Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 34.730
2.
Cell Death Dis ; 15(6): 414, 2024 Jun 13.
Article En | MEDLINE | ID: mdl-38871731

The repurposing of medications developed for central nervous system (CNS) disorders, possessing favorable safety profiles and blood-brain barrier permeability, represents a promising strategy for identifying new therapies to combat glioblastoma (GBM). In this study, we investigated the anti-GBM activity of specific antipsychotics and antidepressants in vitro and in vivo. Our results demonstrate that these compounds share a common mechanism of action in GBM, disrupting lysosomal function and subsequently inducing lysosomal membrane rupture and cell death. Notably, PTEN intact GBMs possess an increased sensitivity to these compounds. The inhibition of lysosomal function synergized with inhibitors targeting the EGFR-PI3K-Akt pathway, leading to an energetic and antioxidant collapse. These findings provide a foundation for the potential clinical application of CNS drugs in GBM treatment. Additionally, this work offers critical insights into the mechanisms and determinants of cytotoxicity for drugs currently undergoing clinical trials as repurposing agents for various cancers, including Fluoxetine, Sertraline, Thioridazine, Chlorpromazine, and Fluphenazine.


Antipsychotic Agents , Glioblastoma , Lysosomes , PTEN Phosphohydrolase , Signal Transduction , Humans , Glioblastoma/drug therapy , Glioblastoma/pathology , Glioblastoma/metabolism , PTEN Phosphohydrolase/metabolism , Lysosomes/metabolism , Lysosomes/drug effects , Signal Transduction/drug effects , Antipsychotic Agents/pharmacology , Antipsychotic Agents/therapeutic use , Animals , Cell Line, Tumor , Mice , Brain Neoplasms/drug therapy , Brain Neoplasms/pathology , Brain Neoplasms/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Mice, Nude , Drug Repositioning , Phosphatidylinositol 3-Kinases/metabolism , ErbB Receptors/metabolism , ErbB Receptors/antagonists & inhibitors , Chlorpromazine/pharmacology
3.
BMC Med Educ ; 24(1): 632, 2024 Jun 06.
Article En | MEDLINE | ID: mdl-38844925

BACKGROUND: This study aims to investigate the benefits of employing a Physical Lifelike Brain (PLB) simulator for training medical students in performing craniotomy for glioblastoma removal and decompressive craniectomy. METHODS: This prospective study included 30 medical clerks (fifth and sixth years in medical school) at a medical university. Before participating in the innovative lesson, all students had completed a standard gross anatomy course as part of their curriculum. The innovative lesson involved PLB Simulator training, after which participants completed the Learning Satisfaction/Confidence Perception Questionnaire and some received qualitative interviews. RESULTS: The average score of students' overall satisfaction with the innovative lesson was 4.71 out of a maximum of 5 (SD = 0.34). After the lesson, students' confidence perception level improved significantly (t = 9.38, p < 0.001, effect size = 1.48), and the average score improved from 2,15 (SD = 1.02) to 3.59 (SD = 0.93). 60% of the students thought that the innovative lesson extremely helped them understand the knowledge of surgical neuroanatomy more, 70% believed it extremely helped them improve their skills in burr hole, and 63% thought it was extremely helpful in improving the patient complications of craniotomy with the removal of glioblastoma and decompressive craniectomy after completing the gross anatomy course. CONCLUSION: This innovative lesson with the PLB simulator successfully improved students' craniotomy knowledge and skills.


Brain Neoplasms , Clinical Competence , Decompressive Craniectomy , Glioblastoma , Simulation Training , Students, Medical , Humans , Glioblastoma/surgery , Prospective Studies , Decompressive Craniectomy/education , Brain Neoplasms/surgery , Male , Female , Education, Medical, Undergraduate/methods , Craniotomy/education , Curriculum
4.
J Transl Med ; 22(1): 540, 2024 Jun 06.
Article En | MEDLINE | ID: mdl-38844944

