Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 920
Filtrar
1.
Cells ; 14(3)2025 Jan 24.
Artigo em Inglês | MEDLINE | ID: mdl-39936970

RESUMO

Glioblastoma (GB) is a highly malignant brain tumor with a poor prognosis, with a median survival of only 14.6 months despite aggressive treatments. Resistance to chemotherapy, particularly temozolomide (TMZ), is a significant challenge. The DNA repair enzyme MGMT and glioblastoma stem cells (GSCs) often mediate this resistance. Recent studies highlight the therapeutic potential of natural compounds, particularly delphinidins, found in deep purple berries. Delphinidins are known for their ability to inhibit NF-κB signaling, a critical pathway for GB progression, chemoresistance, and MGMT expression. Our research demonstrates that glycosylated delphinidins have potential adjuvant use in the treatment of GB, offering a promising natural strategy to combat TMZ resistance. Specifically, we observed that delphinidin 3,5 di-glucoside has potent anticancer effects when used alone. Meanwhile, delphinidin 3 glucoside acted in synergy with temozolomide to decrease cell viability, highlighting its potential as an adjuvant. It also exerted a faster and more sustained inhibition of NF-κB, highlighting its potential for long-lasting therapeutic effects. These findings open new avenues for targeted therapies against glioblastoma, particularly to overcome treatment resistance.


Assuntos
Antocianinas , Resistencia a Medicamentos Antineoplásicos , Glioblastoma , NF-kappa B , Transdução de Sinais , Temozolomida , Glioblastoma/tratamento farmacológico , Glioblastoma/metabolismo , Glioblastoma/patologia , Temozolomida/farmacologia , Temozolomida/uso terapêutico , Humanos , NF-kappa B/metabolismo , Resistencia a Medicamentos Antineoplásicos/efeitos dos fármacos , Transdução de Sinais/efeitos dos fármacos , Linhagem Celular Tumoral , Antocianinas/farmacologia , Antocianinas/uso terapêutico , Metilases de Modificação do DNA/metabolismo , Metilases de Modificação do DNA/genética , Glicosilação/efeitos dos fármacos , Enzimas Reparadoras do DNA/metabolismo , Enzimas Reparadoras do DNA/genética , Neoplasias Encefálicas/tratamento farmacológico , Neoplasias Encefálicas/metabolismo , Neoplasias Encefálicas/patologia , Neoplasias Encefálicas/genética , Proteínas Supressoras de Tumor/metabolismo , Proteínas Supressoras de Tumor/genética , Sobrevivência Celular/efeitos dos fármacos
2.
Int J Mol Sci ; 26(3)2025 Jan 21.
Artigo em Inglês | MEDLINE | ID: mdl-39940647

RESUMO

Glioblastoma is the most common and highly malignant brain tumor in adults. New targeted therapeutic approaches are imperative. EGFRvIII has appealing therapeutic targets using monoclonal antibodies. Thus, endeavors toward developing new mAbs therapies for GBM capable of targeting the tumor EGFRvIII biomarker must prevail to improve the patient's prognosis. Here, we isolated and characterized an anti-EGFRvIII vNAR from a non-immune freshwater stingray mixed library, termed vNAR R426. The vNAR R426 and pEGFRvIII interaction was demonstrated by molecular docking and molecular dynamics, and the recognition of EGFRvIII in vitro was further confirmed by cell immunofluorescence staining. Moreover, the vNAR R426 was shown to be an effective cisplatin drug carrier in the U87-MG glioma cell line. The cisplatin-coupled vNAR demonstrated highly significant differences when compared to free CDDP at 72 h. Notably, the cisplatin-vNAR carrier achieved better efficacy in the U87-MG cell line. Thus, we described the vNAR R426 internalization by receptor-mediated endocytosis and the subsequent COPI-mediated nuclear translocation of EGFRvIII and highlighted the importance of this shuttle mechanism to enhance the targeted delivery of cisplatin within the glioma cell's nucleus and improved cytotoxic effect. In conclusion, vNAR R426 could be a potential therapeutic carrier for EGFRvIII-targeted glioblastoma and cancer therapies.


Assuntos
Cisplatino , Portadores de Fármacos , Receptores ErbB , Glioblastoma , Receptores ErbB/metabolismo , Humanos , Glioblastoma/metabolismo , Glioblastoma/patologia , Glioblastoma/tratamento farmacológico , Animais , Linhagem Celular Tumoral , Cisplatino/farmacologia , Portadores de Fármacos/química , Neoplasias Encefálicas/tratamento farmacológico , Neoplasias Encefálicas/patologia , Neoplasias Encefálicas/metabolismo , Neoplasias Encefálicas/imunologia , Antineoplásicos/farmacologia , Antineoplásicos/química , Simulação de Acoplamento Molecular
3.
Childs Nerv Syst ; 41(1): 91, 2025 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-39812802

RESUMO

INTRODUCTION: Glioblastomas (GBM) are aggressive tumors that make up about 7% of central nervous system tumors in children. Spinal GBMs (sGBMs) are extremely rare, accounting for less than 1% of pediatric spinal tumors. sGBMs are difficult to treat due to their infiltrative nature and cause significant morbidity. While there is extensive literature on treatment outcomes for cranial GBMs, there is limited research on pediatric sGBMs. This meta-analysis aims to assess the impact of available treatments on overall survival (OS) and progression-free survival (PFS) in pediatric sGBM patients and to identify prognostic factors. METHODOLOGY: A comprehensive review of pediatric sGBM cases up to June 2024 was conducted using PubMed and Mendeley. Inclusion criteria were case series and case reports of pediatric sGBM, excluding those with metastatic sGBM or aggregated patient data. A total of 2202 articles were identified, with 46 meeting the inclusion criteria. Data on demographics, tumor characteristics, extent of resection, and treatments were collected. Kaplan-Meier and Cox proportional hazards models were used for statistical analysis. RESULTS: The data was collected from 81 patients, 43 females and 38 males, with an average age of 10.7 years. The majority of tumors were found in the cervical region (32%). Subtotal resection (STR) was performed in 53% of cases, and 59% of patients received both chemotherapy (QT) and radiotherapy (RT). The average progression-free survival (PFS) was 10.95 months, with RT significantly improving PFS (15.2 months vs. 2.1 months, p = 0.001). The average OS was 13.4 months, with RT and QT being significant protective factors (p < 0.05). Age over seven years and cervical tumor location were associated with worse OS. CONCLUSION: This study highlights the significance of radiation therapy and chemotherapy in enhancing overall survival and progression-free survival in pediatric patients with spinal cord glioblastoma. Specifically, RT significantly improves PFS, while advanced age and tumor location in the cervical region are associated with worse outcomes. These findings can help shape treatment approaches and ultimately enhance the quality of life for pediatric sGBM patients.


