OBJECT:Glioblastoma multiforme(GBM)is the most common and lethal primary brain tumor in adults.It isnearly uniformly fatal,with a median survival time of approximately l year,despite modem treatment modalities.Neverth...OBJECT:Glioblastoma multiforme(GBM)is the most common and lethal primary brain tumor in adults.It isnearly uniformly fatal,with a median survival time of approximately l year,despite modem treatment modalities.Nevertheless,a range of survival times exists around this median.Efforts to understand why some patients livelonger or shorter than the average may provide insight into the biology of these neoplasms.The annexin VII(ANX7)gene is located on the human chromosome 10q21,a site long hypothesized to harbor tumor展开更多
Glioblastoma(GBM)is the most common malignant brain tumor.Although current treatment strategies,including surgery,chemotherapy,and radiotherapy,have achieved clinical effects and prolonged the survival of patients,the...Glioblastoma(GBM)is the most common malignant brain tumor.Although current treatment strategies,including surgery,chemotherapy,and radiotherapy,have achieved clinical effects and prolonged the survival of patients,the gradual development of resistance against current therapies has led to a high recurrence rate and treatment failure.Mechanisms underlying the development of resistance involve multiple factors,including drug efflux,DNA damage repair,glioma stem cells,and a hypoxic tumor environment,which are usually correlative and promote each other.As many potential therapeutic targets have been discovered,combination therapy that regulates multiple resistance-related molecule pathways is considered an attractive strategy.In recent years,nanomedicine has revolutionized cancer therapies with optimized accumulation,penetration,internalization,and controlled release.Blood-brain barrier(BBB)penetration efficiency is also significantly improved through modifying ligands on nanomedicine and interacting with the receptors or transporters on the BBB.Moreover,different drugs for combination therapy usually process different pharmacokinetics and biodistribution,which can be further optimized with drug delivery systems to maximize the therapeutic efficiency of combination therapies.Herein the current achievements in nanomedicine-based combination therapy for GBM are discussed.This review aimed to provide a broader understanding of resistance mechanisms and nanomedicine-based combination therapies for future research on GBM treatment.展开更多
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 radioresist...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.展开更多
Recent research has demonstrated the impact of physical activity on the prognosis of glioma patients,with evidence suggesting exercise may reduce mortality risks and aid neural regeneration.The role of the small ubiqu...Recent research has demonstrated the impact of physical activity on the prognosis of glioma patients,with evidence suggesting exercise may reduce mortality risks and aid neural regeneration.The role of the small ubiquitin-like modifier(SUMO)protein,especially post-exercise,in cancer progression,is gaining attention,as are the potential anti-cancer effects of SUMOylation.We used machine learning to create the exercise and SUMO-related gene signature(ESLRS).This signature shows how physical activity might help improve the outlook for low-grade glioma and other cancers.We demonstrated the prognostic and immunotherapeutic significance of ESLRS markers,specifically highlighting how murine double minute 2(MDM2),a component of the ESLRS,can be targeted by nutlin-3.This underscores the intricate relationship between natural compounds such as nutlin-3 and immune regulation.Using comprehensive CRISPR screening,we validated the effects of specific ESLRS genes on low-grade glioma progression.We also revealed insights into the effectiveness of Nutlin-3a as a potent MDM2 inhibitor through molecular docking and dynamic simulation.Nutlin-3a inhibited glioma cell proliferation and activated the p53 pathway.Its efficacy decreased with MDM2 overexpression,and this was reversed by Nutlin-3a or exercise.Experiments using a low-grade glioma mouse model highlighted the effect of physical activity on oxidative stress and molecular pathway regulation.Notably,both physical exercise and Nutlin-3a administration improved physical function in mice bearing tumors derived from MDM2-overexpressing cells.These results suggest the potential for Nutlin-3a,an MDM2 inhibitor,with physical exercise as a therapeutic approach for glioma management.Our research also supports the use of natural products for therapy and sheds light on the interaction of exercise,natural products,and immune regulation in cancer treatment.展开更多
BACKGROUND Glioblastoma multiforme(GBM)is the most aggressive and prevalent primary malignant brain tumor in adults,marked by poor prognosis and high invasiveness.Traditional GBM invasion assays,such as those involvin...BACKGROUND Glioblastoma multiforme(GBM)is the most aggressive and prevalent primary malignant brain tumor in adults,marked by poor prognosis and high invasiveness.Traditional GBM invasion assays,such as those involving mouse brain xenografts,are often time-consuming and limited in efficiency.In this context,stem cell-derived neural organoids(NOs)have emerged as advanced,threedimensional,human-relevant platforms that mimic the cellular architecture and microenvironment of the human brain.These models provide novel opportunities to investigate glioblastoma stem cell invasion,a critical driver of tumor progression and therapeutic resistance.AIM To evaluate studies using stem cell-derived NOs to model glioblastoma migration/invasion,focusing on methodologies,applications and therapeutic implications.METHODS We conducted a systematic review following PRISMA guidelines,searching PubMed and Scopus for studies published between March 2019 and March 2025 that investigated NOs in the context of glioblastoma invasion/migration.After screening 377 articles based on predefined inclusion and exclusion criteria,10 original research articles were selected for analysis.Extracted data were categorized into four analytical domains:(1)Tumor model formation;(2)NO characteristics;(3)NO differentiation protocols;and(4)Invasion/migration assessment methodologies.RESULTS The included studies exhibit significant methodological heterogeneity GBM model development,particularly regarding model type,cell source and culture conditions.Most studies(70%)used suspension cell models,while 30%employed spheroids,with most research focusing on patient-derived glioblastoma stem cells.NOs were predominantly generated from human induced pluripotent stem cells using both guided and unguided differentiation protocols.Confocal fluorescence microscopy was the primary method used for assessing invasion,revealing invasion depths of up to 300μm.Organoid maturity and co-culture duration influenced results,while key factors for model optimization included tumor cell density,organoid age and extracellular matrix composition.Some studies also tested therapeutic strategies such as Zika virus and microRNA modulation.Collectively,findings support the utility of NOs as effective tools for studying GBM behavior and therapeutic responses in a humanized three-dimensional context.CONCLUSION Human NOs represent promising platforms for modeling glioblastoma invasion in a humanized three-dimensional environment.However,a limited number of studies and methodological heterogeneity hinder reproducibility.Protocol standardization is essential to enhance the translational application of these models.展开更多
