Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 54
Filtrar
1.
Mol Cell ; 82(20): 3919-3931.e7, 2022 10 20.
Artigo em Inglês | MEDLINE | ID: mdl-36270249

RESUMO

Cancer-specific TERT promoter mutations have been linked to the reactivation of epigenetically silenced TERT gene by creating de novo binding motifs for E-Twenty-Six transcription factors, especially GABPA. How these mutations switch on TERT from epigenetically repressed states to expressed states have not been defined. Here, we revealed that EGFR activation induces ERK1/2-dependent phosphorylation of argininosuccinate lyase (ASL) at Ser417 (S417), leading to interactions between ASL and GABPA at the mutant regions of TERT promoters. The ASL-generated fumarate inhibits KDM5C, leading to enhanced trimethylation of histone H3 Lys4 (H3K4me3), which in turn promotes the recruitment of c-Myc to TERT promoters for TERT expression. Expression of ASL S417A, which abrogates its binding with GABPA, results in reduced TERT expression, inhibited telomerase activity, shortened telomere length, and impaired brain tumor growth in mice. This study reveals an unrecognized mechanistic insight into epigenetically activation of mutant TERT promoters where GABPA-interacted ASL plays an instrumental role.


Assuntos
Glioblastoma , Telomerase , Animais , Camundongos , Argininossuccinato Liase/genética , Argininossuccinato Liase/metabolismo , Linhagem Celular Tumoral , Receptores ErbB/genética , Fumaratos , Regulação Neoplásica da Expressão Gênica , Glioblastoma/genética , Histonas/genética , Histonas/metabolismo , Mutação , Telomerase/genética , Telomerase/metabolismo , Telômero/metabolismo , Encurtamento do Telômero , Fatores de Transcrição/metabolismo , Regiões Promotoras Genéticas
3.
Anal Chem ; 96(25): 10200-10209, 2024 06 25.
Artigo em Inglês | MEDLINE | ID: mdl-38867357

RESUMO

Rapid tissue differentiation at the molecular level is a prerequisite for precise surgical resection, which is of special value for the treatment of malignant tumors, such as glioblastoma (GBM). Herein, a SERS-active microneedle is prepared by modifying glutathione (GSH)-responsive molecules, 5,5'-dithiobis(2-nitrobenzoic acid) (DTNB), on the surface of Au@Ag substrates for the distinction of different GBM tissues. Since the Raman signals on the surface of the DTNB@Au@Ag microneedle can be collected by both portable and benchtop Raman spectrometers, the distribution of GSH in different tissues at centimeter scale can be displayed through Raman spectroscopy and Raman imaging, and the entire analysis process can be accomplished within 12 min. Accordingly, in vivo brain tissues of orthotopic GBM xenograft mice and ex vivo tissues of GBM patients are accurately differentiated with the microneedle, and the results are well consistent with tissue staining and postoperative pathological reports. In addition, the outline of tumor, peritumoral, and normal tissues can be indicated by the DTNB@Au@Ag microneedle for at least 56 days. Considering that the tumor tissues are quickly discriminated at the molecular level without the restriction of depth, the DTNB@Au@Ag microneedle is promising to be a powerful intraoperative diagnostic tool for surgery navigation.


Assuntos
Neoplasias Encefálicas , Glioblastoma , Glutationa , Ouro , Análise Espectral Raman , Glioblastoma/patologia , Glioblastoma/metabolismo , Glioblastoma/diagnóstico por imagem , Animais , Humanos , Glutationa/análise , Glutationa/metabolismo , Ouro/química , Camundongos , Neoplasias Encefálicas/patologia , Neoplasias Encefálicas/metabolismo , Neoplasias Encefálicas/diagnóstico por imagem , Agulhas , Prata/química , Camundongos Nus , Ácido Ditionitrobenzoico/química , Linhagem Celular Tumoral , Nanopartículas Metálicas/química
4.
Mol Cancer ; 19(1): 28, 2020 02 10.
Artigo em Inglês | MEDLINE | ID: mdl-32039732