Assuntos
Glioblastoma , Neoplasias da Medula Espinal , Humanos , Glioblastoma/terapia , Neoplasias da Medula Espinal/terapia , Criança , Adolescente , Pré-Escolar , Masculino , Feminino , Terapia Combinada/métodos
4.
Molecules ; 30(1)2025 Jan 03.
Artigo em Inglês | MEDLINE | ID: mdl-39795214

RESUMO

Glioblastomas (GBM) are malignant tumours with poor prognosis. Treatment involves chemotherapy and/or radiotherapy; however, there is currently no standard treatment for recurrence, and prognosis remains unfavourable. Inflammatory mediators and microRNAs (miRNAs) influence the aggressiveness of GBM, being involved in the communication with the cells of the tumour parenchyma, including microglia/macrophages, and maintaining an immunosuppressive microenvironment. Hence, the modulation of miRNAs and inflammatory factors may improve GBM treatments. In this study, we investigated the effects of agathisflavone, a biflavonoid purified from Cenostigma pyramidale (Tul.), on the growth and migration of GBM cells, on the expression of inflammatory cytokines and microRNAs, as well on the response of microglia. Agathisflavone (5-30 µM) induced a dose- and time-dependent reduction in the viability of both human GL-15 and rat C6 cells, as determined by the MTT test, and reduced cell migration, as determined by cell scratch assay. RT-qPCR analysis revealed that agathisflavone (5 µM) down-regulated the expression of miR-125b and miR-155 in the secretome derived from GL-15 cells, which was associated with upregulation of the mRNA expression of IL-6 and arginase-1 immunoregulatory factors. Exposure of human microglia/macrophage to the secretome from GL-15 GMB cells modulated proliferation and morphology, effects that were modulated by agathisflavone treatment. These results demonstrate the effect of flavonoids on the growth of GBM cells, which impacts cells in the microenvironment and can be considered for preclinical studies for adjuvant treatments.


Assuntos
Arginase , Biflavonoides , Sobrevivência Celular , Glioblastoma , Interleucina-6 , MicroRNAs , Microglia , MicroRNAs/genética , Glioblastoma/tratamento farmacológico , Glioblastoma/patologia , Glioblastoma/metabolismo , Glioblastoma/genética , Humanos , Microglia/efeitos dos fármacos , Microglia/metabolismo , Arginase/genética , Arginase/metabolismo , Sobrevivência Celular/efeitos dos fármacos , Ratos , Interleucina-6/metabolismo , Interleucina-6/genética , Biflavonoides/farmacologia , Linhagem Celular Tumoral , Animais , Ativação de Macrófagos/efeitos dos fármacos , Movimento Celular/efeitos dos fármacos , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Macrófagos/efeitos dos fármacos , Macrófagos/metabolismo , Proliferação de Células/efeitos dos fármacos
5.
Cochrane Database Syst Rev ; 1: CD012532, 2025 01 08.
Artigo em Inglês | MEDLINE | ID: mdl-39777725