D-D’-A type aza-borondipyrromethenes(aza-BODIPYs)were prepared by Suzuki cross-coupling reaction.Photothermal conversion efficiency of self-assemble aza-BODIPY-based nanoparticles(DA-azaBDP-NPs)with NIR-Ⅱ emission(...D-D’-A type aza-borondipyrromethenes(aza-BODIPYs)were prepared by Suzuki cross-coupling reaction.Photothermal conversion efficiency of self-assemble aza-BODIPY-based nanoparticles(DA-azaBDP-NPs)with NIR-Ⅱ emission(λ_(em)=1065 nm)was 37.2%under near infrared(NIR)irradiation,and the outstanding cytotoxicity was triggered by coexistence of DA-azaBDP-NPs and the NIR irradiation,with the decrease of glioblastoma migration and the inhibition of glioblastoma proliferation.DA-azaBDP-NPs could promote glioblastoma autophagy and accelerate the process of cell death.The photothermal therapy(PTT)of DAazaBDP-NPs can effectively induce glioblastoma death by apoptosis under the NIR irradiation,which is highly promising to be applied in vivo experiments of brain.展开更多
1.HOU J,DU Y,SHAO Y,et al.Identification,Albanol B inhibits glioblastoma progression by inducing senescence and apoptosis via the RNF6/p27 signaling axis.Phytomedicine.DOI:10.1016/j.phymed.2025.157056.(2025)[题目]Alba...1.HOU J,DU Y,SHAO Y,et al.Identification,Albanol B inhibits glioblastoma progression by inducing senescence and apoptosis via the RNF6/p27 signaling axis.Phytomedicine.DOI:10.1016/j.phymed.2025.157056.(2025)[题目]Albanol B通过RNF6/p27信号轴诱导细胞衰老与凋亡,从而抑制胶质母细胞瘤进展。展开更多
Herpes simplex virus thymidine kinase(HSVtk)gene therapy is a promising strategy for glioblastoma therapy.However,delivery of plasmid DNA(pDNA)encoding HSVtk into the brain by systemic administration is a challenge si...Herpes simplex virus thymidine kinase(HSVtk)gene therapy is a promising strategy for glioblastoma therapy.However,delivery of plasmid DNA(pDNA)encoding HSVtk into the brain by systemic administration is a challenge since pDNA can hardly penetrate the bloodbrain barrier.In this study,an exosome-membrane(EM)and polymer-based hybrid complex was developed for systemic delivery of pDNA into the brain.Histidine/arginine-linked polyamidoamine(PHR)was used as a carrier.PHR binds to pDNA by electrostatic interaction.The pDNA/PHR complex was mixed with EM and subjected to extrusion to produce pDNA/PHR-EM hybrid complex.For glioblastoma targeting,T7 peptide was attached to the pDNA/PHR-EM complex.Both pDNA/PHR-EM and T7-decorated pDNA/PHR-EM(pDNA/PHREM-T7)had a surface charge of–5 mV and a size of 280 nm.Transfection assays indicated that pDNA/PHR-EM-T7 enhanced the transfection to C6 cells compared with pDNA/PHREM.Intravenous administration of pHSVtk/PHR-EM-T7 showed that pHSVtk/PHR-EM and pHSVtk/PHR-EM-T7 delivered pHSVtk more efficiently than pHSVtk/lipofectamine and pHSVtk/PHR into glioblastoma in vivo.pHSVtk/PHR-EM-T7 had higher delivery efficiency than pHSVtk/PHR-EM.As a result,the HSVtk expression and apoptosis levels in the tumors of the pHSVtk/PHR-EM-T7 group were higher than those of the other control groups.Therefore,the pDNA/PHR-EM-T7 hybrid complex is a useful carrier for systemic delivery of pHSVtk to glioblastoma.展开更多
Current treatments for glioblastoma face challenges such as the blood-brain barrier and lack of targeted therapy,compounded by the aggressive nature,high invasiveness,and heterogeneity of the disease.Exosomes,a subtyp...Current treatments for glioblastoma face challenges such as the blood-brain barrier and lack of targeted therapy,compounded by the aggressive nature,high invasiveness,and heterogeneity of the disease.Exosomes,a subtype of extracellular vesicles are emerging as promising nanocarrier drug delivery systems to address these limitations.Exosomes released by all cell types can be easily obtained and modified as delivery vehicles or therapeutic agents.A systematic review was conducted to evaluate various methods for exosome isolation,characterization,engineering or modification,drug loading and delivery efficiency,including exosome biodistribution and treatment efficacy.A search of four databases for in vitro and in vivo studies(2000–,2023)identified 6165 records,of which 23 articles were found eligible and included for analyses.Most studies applied ultracentrifugation(UC)for exosomes isolation.Cancer cell lines being the most frequently used source of exosomes,followed by stem cells.The incubation approach was predominantly utilized to modify exosomes for drug loading.In vivo analysis showed that exosome biodistribution was primarily concentrated in the brain region,peaking in the first 6 h and remained moderately high.Compared to native exosomes and untreated control groups,utilizing modified native exosomes(cargo loaded)for treating glioblastoma disease models led to more pronounced suppression of tumor growth and proliferation,enhanced stimulation of immune response and apoptosis,effective restoration of drug chemosensitivity,increased anti-tumor effect and prolonged survival rates.Modified exosomes whether through incubation,sonication,transfection,freeze-thawing or their combination,improve targeted delivery and therapeutic efficacy against glioblastoma.展开更多
Background:Glioblastoma(GBM)is a highly aggressive brain tumor characterized by aberrant angiogenesis and an immunosuppressive microenvironment.Pericytes are aberrantly recruited but their spatiotemporal roles and mol...Background:Glioblastoma(GBM)is a highly aggressive brain tumor characterized by aberrant angiogenesis and an immunosuppressive microenvironment.Pericytes are aberrantly recruited but their spatiotemporal roles and molecular changes remain unclear.This study investigated platelet-derived growth factor receptor beta-positive(Pdgfrb+)pericyte dynamics and reprogramming in GBM vasculature.Methods:We generated GL261-Luc and GL261-CFP glioblastoma cells via lentiviral transduction and established two transgenic models.(1)For pericyte labeling,Ai14 reporter mice was crossed with PDGFRβ-P2A-CreERT2mice for td Tomato-specific lineage tracing(PT mice).(2)For conditional ablation,we generated inducible Pdgfrb-expressing cell ablation models(PT mice was crossed with ROSA-DTA mice).An intravital imaging platform(FITC-dextran/CFP/td Tomato+two-photon microscopy)tracked pericytes,vessels,and tumor cells,while FACSsorted Pdgfrb+cells from GBM and normal brain were analyzed by LC-MS/MS proteomics.Results:Cre-mediated ablation of Pdgfrb-expressing cells revealed stage-dependent effects on GBM growth:early ablation inhibited progression while late ablation promoted it.Pericytes undergo dual spatial reorganization in GBM:regional enrichment with pre-sprouting accumulation at the tumor-brain interface,and focal positioning with preferential localization at vascular branch points.Concurrently,GBM vasculature displayed simplified branching,dilation,and pericyte remodeling(shorter processes,higher density).Proteomics revealed 1426 altered proteins,with upregulated proliferation pathways(e.g.,matrix metallopeptidase 14[Mmp14],lysyl oxidase like 2[Loxl2])and downregulated homeostasis functions(e.g.,transforming growth factor beta 1[Tgfb1]),validated by scRNA-seq in human GBM.Conclusions:This study demonstrates that during early GBM progression,pericytes actively drive tumor angiogenesis through molecular reprogramming toward proliferative and pro-angiogenic phenotypes,with the integrated imaging-proteomics framework revealing potential therapeutic targets for disrupting pericyte-mediated vascular remodeling.展开更多