RESUMO

BACKGROUND: Accumulating evidence shows that long noncoding RNAs (lncRNAs) are important regulator molecules involved in diverse biological processes. Acquired drug resistance is a major challenge in the clinical treatment of glioblastoma (GBM), and lncRNAs have been shown to play a role in chemotherapy resistance. However, the underlying mechanisms by which lncRNA mediates TMZ resistance in GBM remain poorly characterized. METHODS: Quantitative reverse transcription PCR (qRT-PCR) and fluorescence in situ hybridization assays were used to detect small nucleolar RNA host gene 12 (SNHG12) levels in TMZ-sensitive and TMZ-resistant GBM cells and tissues. The effects of SNHG12 on TMZ resistance were investigated through in vitro assays (western blots, colony formation assays, flow cytometry assays, and TUNEL assays). The mechanism mediating the high expression of SNHG12 in TMZ-resistant cells and its relationships with miR-129-5p, mitogen-activated protein kinase 1 (MAPK1), and E2F transcription factor 7 (E2F7) were determined by bioinformatic analysis, bisulfite amplicon sequencing, methylation-specific PCR, dual luciferase reporter assays, chromatin immunoprecipitation assays, RNA immunoprecipitation assays, immunofluorescence, qRT-PCR, and western blot. For in vivo experiments, an intracranial xenograft tumor mouse model was used to investigate SNHG12 function. RESULTS: SNHG12 was upregulated in TMZ-resistant cells and tissues. Overexpression of SNHG12 led to the development of acquired TMZ resistance, while knockdown of SNHG12 restored TMZ sensitivity. An abnormally low level of DNA methylation was detected within the promoter region of SNHG12, and loss of DNA methylation made this region more accessible to the Sp1 transcription factor (SP1); this indicated that methylation and SP1 work together to regulate SNHG12 expression. In the cytoplasm, SNHG12 served as a sponge for miR-129-5p, leading to upregulation of MAPK1 and E2F7 and endowing the GBM cells with TMZ resistance. Disinhibition of MAPK1 regulated TMZ-induced cell apoptosis and the G1/S cell cycle transition by activating the MAPK/ERK pathway, while E2F7 dysregulation was primarily associated with G1/S cell cycle transition. Clinically, SNHG12 overexpression was associated with poor survival of GBM patients undergoing TMZ treatment. CONCLUSION: Our results suggest that SNHG12 could serve as a promising therapeutic target to surmount TMZ resistance, thereby improving the clinical efficacy of TMZ chemotherapy.


Assuntos
Metilação de DNA , Resistencia a Medicamentos Antineoplásicos , Fator de Transcrição E2F7/metabolismo , Glioblastoma/patologia , MicroRNAs/genética , Proteína Quinase 1 Ativada por Mitógeno/metabolismo , RNA Longo não Codificante/genética , Temozolomida/farmacologia , Animais , Antineoplásicos Alquilantes/farmacologia , Apoptose , Biomarcadores Tumorais/genética , Biomarcadores Tumorais/metabolismo , Proliferação de Células , Fator de Transcrição E2F7/genética , Feminino , Regulação Neoplásica da Expressão Gênica , Glioblastoma/tratamento farmacológico , Glioblastoma/metabolismo , Humanos , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Proteína Quinase 1 Ativada por Mitógeno/genética , Células Tumorais Cultivadas , Ensaios Antitumorais Modelo de Xenoenxerto
5.
Cancer Cell Int ; 20: 69, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32158359

RESUMO

BACKGROUND: Glucose metabolic reprogramming is a significant hallmark of malignant tumors including GBM. Previous studies suggest that microRNAs play key roles in modulating this process in GBM cells. miR-181b acts as a tumor suppressor miRNA in influencing glioma tumorigenesis. Our previous results showed that miR-181b was down-regulated in glioma cells and tissues. METHODS: The extracellular acidification rate (ECAR), colony formation assay and levels of Glut1 and PKM2 were measured to assess the glucose metabolic and proliferation changes in GBM cells overexpressing miR-181b. Immunoblotting and luciferase reporter assay were performed to confirm the expression and role of SP1 as a direct target of miR-181b. ChIP assay was used to figure out the transcriptional regulation of SP1 on Glut1 and PKM2. In vivo study was examined for the role of miR-181b in GBM cells. RESULTS: MiR-181b overexpression significantly reduced the glucose metabolic and colony formation ability of GBM cells. And, SP1 was confirmed as a direct target of miR-181b while upregulation of SP1 could reverse the influence of overexpression of miR-181b. Furthermore, Glut1 and PKM2 could be regulated by SP1. Finally, miR-181b could inhibit the tumor growth in vivo. CONCLUSIONS: Our article demonstrated the inhibitory effect of miR-181b on glucose metabolism and proliferation in GBM by suppressing SP1 expression.