RESUMO

BACKGROUND: Glioblastoma multiforme (GBM) is the most common and aggressive adult glioma (16-month median survival). Its immunosuppressive microenvironment limits the efficacy of immune checkpoint inhibitors (ICIs). OBJECTIVES: To assess the effects of the ICIs antibodies anti-programmed cell death 1 (anti-PD-1) and anti-programmed cell death ligand 1 (anti-PD-L1) in treating adults with diffuse glioma. SEARCH METHODS: We searched CENTRAL, MEDLINE, Embase, and clinical trials registers on 8 March 2024. SELECTION CRITERIA: We included randomised controlled trials (RCTs) evaluating adults with diffuse glioma treated with anti-PD-1/PD-L1 compared to placebo or other therapies used alone or with other ICIs. Primary outcomes were overall survival (OS), progression-free survival (PFS), and serious adverse events (SAE). Secondary outcomes were overall response rate (ORR), quality of life (QoL), and less serious AEs. DATA COLLECTION AND ANALYSIS: We followed standard Cochrane methods. MAIN RESULTS: We included seven RCTs evaluating anti-PD-1 treatment in recurrent (N = 4) and newly diagnosed (N = 3) grade 4 glioma participants. The analysis encompassed 1953 participants; sample sizes ranged from 35 to 716. Meta-analyses were not possible due to heterogeneity and the small number of studies. Most trials had high risk of bias. Nivolumab versus bevacizumab in people with recurrent GBM (1 trial, 369 participants) Nivolumab probably does not increase OS (hazard ratio (HR) 1.04, 95% confidence interval (CI) 0.83 to 1.30; 1.3% more, 95% CI 6.30 fewer to 7.80 more; 369 participants; moderate-certainty evidence) or PFS (HR 1.97, 95% CI 1.57 to 2.48; 16.40% more, 95% CI 12.40 more to 19.00 more; 369 participants; moderate-certainty evidence). The evidence for SAE is very uncertain (risk ratio (RR) 1.20, 95% CI 0.74 to 1.92; 347 participants). Nivolumab probably does not increase ORR (RR 0.34, 95% CI 0.18 to 0.63; 309 participants; moderate-certainty evidence), but may not increase less serious AEs (RR 1.03, 95% CI 0.96 to 1.10; 347 participants; low-certainty evidence). Nivolumab plus bevacizumab 10 mg/kg versus nivolumab plus bevacizumab 3 mg/kg in people with recurrent GBM (1 trial, 90 participants) Nivolumab plus bevacizumab 10 mg/kg may not increase OS (HR 1.39, 95% CI 0.86 to 2.25; 9.90% more, 95% CI 5.20 fewer to 18.80 more; 90 participants; low-certainty evidence). The evidence for PFS (HR 1.23, 95% CI 0.78 to 1.93; 5.80% more, 95% CI 8.20 fewer to 14.20 more; 90 participants) and SAE (RR 1.19, 95% CI 0.79 to 1.79; 90 participants) is very uncertain. Nivolumab may not increase less serious AEs (RR 1.02, 95% CI 0.96 to 1.09; low-certainty evidence; 90 participants). Pembrolizumab plus bevacizumab versus pembrolizumab in people with recurrent GBM (1 trial, 80 participants) The evidence for OS (HR 1.03, 95% CI 0.65 to 1.63; 0.30% more, 95% CI 7.60 fewer to 2.90 more; 80 participants), PFS (HR 0.97, 95% CI 0.61 to 1.54: 0.40% fewer, 95% CI 9.20 fewer to 2.80 more; 80 participants), SAE (RR 1.32, 95% CI 0.75 to 2.42; 80 participants), and ORR (RR 12.76, 95% CI 0.77 to 210.27; 80 participants) is very uncertain. Pembrolizumab plus bevacizumab may not increase less serious AEs (RR 1.04, 95% CI 0.96 to 1.13; 80 participants; low-certainty evidence). Neoadjuvant (before surgical resection) and adjuvant (after surgical resection) pembrolizumab versus adjuvant-only pembrolizumab in people with recurrent GBM (1 trial, 35 participants) The evidence for OS (HR 0.39, 95% CI 0.17 to 0.92; 25.20% fewer, 95% CI 37.10 fewer to 2.10 fewer; 35 participants), PFS (HR 0.43, 95% CI 0.20 to 0.91; 30.10% fewer, 95% CI 52.20 fewer to 3.60 fewer; 35 participants), and SAE (RR 1.00, 95% CI 0.31 to 3.28; 32 participants) is very uncertain. Nivolumab plus radiotherapy versus temozolomide plus radiotherapy in people with newly diagnosed unmethylated GBM (1 trial, 560 participants) Nivolumab plus radiotherapy probably does not increase OS (HR 1.31, 95% CI 1.09 to 1.58 months; 8.30% more, 95% CI 2.80 more to 12.90 more; 560 participants) and PFS (HR 1.38, 95% CI 1.15 to 1.65 months; 7.50% more, 95% CI 3.60 more to 10.30 more; 560 participants; moderate-certainty evidence). The evidence for SAE is very uncertain (RR 0.87, 95% CI 0.65 to 1.18; 553 participants). It may not increase ORR (RR 1.08, 95% CI 0.43 to 2.69; 560 participants; low-certainty evidence) and probably does not increase less serious AEs (RR 1.00, 95% CI 0.96 to 1.04; 560 participants; moderate-certainty evidence). The evidence for time to deterioration of QoL is very uncertain (HR 0.76, 95% CI 0.59 to 0.99; 560 participants). Nivolumab plus temozolomide plus radiotherapy versus placebo plus temozolomide plus radiotherapy in people with newly diagnosed methylated GBM (1 trial, 716 participants) Nivolumab plus temozolomide plus radiotherapy probably does not increase OS (HR 1.10, 95% CI 0.92 to 1.32; 3.50 more, 95% CI 3.80 fewer to 9.60 more; 716 participants) and PFS (HR 1.10, 95% CI 0.92 to 1.32; 3.00 more, 95% CI 3.50 fewer to 7.90 more; 716 participants), and probably increases SAE (RR 2.91, 95% CI 2.05 to 4.12; 709 participants; moderate-certainty evidence). It does not increase less serious AEs (RR 1.02, 95% CI 1.00 to 1.04; 709 participants; high-certainty evidence). Adjuvant nivolumab plus temozolomide versus temozolomide in older people with GBM (1 trial, 103 participants) Nivolumab plus temozolomide probably does not increase OS (HR 0.85, 95% CI 0.54 to 1.33; 3.10 fewer, 95% CI 15.80 fewer to 3.60 more; 103 participants; moderate-certainty evidence) and PFS (HR 0.77, 95% CI 0.49 to 1.19; 5.40 fewer, 95% CI 19.10 fewer to 2.40 more; 103 participants; moderate-certainty evidence). The evidence for SAE is very uncertain (RR 1.58, 95% CI 0.88 to 2.81; 103 participants). The evidence for QoL is very uncertain (results only reported graphically; 103 participants). AUTHORS' CONCLUSIONS: In recurrent GBM, nivolumab alone probably has no benefit. Anti-PD1 plus bevacizumab may also be ineffective based on low- to very low-certainty evidence. Neoadjuvant plus adjuvant pembrolizumab may improve OS and PFS, but this was based on only one small trial and very low-certainty evidence. In newly diagnosed GBM, nivolumab plus radiotherapy in unmethylated and plus radiotherapy plus temozolomide in methylated GBM probably has no benefit. In older participants, adjuvant nivolumab probably offers no benefit.


Assuntos
Antineoplásicos Imunológicos , Antígeno B7-H1 , Neoplasias Encefálicas , Glioblastoma , Inibidores de Checkpoint Imunológico , Receptor de Morte Celular Programada 1 , Adulto , Humanos , Anticorpos Monoclonais Humanizados/uso terapêutico , Anticorpos Monoclonais Humanizados/efeitos adversos , Antineoplásicos Imunológicos/uso terapêutico , Antineoplásicos Imunológicos/efeitos adversos , Antígeno B7-H1/antagonistas & inibidores , Bevacizumab/uso terapêutico , Bevacizumab/efeitos adversos , Viés , Neoplasias Encefálicas/tratamento farmacológico , Neoplasias Encefálicas/mortalidade , Neoplasias Encefálicas/imunologia , Glioblastoma/tratamento farmacológico , Glioblastoma/mortalidade , Glioblastoma/imunologia , Inibidores de Checkpoint Imunológico/uso terapêutico , Inibidores de Checkpoint Imunológico/efeitos adversos , Recidiva Local de Neoplasia/tratamento farmacológico , Nivolumabe/uso terapêutico , Nivolumabe/efeitos adversos , Receptor de Morte Celular Programada 1/antagonistas & inibidores , Intervalo Livre de Progressão , Qualidade de Vida , Ensaios Clínicos Controlados Aleatórios como Assunto
6.
Drug Dev Res ; 85(8): e70023, 2024 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-39620407

RESUMO

Glioblastoma multiforme (GBM) is the most prevalent primary brain tumor in adults and is known for its rapid proliferation and infiltrative nature. Current therapeutic strategies include surgical resection followed by radio- and chemotherapy. Still, they are hindered by GBM biological characteristics and physical-chemical properties of chemotherapeutic drugs, leading to limited efficacy and poor prognosis. Cannabinoids have emerged as potential anti-GBM agents, exhibiting antiangiogenic, antimetastatic, and antiproliferative effects. However, their hydrophobicity and poor oral bioavailability pose significant challenges for clinical applications. This study evaluates the potential of nanocarriers in enhancing the solubility and targeted delivery of cannabinoids for GBM therapy. The innovative combination of nanotechnology with cannabinoid-based treatment offers a promising strategy to improve therapeutic outcomes. We addressed the application of nanocarriers to deliver cannabinoids, which can enhance passage across the blood-brain barrier and enable targeted therapy. Studies demonstrate the potential of nanocarriers in improving solubility, stability, and controlled release of cannabinoids, highlighting the advancements in nanocarrier design for optimized delivery to glioma cells. Cannabinoids can exert their antitumor effect, including the induction of apoptosis through the ceramide and p8-regulated pathways and the modulation of immune responses. The evidence found in this study supports the potential of cannabinoid-based nanotechnologies in GBM therapeutic regimens as a strategy to enhance its efficacy and patient outcomes.