Glioblastoma(GBM)remains a major clinical challenge due to limited therapeutic success despite standard treatments including surgery,radiotherapy,and temozolomide(TMZ).Recent evidence links hyperglycemia to cancer pro...Glioblastoma(GBM)remains a major clinical challenge due to limited therapeutic success despite standard treatments including surgery,radiotherapy,and temozolomide(TMZ).Recent evidence links hyperglycemia to cancer progression,and altered glucose metabolism has emerged as a key factor in GBM development.Metformin,an antidiabetic drug,has shown promise in improving survival in GBM patients,possibly due to its ability to cross the blood-brain barrier and target metabolic pathways involved in tumor growth.Preclinical studies suggest metformin may enhance TMZ efficacy by acting on glioma stem cells and overcoming resistance mechanisms.Its activation of AMPK and modulation of Wnt signaling further support its therapeutic potential.However,while early studies and clinical trials have explored metformin’s safety and efficacy,its direct impact on GBM survival remains unclear.Ongoing research aims to clarify its mechanisms and identify responsive patient subgroups.Novel strategies,including PPARγagonists and nanoerythrosome-based drug delivery systems,are also under investigation to improve metformin’s therapeutic profile.Rigorous clinical trials and mechanistic studies are essential to determine the role of metformin as adjunct therapy in GBM treatment.展开更多
Glioblastoma multiforme(GBM)are the most aggressive and common tumors in the central nervous system.GBM are classified as grade IV according to the World Health Organization.The incidence of GBM slightly differs among...Glioblastoma multiforme(GBM)are the most aggressive and common tumors in the central nervous system.GBM are classified as grade IV according to the World Health Organization.The incidence of GBM slightly differs among countries.The etiology of GBM has not been entirely clarified.No risk factors such as smoking,chemicals or dietary can be identified for GBM.Only the exposure to high radiation dose such as radiotherapy of head and neck cancers have been reported to increase the risk of glioma tumors.In this review,the authors attempted to cover several aspects of GBM.This review was based on a collection of recent publications from different research fields but all related to GBM in order to shed the light on this disease.We highlighted the current insights of GBM in the aspects of epidemiology,pathogenesis,etiology,molecular genetics,imaging technologies,artificial intelligence and treatment.A literature review was conducted for GBM with relevant keywords.Although GBM was known since several decades,its causes are still confounding,and its early detection is often unpredictable.Since the hereditary aspect of GBM is very low,there remains as the common symptoms the interference with normal brain function,memory loss,unusual behavior,headaches and seizures.The progress in GBM treatment is not satisfactory even with the deployment of huge efforts and financial costs in many domains like gene therapy,surgery and chemoradiotherapy.Despite the rapid developments of the standard treatment for GBM,the trend of survival rate did not change among years.展开更多
Real-time,noninvasive programmed death-ligand 1(PD-L1)testing using molecular imaging has enhanced our understanding of the immune environments of neoplasms and has served as a guide for immunotherapy.However,the util...Real-time,noninvasive programmed death-ligand 1(PD-L1)testing using molecular imaging has enhanced our understanding of the immune environments of neoplasms and has served as a guide for immunotherapy.However,the utilization of radiotracers in the imaging of human brain tumors using positron emission tomography/computed tomography(PET/CT)remains limited.This investigation involved the synthesis of[18F]AlF-NOTA-PCP2,which is a novel peptide-based radiolabeled tracer that targets PD-L1,and evaluated its imaging capabilities in orthotopic glioblastoma(GBM)models.Using this tracer,we could noninvasively monitor radiation-induced PD-L1 changes in GBM.[18F]AlF-NOTA-PCP2 exhibited high radiochemical purity(>95%)and stability up to 4 h after synthesis.It demonstrated specific,high-affinity binding to PD-L1 in vitro and in vivo,with a dissociation constant of 0.24 nM.PET/CT imaging,integrated with contrast-enhanced magnetic resonance imaging,revealed significant accumulation of[18F]AlF-NOTA-PCP2 in orthotopic tumors,correlating with blood-brain barrier disruption.After radiotherapy(15 Gy),[18F]AlF-NOTA-PCP2 uptake in tumors increased from 9.51%±0.73%to 12.04%±1.43%,indicating enhanced PD-L1 expression consistent with immunohistochemistry findings.Fractionated radiation(5 Gy×3)further amplified PD-L1 upregulation(13.9%±1.54%ID/cc)compared with a single dose(11.48%±1.05%ID/cc).Taken together,[18F]AlF-NOTA-PCP2 may be a valuable tool for noninvasively monitoring PD-L1 expression in brain tumors after radiotherapy.展开更多
Cellular hitchhiking is an emerging therapeutic strategy that uses an endogenous cell migration mechanism to deliver therapeutics to specific sites in the body.Owing to the low permeability and presence of the blood-b...Cellular hitchhiking is an emerging therapeutic strategy that uses an endogenous cell migration mechanism to deliver therapeutics to specific sites in the body.Owing to the low permeability and presence of the blood-brain barrier(BBB),the targeted delivery of therapeutics is limited,leading to inadequate localization in the brain.NCs fail to extravasate significantly into the tumor microenvironment(TME),demonstrating poor accumulation and tumor penetration.The novel cellular hitchhiking concept has been utilized to promote systemic half-life and therapeutic targeting.Neoplastic and neuroinflammatory diseases of the brain,including glioblastoma and neuroinflammation,face critical hurdles for efficiently delivering therapeutic entities owing to the BBB.Cellular hitchhiking can surmount these hurdles by utilizing various cell populations,such as stem cells,monocytes/macrophages,neutrophils,and platelets,as potential functional carriers to deliver the therapeutic cargo through the BBB.These carrier cells have the innate capability to traverse the BBB,transit through the brain parenchyma,and specifically reach disease sites such as inflammatory and neoplastic lesions owing to chemotactic navigation,i.e.,movement attributed to chemical stimuli.Chemotherapeutic drugs delivered by cellular hitchhiking to achieve tumor-specific targeting have been discussed.This article explores various cell types for hitchhiking NCs to the TME with indepth mechanisms and characterization techniques to decipher the backpack dissociation dynamics(nanoparticle payload detachment characteristics from hitchhiked cells)and challenges toward prospective clinical translation.展开更多