6.
J Craniofac Surg ; 30(2): e131-e135, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-30550449

RESUMO

OBJECTIVE: At present, large cranial function area arteriovenous malformation (fAVM) is mainly treated by craniotomy with a high risk of operation and high disability. In recent years, with the continuous improvement of the neural intervention technology, fractionated transcatheter arterial embolization (fTAE) may be used to treat the fAVM instead of surgical treatment. However, its effectiveness for treating fAVM has never been explored. The authors hypothesized that fTAE can be effective in the treatment of fTAE. METHODS: A retrospective study was conducted in 229 cases of large fAVM in multicenter hospitals. Among them, 103 cases were performed fTAE and the other 126 cases were carried on minimally invasive craniotomy (MIC). Clinically relevant symptomatic improvement and complications were compared between 2 groups. RESULTS: Complete resection rate of arteriovenous malformation in MIC group (100%) was significantly higher than that of complete embolization rate in fTAE group (77.7%) (P = 0.000). However, the incidence of postoperative complications includes decreased limb muscle strength (P = 0.001), sensory loss of extremities (P = 0.003), visual field defect (P = 0.025) that were lower in fTAE group and remission rate of headache (P = 0.012) but not epilepsy (P = 0.952) was higher in fTAE group compared with that in MIC group. After 1 year follow-up, the rebleeding rate in the fTAE treatment group was 4.85%, higher than that in MIC group (0%) (P = 0.000). CONCLUSIONS: Fractionated transcatheter arterial embolization therapy is beneficial for reducing the postoperative complications and preoperative symptoms of fAVM, but not for recurrence rate.


Assuntos
Malformações Arteriovenosas/complicações , Malformações Arteriovenosas/terapia , Hemorragia Cerebral/etiologia , Embolização Terapêutica/métodos , Procedimentos Cirúrgicos Vasculares/métodos , Adulto , Malformações Arteriovenosas/cirurgia , Encéfalo/irrigação sanguínea , Craniotomia/efeitos adversos , Embolização Terapêutica/efeitos adversos , Epilepsia/etiologia , Extremidades , Feminino , Cefaleia/etiologia , Humanos , Masculino , Pessoa de Meia-Idade , Procedimentos Cirúrgicos Minimamente Invasivos/efeitos adversos , Procedimentos Cirúrgicos Minimamente Invasivos/métodos , Debilidade Muscular/etiologia , Complicações Pós-Operatórias/etiologia , Recidiva , Estudos Retrospectivos , Distúrbios Somatossensoriais/etiologia , Resultado do Tratamento , Procedimentos Cirúrgicos Vasculares/efeitos adversos , Transtornos da Visão/etiologia
7.
J Recept Signal Transduct Res ; 37(4): 335-340, 2017 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-25424605

RESUMO

IL-27 plays an important role in anti-cancer activity. The -964A/G polymorphism in IL-27 gene has been implicated in susceptibility to cancer, but the results were conflicting. The aim of this study was to assess the association between this polymorphism and cancer risk. Pubmed and Wanfang database were searched for all publications concerning IL-27 -964A/G polymorphism and cancer risk. Odds ratio (OR) and 95% confidence interval (CI) were used to assess the strength of association. Statistical analysis was performed using Stata 11.0 software. A total of eight case-control studies including 2044 cancer cases and 2197 controls were identified. Overall, significant association between IL-27 -964A/G polymorphism and cancer risk was observed (GG versus AA: OR = 1.26, 95% CI = 1.03-1.52; GG versus AG + AA: OR = 1.20, 95% CI = 1.00-1.44). In subgroup analysis based on cancer type, significant association was found in colorectal cancer (GG versus AA: OR = 1.55, 95% CI = 1.07-2.27; AG versus AA: OR = 1.31, 95% CI = 1.02-1.67). The current meta-analysis suggests that IL-27 -964A/G polymorphism might enhance cancer risk. However, large-scale and well-designed studies are still needed to confirm the result of our meta-analysis. The association of IL-27 polymorphism with colorectal cancer may provide insight for future therapies.


Assuntos
Estudos de Associação Genética , Predisposição Genética para Doença , Interleucinas/genética , Neoplasias/genética , China , Feminino , Humanos , Masculino , Neoplasias/patologia , Polimorfismo de Nucleotídeo Único/genética , Fatores de Risco
8.
J Neurooncol ; 133(3): 477-485, 2017 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-28501897