The adaptability of glioblastoma (GBM) cells, encouraged by complex interactions with the tumour microenvironment (TME), currently renders GBM an incurable cancer. Despite intensive research, with many clinical trials, GBM patients rely on standard treatments including surgery followed by radiation and chemotherapy, which have been observed to induce a more aggressive phenotype in recurrent tumours. This failure to improve treatments is undoubtedly a result of insufficient models which fail to incorporate components of the human brain TME. Research has increasingly uncovered mechanisms of tumour-TME interactions that correlate to worsened patient prognoses, including tumour-associated astrocyte mitochondrial transfer, neuronal circuit remodelling and immunosuppression. This tumour hijacked TME is highly implicated in driving therapy resistance, with further alterations within the TME and tumour resulting from therapy exposure inducing increased tumour growth and invasion. Recent developments improving organoid models, including aspects of the TME, are paving an exciting future for the research and drug development for GBM, with the hopes of improving patient survival growing closer. This review focuses on GBMs interactions with the TME and their effect on tumour pathology and treatment efficiency, with a look at challenges GBM models face in sufficiently recapitulating this complex and highly adaptive cancer.


Brain Neoplasms , Drug Resistance, Neoplasm , Glioblastoma , Neoplasm Recurrence, Local , Tumor Microenvironment , Humans , Glioblastoma/pathology , Glioblastoma/therapy , Neoplasm Recurrence, Local/pathology , Brain Neoplasms/pathology , Brain Neoplasms/therapy , Animals
5.
J Cancer Res Clin Oncol ; 150(6): 292, 2024 Jun 06.
Article En | MEDLINE | ID: mdl-38842611

BACKGROUND: Glioblastoma (GBM) is a highly aggressive and prevalent brain tumor that poses significant challenges in treatment. SRSF9, an RNA-binding protein, is essential for cellular processes and implicated in cancer progression. Yet, its function and mechanism in GBM need clarification. METHODS: Bioinformatics analysis was performed to explore differential expression of SRSF9 in GBM and its prognostic relevance to glioma patients. SRSF9 and CDK1 expression in GBM cell lines and patients' tissues were quantified by RT-qPCR, Western blot or immunofluorescence assay. The role of SRSF9 in GBM cell proliferation and migration was assessed by MTT, Transwell and colony formation assays. Additionally, transcriptional regulation of CDK1 by SRSF9 was investigated using ChIP-PCR and dual-luciferase assays. RESULTS: The elevated SRSF9 expression correlates to GBM stages and poor survival of glioma patients. Through gain-of-function and loss-of-function strategies, SRSF9 was demonstrated to promote proliferation and migration of GBM cells. Bioinformatics analysis showed that SRSF9 has an impact on cell growth pathways including cell cycle checkpoints and E2F targets. Mechanistically, SRSF9 appears to bind to the promoter of CDK1 gene and increase its transcription level, thus promoting GBM cell proliferation. CONCLUSIONS: These findings uncover the cellular function of SRSF9 in GBM and highlight its therapeutic potential for GBM.


Brain Neoplasms , CDC2 Protein Kinase , Cell Movement , Cell Proliferation , Glioblastoma , Serine-Arginine Splicing Factors , Humans , Glioblastoma/pathology , Glioblastoma/genetics , Glioblastoma/metabolism , CDC2 Protein Kinase/metabolism , CDC2 Protein Kinase/genetics , Serine-Arginine Splicing Factors/metabolism , Serine-Arginine Splicing Factors/genetics , Brain Neoplasms/pathology , Brain Neoplasms/genetics , Brain Neoplasms/metabolism , Gene Expression Regulation, Neoplastic , Cell Line, Tumor , Prognosis , Female , Male , Middle Aged
6.
Mol Biol Rep ; 51(1): 723, 2024 Jun 04.
Article En | MEDLINE | ID: mdl-38833199

BACKGROUND: Glioblastoma multiforme, a deadly form of brain tumor, is characterized by aggressive growth and poor prognosis. Oxidative stress, a disruption in the balance between antioxidants and oxidants, is a crucial factor in its pathogenesis. Silymarin, a flavonoid extracted from milk thistle, has shown therapeutic potential in inhibiting cancer cell growth, promoting apoptosis, and reducing inflammation. It also regulates oxidative stress. This study aims to investigate the regulatory effects of silymarin on oxidative stress parameters, especially the transcription factor Nrf2 and its related enzymes in GBM cancer cells, to develop a new anti-cancer compound with low toxicity. METHODS AND RESULTS: First, the cytotoxicity of silymarin on U-87 MG cells was investigated by MTT and the results showed an IC50 of 264.6 µM. Then, some parameters of the redox system were measured with commercial kits, and the obtained results showed that silymarin increased the activity of catalase and superoxide dismutase enzymes, as well as the total antioxidant capacity levels; while the malondialdehyde level that is an indicator of lipid peroxidation was decreased by this compound. The expression level of Nrf2 and HO-1 and glutaredoxin and thioredoxin enzymes were checked by real-time PCR method, and the expression level increased significantly after treatment. CONCLUSIONS: Our findings suggest that silymarin may exert its cytotoxic and anticancer effects by enhancing the Nrf2/HO-1 pathway through antioxidant mechanisms in U-87 MG cells.