Assuntos
Neoplasias Encefálicas , Canabinoides , Sistemas de Liberação de Medicamentos , Glioblastoma , Glioblastoma/tratamento farmacológico , Humanos , Canabinoides/administração & dosagem , Canabinoides/química , Canabinoides/uso terapêutico , Canabinoides/farmacologia , Neoplasias Encefálicas/tratamento farmacológico , Nanotecnologia , Antineoplásicos/administração & dosagem , Antineoplásicos/química , Antineoplásicos/uso terapêutico , Animais , Nanopartículas , Portadores de Fármacos/química
7.
BMC Cancer ; 24(1): 1539, 2024 Dec 18.
Artigo em Inglês | MEDLINE | ID: mdl-39695426

RESUMO

BACKGROUND: Glioblastoma (GBM) is an aggressive brain tumor driven by glioblastoma stem cells (GSCs), which represent an appealing target for therapeutic interventions. The cellular prion protein (PrPC), a scaffold protein involved in diverse cellular processes, interacts with various membrane and extracellular matrix molecules, influencing tumor biology. Herein, we investigate the impact of PrPC expression on GBM. METHODS: To address this goal, we employed CRISPR-Cas9 technology to generate PrPC knockout (KO) glioblastoma cell lines, enabling detailed loss-of-function studies. Bulk RNA sequencing followed by differentially expressed gene and pathway enrichment analyses between U87 or U251 PrPC-wild-type (WT) cells and PrPC-knockout (KO) cells were used to identify pathways regulated by PrPC. Immunofluorescence assays were used to evaluate cellular morphology and protein distribution. For assessment of protein levels, Western blot and flow cytometry assays were employed. Transwell and growth curve assays were used to determine the impact of loss-of-PrPC in GBM invasiveness and proliferation, respectively. Single-cell RNA sequencing analysis of data from patient tumors from The Cancer Genome Atlas (TCGA) and the Broad Institute of Single-Cell Data Portal were used to evaluate the correspondence between our in vitro results and patient samples. RESULTS: Transcriptome analysis of PrPC-KO GBM cell lines revealed altered expression of genes associated with crucial tumor progression pathways, including migration, proliferation, and stemness. These findings were corroborated by assays that revealed impaired invasion, migration, proliferation, and self-renewal in PrPC-KO GBM cells, highlighting its critical role in sustaining tumor growth. Notably, loss-of-PrPC disrupted the expression and localization of key stemness markers, particularly CD44. Additionally, the modulation of PrPC levels through CD44 overexpression further emphasizes their regulatory role in these processes. CONCLUSIONS: These findings establish PrPC as a modulator of essential molecules on the cell surface of GSCs, highlighting its potential as a therapeutic target for GBM.


Assuntos
Neoplasias Encefálicas , Glioblastoma , Invasividade Neoplásica , Células-Tronco Neoplásicas , Glioblastoma/patologia , Glioblastoma/metabolismo , Glioblastoma/genética , Humanos , Células-Tronco Neoplásicas/metabolismo , Células-Tronco Neoplásicas/patologia , Linhagem Celular Tumoral , Neoplasias Encefálicas/patologia , Neoplasias Encefálicas/metabolismo , Neoplasias Encefálicas/genética , Proliferação de Células , Técnicas de Inativação de Genes , Proteínas PrPC/metabolismo , Proteínas PrPC/genética , Proteínas Priônicas/metabolismo , Proteínas Priônicas/genética , Regulação Neoplásica da Expressão Gênica , Movimento Celular , Sistemas CRISPR-Cas
8.
FASEB J ; 38(24): e70255, 2024 Dec 13.
Artigo em Inglês | MEDLINE | ID: mdl-39698937

RESUMO

We explored key microRNAs (miRNAs) related to tumorigenesis and immune modulation in glioblastoma (GBM), employing in silico, in vitro, and ex vivo analysis along with an assessment of the cellular impacts resulting from miRNA inhibition. GBM and T cells miRNA expression profiles from public datasets were used to evaluate differentially expressed miRNAs (DEmiRNAs). Some DEmiRNAs were chosen for validation in GBM cell lines, primary cell cultures, and brain tumor patient samples, using RT-qPCR. Target genes and pathways were identified with bioinformatic analyses. In silico functional enrichment analysis revealed that miR-27a-3p and miR-155-5p modulate immune, metabolic, and GBM-related pathways. A172 cells were transfected with miRNA inhibitors and the effects on cellular processes and immunomodulation were analyzed by co-culture assays and flow cytometry. Upon validation, miR-27a-3p and miR-155-5p miRNAs expressions were consistently increased. Inhibiting these two miRNAs reduced cell viability, but only the inhibition of miR-27a-3p led to apoptosis. Co-culture assays showed an increase in Th1 cells along with elevated Th1/Treg and Th17/Treg ratios, and an increase in Th17 cells exclusively with miR-155-5p inhibition. Immune cells' gene expression modulation induced an antitumor profile, concomitant with an increase in the expression of apoptotic genes in cancer cells after co-culture. This study unveils potential targets for immune and tumor regulation, highlighting overexpressed miRNAs modulation as a novel therapeutic approach for GBM.