RESUMO

Temozolomide (TMZ) is a promising chemotherapeutic agent to treat Glioblastoma multiforme (GBM). However, resistance to TMZ develops quickly with a high frequency. The mechanisms underlying GBM cells' resistance to TMZ are not fully understood. MicroRNAs (miRNAs) are small, non-coding RNA molecules that regulate protein expression by cleaving or repressing the translation of target mRNAs. Recently, miRNAs have been discovered to play important roles in drug resistance. A previous study showed that miR-181b in involved in glioma tumorigenesis. Thus, it would be valuable to explore the functions and mechanisms of miR-181b in regulating GMB cells' sensitivity to TMZ. In this study, quantitative real-time reverse transcription PCR (qRT-PCR) data indicated that miR-181b was significantly downregulated in recurrent GBM tissues compared with initial GBM tissues. We also found that miR-181b overexpression increased the chemo-sensitivity of GBM cells to TMZ and potentiated TMZ-induced apoptosis in vitro and in vivo. Moreover, we demonstrated that the epidermal growth factor receptor (EGFR) was a direct target of miR-181b: restoration of EGFR rescued the inhibitory effects of miR-181b and TMZ treatment. Taken together, our data support strongly an important role for miR-181b in conferring TMZ resistance by targeting EGFR expression.


Assuntos
Antineoplásicos Alquilantes/farmacologia , Neoplasias Encefálicas/tratamento farmacológico , Dacarbazina/análogos & derivados , Receptores ErbB/metabolismo , Glioblastoma/tratamento farmacológico , MicroRNAs/metabolismo , Animais , Apoptose/efeitos dos fármacos , Apoptose/fisiologia , Encéfalo/efeitos dos fármacos , Encéfalo/metabolismo , Neoplasias Encefálicas/metabolismo , Linhagem Celular Tumoral , Dacarbazina/farmacologia , Resistencia a Medicamentos Antineoplásicos/fisiologia , Receptores ErbB/antagonistas & inibidores , Glioblastoma/metabolismo , Humanos , Masculino , Camundongos Endogâmicos BALB C , Camundongos Nus , Recidiva Local de Neoplasia , Transplante de Neoplasias , Distribuição Aleatória , Temozolomida
9.
RNA ; 19(4): 552-60, 2013 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-23431408

RESUMO

MicroRNAs (miRNAs) are single-stranded, 18- to 23-nt RNA molecules that function as regulators of gene expression. Previous studies have shown that microRNAs play important roles in human cancers, including gliomas. Here, we found that expression levels of miR-181b were decreased in gliomas, and we identified IGF-1R as a novel direct target of miR-181b. MiR-181b overexpression inhibited cell proliferation, migration, invasion, and tumorigenesis by targeting IGF-1R and its downstream signaling pathways, PI3K/AKT and MAPK/ERK1/2. Overexpression of IGF-1R rescued the inhibitory effects of miR-181b. In clinical specimens, IGF-1R was overexpressed, and its protein levels were inversely correlated with miR-181b expression. Taken together, our results indicate that miR-181b functions in gliomas to suppress growth by targeting the IGF-1R oncogene and that miR-181b may serve as a novel therapeutic target for gliomas.


Assuntos
Proliferação de Células , Transformação Celular Neoplásica , Glioma/metabolismo , Glioma/patologia , MicroRNAs/metabolismo , Receptor IGF Tipo 1/metabolismo , Inibidores da Angiogênese/metabolismo , Animais , Movimento Celular , Genes Supressores de Tumor , Humanos , Masculino , Camundongos , Camundongos Nus , MicroRNAs/genética , Transdução de Sinais
10.
Tumour Biol ; 36(5): 3823-9, 2015 May.
Artigo em Inglês | MEDLINE | ID: mdl-25566967

RESUMO

Fenofibrate, a fibric acid derivative, is known to possess lipid-lowering effects. Although fenofibrate-induced peroxisome proliferator-activated receptor alpha (PPARα) transcriptional activity has been reported to exhibit anticancer effects, the underlying mechanisms are poorly understood. In this study, we investigated the mechanisms behind the antiproliferative effects of fenofibrate in U87MG cells (human glioma cell line) using the WST-8 Cell Proliferation Assay Kit. Furthermore, we examined genome-wide gene expression profiles and molecular networks using the DAVID online software. Fenofibrate reduced the expression of 405 genes and increased the expression of 2280 genes. DAVID analysis suggested that fenofibrate significantly affected cell cycle progression and pathways involved in cancer, including the mTOR signaling pathway and insulin signaling pathway. Results of flow cytometry analysis indicated that fenofibrate induced cell cycle G0/G1 arrest in U87MG cells. Furthermore, we identified the FoxO1-p27(kip) signaling axis to be involved in fenofibrate-induced cell cycle arrest. Our findings suggest that in addition to its known lipid-lowering effects, fenofibrate may be used as an antitumor agent in glioma therapy.