Objective The systemic immune-inflammation index(SII)has recently attracted significant interest as a new biomarker for predicting the prognosis of patients with glioblastoma(GBM).However,the predictive significance o...Objective The systemic immune-inflammation index(SII)has recently attracted significant interest as a new biomarker for predicting the prognosis of patients with glioblastoma(GBM).However,the predictive significance of it is still a subject of debate.This study intended to assess the clinical effectiveness of the SII in GBM and establish a nomogram.Methods Receiver operating characteristic(ROC)curves were utilized to determine the optimal cut-off values of the SII.Kaplan–Meier(KM)survival curves were used to analyze the median overall survival(OS).Cox regression analysis was carried out to evaluate the associations between OS and different clinical factors.Based on the SII and clinical characteristics,a nomogram was constructed,and its value in clinical application was evaluated by means of decision curve analysis.Results The optimal SII cut-off value was 610.13.KM analysis revealed that GBM patients with higher SII values had shorter OS(15.0 vs.34.0 months,P=0.044).Multivariate analysis demonstrated that a high SII was an independent predictor of poor outcome in GBM(HR=1.79,P=0.029).The nomogram incorporating the preoperative SII showed good predictive accuracy for GBM patient prognosis(C-index=0.691).Conclusions The SII is an independent predictive indicator for GBM.Patients with elevated SII levels tend to have a poorer prognosis.A nomogram combining the SII with clinical and molecular pathological features can assist clinicians in assessing the risk of death in GBM patients,providing a basis for individualized treatment decisions.展开更多
Glioblastoma multiforme(GBM)is a highly aggressive and lethal brain tumor with limited treatment options.To improve therapeutic efficacy,we developed a novel multifunctional nanoplatform,GM@P(T/S),comprised of polymer...Glioblastoma multiforme(GBM)is a highly aggressive and lethal brain tumor with limited treatment options.To improve therapeutic efficacy,we developed a novel multifunctional nanoplatform,GM@P(T/S),comprised of polymeric nanoparticles coated with GBM cell membranes as well as co-loaded with temozolomide(TMZ)and superparamagnetic iron oxide(SPIO)nanoparticles.The successful preparation was confirmed in terms of particle size,morphology,stability,the in vitro drug release,and cellular uptake assays.We demonstrated that GM@P(T/S)exhibited the enhanced homotypic targeting,the prolonged blood circulation,and efficient bloodbrain barrier penetration in both in vitro and in vivo studies.The combination of TMZ and SPIO nanoparticles within GM@P(T/S)synergistically improved chemo-radiation therapy,leading to a reduced tumor growth,an increased survival,and minimal systemic toxicity in the orthotopic GBM mouse models.Our findings suggest that GM@P(T/S)holds a great promise as a targeted and efficient therapeutic strategy for GBM.展开更多
Background:To date,there is no effective cure for the highly malignant brain tumor glioblastoma(GBM).GBM is the most common,aggressive central nervous system tumor(CNS).It commonly originates in glial cells such as mi...Background:To date,there is no effective cure for the highly malignant brain tumor glioblastoma(GBM).GBM is the most common,aggressive central nervous system tumor(CNS).It commonly originates in glial cells such as microglia,oligodendroglia,astrocytes,or subpopulations of cancer stem cells(CSCs).Glucose plays an important role in the,which energy metabolism of normal and cancer cells,but cancer cells exhibit an increased demand for glucose is required for their differentiation and proliferation.The main aim of this study is to explore the anti-cancer efficacy of the ketogenic diet against GBM.Also,this research focuses on the identification of the catalytic action of zinc in epigenetic modulators such as oxyresveratrol and ensures the combinatorial effect in the treatment of GBM.Method:In this study,we have evaluated various parameters to understand the therapeutic efficacy of the treatment groups through in vivo experiments against aggressive brain tumors.Intracerebroventricular experiments were performed to induce the tumor in the animals and estimate the tumor burden and proliferative index.Followed by the Morris water maze,an open field test,and rota rod was performed to evaluate the memory and motor coordination.To understand the glucose,and ketone level modification before and after treatment,the level of glucose and ketone was analyzed.Moreover,the zinc level is assessed using flame atomic absorption spectroscopy.Results:The results suggested that the ketogenic diet has an anti-cancer efficacy against C6-induced GBM cell lines.Also,it exerts a synergistic effect with the epigenetic modulator,oxyresveratrol,and zinc against GBM cell lines.Moreover,the treatment groups improved memory and motor coordination and modified the glucose and ketone levels to reduce the tumor burden and Ki-67 proliferative index.Conclusion:This study revealed the therapeutic effect of the ketogenic diet along with its combination such as oxyresveratrol and zinc against the C6-induced GBM in the Wistar rats.Also,it improved memory and motor coordination and reduced tumor growth.It also modified the glucose and ketone levels in the tumor-induced animal and supported to diminish the tumor burden.展开更多
Iron metabolism is a critical factor in tumorigenesis and development. Although TP53 mutations are prevalent in glioblastoma (GBM), the mechanisms by which TP53 regulates iron metabolism remain elusive. We reveal an i...Iron metabolism is a critical factor in tumorigenesis and development. Although TP53 mutations are prevalent in glioblastoma (GBM), the mechanisms by which TP53 regulates iron metabolism remain elusive. We reveal an imbalance iron homeostasis in GBM via TCGA database analysis. TP53 mutations disrupted iron homeostasis in GBM, characterized by elevated total iron levels and reduced ferritin (FTH). The gain-of-function effect triggered by TP53 mutations upregulates itchy E3 ubiquitin-protein ligase (ITCH) protein expression in astrocytes, leading to FTH degradation and an increase in free iron levels. TP53-mut astrocytes were more tolerant to the high iron environment induced by exogenous ferric ammonium citrate (FAC), but the increase in intracellular free iron made them more sensitive to Erastin-induced ferroptosis. Interestingly, we found that Erastin combined with FAC treatment significantly increased ferroptosis. These findings provide new insights for drug development and therapeutic modalities for GBM patients with TP53 mutations from iron metabolism perspectives.展开更多
文摘OBJECT:Glioblastoma multiforme(GBM)is the most common and lethal primary brain tumor in adults.It isnearly uniformly fatal,with a median survival time of approximately l year,despite modem treatment modalities.Nevertheless,a range of survival times exists around this median.Efforts to understand why some patients livelonger or shorter than the average may provide insight into the biology of these neoplasms.The annexin VII(ANX7)gene is located on the human chromosome 10q21,a site long hypothesized to harbor tumor