Antioxidants , Glioblastoma , NF-E2-Related Factor 2 , Oxidation-Reduction , Oxidative Stress , Silymarin , Silymarin/pharmacology , Humans , Glioblastoma/drug therapy , Glioblastoma/metabolism , Glioblastoma/pathology , Cell Line, Tumor , Oxidation-Reduction/drug effects , NF-E2-Related Factor 2/metabolism , Oxidative Stress/drug effects , Antioxidants/pharmacology , Superoxide Dismutase/metabolism , Lipid Peroxidation/drug effects , Cell Survival/drug effects , Apoptosis/drug effects , Brain Neoplasms/drug therapy , Brain Neoplasms/metabolism , Brain Neoplasms/pathology , Catalase/metabolism , Catalase/genetics
7.
Bull Math Biol ; 86(7): 83, 2024 Jun 06.
Article En | MEDLINE | ID: mdl-38842602

5-Aminolevulinic Acid (5-ALA) is the only fluorophore approved by the FDA as an intraoperative optical imaging agent for fluorescence-guided surgery in patients with glioblastoma. The dosing regimen is based on rodent tests where a maximum signal occurs around 6 h after drug administration. Here, we construct a computational framework to simulate the transport of 5-ALA through the stomach, blood, and brain, and the subsequent conversion to the fluorescent agent protoporphyrin IX at the tumor site. The framework combines compartmental models with spatially-resolved partial differential equations, enabling one to address questions regarding quantity and timing of 5-ALA administration before surgery. Numerical tests in two spatial dimensions indicate that, for tumors exceeding the detection threshold, the time to peak fluorescent concentration is 2-7 h, broadly consistent with the current surgical guidelines. Moreover, the framework enables one to examine the specific effects of tumor size and location on the required dose and timing of 5-ALA administration before glioblastoma surgery.


Aminolevulinic Acid , Brain Neoplasms , Computer Simulation , Glioblastoma , Mathematical Concepts , Models, Biological , Protoporphyrins , Surgery, Computer-Assisted , Glioblastoma/surgery , Glioblastoma/drug therapy , Glioblastoma/pathology , Glioblastoma/diagnostic imaging , Aminolevulinic Acid/administration & dosage , Humans , Brain Neoplasms/surgery , Protoporphyrins/administration & dosage , Protoporphyrins/metabolism , Surgery, Computer-Assisted/methods , Animals , Photosensitizing Agents/administration & dosage , Optical Imaging/methods , Fluorescent Dyes/administration & dosage
8.
Lancet Neurol ; 23(7): 740-748, 2024 Jul.
Article En | MEDLINE | ID: mdl-38876751

Despite substantial advances in cancer treatment, for patients with glioblastoma prognosis remains bleak. The emerging field of cancer neuroscience reveals intricate functional interplays between glioblastoma and the cellular architecture of the brain, encompassing neurons, glia, and vessels. New findings underscore the role of structural and functional connections within hierarchical networks, known as the connectome. These connections contribute to the location, spread, and recurrence of a glioblastoma, and a patient's overall survival, revealing a complex interplay between the tumour and the CNS. This mounting evidence prompts a paradigm shift, challenging the perception of glioblastomas as mere foreign bodies within the brain. Instead, these tumours are intricately woven into the structural and functional fabric of the brain. This radical change in thinking holds profound implications for the understanding and treatment of glioblastomas, which could unveil new prognostic factors and surgical strategies and optimise radiotherapy. Additionally, a connectivity approach suggests that non-invasive brain stimulation could disrupt pathological neuron-glioma interactions within specific networks.