Assuntos
Apoptose , Neoplasias Encefálicas , Regulação Neoplásica da Expressão Gênica , Glioblastoma , MicroRNAs , Células Th1 , MicroRNAs/genética , MicroRNAs/metabolismo , Humanos , Glioblastoma/genética , Glioblastoma/patologia , Glioblastoma/metabolismo , Apoptose/genética , Células Th1/metabolismo , Células Th1/imunologia , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/patologia , Neoplasias Encefálicas/metabolismo , Diferenciação Celular , Linhagem Celular Tumoral , Simulação por Computador , Progressão da Doença
9.
Viruses ; 16(11)2024 Nov 12.
Artigo em Inglês | MEDLINE | ID: mdl-39599878

RESUMO

Central nervous system (CNS) cancers are responsible for high rates of morbidity and mortality worldwide. Malignant CNS tumors such as adult Glioblastoma (GBM) and pediatric embryonal CNS tumors such as medulloblastoma (MED) and atypical teratoid rhabdoid tumors (ATRT) present relevant therapeutic challenges due to the lack of response to classic treatment regimens with radio and chemotherapy. Recent findings on the Zika virus' (ZIKV) ability to infect and kill CNS neoplastic cells draw attention to the virus' oncolytic potential. Studies demonstrating the safety of using ZIKV for treating malignant CNS tumors, enabling the translation of this approach to clinical trials, are scarce in the literature. Here we developed a co-culture model of mature human cerebral organoids assembled with GBM, MED or ATRT tumor cells and used these assembloids to test ZIKV oncolytic effect, replication potential and preferential targeting between normal and cancer cells. Our hybrid co-culture models allowed the tracking of tumor cell growth and invasion in cerebral organoids. ZIKV replication and ensuing accumulation in the culture medium was higher in organoids co-cultured with tumor cells than in isolated control organoids without tumor cells. ZIKV infection led to a significant reduction in tumor cell proportion in organoids with GBM and MED cells, but not with ATRT. Tumoroids (3D cultures of tumor cells alone) were efficiently infected by ZIKV. Interestingly, ZIKV rapidly replicated in GBM, MED, and ATRT tumoroids reaching significantly higher viral RNA accumulation levels than co-cultures. Moreover, ZIKV infection reduced viable cells number in MED and ATRT tumoroids but not in GBM tumoroids. Altogether, our findings indicate that ZIKV has greater replication rates in aggressive CNS tumor cells than in normal human cells comprising cerebral organoids. However, such higher ZIKV replication in tumor cells does not necessarily parallels oncolytic effects, suggesting cellular intrinsic and extrinsic factors mediating tumor cell death by ZIKV.


Assuntos
Técnicas de Cocultura , Terapia Viral Oncolítica , Vírus Oncolíticos , Organoides , Replicação Viral , Infecção por Zika virus , Zika virus , Humanos , Zika virus/fisiologia , Zika virus/genética , Organoides/virologia , Terapia Viral Oncolítica/métodos , Vírus Oncolíticos/fisiologia , Vírus Oncolíticos/genética , Infecção por Zika virus/terapia , Infecção por Zika virus/virologia , Glioblastoma/terapia , Glioblastoma/virologia , Neoplasias do Sistema Nervoso Central/terapia , Neoplasias do Sistema Nervoso Central/virologia , Meduloblastoma/terapia , Meduloblastoma/virologia , Linhagem Celular Tumoral , Tumor Rabdoide/terapia , Tumor Rabdoide/virologia
10.
Sci Rep ; 14(1): 27995, 2024 11 14.
Artigo em Inglês | MEDLINE | ID: mdl-39543155

RESUMO

Glioblastoma is an aggressive brain cancer with a poor prognosis. The O6-methylguanine-DNA methyltransferase (MGMT) gene methylation status is crucial for treatment stratification, yet economic constraints often limit access. This study aims to develop an artificial intelligence (AI) framework for predicting MGMT methylation. Diagnostic magnetic resonance (MR) images in public repositories were used for training. The algorithm created was validated in data from a single institution. All images were segmented according to widely used guidelines for radiotherapy planning and combined with clinical evaluations from neuroradiology experts. Radiomic features and clinical impressions were extracted, tabulated, and used for modeling. Feature selection methods were used to identify relevant phenotypes. A total of 100 patients were used for training and 46 for validation. A total of 343 features were extracted. Eight feature selection methods produced seven independent predictive frameworks. The top-performing ML model was a model post-Least Absolute Shrinkage and Selection Operator (LASSO) feature selection reaching accuracy (ACC) of 0.82, an area under the curve (AUC) of 0.81, a recall of 0.75, and a precision of 0.75. This study demonstrates that integrating clinical and radiotherapy-derived AI-driven phenotypes can predict MGMT methylation. The framework addresses constraints that limit molecular diagnosis access.


Assuntos
Inteligência Artificial , Neoplasias Encefálicas , Metilação de DNA , Metilases de Modificação do DNA , Enzimas Reparadoras do DNA , Glioblastoma , Proteínas Supressoras de Tumor , Humanos , Glioblastoma/genética , Glioblastoma/diagnóstico , Metilases de Modificação do DNA/genética , Metilases de Modificação do DNA/metabolismo , Enzimas Reparadoras do DNA/genética , Enzimas Reparadoras do DNA/metabolismo , Proteínas Supressoras de Tumor/genética , Neoplasias Encefálicas/genética , Feminino , Masculino , Pessoa de Meia-Idade , Imageamento por Ressonância Magnética/métodos , Idoso , Sistemas de Apoio a Decisões Clínicas , Adulto , Algoritmos , Prognóstico , Região de Recursos Limitados
11.
Sci Rep ; 14(1): 26003, 2024 10 29.
Artigo em Inglês | MEDLINE | ID: mdl-39472601

RESUMO

We presented a method to find potential cancer attractors using single-cell RNA sequencing (scRNA-seq) data. We tested our method in a Glioblastoma Multiforme (GBM) dataset, an aggressive brain tumor presenting high heterogeneity. Using the cancer attractor concept, we argued that the GBM's underlying dynamics could partially explain the observed heterogeneity, with the dataset covering a representative region around the attractor. Exploratory data analysis revealed promising GBM's cellular clusters within a 3-dimensional marker space. We approximated the clusters' centroid as stable states and each cluster covariance matrix as defining confidence regions. To investigate the presence of attractors inside the confidence regions, we constructed a GBM gene regulatory network, defined a model for the dynamics, and prepared a framework for parameter estimation. An exploration of hyperparameter space allowed us to sample time series intending to simulate myriad variations of the tumor microenvironment. We obtained different densities of stable states across gene expression space and parameters displaying multistability across different clusters. Although we used our methodological approach in studying GBM, we would like to highlight its generality to other types of cancer. Therefore, this report contributes to an advance in the simulation of cancer dynamics and opens avenues to investigate potential therapeutic targets.


Assuntos
Neoplasias Encefálicas , Glioblastoma , Análise de Sequência de RNA , Análise de Célula Única , Glioblastoma/genética , Glioblastoma/patologia , Humanos , Análise de Célula Única/métodos , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/patologia , Análise de Sequência de RNA/métodos , Simulação por Computador , Redes Reguladoras de Genes , Regulação Neoplásica da Expressão Gênica , Microambiente Tumoral/genética , Perfilação da Expressão Gênica/métodos , Biologia Computacional/métodos
12.
Sci Rep ; 14(1): 25435, 2024 10 25.
Artigo em Inglês | MEDLINE | ID: mdl-39455622

RESUMO

Oncostreams are self-organized structures formed by spindle-like, elongated, self-propelled cells recently described in glioblastomas and especially in gliosarcomas. Cells within these structures either move as large clusters in one main direction, flocks, or as linear, intermingling collections of cells advancing in opposite directions, streams. Round, passive cells are also observed, either inside or segregated from the oncostreams. Here we generalize a recently formulated particle-field approach to investigate the genesis and evolution of these structures, first showing that, in systems consisting only of identical self-propelled cells, both flocks and streams emerge as self-organized dynamic configurations. Flocks are the more stable configurations, while streams are transient and usually originate in collisions between flocks. Stream degradation is easier at low self-propulsion speeds. In systems consisting of both motile and passive cells, the latter block stream formation and accelerate their degradation and flock stabilization. Since the flock appears to be the most effective invasive structure, we thus argue that a phenotype mixture (motile and passive cells) may favor glioblastoma invasion. hlBy relating cellular properties to the observed outcome, our model shows that oncostreams are self-organized structures that result from the interplay between speed, shape, and steric repulsion.


Assuntos
Glioma , Humanos , Glioma/patologia , Glioma/metabolismo , Neoplasias Encefálicas/patologia , Glioblastoma/patologia , Modelos Biológicos , Movimento Celular , Invasividade Neoplásica
13.
Biomater Sci ; 12(21): 5547-5561, 2024 Oct 22.
Artigo em Inglês | MEDLINE | ID: mdl-39292186

RESUMO

Carbon-derived compounds are gaining traction in the scientific community because of their unique properties, such as conductivity and strength, and promising innovations in technology and medicine. Graphitic nitride carbon (g-C3N4) stands out among these compounds because of its potential in antitumor therapies. This study aimed to assess g-C3N4's antitumor potential and cytotoxic mechanisms. Prostate cancer (DU-145) and glioblastoma (U87) cell lines were used to evaluate antitumor effects, whereas RAW 264.7 and HFF-1 non-tumor cells were used for selectivity evaluation. The synthesized g-C3N4 particles underwent comprehensive characterization, including the assessment of particle size, morphology, and oxygen content, employing various techniques, such as X-ray diffraction, X-ray photoelectron spectroscopy, scanning electron microscopy, energy dispersive X-ray spectroscopy, transmission electron microscopy, and atomic force microscopy. The results indicated that g-C3N4 significantly affected tumor cell proliferation and viability, exhibiting high cytotoxicity within 48 h. In non-tumor cells, minimal effects on proliferation were observed, except for damage to the cell membranes of RAW 264.7 cells. Moreover, g-C3N4 changed the cell morphology and ultrastructure, affecting cell migration in U87 cells and potentially enhancing migration in RAW 264.7 cells. Biochemical assays in Balb/C mice revealed alterations in alanine aminotransferase, aspartate aminotransferase, and amylase levels. In conclusion, g-C3N4 demonstrated promising antitumor effects with minimal toxicity to non-tumor cells, suggesting its potential in neoplasm treatment.


Assuntos
Antineoplásicos , Proliferação de Células , Glioblastoma , Grafite , Compostos de Nitrogênio , Neoplasias da Próstata , Grafite/química , Grafite/farmacologia , Humanos , Antineoplásicos/farmacologia , Antineoplásicos/química , Antineoplásicos/administração & dosagem , Masculino , Animais , Camundongos , Neoplasias da Próstata/tratamento farmacológico , Neoplasias da Próstata/patologia , Glioblastoma/tratamento farmacológico , Glioblastoma/patologia , Glioblastoma/metabolismo , Compostos de Nitrogênio/química , Compostos de Nitrogênio/farmacologia , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Células RAW 264.7 , Sobrevivência Celular/efeitos dos fármacos , Movimento Celular/efeitos dos fármacos
14.
Oncotarget ; 15: 662-673, 2024 Sep 30.
Artigo em Inglês | MEDLINE | ID: mdl-39347716

RESUMO

INTRODUCTION: Many studies have highlighted the use of oncolytic viruses as a new class of therapeutic agents for central nervous system (CNS) tumors, especially glioblastomas (GMB). Zika Virus (ZIKV) proteins targeted to specific stem cells have been studied in vitro and animal models with promising results. MATERIALS AND METHODS: A systematic review was evaluated the efficacy and safety of the ZIKV use for CNS tumors treatment. Data were extracted and the in vivo studies were evaluated using the Robins-I tool. We assessed bias in each study using criteria such as selection bias, performance bias, detection bias, attrition bias, reporting bias, and others. According to Cochrane guidelines, bias was classified as high, low, or uncertain. High bias occurred when studies did not meet the criteria. Low bias was assigned when criteria were clearly met. Uncertain bias reflected insufficient information for a clear classification. RESULTS: The 14 included studies shown that ZIKV reduced cell viability or inhibited the growth, proliferation of glioma stem cells (GSCs), and Bcl2 expression - which could potentially enhance the effect of chemotherapy/radiotherapy; caused cytopathic effects, induced tumor cell damage, manifested oncolytic properties, and even selectively safely killed GSCs; ultimately, it led to significant tumor remission and enhanced long-term survival through enhanced T-cell response. CONCLUSIONS: Although current evidence suggests ZIKV as a promising treatment for CNS tumors and may improve survival when combined with surgery and radiotherapy. Despite limited human evidence, it shows potential benefits. Further research is needed to confirm safety, efficacy, and optimize treatment in humans.


Assuntos
Neoplasias Encefálicas , Terapia Viral Oncolítica , Zika virus , Animais , Humanos , Neoplasias Encefálicas/terapia , Neoplasias Encefálicas/virologia , Proliferação de Células , Glioblastoma/terapia , Glioblastoma/virologia , Células-Tronco Neoplásicas/virologia , Terapia Viral Oncolítica/métodos , Vírus Oncolíticos
15.
J Cell Biochem ; 125(9): e30637, 2024 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-39150066

RESUMO

Glioblastoma (GBM) aggressiveness is partly driven by the reactivation of signaling pathways such as Sonic hedgehog (SHH) and the interaction with its microenvironment. SHH pathway activation is one of the phenomena behind the glial transformation in response to tumor growth. The reactivation of the SHH signaling cascade during GBM-astrocyte interaction is highly relevant to understanding the mechanisms used by the tumor to modulate the adjacent stroma. The role of reactive astrocytes considering SHH signaling during GBM progression is investigated using a 3D in vitro model. T98G GBM spheroids displayed significant downregulation of SHH (61.4 ± 9.3%), GLI-1 (6.5 ± 3.7%), Ki-67 (33.7 ± 8.1%), and mutant MTp53 (21.3 ± 10.6%) compared to the CONTROL group when incubated with conditioned medium of reactive astrocytes (CM-AST). The SHH pathway inhibitor, GANT-61, significantly reduced previous markers (SHH = 43.0 ± 12.1%; GLI-1 = 9.5 ± 3.4%; Ki-67 = 31.9 ± 4.6%; MTp53 = 6.5 ± 7.5%) compared to the CONTROL, and a synergistic effect could be observed between GANT-61 and CM-AST. The volume (2.0 ± 0.2 × 107 µm³), cell viability (80.4 ± 3.2%), and migration (41 ± 10%) of GBM spheroids were significantly reduced in the presence of GANT-61 and CM-AST when compared to CM-AST after 72 h (volume = 2.3 ± 0.4 × 107 µm³; viability = 92.2 ± 6.5%; migration = 102.5 ± 14.6%). Results demonstrated that factors released by reactive astrocytes promoted a neuroprotective effect preventing GBM progression using a 3D in vitro model potentiated by SHH pathway inhibition.


Assuntos
Astrócitos , Movimento Celular , Proliferação de Células , Glioblastoma , Esferoides Celulares , Proteína Supressora de Tumor p53 , Proteína GLI1 em Dedos de Zinco , Humanos , Proteína GLI1 em Dedos de Zinco/metabolismo , Proteína GLI1 em Dedos de Zinco/genética , Glioblastoma/metabolismo , Glioblastoma/patologia , Glioblastoma/genética , Astrócitos/metabolismo , Meios de Cultivo Condicionados/farmacologia , Proteína Supressora de Tumor p53/metabolismo , Proteína Supressora de Tumor p53/genética , Esferoides Celulares/metabolismo , Proteínas Hedgehog/metabolismo , Proteínas Hedgehog/genética , Regulação para Baixo , Linhagem Celular Tumoral , Piridinas/farmacologia , Regulação Neoplásica da Expressão Gênica , Transdução de Sinais , Mutação , Pirimidinas/farmacologia
16.
Int J Biol Macromol ; 278(Pt 2): 134777, 2024 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-39153669

RESUMO

Glioblastoma (GBM) represents a formidable challenge in oncology, characterized by aggressive proliferation and poor prognosis. Iron metabolism plays a critical player in GBM progression, with dysregulated iron uptake and utilization contributing to tumor growth and therapeutic resistance. Iron's pivotal role in DNA synthesis, oxidative stress, and angiogenesis underscores its significance in GBM pathogenesis. Elevated expression of iron transporters, such as transferrin receptor 1 (TfR1), highlights the tumor's reliance on iron for survival. Innovative treatment strategies targeting iron dysregulation hold promise for overcoming therapeutic challenges in GBM management. Approaches such as iron chelation therapies, induction of ferroptosis to nanoparticle-based drug delivery systems exploit iron-dependent vulnerabilities, offering avenues for enhance treatment efficacy and improve patient outcomes. As research advances, understanding the complexities of iron-mediated carcinogenesis provides a foundation for developing precision medicine approaches tailored to combat GBM effectively. This review explores the intricate relationship between iron metabolism and GBM, elucidating its multifaceted implications and therapeutic opportunities. By consolidating the latest insights into iron metabolism in GBM, this review underscores its potential as a therapeutic target for improving patient care in combination with the standard of care approach.


Assuntos
Ferroptose , Glioblastoma , Ferro , Receptores da Transferrina , Humanos , Receptores da Transferrina/metabolismo , Ferro/metabolismo , Ferroptose/efeitos dos fármacos , Glioblastoma/metabolismo , Glioblastoma/tratamento farmacológico , Glioblastoma/patologia , Animais , Neoplasias Encefálicas/metabolismo , Neoplasias Encefálicas/tratamento farmacológico , Neoplasias Encefálicas/patologia , Antígenos CD/metabolismo , Antígenos CD/genética , Quelantes de Ferro/uso terapêutico , Quelantes de Ferro/farmacologia
17.
Int J Mol Sci ; 25(14)2024 Jul 09.
Artigo em Inglês | MEDLINE | ID: mdl-39062770

RESUMO

Glioblastoma (GBM) is an aggressive brain cancer characterized by significant molecular and cellular heterogeneity, which complicates treatment efforts. Current standard therapies, including surgical resection, radiation, and temozolomide (TMZ) chemotherapy, often fail to achieve long-term remission due to tumor recurrence and resistance. A pro-oxidant environment is involved in glioma progression, with oxidative stress contributing to the genetic instability that leads to gliomagenesis. Evaluating pro-oxidant therapies in brain tumors is crucial due to their potential to selectively target and eradicate cancer cells by exploiting the elevated oxidative stress levels inherent in these malignant cells, thereby offering a novel and effective strategy for overcoming resistance to conventional therapies. This study investigates the therapeutic potential of doxorubicin (DOX) and photodynamic therapy (PDT) with Me-ALA, focusing on their effects on redox homeostasis. Basal ROS levels and antioxidant gene expression (NFE2L2, CAT, GSR) were quantitatively assessed across GBM cell lines, revealing significant variability probably linked to genetic differences. DOX and PDT treatments, both individually and in combination, were analyzed for their efficacy in inducing oxidative stress and cytotoxicity. An in silico analysis further explored the relationship between gene mutations and oxidative stress in GBM patients, providing insights into the molecular mechanisms underlying treatment responses. Our findings suggest that pro-oxidant therapies, such as DOX and PDT in combination, could selectively target GBM cells, highlighting a promising avenue for improving therapeutic outcomes in GBM.


Assuntos
Neoplasias Encefálicas , Doxorrubicina , Glioblastoma , Estresse Oxidativo , Fotoquimioterapia , Espécies Reativas de Oxigênio , Glioblastoma/tratamento farmacológico , Glioblastoma/metabolismo , Glioblastoma/patologia , Glioblastoma/genética , Humanos , Doxorrubicina/farmacologia , Doxorrubicina/uso terapêutico , Fotoquimioterapia/métodos , Estresse Oxidativo/efeitos dos fármacos , Espécies Reativas de Oxigênio/metabolismo , Linhagem Celular Tumoral , Neoplasias Encefálicas/tratamento farmacológico , Neoplasias Encefálicas/patologia , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/metabolismo , Sinergismo Farmacológico , Fármacos Fotossensibilizantes/farmacologia , Fármacos Fotossensibilizantes/uso terapêutico
18.
J Comput Chem ; 45(31): 2666-2677, 2024 Dec 05.
Artigo em Inglês | MEDLINE | ID: mdl-39082832

RESUMO

Recently tetraspanin CD151 has been identified as an important biological target involved in metastatic processes which include cell adhesion, tumor progression processes, and so forth in different types of cancers, such as breast cancer and glioblastoma. This in Silico study considered 1603 compounds from the Food and Drug Administration database, after performing an ADMET analysis; we selected 853 ligands, which were used for docking analysis. The most promising ligands were selected from docking studies, based on two criteria: (a) showed lowest affinity to the CD151 protein and (b) they interact with the QRD motif, located in the second extracellular loop. Furthermore, we investigate the stability of the protein-ligand complexes through MD simulations as well as free energy MM-PBSA calculations. From these results, loperamide and glipizide were identified as the best evaluated drugs. We suggest an in vitro analysis is needed to confirm our in silico prediction studies.


Assuntos
Antineoplásicos , Neoplasias da Mama , Glioblastoma , Tetraspanina 24 , Humanos , Glioblastoma/tratamento farmacológico , Glioblastoma/patologia , Neoplasias da Mama/tratamento farmacológico , Tetraspanina 24/química , Antineoplásicos/química , Antineoplásicos/farmacologia , Ligantes , Feminino , Simulação de Dinâmica Molecular , Simulação por Computador , Simulação de Acoplamento Molecular
19.
Sci Rep ; 14(1): 16721, 2024 07 19.
Artigo em Inglês | MEDLINE | ID: mdl-39030304

RESUMO

Antigen-specific cytotoxic CD8 T cells are extremely effective in controlling tumor growth and have been the focus of immunotherapy approaches. We leverage in silico tools to investigate whether the occurrence of mutations in proteins previously described as immunogenic and highly expressed by glioblastoma multiforme (GBM), such as Epidermal Growth Factor Receptor (EGFR), Isocitrate Dehydrogenase 1 (IDH1), Phosphatase and Tensin homolog (PTEN) and Tumor Protein 53 (TP53), may be contributing to the differential presentation of immunogenic epitopes. We recovered Class I MHC binding information from wild-type and mutated proteins using the Immune Epitope Database (IEDB). After that, we built peptide-MHC (pMHC-I) models in HLA-arena, followed by hierarchical clustering analysis based on electrostatic surface features from each complex. We identified point mutations that are determinants for the presentation of a set of peptides from TP53 protein. We point to structural features in the pMHC-I complexes of wild-type and mutated peptides, which may play a role in the recognition of CD8 T cells. To further explore these features, we performed 100 ns molecular dynamics simulations for the peptide pairs (wt/mut) selected. In pursuit of novel therapeutic targets for GBM treatment, we selected peptides where our predictive results indicated that mutations would not disrupt epitope presentation, thereby maintaining a specific CD8 T cell immune response. These peptides hold potential for future GBM interventions, including peptide-based or mRNA vaccine development applications.


Assuntos
Apresentação de Antígeno , Linfócitos T CD8-Positivos , Glioblastoma , Isocitrato Desidrogenase , Proteína Supressora de Tumor p53 , Glioblastoma/imunologia , Glioblastoma/genética , Glioblastoma/terapia , Humanos , Linfócitos T CD8-Positivos/imunologia , Isocitrato Desidrogenase/genética , Isocitrato Desidrogenase/imunologia , Isocitrato Desidrogenase/química , Proteína Supressora de Tumor p53/genética , Proteína Supressora de Tumor p53/imunologia , Apresentação de Antígeno/imunologia , Mutação , PTEN Fosfo-Hidrolase/genética , PTEN Fosfo-Hidrolase/imunologia , PTEN Fosfo-Hidrolase/química , Receptores ErbB/imunologia , Receptores ErbB/genética , Epitopos de Linfócito T/imunologia , Epitopos de Linfócito T/genética , Antígenos de Histocompatibilidade Classe I/imunologia , Antígenos de Histocompatibilidade Classe I/genética , Neoplasias Encefálicas/imunologia , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/terapia
20.
Braz J Med Biol Res ; 57: e13961, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38985083

RESUMO

Glioblastomas are known for their poor clinical prognosis, with recurrent tumors often exhibiting greater invasiveness and faster growth rates compared to primary tumors. To understand the intratumoral changes driving this phenomenon, we employed single-cell sequencing to analyze the differences between two pairs of primary and recurrent glioblastomas. Our findings revealed an upregulation of ferroptosis in endothelial cells within recurrent tumors, identified by the significant overexpression of the NOX4 gene. Further analysis indicated that knocking down NOX4 in endothelial cells reduced the activity of the ferroptosis pathway. Utilizing conditioned media from endothelial cells with lower ferroptosis activity, we observed a decrease in the growth rate of glioblastoma cells. These results highlighted the complex role of ferroptosis within tumors and suggested that targeting ferroptosis in the treatment of glioblastomas requires careful consideration of its effects on endothelial cells, as it may otherwise produce counterproductive outcomes.


Assuntos
Neoplasias Encefálicas , Células Endoteliais , Ferroptose , Glioblastoma , Isocitrato Desidrogenase , Recidiva Local de Neoplasia , Humanos , Glioblastoma/patologia , Glioblastoma/genética , Ferroptose/genética , Ferroptose/fisiologia , Neoplasias Encefálicas/patologia , Neoplasias Encefálicas/genética , Isocitrato Desidrogenase/genética , Células Endoteliais/patologia , Linhagem Celular Tumoral , Proliferação de Células
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA