a potential target for glioma therapy. Accumulated evidence indicates that alterations in SUMOylation, a reversible post-translational modification, are involved in tumorigenesis and progression. In the present study, we found that UBA2 and RALY were upregulated in glioma tissues and cell lines. Downregulation of UBA2 and RALY inhibited the migration, invasion, and VM of glioma cells. RALY can be SUMOylated by conjugation with SUMO1, which is facilitated by the overexpression of UBA2. The SUMOylation of RALY increases its stability, which in turn increases its expression as well as its promoting effect on FOXD1 mRNA. The overexpression of FOXD1 promotes DKK1 transcription by activating its promoter, thereby promoting glioma cell migration, invasion, and VM. Remarkably, the combined knockdown of UBA2, RALY, and FOXD1 resulted in the smallest tumor volumes and the longest survivals of nude mice in vivo. UBA2/RALY/FOXD1/DKK1 axis may play crucial roles in regulating VM in glioma, which may contribute to the development of potential strategies for the treatment of gliomas.


Assuntos
Neoplasias Encefálicas , Glioma , Camundongos , Animais , Humanos , Neoplasias Encefálicas/metabolismo , Sumoilação , Camundongos Nus , Células Endoteliais/metabolismo , Neovascularização Patológica/genética , Neovascularização Patológica/metabolismo , Glioma/genética , Glioma/metabolismo , Linhagem Celular Tumoral , Ribonucleoproteínas Nucleares Heterogêneas Grupo C/genética , Ribonucleoproteínas Nucleares Heterogêneas Grupo C/metabolismo , Enzimas Ativadoras de Ubiquitina/metabolismo , Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Fatores de Transcrição Forkhead/genética
7.
J Acoust Soc Am ; 154(1): 502-517, 2023 07 01.
Artigo em Inglês | MEDLINE | ID: mdl-37493330

RESUMO

Many odontocetes produce whistles that feature characteristic contour shapes in spectrogram representations of their calls. Automatically extracting the time × frequency tracks of whistle contours has numerous subsequent applications, including species classification, identification, and density estimation. Deep-learning-based methods, which train models using analyst-annotated whistles, offer a promising way to reliably extract whistle contours. However, the application of such methods can be limited by the significant amount of time and labor required for analyst annotation. To overcome this challenge, a technique that learns from automatically generated pseudo-labels has been developed. These annotations are less accurate than those generated by human analysts but more cost-effective to generate. It is shown that standard training methods do not learn effective models from these pseudo-labels. An improved loss function designed to compensate for pseudo-label error that significantly increases whistle extraction performance is introduced. The experiments show that the developed technique performs well when trained with pseudo-labels generated by two different algorithms. Models trained with the generated pseudo-labels can extract whistles with an F1-score (the harmonic mean of precision and recall) of 86.31% and 87.2% for the two sets of pseudo-labels that are considered. This performance is competitive with a model trained with 12 539 expert-annotated whistles (F1-score of 87.47%).


Assuntos
Aprendizado Profundo , Animais , Humanos , Vocalização Animal , Espectrografia do Som , Algoritmos , Baleias
8.
J Acoust Soc Am ; 152(6): 3800, 2022 12.
Artigo em Inglês | MEDLINE | ID: mdl-36586843

RESUMO

This work presents an open-source matlab software package for exploiting recent advances in extracting tonal signals from large acoustic data sets. A whistle extraction algorithm published by Li, Liu, Palmer, Fleishman, Gillespie, Nosal, Shiu, Klinck, Cholewiak, Helble, and Roch [(2020). Proceedings of the International Joint Conference on Neural Networks, July 19-24, Glasgow, Scotland, p. 10] is incorporated into silbido, an established software package for extraction of cetacean tonal calls. The precision and recall of the new system were over 96% and nearly 80%, respectively, when applied to a whistle extraction task on a challenging two-species subset of a conference-benchmark data set. A second data set was examined to assess whether the algorithm generalized to data that were collected across different recording devices and locations. These data included 487 h of weakly labeled, towed array data collected in the Pacific Ocean on two National Oceanographic and Atmospheric Administration (NOAA) cruises. Labels for these data consisted of regions of toothed whale presence for at least 15 species that were based on visual and acoustic observations and not limited to whistles. Although the lack of per whistle-level annotations prevented measurement of precision and recall, there was strong concurrence of automatic detections and the NOAA annotations, suggesting that the algorithm generalizes well to new data.


Assuntos
Aprendizado Profundo , Animais , Vocalização Animal , Espectrografia do Som , Cetáceos , Software
9.
RNA Biol ; 18(1): 47-63, 2021 01.
Artigo em Inglês | MEDLINE | ID: mdl-32618493

RESUMO

RNA-binding proteins (RBPs) are significantly dysregulated in glioma. In this study, we demonstrated the upregulation of Nuclear cap-binding subunit 3 (NCBP3) in glioma tissues and cells. Further, knockdown of NCBP3 inhibited the malignant progression of glioma. NCBP3 directly bound to small nucleolar RNA host gene 6 (SNHG6) and stabilized SNHG6 expression. In contrast, the gastrulation brain homeobox 2 (GBX2) transcription factor was downregulated in glioma tissues and cells. SNHG6 inhibited GBX2 transcription by mediating the H3K27me3 modification induced by polycomb repressive complex 2 (PRC2). Moreover, GBX2 decreased the promoter activities and downregulated the expression of the flotillin protein family 1 (FLOT1) oncogene. In conclusion, NCBP3/SNHG6 inhibits GBX2 transcription in a PRC2-dependent manner to facilitate the malignant progression of gliomas.


Assuntos
Regulação Neoplásica da Expressão Gênica , Glioma/genética , Glioma/metabolismo , Histonas/metabolismo , Proteínas de Homeodomínio/genética , Interferência de RNA , RNA Longo não Codificante/genética , Linhagem Celular Tumoral , Progressão da Doença , Técnicas de Silenciamento de Genes , Glioma/patologia , Humanos , Gradação de Tumores , Estadiamento de Neoplasias , Regiões Promotoras Genéticas , Ligação Proteica , Transcrição Gênica , Proteínas Supressoras de Tumor/genética , Proteínas Supressoras de Tumor/metabolismo
10.
Mol Ther ; 28(2): 613-630, 2020 02 05.
Artigo em Inglês | MEDLINE | ID: mdl-31813799

RESUMO

Studies have found that RNA-binding proteins (RBPs) and long non-coding RNAs (lncRNAs) are dysregulated and play an important regulatory role in the development of tumors. Based on The Cancer Genome Atlas (TCGA) database, our findings from experiments, and the evidence of previous studies, we screened DiGeorge syndrome critical region gene 8 (DGCR8), ZFAT antisense RNA 1 (ZFAT-AS1), and caudal type homeobox 2 (CDX2) as research candidates. In the present study, DGCR8 and CDX2 were highly expressed and ZFAT-AS1 was markedly downregulated in glioma tissues and cells. DGCR8 or CDX2 knockdown or ZFAT-AS1 overexpression suppressed glioma cell proliferation, migration, and invasion and facilitated apoptosis. DGCR8 might decrease ZFAT-AS1 expression by attenuating its stability in a manner of inducing its cleavage. Importantly, ZFAT-AS1 could inhibit CDX2 transcription by mediating the methylation of histone H3 on lysine 27 (H3K27me3) modification induced by PRC2 in the CDX2 promoter region. In addition, CDX2 transcriptionally activated DGCR8 expression by binding to its promoter regions, forming a positive feedback loop of DGCR8/ZFAT-AS1/CDX2. In conclusion, DGCR8/ZFAT-AS1 promotes CDX2 transcription in a PRC2 complex-dependent manner to facilitate the malignant biological behavior of glioma cells.


Assuntos
Fator de Transcrição CDX2/genética , Regulação Neoplásica da Expressão Gênica , Glioma/genética , RNA Antissenso , RNA Longo não Codificante/genética , Proteínas de Ligação a RNA/genética , Fatores de Transcrição/genética , Animais , Apoptose/genética , Linhagem Celular Tumoral , Movimento Celular/genética , Proliferação de Células/genética , Modelos Animais de Doenças , Técnicas de Silenciamento de Genes , Glioma/metabolismo , Glioma/mortalidade , Glioma/patologia , Humanos , Camundongos , Regiões Promotoras Genéticas , Ligação Proteica , Ensaios Antitumorais Modelo de Xenoenxerto
11.
J Cell Mol Med ; 24(11): 6120-6136, 2020 06.
Artigo em Inglês | MEDLINE | ID: mdl-32368853

RESUMO

Increasing evidence has suggested that gliomas can supply blood through vasculogenic mimicry. In this study, the expression and function of ZNRD1-AS1-144aa-uORF (144aa-uORF) and some non-coding RNAs in gliomas were assessed. Real-time quantitative PCR or Western blot was used to discover the expression of 144aa-uORF, ZNRD1-AS1, miR-499a-5p, ELF1 and EMI1 in gliomas. In addition, RIP and RNA pull-down assays were applied to explore the interrelationship between 144aa-uORF and ZNRD1-AS1. The role of the 144aa-uORF\ZNRD1-AS1\miR-499a-5p\ELF1\EMI1 axis in vasculogenic mimicry formation of gliomas was analysed. This study illustrates the reduced expression of the 144aa-uORF in glioma tissues and cells. Up-regulation of 144aa-uORF inhibits proliferation, migration, invasion and vasculogenic mimicry formation within glioma cells. The up-regulated 144aa-uORF can increase the degradation of ZNRD1-AS1 through the nonsense-mediated RNA decay (NMD) pathway. Knockdown of ZNRD1-AS1 inhibits vasculogenic mimicry in glioma cells by modulating miR-499a-5p. At the same time, miR-499a-5p is down-regulated and has a tumour-suppressive effect in gliomas. In addition, ZNRD1-AS1 serves as a competitive endogenous RNA (ceRNA) and regulates the expression of ELF1 by binding to miR-499a-5p. Notably, ELF1 binds to the promoter region of EMI1 and up-regulates EMI1 expression, while simultaneously promoting vasculogenic mimicry in glioma cells. This study suggests that the 144aa-uORF\ZNRD1-AS1\miR-499a-5p\ELF1\EMI1 axis takes key part in regulating the formation of vasculogenic mimicry in gliomas and may provide a potential target for glioma treatment.


Assuntos
Neoplasias Encefálicas/genética , Proteínas de Ciclo Celular/metabolismo , Proteínas F-Box/metabolismo , Glioma/genética , Antígenos de Histocompatibilidade Classe I/metabolismo , MicroRNAs/metabolismo , Proteínas Nucleares/metabolismo , Fases de Leitura Aberta/genética , Transdução de Sinais , Fatores de Transcrição/metabolismo , Animais , Sequência de Bases , Ligação Competitiva , Neoplasias Encefálicas/patologia , Linhagem Celular Tumoral , Proliferação de Células/genética , Regulação para Baixo/genética , Regulação Neoplásica da Expressão Gênica , Glioma/patologia , Células HEK293 , Humanos , Camundongos Nus , MicroRNAs/genética , Regiões Promotoras Genéticas/genética , Ligação Proteica/genética , Estabilidade de RNA/genética , Análise de Sobrevida , Regulação para Cima/genética , Ensaios Antitumorais Modelo de Xenoenxerto
12.
J Cell Mol Med ; 24(1): 342-355, 2020 01.
Artigo em Inglês | MEDLINE | ID: mdl-31654502

RESUMO

Recent studies indicate circular RNAs are related to dysregulation of vascular endothelial cell function, yet the underlying mechanisms have remained elusive. Here, we characterized the functional role of circular RNA USP1 (circ-USP1) in the regulation of the blood-tumour barrier (BTB) permeability and the potential mechanisms. In the current study, the circ-USP1 expressing level was up-regulated in glioma cerebral microvascular endothelial cells (GECs) of the BTB model in vitro. Knockdown of circ-USP1 disrupted the barrier integrity, increased its permeability as well as reduced tight junction-related protein claudin-5, occludin and ZO-1 expressions in GECs. Bioinformatic prediction and luciferase assay indicated that circ-USP1 bound to miR-194-5p and suppressed its activity. MiR-194-5p contributed to circ-USP1 knockdown-induced increase of BTB permeability via targeting and down-regulating transcription factor FLI1. Furthermore, FLI1 regulated the expressions of claudin-5, occludin and ZO-1 in GECs through binding to their promoter regions. Single or combined treatment of circ-USP1 and miR-194-5p effectively promoted anti-tumour drug doxorubicin across BTB to induce apoptosis of glioma cells. Overall, this present study identified the crucial regulation of circ-USP1 on BTB permeability via miR-194-5p/FLI1 axis-mediated regulation of tight junction proteins, which might facilitate the development of therapeutics against human gliomas.


Assuntos
Glioma/sangue , Glioma/patologia , MicroRNAs/metabolismo , Proteína Proto-Oncogênica c-fli-1/metabolismo , RNA Circular/metabolismo , Apoptose/efeitos dos fármacos , Apoptose/genética , Sequência de Bases , Neoplasias Encefálicas/sangue , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/patologia , Linhagem Celular Tumoral , Doxorrubicina/farmacologia , Células Endoteliais/efeitos dos fármacos , Células Endoteliais/metabolismo , Células Endoteliais/patologia , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Inativação Gênica , Glioma/genética , Células HEK293 , Humanos , MicroRNAs/genética , Permeabilidade , Proteínas de Junções Íntimas/metabolismo
13.
Cancer Sci ; 111(3): 891-906, 2020 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-31943575

RESUMO

Upstream ORF (uORF) is a translational initiation element located in the 5'UTR of eukaryotic mRNAs. Studies have found that uORFs play an important regulatory role in many diseases. Based on The Cancer Genome Atlas database, the results of our experiments and previous research evidence, we investigated transcription factor AP-4 (TFAP4) and its uORF, LIM and SH3 protein 1 (LASP1), long noncoding RNA 00520 (LINC00520), and microRNA (miR)-520f-3p as candidates involved in glioma malignancy, which is a poorly understood process. Both TFAP4-66aa-uORF and miR-520f-3p were downregulated, and TFAP4, LASP1, and LINC00520 were highly expressed in glioma tissues and cells. TFAP4-66aa-uORF or miR-520f-3p overexpression or TFAP4, LASP1, or LINC00520 knockdown inhibited glioma cell proliferation, migration, and invasion, but promoted apoptosis. TFAP4-66aa-uORF inhibited the translation of TFAP4 by binding to the TFAP4 mRNA. MicroRNA-520f-3p inhibited TFAP4 expression by binding to its 3'UTR. However, LINC00520 could promote the expression of TFAP4 by competitively binding to miR-520f-3p. In addition, TFAP4 transcriptionally activated LASP1 and LINC00520 expression by binding to their promoter regions, forming a positive feedback loop of TFAP4/LINC00520/miR-520f-3p. Our findings together indicated that TFAP4-66aa-uORF inhibited the TFAP4/LINC00520/miR-520f-3p feedback loop by directly inhibiting TFAP4 expression, subsequently leading to inhibition of glioma malignancy. This provides a basis for developing new therapeutic approaches for glioma treatment.


Assuntos
Movimento Celular/genética , Proteínas de Ligação a DNA/genética , Glioma/genética , MicroRNAs/genética , RNA Longo não Codificante/genética , Proteínas de Ligação a RNA/genética , Regiões 3' não Traduzidas/genética , Animais , Apoptose/genética , Linhagem Celular Tumoral , Proliferação de Células/genética , Proteínas do Citoesqueleto/genética , Regulação para Baixo/genética , Feminino , Regulação Neoplásica da Expressão Gênica/genética , Glioma/patologia , Células HEK293 , Humanos , Proteínas com Domínio LIM/genética , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus
14.
RNA Biol ; 17(9): 1293-1308, 2020 09.
Artigo em Inglês | MEDLINE | ID: mdl-32372707

RESUMO

The blood-brain barrier (BBB) plays a pivotal role in the maintenance and regulation of the neural microenvironment. The BBB breakdown is a pathological change in early Alzheimer's disease (AD). RNA-binding proteins (RBPs) and long non-coding RNAs (lncRNAs) are involved in the regulation of BBB permeability. Our study demonstrates the role of TRA2A/LINC00662/ELK4 axis in regulating BBB permeability in AD microenvironment. In Aß1-42-incubated microvascular endothelial cells (ECs) of the BBB model in vitro, TRA2A and LINC00662 were enriched. TRA2A increased the stability of LINC00662 by binding with it. The knockdown of either TRA2A or LINC00662 decreased BBB permeability due to increased expression of tight junction-related proteins. ELK4 was less expressed in the BBB model in AD microenvironment in vitro. LINC00662 mediated the degradation of ELK4 mRNA by SMD pathway. Downregulation of ELK4 increased BBB permeability by increasing the tight junction-related protein expression.TRA2A/LINC00662/ELK4 axis plays a crucial role in the regulation of BBB permeability in AD microenvironment, which may provide a novel target for the therapy of AD.


Assuntos
Barreira Hematoencefálica/metabolismo , Microambiente Celular/genética , Regulação da Expressão Gênica , RNA Longo não Codificante/metabolismo , Proteínas de Ligação a RNA/metabolismo , Proteínas Elk-4 do Domínio ets/genética , Doença de Alzheimer/etiologia , Doença de Alzheimer/metabolismo , Doença de Alzheimer/patologia , Peptídeos beta-Amiloides/metabolismo , Biomarcadores , Células Endoteliais/metabolismo , Técnicas de Silenciamento de Genes , Humanos , Fragmentos de Peptídeos/metabolismo , Permeabilidade , Regiões Promotoras Genéticas , Ligação Proteica , Interferência de RNA , Estabilidade de RNA , RNA Longo não Codificante/genética , Proteínas de Junções Íntimas/genética , Proteínas de Junções Íntimas/metabolismo
15.
RNA Biol ; 17(12): 1777-1788, 2020 12.
Artigo em Inglês | MEDLINE | ID: mdl-32713259

RESUMO

Blood-tumour barrier (BTB) has been known to significantly attenuate the efficacy of chemotherapy for glioma. In this report, we identified that insulin-like grown factor 2 mRNA-binding protein 2 (IGF2BP2) was over-expressed in glioma microvessel and glioma endothelial cells (GECs). Knockdown of IGF2BP2 decreased the expression of lncRNA FBXL19-AS1 and tight junction-related proteins, thereby promoting BTB permeability. FBXL19-AS1 was over-expressed and more enriched in the cytoplasm of GECs. In addition, FBXL19-AS1 could bind to 3'-UTR of ZNF765 mRNA and down-regulate ZNF765 mRNA expression through STAU1-mediated mRNA decay (SMD). The low expression of ZNF765 was discovered in GECs and verified to increase BTB permeability by inhibiting the promoter activities of tight junction-related proteins. Meanwhile, ZNF765 also inhibited the transcriptional activity of IGF2BP2, thereby forming a feedback loop in regulating the BTB permeability. Single or combined application of silenced IGF2BP2 and FBXL19-AS1 improved the delivery and antitumor efficiency of doxorubicin (DOX). In general, our study revealed the regulation mechanism of IGF2BP2/FBXL19-AS1/ZNF765 axis on BTB permeability, which may provide valuable insight into treatment strategy for glioma.


Assuntos
Proteínas do Citoesqueleto/metabolismo , Proteínas de Ligação a DNA/genética , Proteínas F-Box/genética , Regulação Neoplásica da Expressão Gênica , RNA Longo não Codificante/genética , Proteínas de Ligação a RNA/genética , Proteínas de Ligação a RNA/metabolismo , Microambiente Tumoral , Linhagem Celular Tumoral , Células Endoteliais/metabolismo , Técnicas de Silenciamento de Genes , Humanos , Degradação do RNAm Mediada por Códon sem Sentido , Permeabilidade , Ligação Proteica , Estabilidade de RNA , Transcriptoma , Microambiente Tumoral/genética
16.
J Cell Mol Med ; 23(8): 5048-5062, 2019 08.
Artigo em Inglês | MEDLINE | ID: mdl-31207033

RESUMO

Dysregulation of long non-coding RNAs (lncRNAs) confirm that it plays a crucial role in tumourigenesis and malignant progression of glioma. The present study demonstrated that LncRNA secretory carrier membrane protein 1 (SCAMP1) was up-regulated and functioned as an oncogene in glioma cells. In addition, miR-499a-5p was down-regulated meanwhile exerted tumour-suppressive function in glioma cells. Subsequently, inhibition of SCAMP1 significantly restrained the cell proliferation, migration and invasion, as well as promoted apoptosis by acting as a molecular sponge of miR-499a-5p. Transcription factor LIM homeobox transcription factor 1, alpha (LMX1A) was overexpressed in glioma tissues and cells. Moreover, miR-499a-5p targeted LMX1A 3'-UTR in a sequence-specific manner. Hence, down-regulation of SCAMP1 remarkably reduced the expression level of LMX1A, indicating that LMX1A participated in miR-499a-5p-induced tumour-suppressive effects on glioma cells. Furthermore, knockdown of LMX1A decreased NLR family, CARD domain containing 5 (NLRC5) mRNA and protein expression levels through directly binding to the NLRC5 promoter region. Down-regulation of NLRC5 obviously inhibited malignant biological behaviours of glioma cells through attenuating the activity of Wnt/ß-catenin signalling pathway. In conclusion, our study clarifies that SCAMP1/miR-499a-5p/LMX1A/NLRC5 axis plays a critical role in modulating malignant progression of glioma cells, which provide a novel therapeutic strategy for glioma treatment.


Assuntos
Neoplasias Encefálicas/metabolismo , Glioma/metabolismo , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Proteínas com Homeodomínio LIM/genética , MicroRNAs/metabolismo , RNA Longo não Codificante/metabolismo , Fatores de Transcrição/genética , Proteínas de Transporte Vesicular/metabolismo , Regiões 3' não Traduzidas , Animais , Apoptose/genética , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/patologia , Linhagem Celular Tumoral , Movimento Celular/genética , Proliferação de Células/genética , Imunoprecipitação da Cromatina , Regulação Neoplásica da Expressão Gênica , Glioma/genética , Glioma/patologia , Humanos , Peptídeos e Proteínas de Sinalização Intracelular/genética , Proteínas com Homeodomínio LIM/metabolismo , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , MicroRNAs/genética , Análise de Sequência com Séries de Oligonucleotídeos , Prognóstico , Regiões Promotoras Genéticas , RNA Longo não Codificante/genética , Fatores de Transcrição/metabolismo , Proteínas de Transporte Vesicular/genética , Via de Sinalização Wnt/genética
17.
Biochim Biophys Acta Mol Cell Res ; 1864(10): 1605-1617, 2017 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-28666797

RESUMO

Glioma stem cells (GSCs) make up highly tumorigenic subpopulations within gliomas, and aberrant expression of GSC genes is a major underlying cause of glioma pathogenesis and treatment failure. The present study characterized the expression and function of long non-coding RNA growth arrest specific 5 (GAS5) in GSCs in order to elucidate the molecular mechanisms by which GAS5 contributes to glioma pathogenesis. We demonstrate that GAS5 suppresses GSC malignancy by binding to miR-196a-5p. miR-196a-5p, an onco-miRNA, stimulates GSC proliferation, migration, and invasion, in addition to reducing levels of apoptosis. miR-196a-5p specifically downregulates the expression of forkhead box protein O1 (FOXO1) by targeting its 3' untranslated region (3'-UTR). FOXO1 upregulates expression of phosphotyrosine interaction domain containing 1 (PID1), thereby inhibiting GSC tumorigenicity and growth. FOXO1 also upregulates migration and invasion inhibitory protein (MIIP), resulting in attenuation of migration and invasion activities. Interestingly, we also show that FOXO1 promotes GAS5 transcription, thus forminga positive feedback loop. These data provide insights into potential new pathways for GSC molecular therapy and suggest that GAS5 may be an efficacious target for glioma treatments.


Assuntos
Proteína Forkhead Box O1/genética , Glioma/genética , MicroRNAs/genética , RNA Longo não Codificante/genética , Animais , Apoptose/genética , Carcinogênese/genética , Proliferação de Células/genética , Regulação Neoplásica da Expressão Gênica , Glioma/patologia , Humanos , Camundongos , Células-Tronco Neoplásicas/patologia , Ensaios Antitumorais Modelo de Xenoenxerto
18.
Cell Physiol Biochem ; 45(1): 131-147, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29310118

RESUMO

BACKGROUND/AIMS: Vasculogenic mimicry (VM) has been reported to be a novel glioma neovascularization process. Anti-VM therapy provides new insight into glioma clinical management. In this study, we revealed the role of the long non-coding RNA HOXA cluster antisense RNA 2 (HOXA-AS2) in malignant glioma behaviors and VM formation. METHODS: Quantitative real-time PCR was performed to determine the expression levels of HOXA-AS2 in glioma samples and glioblastoma cell lines. CD34-periodic acid-Schiff dual-staining was performed to assess VM in glioma samples. CCK-8, transwell, and Matrigel tube formation assays were performed to measure the effects of HOXA-AS2 knockdown on cell viability, migration, invasion, and VM tube formation, respectively. RNA immunoprecipitation, dual-luciferase reporter and Western blot assays were performed to explore the molecular mechanisms underlying the functions of HOXS-AS2 in glioblastoma cells. A nude mouse xenograft model was used to investigate the role of HOXA-AS2 in xenograft glioma growth and VM density. Student's t-tests, one-way ANOVAs followed by Bonferroni posthoc tests, and chi-square tests were used for the statistical analyses. RESULTS: HOXA-AS2 was upregulated in glioma samples and cell lines and was positively correlated with VM. HOXA-AS2 knockdown attenuated cell viability, migration, invasion, and VM formation in glioma cells and inhibited the expression of vascular endothelial-cadherin (VE-cadherin), as well as the expression and activity of matrix metalloproteinase matrix metalloproteinase (MMP)-2 and MMP-9. miR-373 was downregulated in glioma samples and cell lines and suppressed malignancy in glioblastoma cells. HOXA-AS2 bound to miR-373 and negatively regulated its expression. Epidermal growth factor receptor (EGFR), a target of miR-373, increased the expression levels of VE-cadherin, as well as the expression and activity levels of MMP-2 and MMP-9, via activating phosphatidylinositol 3-kinase/serine/threonine kinase pathways. HOXA-AS2 knockdown combined with miR-373 overexpression yielded optimal tumor suppressive effects and the lowest VM density in vivo. CONCLUSION: HOXA-AS2 knockdown inhibited malignant glioma behaviors and VM formation via the miR-373/EGFR axis.


Assuntos
Neoplasias Encefálicas/patologia , Glioma/patologia , MicroRNAs/metabolismo , Neovascularização Patológica , RNA Longo não Codificante/metabolismo , Animais , Encéfalo/metabolismo , Encéfalo/patologia , Neoplasias Encefálicas/irrigação sanguínea , Neoplasias Encefálicas/metabolismo , Linhagem Celular Tumoral , Movimento Celular , Proliferação de Células , Receptores ErbB/antagonistas & inibidores , Receptores ErbB/genética , Receptores ErbB/metabolismo , Glioma/irrigação sanguínea , Glioma/metabolismo , Humanos , Masculino , Metaloproteinase 2 da Matriz/metabolismo , Metaloproteinase 9 da Matriz/metabolismo , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , MicroRNAs/genética , Fosfatidilinositol 3-Quinases/metabolismo , Interferência de RNA , RNA Longo não Codificante/antagonistas & inibidores , RNA Longo não Codificante/genética , Regulação para Cima
19.
Biochim Biophys Acta Mol Basis Dis ; 1864(5 Pt A): 1783-1794, 2018 May.
Artigo em Inglês | MEDLINE | ID: mdl-29501773

RESUMO

The long non-coding RNA (lncRNA) PVT1 is reported to be involved in tumorigenesis and the progression of many malignancies. However, the function of PVT1 in gliomas remains unclarified. The present study demonstrated the expression level of PVT1 using qRT-PCR. The role of PVT1 in the regulation of biological behaviors of glioma cells was investigated using CCK-8 assay, Transwell assay and flow cytometry. The possible molecular mechanisms were also elucidated. In our results, PVT1 was up-regulated in glioma specimens and cell lines. Knockdown of PVT1 impaired the malignant behaviors of glioma cells via the suppression of proliferation, migration and invasion, as well as through promotion of apoptosis. Furthermore, PVT1 was identified to affect the glioma cells via binding to miR-190a-5p and miR-488-3p, which were down-regulated and played tumor suppressor roles in glioma cells. Up-regulated miR-190a-5p or miR-488-3p partially rescued the suppressive effect induced by PVT1 knockdown. Myocyte enhancer factor 2C (MEF2C) was a direct downstream target of miR-190a-5p and miR-488-3p, which was proved to be an oncogene and involved in the PVT1 knockdown induced regulation of biological behaviors of glioma cells. Over-expression of MEF2C up-regulated JAGGED1 by increasing the promoter activity of JAGGED1. PVT1 knockdown combined with miR-190a-5p and miR-488-3p over-expression contributed to the smallest tumor volume and the longest survivals in nude mice. In conclusion, PVT1-miR-190a-5p/miR-488-3p-MEF2C-JAGGED1 axis is involved in proliferation and progression of glioma. Thus, PVT1 may become a novel target in glioma therapy.


Assuntos
Proliferação de Células , Glioma/metabolismo , MicroRNAs/metabolismo , RNA Longo não Codificante/metabolismo , RNA Neoplásico/metabolismo , Animais , Linhagem Celular Tumoral , Glioma/genética , Humanos , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , MicroRNAs/genética , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/metabolismo , RNA Longo não Codificante/genética , RNA Neoplásico/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA