Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 40
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Cell Mol Life Sci ; 81(1): 247, 2024 Jun 03.
Artigo em Inglês | MEDLINE | ID: mdl-38829550

RESUMO

BACKGROUND: The high degree of intratumoral genomic heterogeneity is a major obstacle for glioblastoma (GBM) tumors, one of the most lethal human malignancies, and is thought to influence conventional therapeutic outcomes negatively. The proneural-to-mesenchymal transition (PMT) of glioma stem cells (GSCs) confers resistance to radiation therapy in glioblastoma patients. POLD4 is associated with cancer progression, while the mechanisms underlying PMT and tumor radiation resistance have remained elusive. METHOD: Expression and prognosis of the POLD family were analyzed in TCGA, the Chinese Glioma Genome Atlas (CGGA) and GEO datasets. Tumorsphere formation and in vitro limiting dilution assay were performed to investigate the effect of UCHL3-POLD4 on GSC self-renewal. Apoptosis, TUNEL, cell cycle phase distribution, modification of the Single Cell Gel Electrophoresis (Comet), γ-H2AX immunofluorescence, and colony formation assays were conducted to evaluate the influence of UCHL3-POLD4 on GSC in ionizing radiation. Coimmunoprecipitation and GST pull-down assays were performed to identify POLD4 protein interactors. In vivo, intracranial xenograft mouse models were used to investigate the molecular effect of UCHL3, POLD4 or TCID on GCS. RESULT: We determined that POLD4 was considerably upregulated in MES-GSCs and was associated with a meagre prognosis. Ubiquitin carboxyl terminal hydrolase L3 (UCHL3), a DUB enzyme in the UCH protease family, is a bona fide deubiquitinase of POLD4 in GSCs. UCHL3 interacted with, depolyubiquitinated, and stabilized POLD4. Both in vitro and in vivo assays indicated that targeted depletion of the UCHL3-POLD4 axis reduced GSC self-renewal and tumorigenic capacity and resistance to IR treatment by impairing homologous recombination (HR) and nonhomologous end joining (NHEJ). Additionally, we proved that the UCHL3 inhibitor TCID induced POLD4 degradation and can significantly enhance the therapeutic effect of IR in a gsc-derived in situ xenograft model. CONCLUSION: These findings reveal a new signaling axis for GSC PMT regulation and highlight UCHL3-POLD4 as a potential therapeutic target in GBM. TCID, targeted for reducing the deubiquitinase activity of UCHL3, exhibited significant synergy against MES GSCs in combination with radiation.


Assuntos
Células-Tronco Neoplásicas , Tolerância a Radiação , Ubiquitina Tiolesterase , Humanos , Ubiquitina Tiolesterase/metabolismo , Ubiquitina Tiolesterase/genética , Tolerância a Radiação/genética , Células-Tronco Neoplásicas/metabolismo , Células-Tronco Neoplásicas/patologia , Células-Tronco Neoplásicas/efeitos da radiação , Animais , Camundongos , Linhagem Celular Tumoral , Glioma/patologia , Glioma/genética , Glioma/radioterapia , Glioma/metabolismo , Apoptose/genética , Apoptose/efeitos da radiação , Ubiquitinação , Neoplasias Encefálicas/patologia , Neoplasias Encefálicas/metabolismo , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/radioterapia , Camundongos Nus , Fenótipo , Regulação Neoplásica da Expressão Gênica , Prognóstico
2.
Cancer Lett ; 593: 216875, 2024 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-38643837

RESUMO

Mesenchymal glioma stem cells (MES GSCs) are a subpopulation of cells in glioblastoma (GBM) that contribute to a worse prognosis owing to their highly aggressive nature and resistance to radiation therapy. Here, OCT4 is characterized as a critical factor in sustaining the stemness phenotype of MES GSC. We find that OCT4 is expressed intensively in MES GSC and is intimately associated with poor prognosis, moreover, OCT4 depletion leads to diminished invasive capacity and impairment of the stem phenotype in MES GSC. Subsequently, we demonstrated that USP5 is a deubiquitinating enzyme which directly interacts with OCT4 and preserves OCT4 stability through its deubiquitination. USP5 was additionally proven to be aberrantly over-expressed in MES GSCs, and its depletion resulted in a noticeable diminution of OCT4 and consequently a reduced self-renewal and tumorigenic capacity of MES GSCs, which can be substantially restored by ectopic expression of OCT4. In addition, we detected the dominant molecule that regulates USP5 transcription, E2F1, with dual luciferase reporter gene analysis. In combination, targeting the E2F1-USP5-OCT4 axis is a potentially emerging strategy for the therapy of GBM.


Assuntos
Neoplasias Encefálicas , Fator de Transcrição E2F1 , Células-Tronco Neoplásicas , Fator 3 de Transcrição de Octâmero , Proteases Específicas de Ubiquitina , Humanos , Fator 3 de Transcrição de Octâmero/genética , Fator 3 de Transcrição de Octâmero/metabolismo , Células-Tronco Neoplásicas/patologia , Células-Tronco Neoplásicas/metabolismo , Animais , Fator de Transcrição E2F1/metabolismo , Fator de Transcrição E2F1/genética , Neoplasias Encefálicas/patologia , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/metabolismo , Proteases Específicas de Ubiquitina/genética , Proteases Específicas de Ubiquitina/metabolismo , Glioma/patologia , Glioma/genética , Glioma/metabolismo , Linhagem Celular Tumoral , Regulação Neoplásica da Expressão Gênica , Camundongos , Estabilidade Proteica , Glioblastoma/patologia , Glioblastoma/genética , Glioblastoma/metabolismo , Ubiquitinação
5.
Oper Neurosurg (Hagerstown) ; 22(5): 290-297, 2022 05 01.
Artigo em Inglês | MEDLINE | ID: mdl-35315837

RESUMO

BACKGROUND: The management of petroclival region meningioma remains the ultimate achievement in neurosurgery, because of the formidable technical challenges involved. OBJECTIVE: To describe the technique and feasibility of the purely endoscopic far-lateral supracerebellar infratentorial approach (EF-SCITA) for the treatment of petroclival region meningiomas. METHODS: We reviewed the clinical data of 10 consecutive cases of petroclival region meningiomas treated with the EF-SCITA from August 2018 to August 2020. The clinical outcomes were analyzed. The patient was placed in the lateral position, and then, a "C" shaped incision and craniotomy with exposed sigmoid and transverse sinuses were performed. With the endoscopic holder, endoscopic procedures were performed using standard 2-hand microsurgical techniques. Whether the tentorium or Meckel cave was handled depended on the tumor extension. RESULTS: The mean diameter was 45 × 25 mm. Dizziness and headache were the main symptoms. All 10 patients achieved gross total or subtotal resection (Petroclival Meningioma Grade I-III) with good neurological outcomes. The EF-SCITA provides satisfactory, direct exposure to the petroclival region. Cranial nerve deficits are the main postoperative complications. Two patients had a trochlear nerve injury, 3 patients had transient facial paralysis, and 2 patients had oculomotor paralysis (1 total and 1 incomplete), but both of them recovered during the follow-up period. One patient experienced an ipsilateral superior cerebellar artery infarction, and another patient had transient hemiparesis. CONCLUSION: The EF-SCITA is effective for most petroclival region meningiomas, except for the cavernous sinus type. This approach simplifies craniotomy procedures, omits burdensome petrosectomy, and avoids crossing posterior neurovascular structures.


Assuntos
Neoplasias Meníngeas , Meningioma , Neoplasias da Base do Crânio , Craniotomia/métodos , Humanos , Neoplasias Meníngeas/diagnóstico por imagem , Neoplasias Meníngeas/patologia , Neoplasias Meníngeas/cirurgia , Meningioma/diagnóstico por imagem , Meningioma/patologia , Meningioma/cirurgia , Procedimentos Neurocirúrgicos/métodos , Neoplasias da Base do Crânio/cirurgia
6.
Cancer Cell Int ; 21(1): 347, 2021 Jul 05.
Artigo em Inglês | MEDLINE | ID: mdl-34225723

RESUMO

BACKGROUND: Glioblastoma remains one of the most lethal brain cancers. T-cell immunoglobulin and mucin domain 1 (Tim-1) is associated with various immune diseases. The molecular mechanism of Tim-1 in regulating glioblastoma cell proliferation, invasion, and migration is still unknown. Moreover, it has shown that miR-133a plays an important role in glioblastoma. However, little is known about the interaction between Tim-1 and miR-133a in glioblastoma. METHODS: Tim-1 expression in glioblastoma and normal brain tissues was detected by qPCR, Western Blot and IHC. After Tim-1 knockdown in U251 and U87 cells, genes showing significantly differential expression, along with the significant differential miRNAs were analyzed using RNA-seq analysis. The binding sites were verified using dual-luciferase reporter gene assay. U251 and U87 cells were allocated into the small harpin-negative control (sh-NC), sh-Tim-1, sh-Tim-1 + inhibitor NC, and sh-Tim-1 + miR-133a inhibitor group. Cell proliferation, migration, and invasion were determined by CCK-8, flow cytometry, wound-healing and Transwell assays, respectively. Next, U251 and U87 cells were allocated into the mimic NC, miR-133a mimic, miR-133a mimic + pcDNA3.1, and miR-133a mimic + pcDNA3.1-TGFBR1 groups, followed by the detection of cell proliferation, migration, and invasion. Western blot was used to identify the expression of vital kinases in the Wnt/ß-catenin pathway. RESULTS: Tim-1 was highly expressed in glioblastoma tissues compared with that in normal brain tissues. RNA-seq analysis showed that Tim-1 knockdown could lead to the downregulation of TGFBR1 and the upregulation of miR-133a. The binding sites between TGFBR1 and miR-133a were confirmed. Tim-1 knockdown impaired the invasion, migration, proliferation of U251 and U87 cells, which could be reversed by miR-133a downregulation. miR-133a upregulation inhibited the proliferation, invasion, and migration of U251 and U87 cells, which could be reversed by TGFBR1 upregulation. Tim-1 knockdown and miR-133a upregulation could inhibit the activation of the Wnt/ß-catenin pathway, while the elevation of TGFBR1 showed opposite effects. CONCLUSION: Tim-1 knockdown inhibited glioblastoma cell proliferation, invasion, and migration through the miR-133a/TGFBR1 axis and restrained the activation of the Wnt/ß-catenin pathway.

7.
Exp Ther Med ; 21(5): 491, 2021 May.
Artigo em Inglês | MEDLINE | ID: mdl-33791000

RESUMO

Gliomas account for 50% of primary brain tumours in the central nervous system. Small ubiquitin-like modifier 1 pseudogene 3 (SUMO1P3), a newly identified long non-coding RNA (lncRNA), serves an oncogenic role in various types of cancer. The aim of the present study was to investigate the effect of SUMO1P3 on glioma progression. The results demonstrated that SUMO1P3 expression was upregulated in glioma tissues and cell lines. Furthermore, SUMO1P3 was associated with a poor overall survival of patients with glioma. The results of the in vitro cell proliferation and flow cytometry assays demonstrated that SUMO1P3-knockdown suppressed cell proliferation and cell cycle. The results of the wound healing and Transwell assays demonstrated that SUMO1P3-knockdown significantly repressed cell migration and invasion. In addition, SUMO1P3 promoted glioma by regulating the expression levels of ß-catenin, cyclin-D1, N-cadherin and E-cadherin. Overall, the results of the present study suggested that SUMO1P3 may act as an oncogene by regulating cell proliferation, cell cycle, cell migration and invasion in glioma, and may represent a novel diagnostic biomarker and therapeutic target for glioma.

8.
Neoplasma ; 68(4): 732-741, 2021 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-33847130

RESUMO

Protein- or peptide-based therapeutics have emerged as an innovative strategy for the treatment of cancer. Our previous research demonstrated that tripartite motif 9 short isoform (TRIM9s) is a tumor suppressor in glioma. In this report, we investigated whether a new peptide derived from TRIM9s, named T9sP, inhibits glioma progression and determined the possible molecular mechanism. The CCK-8 proliferation assay was performed in LN229 and U251 glioma cells. The scratch-wound assay was used to determine the migration of the cells. Apoptosis was assessed by flow cytometry using Annexin V-FITC/PI double staining method. The relative protein expression levels were detected by immunoblot analysis. The cell-penetrating peptide TAT was fused with T9sP to form TAT-T9sP. TAT-T9sP efficiently penetrated through the cell membrane of both LN229 and U251 cells. TAT-T9sP inhibited proliferation and migration and promoted apoptosis of glioma cells. TAT-T9sP activated p38 signaling by upregulating MKK6, and a p38 inhibitor, SB203580, reversed the inhibitory effects of TAT-T9sP on glioma cells. These results indicated the potential of TAT-T9sP for the development of a new anti-glioma medicine.


Assuntos
Neoplasias Encefálicas , Glioma , Apoptose , Neoplasias Encefálicas/tratamento farmacológico , Linhagem Celular Tumoral , Movimento Celular , Proliferação de Células , Glioma/tratamento farmacológico , Humanos , Peptídeos/farmacologia , Transdução de Sinais
9.
J Cancer ; 12(6): 1592-1603, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33613746

RESUMO

Background: Invasive growth is one of the most typical features of aggressive types of malignant cancer, including glioblastoma. Lysosomal cysteine protease-cathepsin S (CTSS), has been reported to be involved in invasive growth and distant metastasis of cancer cells. However, the underlying mechanisms remained elusive. Methods: U87 and U251 human glioblastoma cell lines were applied in this study. Cell migration and invasion ability were measured by wound healing assay and transwell assay. Western blot was employed to detect the expression levels of proteins. Immunofluorescence assays of cells and tissues were used to visualize the localization and expression of proteins. The SPSS software was used for statistical analysis. Results: Our results showed that the high expression of CTSS was link with the grades of glioma tissues. The CTSS inhibitor-Z-FL-COCHO (ZFL), could attenuate TGF-ß-induced invasive growth as proven by wound healing and transwell assays. Furthermore, inhibition of CTSS could reverse TGF-ß-induced epithelial-to-mesenchymal transition (EMT) and restore TGF-ß-triggered tight junction proteins turnover, thus decreasing glioblastoma cell mobility. We also observed that TGF-ß could change the morphology of glioblastoma cells, redistribute intermediate-filament, vimentin, which was highly relevant to mesenchymal type cells and enhanced mobility. However, inhibition of CTSS could significantly restore this transformation. Our results proved that PI3K/AKT/mTOR pathway was significantly suppressed in the TGF-ß+ZFL (CTSS inhibitor) groups, and AKT activator-SC79, could reverse the anti-invasion effect of CTSS, indicating an important role of PI3K/AKT/mTOR pathway in this process. Conclusion: Z-FL-COCHO (ZFL), a CTSS inhibitor, could reverse TGF-ß-induced EMT and change of tight junction proteins via PI3K/AKT/mTOR pathway.

10.
J Craniofac Surg ; 32(1): e19-e20, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-32858607

RESUMO

OBJECTIVES: The aim of this study was to study and explore the genetic mechanism of familial meningiomas through 3 cases of familial tuberculum sellae meningioma. METHODS: A retrospective analysis of clinical data of 3 cases of familial tuberculum sellae meningioma patients, and the pathological results of types and immunohistochemical results of the 3 patients were compare. REULTS: Three cases of postoperative pathology were meningiomas (mixed type), immunohistochemical examination showed that Vimentin, epithelial membrain antigen , and Ki67 were positive. CONCLUSIONS: The occurrence of meningiomas is associated with ≥1 chromosomal deletions, and the absence of certain tumor suppressor genes may be the genetic basis for the familial growth of meningiomas.


Assuntos
Neoplasias Meníngeas , Meningioma , Neoplasias da Base do Crânio , Idoso , Feminino , Humanos , Neoplasias Meníngeas/genética , Neoplasias Meníngeas/cirurgia , Meningioma/genética , Meningioma/cirurgia , Procedimentos Neurocirúrgicos , Estudos Retrospectivos , Sela Túrcica , Neoplasias da Base do Crânio/cirurgia , Resultado do Tratamento
11.
Aging (Albany NY) ; 12(24): 25035-25059, 2020 11 10.
Artigo em Inglês | MEDLINE | ID: mdl-33197884

RESUMO

Our previous studies have shown that the δ-opioid receptor (DOR) is an important neuroprotector via the regulation of PTEN-induced kinase 1 (PINK1), a mitochondria-related molecule, under hypoxic and MPP+ insults. Since mitochondrial dysfunctions are observed in both hypoxia and MPP+ insults, this study further investigated whether DOR is cytoprotective against these insults by targeting mitochondria. Through comparing DOR-induced responses to hypoxia versus MPP+-induced parkinsonian insult in PC12 cells, we found that both hypoxia and MPP+ caused a collapse of mitochondrial membrane potential and severe mitochondrial dysfunction. In sharp contrast to its inappreciable effect on mitochondria in hypoxic conditions, DOR activation with UFP-512, a specific agonist, significantly attenuated the MPP+-induced mitochondrial injury. Mechanistically, DOR activation effectively upregulated PINK1 expression and promoted Parkin's mitochondrial translocation and modification, thus enhancing the PINK1-Parkin mediated mitophagy. Either PINK1 knockdown or DOR knockdown largely interfered with the DOR-mediated mitoprotection in MPP+ conditions. Moreover, there was a major difference between hypoxia versus MPP+ in terms of the regulation of mitophagy with hypoxia-induced mitophagy being independent from DOR-PINK1 signaling. Taken together, our novel data suggest that DOR activation is neuroprotective against parkinsonian injury by specifically promoting mitophagy in a PINK1-dependent pathway and thus attenuating mitochondrial damage.


Assuntos
Hipóxia Celular/genética , Potencial da Membrana Mitocondrial/genética , Mitocôndrias/metabolismo , Mitofagia/genética , Proteínas Quinases/genética , Receptores Opioides delta/genética , Ubiquitina-Proteína Ligases/genética , 1-Metil-4-fenilpiridínio/toxicidade , Animais , Benzimidazóis/farmacologia , Técnicas de Silenciamento de Genes , Herbicidas/toxicidade , Potencial da Membrana Mitocondrial/efeitos dos fármacos , Mitocôndrias/efeitos dos fármacos , Mitofagia/efeitos dos fármacos , Oligopeptídeos/farmacologia , Células PC12 , Transtornos Parkinsonianos/induzido quimicamente , Transtornos Parkinsonianos/genética , Transtornos Parkinsonianos/metabolismo , Proteínas Quinases/efeitos dos fármacos , Proteínas Quinases/metabolismo , Ratos , Receptores Opioides delta/agonistas , Receptores Opioides delta/metabolismo , Superóxidos/metabolismo , Ubiquitina-Proteína Ligases/efeitos dos fármacos , Ubiquitina-Proteína Ligases/metabolismo , Regulação para Cima
12.
Cancer Cell Int ; 19: 280, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31708690

RESUMO

BACKGROUND: Glioma is the most common central nervous system (CNS) tumour. p62, an important autophagy adaptor, plays a crucial role in cancer. However, the role of p62 in the progression of glioma is poorly characterized. METHODS: We examined the expression of p62 in glioma tissues and cell lines. Then we investigated the function of p62 in vitro, and clarified the mechanism underlying the regulation of p62 expression. RESULTS: We revealed that p62 was upregulated at both the mRNA and protein levels in human glioma tissues irrelevant to isocitrate dehydrogenase (IDH) status. Then, we found that overexpression of p62 promoted glioma progression by promoting proliferation, migration, glycolysis, temozolomide (TMZ) resistance and nuclear factor κB (NF-κB) signalling pathway, and repressing autophagic flux and reactive oxygen species (ROS) in vitro. In accordance with p62 overexpression, knockdown of p62 exerted anti-tumour effects in glioma cells. Subsequently, we demonstrated that miR-124-3p directly targeted the 3'-UTR of p62 mRNA, leading to the downregulation of p62. Finally, we found that p62 function could be partially reversed by miR-124-3p overexpression. CONCLUSIONS: Our results demonstrate that p62 can be targeted by miR-124-3p and acts as an oncogene in glioma, suggesting the potential value of p62 as a novel therapeutic target for glioma.

13.
Onco Targets Ther ; 12: 2383-2396, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-30992674

RESUMO

PURPOSE: Glioma is a malignant tumor that originates in the brain and spine and is difficult to be completely removed. Though glioma patients receive active treatment, the survival rate is still poor. Therefore, it is urgent to discover a new medicine to treat glioma patients in order to improve the survival rate. In this study, we explored the anticancer effect and the potential mechanism of luteolin on glioma in vitro. MATERIALS AND METHODS: Cell viability was determined by Cell Counting Kit-8 (CCK-8) assay. Fluorescent microscopy and flow cytometry analysis were used to determine the cellular apoptosis. Western blot analysis was performed to explore the changes in protein expression. Quantitative reverse transcription-PCR (qRT-PCR) analysis was utilized to evaluate the expression level of the tumor suppressor miR-124-3p. RESULTS: CCK-8 assays indicated that luteolin significantly inhibited glioma cell proliferation in a time- and dose-dependent manner. Fluorescent microscopy and flow cytometry analysis confirmed that luteolin induced glioma cell apoptosis. Western blot analysis showed that luteolin induced cellular apoptosis in glioma cells via MAPK activation (JNK, ERK, and p38). Luteolin stimulated the death receptor (FADD) to regulate the apoptosis proteins (Caspase-8, Caspase-3, and PARP). Luteolin increased the expression levels of LC3B II/I and downregulated the level of p62 that promotes cell autophagy. Finally, qRT-PCR confirmed that luteolin upregulated the expression levels of miR-124-3p. CONCLUSION: These findings illustrate that luteolin may be a potential drug for glioma treatment.

14.
Mol Neurobiol ; 56(1): 252-266, 2019 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-29687347

RESUMO

There is emerging evidence suggesting that neurotoxic insults and hypoxic/ischemic injury are underlying causes of Parkinson's disease (PD). Since PTEN-induced kinase 1 (PINK1) dysfunction is involved in the molecular genesis of PD and since our recent studies have demonstrated that the δ-opioid receptor (DOR) induced neuroprotection against hypoxic and 1-methyl-4-phenyl-pyridimium (MPP+) insults, we sought to explore whether DOR protects neuronal cells from hypoxic and/or MPP+ injury via the regulation of PINK1-related pathways. Using highly differentiated rat PC12 cells exposed to either severe hypoxia (0.5-1% O2) for 24-48 h or varying concentrations of MPP+, we found that both hypoxic and MPP+ stress reduced the level of PINK1 expression, while incubation with the specific DOR agonist UFP-512 reversed this reduction and protected the cells from hypoxia and/or MPP+-induced injury. However, the DOR-mediated cytoprotection largely disappeared after knocking down PINK1 by PINK1 small interfering RNA. Moreover, we examined several important signaling molecules related to cell survival and apoptosis and found that DOR activation attenuated the hypoxic and/or MPP+-induced reduction in phosphorylated Akt and inhibited the activation of cleaved caspase-3, whereas PINK1 knockdown largely deprived the cell of the DOR-induced effects. Our novel data suggests a unique mechanism underlying DOR-mediated cytoprotection against hypoxic and MPP+ stress via a PINK1-mediated regulation of signaling.


Assuntos
Regulação para Baixo , Hipóxia/patologia , Neuroproteção , Doença de Parkinson/patologia , Doença de Parkinson/prevenção & controle , Proteínas Quinases/metabolismo , Receptores Opioides delta/metabolismo , 1-Metil-4-fenilpiridínio , Animais , Caspase 3/metabolismo , Sobrevivência Celular , Ativação Enzimática , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Técnicas de Silenciamento de Genes , Células PC12 , Fosforilação , Proteínas Proto-Oncogênicas c-akt/metabolismo , RNA Interferente Pequeno/metabolismo , Ratos , Transdução de Sinais , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo
15.
Mol Cancer Ther ; 18(2): 459-469, 2019 02.
Artigo em Inglês | MEDLINE | ID: mdl-30413650

RESUMO

Glioma is the most common type of primary malignant brain tumor in adults. Our previous work discovered that plasma miR-454-3p may have some advantages in glioma prognosis, but the clinical significance and the regulatory mechanism of miR-454-3p in glioma have not been systematically investigated, especially regarding the relationship between circulating and tissue miR-454-3p. The expression level of miR-454-3p in glioma serum and tissues was analyzed through quantitative real-time PCR (qRT-PCR). Cell-Counting Kit 8 (CCK-8), wound healing, transwell invasion, apoptosis, and immunofluorescence assays were used to assess the role of miR-454-3p in glioma cancer cells. ATG12 was selected as the target gene of miR-454-3p by bioinformatic analysis. The relationship between ATG12 and miR-454-3p was further validated by luciferase reporter assays and Western blot analysis. miR-454-3p was significantly downregulated in tumor tissues, while it was remarkably upregulated in exosomes from the same patients with glioma. The area under curve (AUC) of exosomal miR-454-3p for glioma diagnosis was 0.8663. The exosomal miR-454-3p was prominently lower in the postoperative serums than that in the preoperative serums. High miR-454-3p expression in exosomes or low miR-454-3p expression in tissue was associated with poor prognosis. Restored expression of miR-454-3p suppressed cell proliferation, migration, invasion, and autophagy in glioma. ATG12 was validated as a direct target of miR-454-3p. The overexpression of ATG12 could partially reverse the effects induced by miR-454-3p suppression. Our data indicate that miR-454-3p may serve as an exosomal biomarker and may be developed into a novel treatment for glioma.


Assuntos
Proteína 12 Relacionada à Autofagia/genética , Biomarcadores Tumorais/genética , Neoplasias Encefálicas/diagnóstico , Exossomos/genética , Glioma/diagnóstico , MicroRNAs/genética , Regiões 3' não Traduzidas , Neoplasias Encefálicas/genética , Linhagem Celular Tumoral , Movimento Celular , Proliferação de Células , Regulação para Baixo , Regulação Neoplásica da Expressão Gênica , Glioma/genética , Humanos , Gradação de Tumores , Prognóstico , Análise de Sobrevida , Regulação para Cima
16.
J Neurooncol ; 141(2): 277-288, 2019 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-30460630

RESUMO

OBJECTIVE: Malignant glioma is a lethal brain tumor with a low survival rate and poor prognosis. New strategies are urgently needed to augment the chemotherapeutic effects of temozolomide (TMZ), the standard drug in glioma treatment. Carnosic acid (CA) has been reported to have anticancer, antioxidant and anti-infectious properties. In this study, we aimed to investigate the anticancer effects and the underlying mechanisms of CA in combination with TMZ in glioma cancer cells. METHODS: The glioma cancer cells were treated with TMZ, CA, or TMZ + CA. We evaluated cell survival by CCK-8 assay, cell anchorage-independent survival by colony formation assay, cell migration by wound-healing assay, cell cycle and cell apoptosis by flow cytometry, and protein expression by western blot. RESULTS: CA enhanced the cytotoxic effect of TMZ in glioma cancer cells. CA enhanced TMZ-induced inhibition of colony formation and cell migration and enhanced TMZ-induced cell cycle arrest and cellular apoptosis. Immunofluorescence suggested that CA in combination with TMZ triggered autophagy. Furthermore, CA promoted TMZ-induced cell cycle arrest and cellular apoptosis by Cyclin B1 inhibition and activation of PARP and Caspase-3, while CA promoted TMZ-induced cellular autophagy by p-AKT inhibition, p62 downregulation and LC3-I to LC3-II transition. CONCLUSION: These data suggest that the combination therapy of CA and TMZ strengthens the anticancer effect of TMZ by enhancing apoptosis and autophagy.


Assuntos
Abietanos/administração & dosagem , Antineoplásicos Alquilantes/administração & dosagem , Apoptose/efeitos dos fármacos , Autofagia/efeitos dos fármacos , Neoplasias Encefálicas/tratamento farmacológico , Glioma/tratamento farmacológico , Temozolomida/administração & dosagem , Pontos de Checagem do Ciclo Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Movimento Celular/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Sinergismo Farmacológico , Humanos
17.
Cell Physiol Biochem ; 48(2): 450-460, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30016785

RESUMO

BACKGROUND/AIMS: Glioma causes significant human mortalities annually. Molecularly-targeted therapy is a focus of glioma research. METHODS: Grb2-associated binding 1 (Gab1) expression and microRNA-29a-3p ("miR-29a-3p") expression in human glioma cells and tissues were tested by Western blotting assay and qRT-PCR assay. shRNA/siRNA strategy was applied to silence Gab1 in human glioma cells. miR-29a or anti-sense miR-29a construct was transfected to human glioma cells. Cell proliferation was tested by BrdU ELISA assay and cell counting assay. RESULTS: We show that expression of Gab1 was significantly elevated in human glioma tissues and cells, which correlated with downregulation of its putative microRNA: miR-29a-3p. In A172 glioma cells and primary human glioma cells, Gab1 shRNA/siRNA inhibited Akt-Erk activation and cell proliferation. Forced-expression of miR-29a-3p downregulated Gab1, inhibiting glioma cell proliferation, whereas miR-29a-3p was in-effective on cell proliferation in Gab1-silenced A172 cells. Furthermore, introduction of a 3'-untranslated region (3'-UTR) mutant Gab1 (UTR-G160A) blocked miR-29a-3p-induced inhibition on Akt signaling and A172 cell proliferation. CONCLUSIONS: miR-29a-3p downregulation leads to Gab1 upregulation to promote glioma cell proliferation.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Glioma/patologia , MicroRNAs/metabolismo , Regiões 3' não Traduzidas , Proteínas Adaptadoras de Transdução de Sinal/antagonistas & inibidores , Proteínas Adaptadoras de Transdução de Sinal/genética , Antagomirs/metabolismo , Linhagem Celular Tumoral , Proliferação de Células , Regulação para Baixo , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Glioma/metabolismo , Humanos , MicroRNAs/antagonistas & inibidores , MicroRNAs/genética , Proteínas Proto-Oncogênicas c-akt/metabolismo , Interferência de RNA , RNA Interferente Pequeno/metabolismo , Transdução de Sinais
18.
Oncotarget ; 9(15): 12112-12124, 2018 Feb 23.
Artigo em Inglês | MEDLINE | ID: mdl-29552296

RESUMO

Malignant glioma is one of the most common types of primary brain tumours. Long non-coding RNAs (lncRNAs) have recently emerged as a new class of therapeutic targets for many cancers. In this study, we aimed to explore the functional involvement of small nucleolar RNA host gene 14 (SNHG14) and its potential regulatory mechanism in glioma progression. SNHG14 was found to be downregulated in human glioma tissues and cell lines. SNHG14 significantly inhibited cell viability, reduced cell invasion, and induced apoptosis in glioma cell lines. Furthermore, a correlation analysis demonstrated that there was a negative correlation between SNHG14 expression and miR-92a-3p expression. Bioinformatics prediction and luciferase reporter assays demonstrated that miR-92a-3p could directly bind to SNHG14. miR-92a-3p was significantly upregulated in glioma and acted as an oncogene in glioma cells by inhibiting Bim. Moreover, mechanistic investigations showed that miR-92a-3p could reverse the tumour suppressive effects induced by SNHG14 in glioma, indicating that SNHG14 may act as an endogenous sponge that competes for binding to miR-92a-3p. Our results suggest that SNHG14 and miR-92a-3p may be promising molecular targets for glioma therapy.

19.
Onco Targets Ther ; 11: 1183-1192, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29535541

RESUMO

BACKGROUND: Glioblastoma multiforme (GBM) is the most malignant primary tumor of the central nervous system and is associated with a very poor prognosis. No further improvements in outcomes have been reported since radiotherapy-temozolomide therapy was introduced. Therefore, developing new agents to treat GBM is important. AIM: This study aimed to evaluate the anti-tumor effect of evodiamine (Evo) on GBM cells, and to determine the underlying mechanisms involved. RESULTS: According to MTT assay results, Evo significantly inhibited the cell proliferation in a time- and dose-dependent manner. Fluorescence microscopy and flow cytometry analyses revealed that Evo induced cell apoptosis in a concentration-dependent manner. Moreover, Evo induced reactive oxygen species (ROS) production and mitochondrial membrane potential (MMP) disruption. Finally, Evo induced apoptosis in cancer cells by suppressing PI3K/AKT signaling and inducing MAPK phosphorylation (p38 and JNK, but not ERK) to regulate apoptotic proteins (Bax, Bcl-2, Cytochrome c, Caspase-3, and PARP). CONCLUSION: In summary, Evo inhibits cell proliferation by inducing cellular apoptosis via suppressing PI3K/AKT and activating MAPK in GBM; these results indicate that Evo may be regarded as a new approach for GBM treatment.

20.
Cell Physiol Biochem ; 44(6): 2296-2309, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-29262398

RESUMO

BACKGROUND/AIMS: Hypoxic/ischemic injury to the liver is a frequently encountered clinical problem with limited therapeutic options. Since microRNAs (miRNAs) are involved in hypoxic/ ischemic events, and δ-opioid receptor (DOR) is protective against hypoxic/ischemic injury, we asked if pharmacological activation of DOR can alter hypoxic events by regulating miRNA expression in the liver. As the first step, the present work aimed at testing the effect of DOR activation on hepatic miRNA expression in hypoxia. METHODS: Male Sprague Dawley rats were exposed to hypoxia (9.5-10% O2) for 1, 5, or 10 days with or without DOR activation. The target miRNAs were selected according to TaqMan low-density array (TLDA) data and analyzed by quantitative real-time PCR. RESULTS: We found that: 1) 1-day hypoxia caused the upregulation of 9 miRNAs (miR-7a-5p, miR-10a-5p, miR-25-3p, miR-26b-5p, miR-122-5p, miR-128a-3p, miR-135b-5p, miR-145-5p, and miR-181a-5p) and the downregulation of 2 miRNAs (miR-34a-5p and miR-182); 2) 5 and 10-days hypoxia altered the expression of 4 miRNAs (miR-34c-5p, miR-184, miR-107-3p and miR192-5p); 3) DOR activation shifted the expression of 8 miRNAs (miR-122-5p, miR-146a-5p, miR-30e-5p, miR-128a-3p, miR-182, miR-192-5p miR-107-3p and miR-184) in normoxic condition; and 4) DOR activation modified hypoxia-induced changes in 6 miRNAs (miR-142-5p, miR-145-5p, miR-146a-5p, miR-204-5p, miR-34a-5p and miR-192-5p). CONCLUSION: Hypoxia significantly modifies the miRNA profile in the liver, while DOR activation can modify the hypoxic modification. Therefore, it is potentially possible to alter hypoxic/ischemic pathophysiology in the liver through DOR pharmacotherapy.


Assuntos
Regulação da Expressão Gênica , Hipóxia/genética , Fígado/metabolismo , MicroRNAs/genética , Receptores Opioides delta/genética , Animais , Perfilação da Expressão Gênica , Hipóxia/patologia , Fígado/patologia , Masculino , Ratos Sprague-Dawley
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...