基金supported by the National Key Research and Development Programs of China(Grant No.2018YFA0209700)National Natural Science Foundation of China(Grant No.22077073)+1 种基金Frontiers Science Center for New Organic Matter,Nankai University(Grant No.63181206)the Fundamental Research Funds for the Central Universities,Nankai University(Grant No.63206015)。
文摘Glioblastoma(GBM)is the most common malignant brain tumor.Although current treatment strategies,including surgery,chemotherapy,and radiotherapy,have achieved clinical effects and prolonged the survival of patients,the gradual development of resistance against current therapies has led to a high recurrence rate and treatment failure.Mechanisms underlying the development of resistance involve multiple factors,including drug efflux,DNA damage repair,glioma stem cells,and a hypoxic tumor environment,which are usually correlative and promote each other.As many potential therapeutic targets have been discovered,combination therapy that regulates multiple resistance-related molecule pathways is considered an attractive strategy.In recent years,nanomedicine has revolutionized cancer therapies with optimized accumulation,penetration,internalization,and controlled release.Blood-brain barrier(BBB)penetration efficiency is also significantly improved through modifying ligands on nanomedicine and interacting with the receptors or transporters on the BBB.Moreover,different drugs for combination therapy usually process different pharmacokinetics and biodistribution,which can be further optimized with drug delivery systems to maximize the therapeutic efficiency of combination therapies.Herein the current achievements in nanomedicine-based combination therapy for GBM are discussed.This review aimed to provide a broader understanding of resistance mechanisms and nanomedicine-based combination therapies for future research on GBM treatment.
基金supported by the National Natural Science Foundation of China(Grant Nos.82072765 to X.Q.and 82172667 to X.W.).
文摘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.
基金supported by Project of the Health Shanghai Initiative Special Fund(Medical-Sports Integration,Creating a New Model of Exercise for Health),No.JKSHZX-2022-02(to SC).
文摘Recent research has demonstrated the impact of physical activity on the prognosis of glioma patients,with evidence suggesting exercise may reduce mortality risks and aid neural regeneration.The role of the small ubiquitin-like modifier(SUMO)protein,especially post-exercise,in cancer progression,is gaining attention,as are the potential anti-cancer effects of SUMOylation.We used machine learning to create the exercise and SUMO-related gene signature(ESLRS).This signature shows how physical activity might help improve the outlook for low-grade glioma and other cancers.We demonstrated the prognostic and immunotherapeutic significance of ESLRS markers,specifically highlighting how murine double minute 2(MDM2),a component of the ESLRS,can be targeted by nutlin-3.This underscores the intricate relationship between natural compounds such as nutlin-3 and immune regulation.Using comprehensive CRISPR screening,we validated the effects of specific ESLRS genes on low-grade glioma progression.We also revealed insights into the effectiveness of Nutlin-3a as a potent MDM2 inhibitor through molecular docking and dynamic simulation.Nutlin-3a inhibited glioma cell proliferation and activated the p53 pathway.Its efficacy decreased with MDM2 overexpression,and this was reversed by Nutlin-3a or exercise.Experiments using a low-grade glioma mouse model highlighted the effect of physical activity on oxidative stress and molecular pathway regulation.Notably,both physical exercise and Nutlin-3a administration improved physical function in mice bearing tumors derived from MDM2-overexpressing cells.These results suggest the potential for Nutlin-3a,an MDM2 inhibitor,with physical exercise as a therapeutic approach for glioma management.Our research also supports the use of natural products for therapy and sheds light on the interaction of exercise,natural products,and immune regulation in cancer treatment.
基金Supported by Conselho Nacional de Desenvolvimento Científico e Tecnológico,No.307318/2023-0 and No.102035/2024-5Fundação de AmparoàPesquisa do Estado de São Paulo,No.2023/10843-7 and No 2019/21070-3Nanotechnology National Laboratory System 2.0,Ministry of Science,Technology,Innovation and Communication,No.442539/2019-3.
文摘BACKGROUND Glioblastoma multiforme(GBM)is the most aggressive and prevalent primary malignant brain tumor in adults,marked by poor prognosis and high invasiveness.Traditional GBM invasion assays,such as those involving mouse brain xenografts,are often time-consuming and limited in efficiency.In this context,stem cell-derived neural organoids(NOs)have emerged as advanced,threedimensional,human-relevant platforms that mimic the cellular architecture and microenvironment of the human brain.These models provide novel opportunities to investigate glioblastoma stem cell invasion,a critical driver of tumor progression and therapeutic resistance.AIM To evaluate studies using stem cell-derived NOs to model glioblastoma migration/invasion,focusing on methodologies,applications and therapeutic implications.METHODS We conducted a systematic review following PRISMA guidelines,searching PubMed and Scopus for studies published between March 2019 and March 2025 that investigated NOs in the context of glioblastoma invasion/migration.After screening 377 articles based on predefined inclusion and exclusion criteria,10 original research articles were selected for analysis.Extracted data were categorized into four analytical domains:(1)Tumor model formation;(2)NO characteristics;(3)NO differentiation protocols;and(4)Invasion/migration assessment methodologies.RESULTS The included studies exhibit significant methodological heterogeneity GBM model development,particularly regarding model type,cell source and culture conditions.Most studies(70%)used suspension cell models,while 30%employed spheroids,with most research focusing on patient-derived glioblastoma stem cells.NOs were predominantly generated from human induced pluripotent stem cells using both guided and unguided differentiation protocols.Confocal fluorescence microscopy was the primary method used for assessing invasion,revealing invasion depths of up to 300μm.Organoid maturity and co-culture duration influenced results,while key factors for model optimization included tumor cell density,organoid age and extracellular matrix composition.Some studies also tested therapeutic strategies such as Zika virus and microRNA modulation.Collectively,findings support the utility of NOs as effective tools for studying GBM behavior and therapeutic responses in a humanized three-dimensional context.CONCLUSION Human NOs represent promising platforms for modeling glioblastoma invasion in a humanized three-dimensional environment.However,a limited number of studies and methodological heterogeneity hinder reproducibility.Protocol standardization is essential to enhance the translational application of these models.
基金supported by the National Natural Science Foundation of China(Nos.22078201,U1908202)Liaoning&Shenyang Key Laboratory of Functional Dye and Pigment(Nos.2021JH13/10200018,21-104-0-23).
文摘D-D’-A type aza-borondipyrromethenes(aza-BODIPYs)were prepared by Suzuki cross-coupling reaction.Photothermal conversion efficiency of self-assemble aza-BODIPY-based nanoparticles(DA-azaBDP-NPs)with NIR-Ⅱ emission(λ_(em)=1065 nm)was 37.2%under near infrared(NIR)irradiation,and the outstanding cytotoxicity was triggered by coexistence of DA-azaBDP-NPs and the NIR irradiation,with the decrease of glioblastoma migration and the inhibition of glioblastoma proliferation.DA-azaBDP-NPs could promote glioblastoma autophagy and accelerate the process of cell death.The photothermal therapy(PTT)of DAazaBDP-NPs can effectively induce glioblastoma death by apoptosis under the NIR irradiation,which is highly promising to be applied in vivo experiments of brain.
文摘1.HOU J,DU Y,SHAO Y,et al.Identification,Albanol B inhibits glioblastoma progression by inducing senescence and apoptosis via the RNF6/p27 signaling axis.Phytomedicine.DOI:10.1016/j.phymed.2025.157056.(2025)[题目]Albanol B通过RNF6/p27信号轴诱导细胞衰老与凋亡,从而抑制胶质母细胞瘤进展。
基金supported by the Individual Basic Science&Engineering Research Program(NRF-2022R1A2B5B01001920)through the National Research Foundation,funded by the Ministry of Science and ICT in Korea.
文摘Herpes simplex virus thymidine kinase(HSVtk)gene therapy is a promising strategy for glioblastoma therapy.However,delivery of plasmid DNA(pDNA)encoding HSVtk into the brain by systemic administration is a challenge since pDNA can hardly penetrate the bloodbrain barrier.In this study,an exosome-membrane(EM)and polymer-based hybrid complex was developed for systemic delivery of pDNA into the brain.Histidine/arginine-linked polyamidoamine(PHR)was used as a carrier.PHR binds to pDNA by electrostatic interaction.The pDNA/PHR complex was mixed with EM and subjected to extrusion to produce pDNA/PHR-EM hybrid complex.For glioblastoma targeting,T7 peptide was attached to the pDNA/PHR-EM complex.Both pDNA/PHR-EM and T7-decorated pDNA/PHR-EM(pDNA/PHREM-T7)had a surface charge of–5 mV and a size of 280 nm.Transfection assays indicated that pDNA/PHR-EM-T7 enhanced the transfection to C6 cells compared with pDNA/PHREM.Intravenous administration of pHSVtk/PHR-EM-T7 showed that pHSVtk/PHR-EM and pHSVtk/PHR-EM-T7 delivered pHSVtk more efficiently than pHSVtk/lipofectamine and pHSVtk/PHR into glioblastoma in vivo.pHSVtk/PHR-EM-T7 had higher delivery efficiency than pHSVtk/PHR-EM.As a result,the HSVtk expression and apoptosis levels in the tumors of the pHSVtk/PHR-EM-T7 group were higher than those of the other control groups.Therefore,the pDNA/PHR-EM-T7 hybrid complex is a useful carrier for systemic delivery of pHSVtk to glioblastoma.
基金supported by the Bridging Grant from Universiti Sains Malaysia (R501LR-RND003–0000001319–0000)funding through the Fundamental Research Grant Scheme (FRGS/1/2020/TK0/USM/02/32–6171275) awarded by the Ministry of Higher Education Malaysia
文摘Current treatments for glioblastoma face challenges such as the blood-brain barrier and lack of targeted therapy,compounded by the aggressive nature,high invasiveness,and heterogeneity of the disease.Exosomes,a subtype of extracellular vesicles are emerging as promising nanocarrier drug delivery systems to address these limitations.Exosomes released by all cell types can be easily obtained and modified as delivery vehicles or therapeutic agents.A systematic review was conducted to evaluate various methods for exosome isolation,characterization,engineering or modification,drug loading and delivery efficiency,including exosome biodistribution and treatment efficacy.A search of four databases for in vitro and in vivo studies(2000–,2023)identified 6165 records,of which 23 articles were found eligible and included for analyses.Most studies applied ultracentrifugation(UC)for exosomes isolation.Cancer cell lines being the most frequently used source of exosomes,followed by stem cells.The incubation approach was predominantly utilized to modify exosomes for drug loading.In vivo analysis showed that exosome biodistribution was primarily concentrated in the brain region,peaking in the first 6 h and remained moderately high.Compared to native exosomes and untreated control groups,utilizing modified native exosomes(cargo loaded)for treating glioblastoma disease models led to more pronounced suppression of tumor growth and proliferation,enhanced stimulation of immune response and apoptosis,effective restoration of drug chemosensitivity,increased anti-tumor effect and prolonged survival rates.Modified exosomes whether through incubation,sonication,transfection,freeze-thawing or their combination,improve targeted delivery and therapeutic efficacy against glioblastoma.
基金The National Key Research and Development Program of China,Grant/Award Number:2022YFF0710700Nonprofit Central Research Institute Fund of Chinese Academy of Medical Sciences,Grant/Award Number:2023-PT180-01。
文摘Background:Glioblastoma(GBM)is a highly aggressive brain tumor characterized by aberrant angiogenesis and an immunosuppressive microenvironment.Pericytes are aberrantly recruited but their spatiotemporal roles and molecular changes remain unclear.This study investigated platelet-derived growth factor receptor beta-positive(Pdgfrb+)pericyte dynamics and reprogramming in GBM vasculature.Methods:We generated GL261-Luc and GL261-CFP glioblastoma cells via lentiviral transduction and established two transgenic models.(1)For pericyte labeling,Ai14 reporter mice was crossed with PDGFRβ-P2A-CreERT2mice for td Tomato-specific lineage tracing(PT mice).(2)For conditional ablation,we generated inducible Pdgfrb-expressing cell ablation models(PT mice was crossed with ROSA-DTA mice).An intravital imaging platform(FITC-dextran/CFP/td Tomato+two-photon microscopy)tracked pericytes,vessels,and tumor cells,while FACSsorted Pdgfrb+cells from GBM and normal brain were analyzed by LC-MS/MS proteomics.Results:Cre-mediated ablation of Pdgfrb-expressing cells revealed stage-dependent effects on GBM growth:early ablation inhibited progression while late ablation promoted it.Pericytes undergo dual spatial reorganization in GBM:regional enrichment with pre-sprouting accumulation at the tumor-brain interface,and focal positioning with preferential localization at vascular branch points.Concurrently,GBM vasculature displayed simplified branching,dilation,and pericyte remodeling(shorter processes,higher density).Proteomics revealed 1426 altered proteins,with upregulated proliferation pathways(e.g.,matrix metallopeptidase 14[Mmp14],lysyl oxidase like 2[Loxl2])and downregulated homeostasis functions(e.g.,transforming growth factor beta 1[Tgfb1]),validated by scRNA-seq in human GBM.Conclusions:This study demonstrates that during early GBM progression,pericytes actively drive tumor angiogenesis through molecular reprogramming toward proliferative and pro-angiogenic phenotypes,with the integrated imaging-proteomics framework revealing potential therapeutic targets for disrupting pericyte-mediated vascular remodeling.
文摘Glioblastoma(GBM)remains a major clinical challenge due to limited therapeutic success despite standard treatments including surgery,radiotherapy,and temozolomide(TMZ).Recent evidence links hyperglycemia to cancer progression,and altered glucose metabolism has emerged as a key factor in GBM development.Metformin,an antidiabetic drug,has shown promise in improving survival in GBM patients,possibly due to its ability to cross the blood-brain barrier and target metabolic pathways involved in tumor growth.Preclinical studies suggest metformin may enhance TMZ efficacy by acting on glioma stem cells and overcoming resistance mechanisms.Its activation of AMPK and modulation of Wnt signaling further support its therapeutic potential.However,while early studies and clinical trials have explored metformin’s safety and efficacy,its direct impact on GBM survival remains unclear.Ongoing research aims to clarify its mechanisms and identify responsive patient subgroups.Novel strategies,including PPARγagonists and nanoerythrosome-based drug delivery systems,are also under investigation to improve metformin’s therapeutic profile.Rigorous clinical trials and mechanistic studies are essential to determine the role of metformin as adjunct therapy in GBM treatment.
文摘Glioblastoma multiforme(GBM)are the most aggressive and common tumors in the central nervous system.GBM are classified as grade IV according to the World Health Organization.The incidence of GBM slightly differs among countries.The etiology of GBM has not been entirely clarified.No risk factors such as smoking,chemicals or dietary can be identified for GBM.Only the exposure to high radiation dose such as radiotherapy of head and neck cancers have been reported to increase the risk of glioma tumors.In this review,the authors attempted to cover several aspects of GBM.This review was based on a collection of recent publications from different research fields but all related to GBM in order to shed the light on this disease.We highlighted the current insights of GBM in the aspects of epidemiology,pathogenesis,etiology,molecular genetics,imaging technologies,artificial intelligence and treatment.A literature review was conducted for GBM with relevant keywords.Although GBM was known since several decades,its causes are still confounding,and its early detection is often unpredictable.Since the hereditary aspect of GBM is very low,there remains as the common symptoms the interference with normal brain function,memory loss,unusual behavior,headaches and seizures.The progress in GBM treatment is not satisfactory even with the deployment of huge efforts and financial costs in many domains like gene therapy,surgery and chemoradiotherapy.Despite the rapid developments of the standard treatment for GBM,the trend of survival rate did not change among years.
基金support from the National Natural Science Foundation of China(Grant Nos.:82272751 and 82202958)the Natural Science Foundation of Shandong,China(Grant No.:ZR2021LSW002)+6 种基金the Science and Technology Program of Jinan,China(Grant Nos.:202225019 and 202225013)to Man Huthe Shandong Postdoctoral Innovation Program,China(Grant No.:SDCX-ZG-202302011)Beijing Science and Technology Innovation Medical Development Foundation,China(Grant No.:KC2023-JX-0288-BQ26)to Yong Wangthe Natural Science Foundation of China(Grant No.:NSFC82303676)the Natural Science Foundation of Shandong(Grant No.:ZR2023QH208)the China Postdoctoral Science Foundation(Grant No.:2023M732125)the Taishan Scholar Project Special Fund(Grant No.:tsqn202312368)to Kewen He.
文摘Real-time,noninvasive programmed death-ligand 1(PD-L1)testing using molecular imaging has enhanced our understanding of the immune environments of neoplasms and has served as a guide for immunotherapy.However,the utilization of radiotracers in the imaging of human brain tumors using positron emission tomography/computed tomography(PET/CT)remains limited.This investigation involved the synthesis of[18F]AlF-NOTA-PCP2,which is a novel peptide-based radiolabeled tracer that targets PD-L1,and evaluated its imaging capabilities in orthotopic glioblastoma(GBM)models.Using this tracer,we could noninvasively monitor radiation-induced PD-L1 changes in GBM.[18F]AlF-NOTA-PCP2 exhibited high radiochemical purity(>95%)and stability up to 4 h after synthesis.It demonstrated specific,high-affinity binding to PD-L1 in vitro and in vivo,with a dissociation constant of 0.24 nM.PET/CT imaging,integrated with contrast-enhanced magnetic resonance imaging,revealed significant accumulation of[18F]AlF-NOTA-PCP2 in orthotopic tumors,correlating with blood-brain barrier disruption.After radiotherapy(15 Gy),[18F]AlF-NOTA-PCP2 uptake in tumors increased from 9.51%±0.73%to 12.04%±1.43%,indicating enhanced PD-L1 expression consistent with immunohistochemistry findings.Fractionated radiation(5 Gy×3)further amplified PD-L1 upregulation(13.9%±1.54%ID/cc)compared with a single dose(11.48%±1.05%ID/cc).Taken together,[18F]AlF-NOTA-PCP2 may be a valuable tool for noninvasively monitoring PD-L1 expression in brain tumors after radiotherapy.
基金the research funding support by the Department of Pharmaceuticals(DoP),Ministry of Chemicals and Fertilizers,Govt.of India to“Pharmaceutical Innovation and Translational Research Lab”(PITRL),Department of Pharmaceutics,National Institute of Pharmaceutical Education and Research(NIPER)Hyderabad,INDIA.
文摘Cellular hitchhiking is an emerging therapeutic strategy that uses an endogenous cell migration mechanism to deliver therapeutics to specific sites in the body.Owing to the low permeability and presence of the blood-brain barrier(BBB),the targeted delivery of therapeutics is limited,leading to inadequate localization in the brain.NCs fail to extravasate significantly into the tumor microenvironment(TME),demonstrating poor accumulation and tumor penetration.The novel cellular hitchhiking concept has been utilized to promote systemic half-life and therapeutic targeting.Neoplastic and neuroinflammatory diseases of the brain,including glioblastoma and neuroinflammation,face critical hurdles for efficiently delivering therapeutic entities owing to the BBB.Cellular hitchhiking can surmount these hurdles by utilizing various cell populations,such as stem cells,monocytes/macrophages,neutrophils,and platelets,as potential functional carriers to deliver the therapeutic cargo through the BBB.These carrier cells have the innate capability to traverse the BBB,transit through the brain parenchyma,and specifically reach disease sites such as inflammatory and neoplastic lesions owing to chemotactic navigation,i.e.,movement attributed to chemical stimuli.Chemotherapeutic drugs delivered by cellular hitchhiking to achieve tumor-specific targeting have been discussed.This article explores various cell types for hitchhiking NCs to the TME with indepth mechanisms and characterization techniques to decipher the backpack dissociation dynamics(nanoparticle payload detachment characteristics from hitchhiked cells)and challenges toward prospective clinical translation.
基金funded by National Natural Science Foundation of China,grant number 82203007.
文摘Objective The systemic immune-inflammation index(SII)has recently attracted significant interest as a new biomarker for predicting the prognosis of patients with glioblastoma(GBM).However,the predictive significance of it is still a subject of debate.This study intended to assess the clinical effectiveness of the SII in GBM and establish a nomogram.Methods Receiver operating characteristic(ROC)curves were utilized to determine the optimal cut-off values of the SII.Kaplan–Meier(KM)survival curves were used to analyze the median overall survival(OS).Cox regression analysis was carried out to evaluate the associations between OS and different clinical factors.Based on the SII and clinical characteristics,a nomogram was constructed,and its value in clinical application was evaluated by means of decision curve analysis.Results The optimal SII cut-off value was 610.13.KM analysis revealed that GBM patients with higher SII values had shorter OS(15.0 vs.34.0 months,P=0.044).Multivariate analysis demonstrated that a high SII was an independent predictor of poor outcome in GBM(HR=1.79,P=0.029).The nomogram incorporating the preoperative SII showed good predictive accuracy for GBM patient prognosis(C-index=0.691).Conclusions The SII is an independent predictive indicator for GBM.Patients with elevated SII levels tend to have a poorer prognosis.A nomogram combining the SII with clinical and molecular pathological features can assist clinicians in assessing the risk of death in GBM patients,providing a basis for individualized treatment decisions.
基金supported by the National Natural Science Foundation of China(Grant Nos.82073308 and 82104089)。
文摘Glioblastoma multiforme(GBM)is a highly aggressive and lethal brain tumor with limited treatment options.To improve therapeutic efficacy,we developed a novel multifunctional nanoplatform,GM@P(T/S),comprised of polymeric nanoparticles coated with GBM cell membranes as well as co-loaded with temozolomide(TMZ)and superparamagnetic iron oxide(SPIO)nanoparticles.The successful preparation was confirmed in terms of particle size,morphology,stability,the in vitro drug release,and cellular uptake assays.We demonstrated that GM@P(T/S)exhibited the enhanced homotypic targeting,the prolonged blood circulation,and efficient bloodbrain barrier penetration in both in vitro and in vivo studies.The combination of TMZ and SPIO nanoparticles within GM@P(T/S)synergistically improved chemo-radiation therapy,leading to a reduced tumor growth,an increased survival,and minimal systemic toxicity in the orthotopic GBM mouse models.Our findings suggest that GM@P(T/S)holds a great promise as a targeted and efficient therapeutic strategy for GBM.
文摘Background:To date,there is no effective cure for the highly malignant brain tumor glioblastoma(GBM).GBM is the most common,aggressive central nervous system tumor(CNS).It commonly originates in glial cells such as microglia,oligodendroglia,astrocytes,or subpopulations of cancer stem cells(CSCs).Glucose plays an important role in the,which energy metabolism of normal and cancer cells,but cancer cells exhibit an increased demand for glucose is required for their differentiation and proliferation.The main aim of this study is to explore the anti-cancer efficacy of the ketogenic diet against GBM.Also,this research focuses on the identification of the catalytic action of zinc in epigenetic modulators such as oxyresveratrol and ensures the combinatorial effect in the treatment of GBM.Method:In this study,we have evaluated various parameters to understand the therapeutic efficacy of the treatment groups through in vivo experiments against aggressive brain tumors.Intracerebroventricular experiments were performed to induce the tumor in the animals and estimate the tumor burden and proliferative index.Followed by the Morris water maze,an open field test,and rota rod was performed to evaluate the memory and motor coordination.To understand the glucose,and ketone level modification before and after treatment,the level of glucose and ketone was analyzed.Moreover,the zinc level is assessed using flame atomic absorption spectroscopy.Results:The results suggested that the ketogenic diet has an anti-cancer efficacy against C6-induced GBM cell lines.Also,it exerts a synergistic effect with the epigenetic modulator,oxyresveratrol,and zinc against GBM cell lines.Moreover,the treatment groups improved memory and motor coordination and modified the glucose and ketone levels to reduce the tumor burden and Ki-67 proliferative index.Conclusion:This study revealed the therapeutic effect of the ketogenic diet along with its combination such as oxyresveratrol and zinc against the C6-induced GBM in the Wistar rats.Also,it improved memory and motor coordination and reduced tumor growth.It also modified the glucose and ketone levels in the tumor-induced animal and supported to diminish the tumor burden.
基金supported by the National Natural Science Foundation of China(32171131,32371013,82071429,82471274,and 32371181)Shandong Province Natural Science Foundation(2021ZDSYS11,ZR2019ZD31,and ZR2022MC098)the Taishan Scholars Construction Project.
文摘Iron metabolism is a critical factor in tumorigenesis and development. Although TP53 mutations are prevalent in glioblastoma (GBM), the mechanisms by which TP53 regulates iron metabolism remain elusive. We reveal an imbalance iron homeostasis in GBM via TCGA database analysis. TP53 mutations disrupted iron homeostasis in GBM, characterized by elevated total iron levels and reduced ferritin (FTH). The gain-of-function effect triggered by TP53 mutations upregulates itchy E3 ubiquitin-protein ligase (ITCH) protein expression in astrocytes, leading to FTH degradation and an increase in free iron levels. TP53-mut astrocytes were more tolerant to the high iron environment induced by exogenous ferric ammonium citrate (FAC), but the increase in intracellular free iron made them more sensitive to Erastin-induced ferroptosis. Interestingly, we found that Erastin combined with FAC treatment significantly increased ferroptosis. These findings provide new insights for drug development and therapeutic modalities for GBM patients with TP53 mutations from iron metabolism perspectives.