Assuntos
Inibidor de Quinase Dependente de Ciclina p27/biossíntese , Fenofibrato/administração & dosagem , Fatores de Transcrição Forkhead/biossíntese , Glioblastoma/tratamento farmacológico , PPAR alfa/biossíntese , Pontos de Checagem do Ciclo Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Proteína Forkhead Box O1 , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Genoma Humano , Glioblastoma/genética , Glioblastoma/patologia , Humanos , Proteínas de Neoplasias/biossíntese , Transdução de Sinais/efeitos dos fármacos , Serina-Treonina Quinases TOR/biossíntese
11.
Huan Jing Ke Xue ; 45(1): 543-554, 2024 Jan 08.
Artigo em Chinês | MEDLINE | ID: mdl-38216503

RESUMO

This study aimed to clarify the effect of long-term continuous cropping of pepper on soil fungal community structure, reveal the mechanism of continuous cropping obstacles, and provide a theoretical basis for the ecological safety and sustainable development of pepper industry. We took the pepper continuous cropping soil in the vegetable greenhouse planting base of Tongren City as the research object. The diversity and community structure of fungi in farmland soil were analyzed using Illumina MiSeq high-throughput sequencing, the responses of soil physio-chemical properties and fungal community characteristics to long-term continuous pepper cropping were discussed, and the relationships between the characteristics of fungal community structure and environmental factors were determined using CCA and correlation network analysis. The results showed that with the extension of pepper continuous cropping years, the soil pH value and organic matter (OM) content decreased, total phosphorus (TP) and available phosphorus (AP) contents increased, hydrolyzed nitrogen (AN) and available potassium (AK) contents decreased first and then increased, and total nitrogen (TN) and total potassium (TK) contents did not change significantly. Long-term continuous cropping decreased the Chao1 index and observed species index and decreased the Shannon index and Simpson index. The change in continuous cropping years had a significant effect on the relative abundance of soil fungal dominant flora. At the phylum level, the relative abundance of Mortierellomycota decreased with the extension of pepper continuous cropping years, the relative abundance of Ascomycota increased first and then decreased, and the relative abundance of Basidiomycota decreased first and then increased. At the genus level, with the increasing of pepper continuous cropping years, the relative abundance of Fusarium increased, and the relative abundance of Mortierella and Penicillium decreased. In addition, long-term continuous cropping simplified the soil fungal symbiosis network. CCA analysis indicated that pH, OM, TN, AN, AP, and AK were the driving factors of soil fungal community structure, and correlation network analysis showed that pH, OM, TN, TP, TK, AN, AP, and AK were the driving factors of soil fungal community structure, including Fusarium, Lophotrichus, Penicillium, Mortierella, Botryotrichum, Staphylotrichum, Plectosphaerella, and Acremonium. In conclusion, continuous cropping changed the soil physical and chemical properties, affected the diversity and community structure of the soil fungal community, changed the interaction between microorganisms, and destroyed the microecological balance of the soil, which might explain obstacles associated with continuous cropped pepper.


Assuntos
Fusarium , Micobioma , Penicillium , Solo/química , Microbiologia do Solo , Produtos Agrícolas , Nitrogênio , Fósforo , Potássio
12.
Neuro Oncol ; 2024 May 09.
Artigo em Inglês | MEDLINE | ID: mdl-38721826

RESUMO

BACKGROUND: The high fatality rate of glioblastoma (GBM) is attributed to glioblastoma stem cells (GSCs), which exhibit heterogeneity and therapeutic resistance. Metabolic plasticity of mitochondria is the hallmark of GSCs. Targeting mitochondrial biogenesis of GSCs is crucial for improving clinical prognosis in GBM patients. METHODS: SMYD2-induced PGC1α methylation and followed nuclear export is confirmed by co-immunoprecipitation, cellular fractionation, and immunofluorescence. The effects of SMYD2/PGC1α/CRM1 axis on GSCs mitochondrial biogenesis is validated by OCR, ECAR and intracranial glioma model. RESULTS: PGC1α methylation causes disabled mitochondrial function to maintain the stemness, thereby enhancing radio-resistance of GSCs. SMYD2 drives PGC1α K224 methylation (K224me), which is essential for promoting the stem-like characteristics of GSCs. PGC1α K224me is preferred binding with CRM1, accelerating PGC1α nuclear export and subsequent dysfunction. Targeting PGC1α methylation exhibits significant radiotherapeutic efficacy and prolongs patient survival. CONCLUSIONS: These findings unveil a novel regulatory pathway involving mitochondria that governs stemness in GSCs, thereby emphasizing promising therapeutic strategies targeting PGC1α and mitochondria for the treatment of GBM.

13.
Sci Transl Med ; 16(739): eadg5553, 2024 Mar 20.
Artigo em Inglês | MEDLINE | ID: mdl-38507470

RESUMO

Glioblastoma, the most lethal primary brain tumor, harbors glioma stem cells (GSCs) that not only initiate and maintain malignant phenotypes but also enhance therapeutic resistance. Although frequently mutated in glioblastomas, the function and regulation of PTEN in PTEN-intact GSCs are unknown. Here, we found that PTEN directly interacted with MMS19 and competitively disrupted MMS19-based cytosolic iron-sulfur (Fe-S) cluster assembly (CIA) machinery in differentiated glioma cells. PTEN was specifically succinated at cysteine (C) 211 in GSCs compared with matched differentiated glioma cells. Isotope tracing coupled with mass spectrometry analysis confirmed that fumarate, generated by adenylosuccinate lyase (ADSL) in the de novo purine synthesis pathway that is highly activated in GSCs, promoted PTEN C211 succination. This modification abrogated the interaction between PTEN and MMS19, reactivating the CIA machinery pathway in GSCs. Functionally, inhibiting PTEN C211 succination by reexpressing a PTEN C211S mutant, depleting ADSL by shRNAs, or consuming fumarate by the US Food and Drug Administration-approved prescription drug N-acetylcysteine (NAC) impaired GSC maintenance. Reexpressing PTEN C211S or treating with NAC sensitized GSC-derived brain tumors to temozolomide and irradiation, the standard-of-care treatments for patients with glioblastoma, by slowing CIA machinery-mediated DNA damage repair. These findings reveal an immediately practicable strategy to target GSCs to treat glioblastoma by combination therapy with repurposed NAC.


Assuntos
Neoplasias Encefálicas , Glioblastoma , Glioma , Humanos , Glioblastoma/tratamento farmacológico , Ferro/metabolismo , Glioma/tratamento farmacológico , Neoplasias Encefálicas/tratamento farmacológico , Células-Tronco Neoplásicas/patologia , Enxofre/metabolismo , Enxofre/uso terapêutico , Fumaratos , Linhagem Celular Tumoral , PTEN Fosfo-Hidrolase/metabolismo
14.
Cell Death Differ ; 31(6): 738-752, 2024 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-38594444

RESUMO

Glioblastoma (GBM) is the most aggressive malignant primary brain tumor characterized by a highly heterogeneous and immunosuppressive tumor microenvironment (TME). The symbiotic interactions between glioblastoma stem cells (GSCs) and tumor-associated macrophages (TAM) in the TME are critical for tumor progression. Here, we identified that IFI35, a transcriptional regulatory factor, plays both cell-intrinsic and cell-extrinsic roles in maintaining GSCs and the immunosuppressive TME. IFI35 induced non-canonical NF-kB signaling through proteasomal processing of p105 to the DNA-binding transcription factor p50, which heterodimerizes with RELB (RELB/p50), and activated cell chemotaxis in a cell-autonomous manner. Further, IFI35 induced recruitment and maintenance of M2-like TAMs in TME in a paracrine manner. Targeting IFI35 effectively suppressed in vivo tumor growth and prolonged survival of orthotopic xenograft-bearing mice. Collectively, these findings reveal the tumor-promoting functions of IFI35 and suggest that targeting IFI35 or its downstream effectors may provide effective approaches to improve GBM treatment.


Assuntos
Glioblastoma , NF-kappa B , Células-Tronco Neoplásicas , Transdução de Sinais , Macrófagos Associados a Tumor , Glioblastoma/metabolismo , Glioblastoma/patologia , Glioblastoma/genética , Humanos , Animais , Camundongos , Células-Tronco Neoplásicas/metabolismo , Células-Tronco Neoplásicas/patologia , Macrófagos Associados a Tumor/metabolismo , Macrófagos Associados a Tumor/patologia , NF-kappa B/metabolismo , Neoplasias Encefálicas/patologia , Neoplasias Encefálicas/metabolismo , Neoplasias Encefálicas/genética , Linhagem Celular Tumoral , Microambiente Tumoral
15.
Front Oncol ; 13: 1222581, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37564933

RESUMO

Objectives: Is intradural fat graft packing indispensable in preventing postoperative cerebrospinal fluid leakage in endoscopic endonasal pituitary adenoma surgeries? This study aimed to review the methods and outcomes of our graded sellar floor reconstruction strategy without fat graft packing in endoscopic endonasal pituitary adenoma surgeries. Methods: From March 2018 to December 2022, 200 patients underwent endoscopic endonasal pituitary adenoma resection by a single author in our institute. We applied different graded skull base reconstruction strategies in different periods. Intradural fat graft packing was used to reconstruct the skull base in the early period, from March 2018 to June 2019, but fat graft was not used in the late period, from January 2020 to December 2022. The effect of these different graded skull base reconstruction strategies and whether intradural fat graft packing is necessary were evaluated by observing the incidence of postoperative cerebrospinal fluid leak. Results: In the early period, fat graft was used to reconstruct skull base when the intraoperative cerebrospinal fluid (CSF) leakage existed. There were two patients who suffered from postoperative cerebrospinal fluid leak in this group. In the late period, fat graft was not used to reconstruct the skull base, and no patient suffered from postoperative cerebrospinal fluid leakage in this group. Conclusions: Intradural fat graft packing is unnecessary in the endoscopic endonasal pituitary adenoma resection. The outcome of our graded sellar floor reconstruction strategy is satisfactory.

16.
J Mol Neurosci ; 73(4-5): 259-268, 2023 May.
Artigo em Inglês | MEDLINE | ID: mdl-37014544

RESUMO

Temozolomide (TMZ)-based chemotherapy plays a central part in glioma treatment. However, prominent resistance to TMZ is a major change by now. In this study, expression and prognosis of SRSF4 were analyzed using multiple public datasets. Therapeutic efficacy against TMZ resistance was determined by assessing colony formation, flow cytometry, and western blot assays. Bio-informational analysis, immunofluorescence (IF), and western blot assays were performed to evaluate double strand break repair. An orthotopic xenograft model was used to exam the functional role of SRSF4. Here, we found that SRSF4 expression was associated with histological grade, IDH1 status, 1p/19q codeletion, molecular subtype, tumor recurrence, and poor prognosis. SRSF4 promotes TMZ resistance through positively regulating MDC1, thereby accelerating double strand break repair. Targeting SRSF4 could significantly improve chemosensitivity. Taken together, our collective findings highlight an important role of SRSF4 in the regulation of TMZ resistance by modulation of double strand break repair.


Assuntos
Neoplasias Encefálicas , Glioma , Humanos , Temozolomida/farmacologia , Resistencia a Medicamentos Antineoplásicos , Recidiva Local de Neoplasia/tratamento farmacológico , Glioma/tratamento farmacológico , Glioma/genética , Glioma/metabolismo , Reparo do DNA , Linhagem Celular Tumoral , Neoplasias Encefálicas/tratamento farmacológico , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/metabolismo , Antineoplásicos Alquilantes/farmacologia , Antineoplásicos Alquilantes/uso terapêutico , Fatores de Processamento de Serina-Arginina/genética , Fatores de Processamento de Serina-Arginina/metabolismo
17.
J Biomed Res ; 37(5): 326-339, 2023 Sep 28.
Artigo em Inglês | MEDLINE | ID: mdl-37750323

RESUMO

Ionizing radiation is a popular and effective treatment option for glioblastoma (GBM). However, resistance to radiation therapy inevitably occurs during treatment. It is urgent to investigate the mechanisms of radioresistance in GBM and to find ways to improve radiosensitivity. Here, we found that heat shock protein 90 beta family member 1 (HSP90B1) was significantly upregulated in radioresistant GBM cell lines. More importantly, HSP90B1 promoted the localization of glucose transporter type 1, a key rate-limiting factor of glycolysis, on the plasma membrane, which in turn enhanced glycolytic activity and subsequently tumor growth and radioresistance of GBM cells. These findings imply that targeting HSP90B1 may effectively improve the efficacy of radiotherapy for GBM patients, a potential new approach to the treatment of glioblastoma.

18.
Cancer Lett ; 573: 216380, 2023 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-37660885

RESUMO

Preoperative MRI is an essential diagnostic and therapeutic reference for gliomas. This study aims to evaluate the prognostic aspect of a radiomics biomarker for glioma and further investigate its relationship with tumor microenvironment and macrophage infiltration. We covered preoperative MRI of 664 glioma patients from three independent datasets: Jiangsu Province Hospital (JSPH, n = 338), The Cancer Genome Atlas dataset (TCGA, n = 252), and Repository of Molecular Brain Neoplasia Data (REMBRANDT, n = 74). Incorporating a multistep post-processing workflow, 20 radiomics features (Rads) were selected and a radiomics survival biomarker (RadSurv) was developed, proving highly efficient in risk stratification of gliomas (cut-off = 1.06), as well as lower-grade gliomas (cut-off = 0.64) and glioblastomas (cut-off = 1.80) through three fixed cut-off values. Through immune infiltration analysis, we found a positive correlation between RadSurv and macrophage infiltration (RMΦ = 0.297, p < 0.001; RM2Φ = 0.241, p < 0.001), further confirmed by immunohistochemical-staining (glioblastomas, n = 32) and single-cell sequencing (multifocal glioblastomas, n = 2). In conclusion, RadSurv acts as a strong prognostic biomarker for gliomas, exhibiting a non-negligible positive correlation with macrophage infiltration, especially with M2 macrophage, which strongly suggests the promise of radiomics-based models as a preoperative alternative to conventional genomics for predicting tumor macrophage infiltration and provides clinical guidance for immunotherapy.


Assuntos
Neoplasias Encefálicas , Glioblastoma , Glioma , Humanos , Glioblastoma/diagnóstico por imagem , Glioblastoma/genética , Glioma/diagnóstico por imagem , Glioma/genética , Glioma/terapia , Neoplasias Encefálicas/diagnóstico por imagem , Neoplasias Encefálicas/genética , Genômica , Macrófagos , Microambiente Tumoral
19.
Cell Death Dis ; 14(7): 417, 2023 07 12.
Artigo em Inglês | MEDLINE | ID: mdl-37438359

RESUMO

Long noncoding RNAs (lncRNAs) are involved in glioma initiation and progression. Glioma stem cells (GSCs) are essential for tumor initiation, maintenance, and therapeutic resistance. However, the biological functions and underlying mechanisms of lncRNAs in GSCs remain poorly understood. Here, we identified that LINC00839 was overexpressed in GSCs. A high level of LINC00839 was associated with GBM progression and radiation resistance. METTL3-mediated m6A modification on LINC00839 enhanced its expression in a YTHDF2-dependent manner. Mechanistically, LINC00839 functioned as a scaffold promoting c-Src-mediated phosphorylation of ß-catenin, thereby inducing Wnt/ß-catenin activation. Combinational use of celecoxib, an inhibitor of Wnt/ß-catenin signaling, greatly sensitized GSCs to radiation. Taken together, our results showed that LINC00839, modified by METTL3-mediated m6A, exerts tumor progression and radiation resistance by activating Wnt/ß-catenin signaling.


Assuntos
Glioma , RNA Longo não Codificante , Via de Sinalização Wnt , Humanos , beta Catenina/genética , Transformação Celular Neoplásica , Glioma/genética , Glioma/radioterapia , Metiltransferases/genética , Células-Tronco Neoplásicas , RNA Longo não Codificante/genética
20.
Brain Behav ; 13(5): e2969, 2023 05.
Artigo em Inglês | MEDLINE | ID: mdl-36978245

RESUMO

OBJECTIVE: The structural alteration that occurs within the salience network (SN) in patients with insular glioma is unclear. Therefore, we aimed to investigate the changes in the topological network and brain structure alterations within the SN in patients with insular glioma. METHODS: We enrolled 46 patients with left insular glioma, 39 patients with right insular glioma, and 21 demographically matched healthy controls (HCs). We compared the topological network, gray matter (GM) volume, and fractional anisotropy (FA) between HCs and patients after controlling for the effects of age and gender. RESULTS: Patients with insular glioma showed topological network decline mainly in the insula, basal ganglia region, and anterior cingulate cortex (ACC). Compared with HCs, patients primarily showed GM volume increased in the ACC, inferior temporal gyrus (ITG), superior temporal gyrus (STG), temporal pole: middle temporal gyrus (TPOmid), insula, middle temporal gyrus (MTG), middle frontal gyrus, and superior occipital gyrus (SOG), but decreased in TPOmid, ITG, temporal pole: superior temporal gyrus, and SOG. FA declined mainly in the STG, MTG, ACC, superior frontal gyrus, and SOG, and also showed an increased cluster in SOG. CONCLUSIONS: FA represents the integrity of the white matter. In patients with insular glioma, decreased FA may lead to the destruction of the topological network within the SN, which in turn may lead to the decrease of network efficiency and brain function, and the increase of GM volume may compensate for these changes. Overall, this pattern of structural changes provides new insight into the compensation model of insular glioma.


Assuntos
Imageamento por Ressonância Magnética , Substância Branca , Humanos , Encéfalo , Substância Cinzenta/diagnóstico por imagem , Mapeamento Encefálico , Substância Branca/diagnóstico por imagem
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa