Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
2.
Clin Cancer Res ; 29(16): 3172-3188, 2023 08 15.
Artigo em Inglês | MEDLINE | ID: mdl-37249604

RESUMO

PURPOSE: Temozolomide resistance remains a major obstacle in the treatment of glioblastoma (GBM). The combination of temozolomide with another agent could offer an improved treatment option if it could overcome chemoresistance and prevent side effects. Here, we determined the critical drug that cause ferroptosis in GBM cells and elucidated the possible mechanism by which drug combination overcomes chemoresistance. EXPERIMENTAL DESIGN: Haloperidol/temozolomide synergism was assessed in GBM cell lines with different dopamine D2 receptor (DRD2) expression in vitro and in vivo. Inhibitors of ferroptosis, autophagy, endoplasmic reticulum (ER) stress and cyclic adenosine monophosphate (cAMP)/protein kinase A (PKA) were used to validate the specific mechanisms by which haloperidol and temozolomide induce ferroptosis in GBM cells. RESULTS: In the present work, we demonstrate that the DRD2 level is increased by temozolomide in a time-dependent manner and is inversely correlated with temozolomide sensitivity in GBM. The DRD2 antagonist haloperidol, a butylbenzene antipsychotic, markedly induces ferroptosis and effectively enhances temozolomide efficacy in vivo and in vitro. Mechanistically, haloperidol suppressed the effect of temozolomide on cAMP by antagonizing DRD2 receptor activity, and the increases in cAMP/PKA triggered ER stress, which led to autophagy and ferroptosis. Furthermore, elevated autophagy mediates downregulation of FTH1 expression at the posttranslational level in an autophagy-dependent manner and ultimately leads to ferroptosis. CONCLUSIONS: Our results provide experimental evidence for repurposing haloperidol as an effective adjunct therapy to inhibit adaptive temozolomide resistance to enhance the efficacy of chemoradiotherapy in GBM, a strategy that may have broad prospects for clinical application.


Assuntos
Neoplasias Encefálicas , Ferroptose , Glioblastoma , Humanos , Temozolomida/farmacologia , Temozolomida/uso terapêutico , Glioblastoma/tratamento farmacológico , Glioblastoma/genética , Glioblastoma/metabolismo , Haloperidol/farmacologia , Haloperidol/uso terapêutico , Antagonistas dos Receptores de Dopamina D2/farmacologia , Linhagem Celular Tumoral , Autofagia , Estresse do Retículo Endoplasmático , Neoplasias Encefálicas/tratamento farmacológico , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/metabolismo , Resistencia a Medicamentos Antineoplásicos/genética , Receptores de Dopamina D2/genética
3.
Cancer Lett ; 557: 216078, 2023 03 31.
Artigo em Inglês | MEDLINE | ID: mdl-36736531

RESUMO

For treatment of glioblastoma (GBM), temozolomide (TMZ) and radiotherapy (RT) exert antitumor effects by inducing DNA double-strand breaks (DSBs), mainly via futile DNA mismatch repair (MMR) and inducing apoptosis. Here, we provide evidence that RBBP4 modulates glioblastoma resistance to chemotherapy and radiotherapy by recruiting transcription factors and epigenetic regulators that bind to their promoters to regulate the expression of the Mre11-Rad50-NBS1(MRN) complex and the level of DNA-DSB repair, which are closely associated with recovery from TMZ- and radiotherapy-induced DNA damage in U87MG and LN229 glioblastoma cells, which have negative MGMT expression. Disruption of RBBP4 induced GBM cell DNA damage and apoptosis in response to TMZ and radiotherapy and enhanced radiotherapy and chemotherapy sensitivity by the independent pathway of MGMT. These results displayed a possible chemo-radioresistant mechanism in MGMT negative GBM. In addition, the RBBP4-MRN complex regulation axis may provide an interesting target for developing therapy-sensitizing strategies for GBM.


Assuntos
Quebras de DNA de Cadeia Dupla , Glioblastoma , Humanos , Glioblastoma/patologia , Enzimas Reparadoras do DNA/genética , Proteína Homóloga a MRE11/genética , Reparo do DNA , Temozolomida/uso terapêutico , Fatores de Transcrição/genética , DNA , Quimiorradioterapia , Proteínas de Ciclo Celular/metabolismo , Proteínas de Ligação a DNA/genética , Hidrolases Anidrido Ácido/metabolismo , Proteína 4 de Ligação ao Retinoblastoma/genética , Proteína 4 de Ligação ao Retinoblastoma/metabolismo
5.
Cell Death Dis ; 13(11): 990, 2022 11 23.
Artigo em Inglês | MEDLINE | ID: mdl-36418306

RESUMO

Switch/sucrose-nonfermenting (SWI/SNF) complexes play a key role in chromatin remodeling. Recent studies have found that SMARCC2, as the core subunit of the fundamental module of the complex, plays a key role in its early assembly. In this study, we found a unique function of SMARCC2 in inhibiting the progression of glioblastoma by targeting the DKK1 signaling axis. Low expression of SMARCC2 is found in malignant glioblastoma (GBM) compared with low-grade gliomas. SMARCC2 knockout promoted the proliferation of glioblastoma cells, while its overexpression showed the opposite effect. Mechanistically, SMARCC2 negatively regulates transcription by dynamically regulating the chromatin structure and closing the promoter region of the target gene DKK1, which can be bound by the transcription factor EGR1. DKK1 knockdown significantly reduced the proliferation of glioblastoma cell lines by inhibiting the PI3K-AKT pathway. We also studied the functions of the SWIRM and SANT domains of SMARCC2 and found that the SWIRM domain plays a more important role in the complete chromatin remodeling function of SMARCC2. In addition, in vivo studies confirmed that overexpression of SMARCC2 could significantly inhibit the size of intracranial gliomas in situ in nude mice. Overall, this study shows that SMARCC2, as a tumor suppressor, inhibits the proliferation of glioblastoma by targeting the transcription of the oncogene DKK1 through chromatin remodeling, indicating that SMARCC2 is a potentially attractive therapeutic target in glioblastoma.


Assuntos
Montagem e Desmontagem da Cromatina , Glioblastoma , Animais , Camundongos , Montagem e Desmontagem da Cromatina/genética , Proteína 1 de Resposta de Crescimento Precoce/genética , Proteína 1 de Resposta de Crescimento Precoce/metabolismo , Glioblastoma/genética , Camundongos Nus , Fosfatidilinositol 3-Quinases/genética , Fosfatidilinositol 3-Quinases/metabolismo , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo
6.
Front Cell Neurosci ; 16: 971100, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-36072565

RESUMO

This study aimed to explore the mechanism underlying cognitive dysfunction mediated by the lateral hypothalamic area (LHA) in a hypothalamic-hippocampal circuit in rats with lesion-induced hypothalamic obesity (HO). The HO model was established by electrically lesioning the hypothalamic nuclei. The open field (OP) test, Morris water maze (MWM), novel object recognition (NOR), and novel object location memory (NLM) tests were used to evaluate changes in cognition due to alterations in the hypothalamic-hippocampal circuit. Western blotting, immunohistochemical staining, and cholera toxin subunit B conjugated with Alexa Fluor 488 (CTB488) reverse tracer technology were used to determine synaptophysin (SYN), postsynaptic density protein 95 (PSD95), ionized calcium binding adaptor molecule 1 (Iba1), neuronal nuclear protein (NeuN), and Caspase3 expression levels and the hypothalamic-hippocampal circuit. In HO rats, severe obesity was associated with cognitive dysfunction after the lesion of the hypothalamus. Furthermore, neuronal apoptosis and activated microglia in the downstream of the lesion area (the LHA) induced microglial infiltration into the intact hippocampus via the LHA-hippocampal circuit, and the synapses engulfment in the hippocampus may be the underlying mechanism by which the remodeled microglial mediates memory impairments in HO rats. The HO rats exhibited microglial infiltration and synapse loss into the hippocampus from the lesioned LHA via the hypothalamic-hippocampal circuit. The underlying mechanisms of memory function may be related to the circuit.

7.
World Neurosurg ; 164: e724-e740, 2022 08.
Artigo em Inglês | MEDLINE | ID: mdl-35595047

RESUMO

BACKGROUND: Treatment of Chiari malformation (CM) is controversial, especially when it coexists with "stable" or Type II basilar invagination (CM + II-BI). Precise evaluation of craniovertebral junction (CVJ) stability is crucial in such patients; however, this has never been validated. This study aimed to dynamically evaluate atlanto-condyle and atlantoaxial stability by kinematic computed tomography (CT) and report its surgical treatment. METHODS: The study recruited 101 patients (control, CM, and CM + II-BI groups: 48, 34, and 19 patients, respectively). During kinematic CT, the CVJ stability-related parameters were measured and compared between the 3 groups. The surgical strategy for treating CM + II-BI was based on these results. Preoperative and postoperative images were acquired, and functional scores were used to assess the outcome. RESULTS: Among the 3 groups, the length of the clivus and the height of the condyle were the shortest in the CM + II-BI group, which was accompanied by the greatest rotation of the atlas and atlanto-condyle facet movement on cervical flexion and extension. Moreover, in such patients, increased Chamberlain's baseline violation indicated the aggregate invagination of the odontoid in the flexed position, and asymmetric displacement of atlantoaxial facets was observed. Seventeen CM + II-BI patients underwent surgical treatment with atlantoaxial distraction and occipitocervical fusion. The syringomyelia width and tonsillar herniation decreased significantly, and functional scores indicated symptom relief and good outcomes. CONCLUSIONS: CVJ instability, especially the ultramovement of atlanto-condyle facets, commonly exists in II-BI as evaluated using kinematic CT. The surgical strategy of atlantoaxial distraction and occipitocervical fusion should be considered to treat such patients.


Assuntos
Malformação de Arnold-Chiari , Articulação Atlantoaxial , Platibasia , Fusão Vertebral , Malformação de Arnold-Chiari/complicações , Malformação de Arnold-Chiari/diagnóstico por imagem , Malformação de Arnold-Chiari/cirurgia , Articulação Atlantoaxial/anormalidades , Articulação Atlantoaxial/diagnóstico por imagem , Articulação Atlantoaxial/cirurgia , Fenômenos Biomecânicos , Descompressão Cirúrgica/métodos , Humanos , Platibasia/complicações , Platibasia/diagnóstico por imagem , Platibasia/cirurgia , Fusão Vertebral/métodos , Tomografia Computadorizada por Raios X
8.
Front Oncol ; 12: 1024208, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-36601479

RESUMO

Introduction: Necroptosis-related genes are essential for the advancement of IDH-wild-type GBM. However, the putative effects of necroptosis-related lncRNAs (nrlncRNAs) in IDH-wild-type GBM remain unknown. Methods: By using the TCGA and GTEx databases, a nrlncRNA prognostic signature was created using LASSO Cox regression. The median risk score was used to categorize the patients into low and high-risk groups. To confirm the validity, univariate, multivariate Cox regression and ROC curves were used. Furthermore, by enrichment analysis, immune correlation analysis, and drug sensitivity analysis, the targeted lncRNAs were selected for further verification. As the highest upregulated expression in tumor than peritumor specimens, RP11-131L12.4 was selected for phenotype and functional experiments in primary GBM cells. Results: Six lncRNAs were proved to be closely related to necroptosis in IDH-1-wild-type GBM, which were used to create a new signature. For 1-, 2-, and 3-year OS, the AUCs were 0.709, 0.645 and 0.694, respectively. Patients in the low-risk group had a better prognosis, stronger immune function activity, and more immune cell infiltration. In contrast, enrichment analysis revealed that the malignant phenotype was more prevalent in the high-risk group. In vitro experiments indicated that RP11-131L12.4 increased the tumor proliferation, migration and invasion, but decreased the necroptosis. Moreover, this nrlncRNA was also proved to be negatively associated with patient prognosis. Conclusion: The signature of nrlncRNAs may aid in the formulation of tailored and precise treatment for individuals with IDH-wild-type GBM. RP11-131L12.4 may play indispensable role in necroptosis suppression.

9.
Mol Med Rep ; 24(2)2021 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-34080022

RESUMO

Glioma is the most common type of central nervous system tumor. SWItch/sucrose non­fermentable (SWI/SNF) is a tumor suppressor that serves an important role in epithelial­mesenchymal transition (EMT). The present study aimed to identify key molecules involved in the EMT process. SWI/SNF related, matrix associated, actin dependent regulator of chromatin subfamily c member 2 (SMARCC2) is mutated in and its expression is low in multiple types of cancer. SMARCC2 is the core subunit of the chromatin­remodeling complex, SWI/SNF. Relative mRNA SMARCC2 expression levels in human glioma tissue were analyzed via reverse transcription­quantitative PCR, whereas the protein expression levels were determined via immunohistochemistry staining. SMARCC2 expression was knocked down in glioma cells using small interfering RNA (si) and overexpressed by infection with adenovirus vectors carrying SMARCC2 cDNA. Wound healing and Transwell assays were performed to assess cell migration and invasion, respectively. Subsequently, immunofluorescence and western blotting were performed to analyze the expression levels of the oncogene c­Myc, which is associated with SMARCC2. SMARCC2 combines with C­MYC to downregulate its expression. Consistent with the results of the bioinformatics analysis, which revealed that the upregulated expression levels of SMARCC2 were associated with a more favorable prognosis in patients with glioma, the mRNA and protein expression levels of SMARCC2 were significantly upregulated in low­grade glioma tissues compared with high­grade glioma tissues. The results of the wound healing assay demonstrated that cell migration was significantly increased in the siSMARCC2­1/3 groups compared with the negative control (NC) group. By contrast, the migratory ability of cells was significantly reduced following transduction with adenovirus overexpressing SMARCC2, which upregulated the expression of SMARCC2, compared with the lentiviral vector­non­specific control (LVS­NC) group. The Transwell assay results further showed that SMARCC2 overexpression significantly inhibited the migratory and invasive abilities of U87MG and LN229 cells compared with the LVS­NC group. Co­immunoprecipitation assays were subsequently conducted to validate the binding of SMARCC2 and c­Myc; the results demonstrated that the expression of c­Myc was downregulated in adenovirus­transfected cells compared with LVS­NC­transfected cells. The results of the western blotting experiments demonstrated that the expression levels of N­cadherin, vimentin, snail family transcriptional repressor 1 and ß­catenin were notably downregulated, whereas the expression levels of T­cadherin were markedly upregulated in cell lines stably overexpressing SMARCC2 compared with the LVS­NC group. In conclusion, the results of the present study suggested that SMARCC2 may inhibit Wnt/ß­catenin signaling by regulating c­Myc expression in glioma. SMARCC2 regulates the EMT status of the glioblastoma cell line by mediating the expression of the oncogene C­MYC to inhibit its migration and invasion ability. Thus, SMARCC2 may function as a tumor suppressor or oncogene by regulating associated oncogenes or tumor suppressor genes.


Assuntos
Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/metabolismo , Glioma/genética , Proteínas Proto-Oncogênicas c-myc/genética , Proteínas Proto-Oncogênicas c-myc/metabolismo , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo , Via de Sinalização Wnt/genética , Adulto , Idoso , Caderinas/metabolismo , Linhagem Celular Tumoral , Movimento Celular/genética , Transição Epitelial-Mesenquimal/genética , Feminino , Técnicas de Silenciamento de Genes , Glioma/metabolismo , Humanos , Masculino , Pessoa de Meia-Idade , Invasividade Neoplásica/genética , Fatores de Transcrição da Família Snail/metabolismo , Análise de Sobrevida , Vimentina/metabolismo , Adulto Jovem , beta Catenina/metabolismo
10.
Nan Fang Yi Ke Da Xue Xue Bao ; 41(1): 69-74, 2021 Jan 30.
Artigo em Chinês | MEDLINE | ID: mdl-33509755

RESUMO

OBJECTIVE: To establish a mouse model bearing orthotopic temozolomide (TMZ)-resistant glioma that mimics the development of drug resistance in gliomas in vivo. METHODS: Seventy-eight adult C57BL/6 mice were randomly divided into 6 groups (n=13), including 3 TMZ induced groups with low, medium and high doses (5, 25, and 50 mg/kg, respectively) and 3 control groups. In each group, 5 mice were used for evaluating tumor size, 5 for observing survival, and 3 for collecting tumor tissues for primary cell culture. In low-dose TMZ induced group, 3 mice bearing orthotopic murine glioma GL261 cell xenografts received intraperitoneal injections of 5 mg/kg TMZ for 5 days followed by a 10-day washout period before collecting glioma tissues. Tumor cell suspensions were prepared and injected in the mice in the medium-dose group, which were treated with the same protocol but with an increased TMZ dose, and the tumor cells harvested from 3 mice were injected in the high-dose group. The mice bearing GL261 cell xenografts in the 3 control groups received no treatment or were injected with medium- or high-dose TMZ. Cell colony forming assay was used to assess TMZ resistance of each generation of the tumor cells; CCK8 assay was used to determine drug resistance index of the cells. RESULTS: The mouse models bearing TMZresistant glioma was successfully established. The cells from the high-dose induced group showed a significantly higher colony-forming rate than those from the high-dose control group (P < 0.05), and had a drug resistance 4.25 times higher than that of the cells from untreated control group. High-dose TMZ significantly reduced the tumor volume in the control group (P < 0.05) but not in the high-dose induced group (P < 0.01). The survival time of the tumor-bearing mice was significantly shortened in the high-dose induced group (P=0.0018). CONCLUSIONS: Progressive increase of TMZ doses in mice bearing orthotopic gliomas can effectively induce TMZ resistance of the gliomas.


Assuntos
Neoplasias Encefálicas , Glioma , Animais , Antineoplásicos Alquilantes/farmacologia , Neoplasias Encefálicas/tratamento farmacológico , Linhagem Celular Tumoral , Modelos Animais de Doenças , Resistencia a Medicamentos Antineoplásicos , Glioma/tratamento farmacológico , Camundongos , Camundongos Endogâmicos C57BL , Temozolomida/farmacologia , Temozolomida/uso terapêutico
11.
J Neurosci Methods ; 346: 108928, 2020 12 01.
Artigo em Inglês | MEDLINE | ID: mdl-32898574

RESUMO

BACKGROUND: Spinal glioma is a nervous system tumor that tends to relapse and has no specific prognostic molecular biomarkers. Thus, a stable and reproduceable animal research model of spinal glioma is urgently needed. NEW METHOD: We established a new in situ tumor xenograft model of spinal glioma using nude mice. In this study, we implanted tumors into the cervical spinal cord of nude mice to mimic the pathological characteristics of the original tumors. RESULTS: Through anatomical experiments, we found that the cervical lamina of mice was thinner, the intervertebral space was much wider, and the adhesion muscles were more easily separated. According to the examination of spinal cord sections, the best puncture point we identified was located 0.9 mm lateral to the posterior median line at the level of the line between the midpoints of the scapulae and at a depth of 0.9 mm. In the nude mouse xenograft experiment, the implanted tumor tissue retained the pathological characteristics of the original tumor. COMPARISON WITH EXISTING METHOD(S): This model used the cervical spinal cord as the puncture site and patient-derived primary tumor cells, which has never been performed before. Tumor cells could be injected directly without damaging the lamina. Thus, we could reduce the risk of man-made spinal cord injury and infection and avoid destroying the stability and integrity of the spine. CONCLUSIONS: This study established a stable and reliable animal model of spinal glioma for further molecular research and targeted therapy development.


Assuntos
Glioma , Recidiva Local de Neoplasia , Animais , Modelos Animais de Doenças , Xenoenxertos , Humanos , Camundongos , Camundongos Nus
12.
Neurosurgery ; 87(6): 1206-1222, 2020 11 16.
Artigo em Inglês | MEDLINE | ID: mdl-32691825

RESUMO

BACKGROUND: Spinal schwannoma is a common benign tumor. However, the high recurrence rate and incidence of surgical complications are unsolved problems. OBJECTIVE: To propose a morphological classification of spinal schwannoma based on tumor-membrane relationships to increase the gross total resection (GTR) rate and to decrease the incidence of surgical complications. METHODS: Histological techniques were used to study 7 adult cadavers. Following picrosirius staining, the membranes around the nerve root were observed under a microscope. Data from 101 patients with spinal schwannoma were also collected for clinical analysis. RESULTS: The sleeve around the spinal nerve root consisted of dura and arachnoid tissues. The space between them gradually narrowed and fused at the proximal pole of the nerve root ganglion. Spinal schwannomas were divided into 4 types based on membranous structure: intrapial (type I), subarachnoidal (type II), intra- and extradural (type III), and extradural growth (type IV). Types II and III were further subdivided into 2 subtypes. GTR was achieved in all patients (100%), with no tumor recurrence during follow-up. Overall functional status significantly improved postoperatively. A total of 59 patients (92%) showed improvement or significant improvement postoperatively. There was no difference in surgical outcomes among the tumor classifications (P = .618). No intraoperative vertebral artery injuries or postoperative cerebrospinal fluid fistula occurred. CONCLUSION: Spinal schwannoma classification based on a membranous hierarchy provides an intuitive platform for preoperative planning and intraoperative safety. This classification scheme may help surgeons better define surgical goals and anticipate or even avoid complications from resection.


Assuntos
Neurilemoma , Adulto , Humanos , Neurilemoma/cirurgia , Complicações Pós-Operatórias/epidemiologia , Estudos Retrospectivos , Raízes Nervosas Espinhais/cirurgia , Resultado do Tratamento
13.
Theranostics ; 9(7): 1909-1922, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31037147

RESUMO

Rationale: Glioblastoma (GBM) is the most common and aggressive brain tumor, characterized by its propensity to invade the surrounding brain parenchyma. The effect of extracellular high-mobility group box 1 (HMGB1) protein on glioblastoma (GBM) progression is still controversial. p62 is overexpressed in glioma cells, and has been associated with the malignant features and poor prognosis of GBM patients. Hence, this study aimed to clarify the role of p62 in HMGB1-induced epithelial-mesenchymal transition (EMT) of GBM both in vitro and in vivo. Methods: Immunoblotting, immunofluorescence and qRT-PCR were performed to evaluate EMT progression in both human GBM cell line and primary GBM cells. Transwell and wound healing assays were used to assess the invasion and migration of GBM cells. shRNA technique was used to investigate the role of p62 in HMGB1-induced EMT both in vitro and in vivo orthotopic tumor model. Co-immunoprecipitation assay was used to reveal the interaction between p62 and GSK-3ß (glycogen synthase kinase 3 beta). Immunohistochemistry was performed to detect the expression levels of proteins in human GBM tissues. Results: In this study, GBM cells treated with recombinant human HMGB1 (rhHMGB1) underwent spontaneous EMT through GSK-3ß/Snail signaling pathway. In addition, our study revealed that rhHMGB1-induced EMT of GBM cells was accompanied by p62 overexpression, which was mediated by the activation of TLR4-p38-Nrf2 signaling pathway. Moreover, the results demonstrated that p62 knockdown impaired rhHMGB1-induced EMT both in vitro and in vivo. Subsequent mechanistic investigations showed that p62 served as a shuttling factor for the interaction of GSK-3ß with proteasome, and ultimately activated GSK-3ß/Snail signaling pathway by augmenting the degradation of GSK-3ß. Furthermore, immunohistochemistry analysis revealed a significant inverse correlation between p62 and GSK-3ß, and a combination of the both might serve as a more powerful predictor of poor survival in GBM patients. Conclusions: This study suggests that p62 is an effector for HMGB1-induced EMT, and may represent a novel therapeutic target in GBM.


Assuntos
Transição Epitelial-Mesenquimal/fisiologia , Glioblastoma/metabolismo , Glicogênio Sintase Quinase 3 beta/metabolismo , Proteína HMGB1/metabolismo , Proteína Sequestossoma-1/metabolismo , Fatores de Transcrição da Família Snail/metabolismo , Linhagem Celular , Linhagem Celular Tumoral , Movimento Celular/fisiologia , Glioma/metabolismo , Células HEK293 , Humanos , Complexo de Endopeptidases do Proteassoma/metabolismo , Transdução de Sinais/fisiologia , Fatores de Transcrição/metabolismo
14.
Clin Cancer Res ; 25(12): 3602-3616, 2019 06 15.
Artigo em Inglês | MEDLINE | ID: mdl-30862693

RESUMO

PURPOSE: Glioblastoma, a common malignant intracranial tumor, has the most dismal prognosis. Autophagy was reported to act as a survival-promoting mechanism in gliomas by inducing epithelial-to-mesenchymal transition (EMT). Here, we determined the critical molecules involved in autophagy-induced EMT and elucidated the possible mechanism of chemoradiotherapy resistance and tumor recurrence. EXPERIMENTAL DESIGN: We used isobaric tags for relative and absolute quantitation to identify the critical proteins and pathway mediating EMT via autophagy inducer treatment, and tested the expression of these proteins using tissue microarray of gliomas and clinical glioblastoma samples as well as tissues and cells separated from the core lesion and tumor-peripheral region. Analysis of the Cancer Genome Atlas database and 110 glioblastoma cases revealed the prognostic value of these molecules. The functional role of these critical molecules was further confirmed by in vitro experiments and intracranial xenograft in nude mice. RESULTS: Autophagy inducers significantly upregulated the expression of HERC3, which promotes ubiquitination-mediated degradation of SMAD7 in an autolysosome-dependent manner. The corresponding increase in p-SMAD2/3 level and TGFß pathway activation finally induced EMT in cell lines and primary glioblastoma cells. Moreover, HERC3 overexpression was observed in pseudo-palisade cells surrounding tumor necrosis and in tumor-adjacent tissue; high HERC3 and low SMAD7 levels predicted poor clinical outcome in glioblastoma; xenograft of nude mice and in vitro experiments confirmed these findings. CONCLUSIONS: Together, our findings reveal the indispensable role of HERC3 in regulating canonical SMAD2/3-dependent TGFß pathway involvement in autophagy-induced EMT, providing insights toward a better understanding of the mechanism of resistance to temozolomide and peripheral recurrence of glioblastoma.


Assuntos
Neoplasias Encefálicas/metabolismo , Neoplasias Encefálicas/patologia , Glioblastoma/metabolismo , Glioblastoma/patologia , Proteína Smad7/metabolismo , Temozolomida/farmacologia , Ubiquitina-Proteína Ligases/metabolismo , Animais , Antineoplásicos Alquilantes/farmacologia , Autofagia , Neoplasias Encefálicas/tratamento farmacológico , Linhagem Celular Tumoral , Resistencia a Medicamentos Antineoplásicos , Transição Epitelial-Mesenquimal , Glioblastoma/tratamento farmacológico , Células HEK293 , Humanos , Camundongos , Camundongos Nus , Prognóstico , Proteólise , Transdução de Sinais , Taxa de Sobrevida , Fator de Crescimento Transformador beta/metabolismo , Ubiquitina-Proteína Ligases/genética , Ubiquitinação , Ensaios Antitumorais Modelo de Xenoenxerto
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...