Brain Neoplasms , Brain , Connectome , Glioblastoma , Humans , Glioblastoma/therapy , Glioblastoma/pathology , Brain Neoplasms/therapy , Brain Neoplasms/pathology , Brain Neoplasms/physiopathology , Brain/pathology , Brain/physiopathology , Nerve Net/physiopathology , Nerve Net/pathology
9.
BMC Cancer ; 24(1): 736, 2024 Jun 15.
Article En | MEDLINE | ID: mdl-38879476

BACKGROUND: Glioblastoma (GBM) is the most common and aggressive primary brain cancer. The treatment of GBM consists of a combination of surgery and subsequent oncological therapy, i.e., radiotherapy, chemotherapy, or their combination. If postoperative oncological therapy involves irradiation, magnetic resonance imaging (MRI) is used for radiotherapy treatment planning. Unfortunately, in some cases, a very early worsening (progression) or return (recurrence) of the disease is observed several weeks after the surgery and is called rapid early progression (REP). Radiotherapy planning is currently based on MRI for target volumes definitions in many radiotherapy facilities. However, patients with REP may benefit from targeting radiotherapy with other imaging modalities. The purpose of the presented clinical trial is to evaluate the utility of 11C-methionine in optimizing radiotherapy for glioblastoma patients with REP. METHODS: This study is a nonrandomized, open-label, parallel-setting, prospective, monocentric clinical trial. The main aim of this study was to refine the diagnosis in patients with GBM with REP and to optimize subsequent radiotherapy planning. Glioblastoma patients who develop REP within approximately 6 weeks after surgery will undergo 11C-methionine positron emission tomography (PET/CT) examinations. Target volumes for radiotherapy are defined using both standard planning T1-weighted contrast-enhanced MRI and PET/CT. The primary outcome is progression-free survival defined using RANO criteria and compared to a historical cohort with REP treated without PET/CT optimization of radiotherapy. DISCUSSION: PET is one of the most modern methods of molecular imaging. 11C-Methionine is an example of a radiolabelled (carbon 11) amino acid commonly used in the diagnosis of brain tumors and in the evaluation of response to treatment. Optimized radiotherapy may also have the potential to cover those regions with a high risk of subsequent progression, which would not be identified using standard-of-care MRI for radiotherapy planning. This is one of the first study focused on radiotherapy optimization for subgroup of patinets with REP. TRIAL REGISTRATION: NCT05608395, registered on 8.11.2022 in clinicaltrials.gov; EudraCT Number: 2020-000640-64, registered on 26.5.2020 in clinicaltrialsregister.eu. Protocol ID: MOU-2020-01, version 3.2, date 18.09.2020.


Brain Neoplasms , Disease Progression , Glioblastoma , Methionine , Humans , Glioblastoma/diagnostic imaging , Glioblastoma/therapy , Glioblastoma/diagnosis , Glioblastoma/radiotherapy , Brain Neoplasms/diagnostic imaging , Brain Neoplasms/therapy , Brain Neoplasms/radiotherapy , Brain Neoplasms/diagnosis , Prospective Studies , Positron Emission Tomography Computed Tomography/methods , Carbon Radioisotopes , Magnetic Resonance Imaging/methods , Male , Female , Adult , Middle Aged , Radiopharmaceuticals/therapeutic use , Radiotherapy Planning, Computer-Assisted/methods , Aged
10.
Neoplasia ; 54: 101008, 2024 Aug.
Article En | MEDLINE | ID: mdl-38823209

Successful treatment of glioblastoma multiforme (GBM), an aggressive form of primary brain neoplasm, mandates the need to develop new therapeutic strategies. In this study, we investigated the potential of PBI-05204 in targeting GBM stem cells (GSCs) and the underlying mechanisms. Treatment with PBI-05204 significantly reduced both the number and size of tumor spheres derived from patient-derived GSCs (GBM9, GSC28 and TS543), and suppressed the tumorigenesis of GBM9 xenografts. Moreover, PBI-05204 treatment led to a significant decrease in the expression of CD44 and NANOG, crucial markers of progenitor stem cells, in GBM9 and GSC28 GSCs. This treatment also down-regulated GRP78 expression in both GSC types. Knocking down GRP78 expression through GRP78 siRNA transfection in GBM9 and GSC28 GSCs also resulted in reduced spheroid size and CD44 expression. Combining PBI-05204 with GRP78 siRNA further decreased spheroid numbers compared to GRP78 siRNA treatment alone. PBI-05204 treatment led to increased expression of pRIP1K and pRIP3K, along with enhanced binding of RIPK1/RIPK3 in GBM9 and GSC28 cells, resembling the effects observed in GRP78-silenced GSCs, suggesting that PBI-05204 induced necroptosis in these cells. Furthermore, oleandrin, a principle active cardiac glycoside component of PBI-05204, showed the ability to inhibit the self-renewal capacity in GSCs. These findings highlight the potential of PBI-05204 as a promising candidate for the development of novel therapies that target GBM stem cells.


Endoplasmic Reticulum Chaperone BiP , Glioblastoma , Heat-Shock Proteins , Neoplastic Stem Cells , Xenograft Model Antitumor Assays , Humans , Glioblastoma/pathology , Glioblastoma/drug therapy , Glioblastoma/metabolism , Glioblastoma/genetics , Neoplastic Stem Cells/drug effects , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology , Animals , Mice , Heat-Shock Proteins/metabolism , Heat-Shock Proteins/genetics , Cell Line, Tumor , Plant Extracts/pharmacology , Necroptosis/drug effects , Brain Neoplasms/pathology , Brain Neoplasms/drug therapy , Brain Neoplasms/metabolism , Brain Neoplasms/genetics , Cell Proliferation/drug effects , Apoptosis/drug effects , Disease Models, Animal , Hyaluronan Receptors/metabolism , Hyaluronan Receptors/genetics
11.
Endocr Regul ; 58(1): 144-152, 2024 Jan 01.
Article En | MEDLINE | ID: mdl-38861539

Objective. Serine hydroxymethyltransferase (SHMT2) plays a multifunctional role in mitochondria (folate-dependent tRNA methylation, translation, and thymidylate synthesis). The endoplasmic reticulum stress, hypoxia, and glucose and glutamine supply are significant factors of malignant tumor growth including glioblastoma. Previous studies have shown that the knockdown of the endoplasmic reticulum to nucleus signaling 1 (ERN1) pathway of endoplasmic reticulum stress strongly suppressed glioblastoma cell proliferation and modified the sensitivity of these cells to hypoxia and glucose or glutamine deprivations. The present study aimed to investigate the regulation of the SHMT2 gene in U87MG glioblastoma cells by ERN1 knockdown, hypoxia, and glucose or glutamine deprivations with the intent to reveal the role of ERN1 signaling in sensitivity of this gene expression to hypoxia and nutrient supply. Methods. The control U87MG glioblastoma cells (transfected by an empty vector) and ERN1 knockdown cells with inhibited ERN1 endoribonuclease and protein kinase (dnERN1) or only ERN1 endoribonuclease (dnrERN1) were used. Hypoxia was introduced by dimethyloxalylglycine (500 ng/ml for 4 h). For glucose and glutamine deprivations, cells were exposed in DMEM without glucose and glutamine, respectively for 16 h. RNA was extracted from cells and reverse transcribed. The expression level of the SHMT2 gene was studied by real-time qPCR and normalized to ACTB. Results. It was found that inhibition of ERN1 endoribonuclease and protein kinase in glioblastoma cells led to a down-regulation of SHMT2 gene expression in U87MG cells. At the same time, the expression of this gene did not significantly change in cells with inhibited ERN1 endoribonuclease, but tunicamycin strongly increased its expression. Moreover, the expression of the SHMT2 gene was not affected in U87MG cells after silencing of XBP1. Hypoxia up-regulated the expression level of the SHMT2 gene in both control and ERN1 knockdown U87MG cells. The expression of this gene was significantly up-regulated in glioblastoma cells under glucose and glutamine deprivations and ERN1 knockdown significantly increased the sensitivity of the SHMT2 gene to these nutrient deprivation conditions. Conclusion. The results of the present study demonstrate that the expression of the SHMT2 gene responsible for serine metabolism and formation of folate one-carbon is controlled by ERN1 protein kinase and induced by hypoxia as well as glutamine and glucose deprivation conditions in glioblastoma cells and reflects the ERN1-mediated reprogramming of sensitivity this gene expression to nutrient deprivation.


Endoplasmic Reticulum Stress , Endoribonucleases , Gene Expression Regulation, Neoplastic , Glioblastoma , Glycine Hydroxymethyltransferase , Humans , Glycine Hydroxymethyltransferase/genetics , Glycine Hydroxymethyltransferase/metabolism , Glioblastoma/genetics , Glioblastoma/metabolism , Glioblastoma/pathology , Endoplasmic Reticulum Stress/physiology , Endoplasmic Reticulum Stress/genetics , Cell Line, Tumor , Endoribonucleases/genetics , Endoribonucleases/metabolism , Glucose/metabolism , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Brain Neoplasms/genetics , Brain Neoplasms/metabolism , Brain Neoplasms/pathology , Cell Hypoxia/physiology , Cell Hypoxia/genetics , Glutamine/metabolism , Gene Knockdown Techniques
12.
Cancer Cell ; 42(6): 934-936, 2024 Jun 10.
Article En | MEDLINE | ID: mdl-38861929

In this issue of Cancer Cell, Zhong et al. explore the dual role of TREM2 in glioblastoma-associated myeloid cells, demonstrating its function in promoting inflammation at the tumor-neural interface and suppression within the tumor core, influenced by the local microenvironment. These findings open up promising prospects for advancements in neuro-oncological immunotherapy.


Glioblastoma , Membrane Glycoproteins , Myeloid Cells , Tumor Microenvironment , Humans , Tumor Microenvironment/immunology , Myeloid Cells/immunology , Myeloid Cells/pathology , Myeloid Cells/metabolism , Membrane Glycoproteins/metabolism , Glioblastoma/pathology , Glioblastoma/immunology , Glioblastoma/metabolism , Receptors, Immunologic/metabolism , Animals , Brain Neoplasms/pathology , Brain Neoplasms/immunology , Brain Neoplasms/metabolism , Neurons/metabolism , Neurons/pathology
13.
PLoS One ; 19(6): e0305042, 2024.
Article En | MEDLINE | ID: mdl-38861512

Multiple sclerosis (MS) and glioblastoma (GBM) are CNS diseases in whose development and progression immune privilege is intimately important, but in a relatively opposite manner. Maintenance and strengthening of immune privilege have been shown to be an important mechanism in glioblastoma immune evasion, while the breakdown of immune privilege leads to MS initiation and exacerbation. We hypothesize that molecular signaling pathways can be oppositely regulated in peripheral blood CD8+ T cells of MS and glioblastoma patients at a transcriptional level. We analyzed publicly available data of the peripheral blood CD8+ T cell MS vs. control (MSvsCTRL) and GBM vs. control (GBMvsCTRL) differentially expressed gene (DEG) contrasts with Qiagen's Ingenuity pathway analysis software (IPA). We have identified sphingolipid signaling pathway which was significantly downregulated in the GBMvsCTRL and upregulated in the MSvsCTRL. As the pathway is important for the CD8+ T lymphocytes CNS infiltration, this result is in line with our previously stated hypothesis. Comparing publicly available lists of differentially expressed serum exosomal miRNAs from MSvsCTRL and GBMvsCTRL contrasts, we have identified that hsa-miR-182-5p has the greatest potential effect on sphingolipid signaling regarding the number of regulated DEGs in the GBMvsCTRL contrast, while not being able to find any relevant potential sphingolipid signaling target transcripts in the MSvsCTRL contrast. We conclude that the sphingolipid signaling pathway is a top oppositely regulated pathway in peripheral blood CD8+ T cells from GBM and MS, and might be crucial for the differences in CNS immune privilege maintenance of investigated diseases, but further experimental research is necessary.


CD8-Positive T-Lymphocytes , Glioblastoma , MicroRNAs , Multiple Sclerosis , Signal Transduction , Sphingolipids , Transcriptome , Humans , Glioblastoma/immunology , Glioblastoma/genetics , Glioblastoma/blood , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Sphingolipids/blood , Sphingolipids/metabolism , Multiple Sclerosis/immunology , Multiple Sclerosis/blood , Multiple Sclerosis/genetics , MicroRNAs/genetics , MicroRNAs/blood , Gene Expression Profiling , Brain Neoplasms/immunology , Brain Neoplasms/genetics , Brain Neoplasms/blood , Brain Neoplasms/pathology , Gene Expression Regulation, Neoplastic
14.
PLoS One ; 19(6): e0304149, 2024.
Article En | MEDLINE | ID: mdl-38848430

Glioblastoma, the most aggressive form of brain cancer, poses a significant global health challenge with a considerable mortality rate. With the predicted increase in glioblastoma incidence, there is an urgent need for more effective treatment strategies. In this study, we explore the potential of caerin 1.1 and 1.9, host defence peptides derived from an Australian tree frog, in inhibiting glioblastoma U87 and U118 cell growth. Our findings demonstrate the inhibitory impact of caerin 1.1 and 1.9 on cell growth through CCK8 assays. Additionally, these peptides effectively curtail the migration of glioblastoma cells in a cell scratch assay, exhibiting varying inhibitory effects among different cell lines. Notably, the peptides hinder the G0/S phase replication in both U87 and U118 cells, pointing to their impact on the cell cycle. Furthermore, caerin 1.1 and 1.9 show the ability to enter the cytoplasm of glioblastoma cells, influencing the morphology of mitochondria. Proteomics experiments reveal intriguing insights, with a decrease in CHI3L1 expression and an increase in PZP and JUNB expression after peptide treatment. These proteins play roles in cell energy metabolism and inflammatory response, suggesting a multifaceted impact on glioblastoma cells. In conclusion, our study underscores the substantial anticancer potential of caerin 1.1 and 1.9 against glioblastoma cells. These findings propose the peptides as promising candidates for further exploration in the realm of glioblastoma management, offering new avenues for developing effective treatment strategies.


Cell Proliferation , Down-Regulation , Glioblastoma , Mitochondria , Glioblastoma/metabolism , Glioblastoma/pathology , Humans , Cell Proliferation/drug effects , Mitochondria/metabolism , Mitochondria/drug effects , Cell Line, Tumor , Down-Regulation/drug effects , Cell Respiration/drug effects , Animals , Brain Neoplasms/metabolism , Brain Neoplasms/pathology , Antimicrobial Cationic Peptides/pharmacology , Antimicrobial Cationic Peptides/metabolism , Cell Movement/drug effects
15.
Trials ; 25(1): 366, 2024 Jun 07.
Article En | MEDLINE | ID: mdl-38849943

BACKGROUND: Chemotherapy with lomustine is widely considered as standard treatment option for progressive glioblastoma. The value of adding radiotherapy to second-line chemotherapy is not known. METHODS: EORTC-2227-BTG (LEGATO, NCT05904119) is an investigator-initiated, pragmatic (PRECIS-2 score: 34 out of 45), randomized, multicenter phase III trial in patients with first progression of glioblastoma. A total of 411 patients will be randomized in a 1:1 ratio to lomustine (110 mg/m2 every 6 weeks) or lomustine (110 mg/m2 every 6weeks) plus radiotherapy (35 Gy in 10 fractions). Main eligibility criteria include histologic confirmation of glioblastoma, isocitrate dehydrogenase gene (IDH) wild-type per WHO 2021 classification, first progression at least 6 months after the end of prior radiotherapy, radiologically measurable disease according to RANO criteria with a maximum tumor diameter of 5 cm, and WHO performance status of 0-2. The primary efficacy endpoint is overall survival (OS) and secondary endpoints include progression-free survival, response rate, neurocognitive function, health-related quality of life, and health economic parameters. LEGATO is funded by the European Union's Horizon Europe Research program, was activated in March 2024 and will enroll patients in 43 sites in 11 countries across Europe with study completion projected in 2028. DISCUSSION: EORTC-2227-BTG (LEGATO) is a publicly funded pragmatic phase III trial designed to clarify the efficacy of adding reirradiation to chemotherapy with lomustine for the treatment of patients with first progression of glioblastoma. TRIAL REGISTRATION: ClinicalTrials.gov NCT05904119. Registered before start of inclusion, 23 May 2023.


Antineoplastic Agents, Alkylating , Brain Neoplasms , Disease Progression , Glioblastoma , Lomustine , Multicenter Studies as Topic , Progression-Free Survival , Glioblastoma/pathology , Glioblastoma/drug therapy , Glioblastoma/mortality , Glioblastoma/radiotherapy , Glioblastoma/therapy , Humans , Lomustine/administration & dosage , Lomustine/therapeutic use , Lomustine/adverse effects , Brain Neoplasms/radiotherapy , Brain Neoplasms/pathology , Brain Neoplasms/mortality , Brain Neoplasms/therapy , Antineoplastic Agents, Alkylating/therapeutic use , Quality of Life , Randomized Controlled Trials as Topic , Chemoradiotherapy/methods , Clinical Trials, Phase III as Topic , Pragmatic Clinical Trials as Topic , Time Factors
16.
Comput Biol Med ; 177: 108678, 2024 Jul.
Article En | MEDLINE | ID: mdl-38833796

Cells exposed to a pulsed electric field undergo electroporation(EP) and electrodeformation(ED) under electric field stress, and a coupled model of EP and ED of glioblastoma(GBM) taking into account Joule heating is proposed. The model geometry is extracted from real cell boundaries, and the effects of Joule heating-induced temperature rise on the EP and ED processes are considered. The results show that the temperature rise will increase the cell's local conductivity, leading to a decrease in the transmembrane potential(TMP). The temperature rise also causes a decrease in the dynamic Young's modulus of the cell membrane, making the cell less resistant to deformation. In addition, GBM cells are more susceptible to EP in the middle portion of the cell and ED in the three tentacle portions under pulsed electric fields, and the cells undergo significant positional shifts. The ED of the nucleus is similar to spherical cells, but the degree of ED is smaller.


Electroporation , Glioblastoma , Models, Biological , Humans , Electroporation/methods , Hot Temperature , Cell Line, Tumor , Brain Neoplasms , Membrane Potentials/physiology , Cell Membrane
17.
Sci Adv ; 10(23): eadj7706, 2024 Jun 07.
Article En | MEDLINE | ID: mdl-38848360

Poor prognosis and drug resistance in glioblastoma (GBM) can result from cellular heterogeneity and treatment-induced shifts in phenotypic states of tumor cells, including dedifferentiation into glioma stem-like cells (GSCs). This rare tumorigenic cell subpopulation resists temozolomide, undergoes proneural-to-mesenchymal transition (PMT) to evade therapy, and drives recurrence. Through inference of transcriptional regulatory networks (TRNs) of patient-derived GSCs (PD-GSCs) at single-cell resolution, we demonstrate how the topology of transcription factor interaction networks drives distinct trajectories of cell-state transitions in PD-GSCs resistant or susceptible to cytotoxic drug treatment. By experimentally testing predictions based on TRN simulations, we show that drug treatment drives surviving PD-GSCs along a trajectory of intermediate states, exposing vulnerability to potentiated killing by siRNA or a second drug targeting treatment-induced transcriptional programs governing nongenetic cell plasticity. Our findings demonstrate an approach to uncover TRN topology and use it to rationally predict combinatorial treatments that disrupt acquired resistance in GBM.


Drug Resistance, Neoplasm , Gene Expression Regulation, Neoplastic , Gene Regulatory Networks , Glioma , Neoplastic Stem Cells , Humans , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/drug effects , Neoplastic Stem Cells/pathology , Drug Resistance, Neoplasm/genetics , Glioma/genetics , Glioma/pathology , Glioma/metabolism , Glioma/drug therapy , Temozolomide/pharmacology , Brain Neoplasms/genetics , Brain Neoplasms/pathology , Brain Neoplasms/metabolism , Brain Neoplasms/drug therapy , Cell Line, Tumor , Glioblastoma/genetics , Glioblastoma/pathology , Glioblastoma/metabolism , Glioblastoma/drug therapy
18.
Brief Bioinform ; 25(4)2024 May 23.
Article En | MEDLINE | ID: mdl-38855914

Cluster analysis, a pivotal step in single-cell sequencing data analysis, presents substantial opportunities to effectively unveil the molecular mechanisms underlying cellular heterogeneity and intercellular phenotypic variations. However, the inherent imperfections arise as different clustering algorithms yield diverse estimates of cluster numbers and cluster assignments. This study introduces Single Cell Consistent Clustering based on Spectral Matrix Decomposition (SCSMD), a comprehensive clustering approach that integrates the strengths of multiple methods to determine the optimal clustering scheme. Testing the performance of SCSMD across different distances and employing the bespoke evaluation metric, the methodological selection undergoes validation to ensure the optimal efficacy of the SCSMD. A consistent clustering test is conducted on 15 authentic scRNA-seq datasets. The application of SCSMD to human embryonic stem cell scRNA-seq data successfully identifies known cell types and delineates their developmental trajectories. Similarly, when applied to glioblastoma cells, SCSMD accurately detects pre-existing cell types and provides finer sub-division within one of the original clusters. The results affirm the robust performance of our SCSMD method in terms of both the number of clusters and cluster assignments. Moreover, we have broadened the application scope of SCSMD to encompass larger datasets, thereby furnishing additional evidence of its superiority. The findings suggest that SCSMD is poised for application to additional scRNA-seq datasets and for further downstream analyses.


Algorithms , Single-Cell Analysis , Humans , Single-Cell Analysis/methods , Cluster Analysis , Computational Biology/methods , Glioblastoma/genetics , Glioblastoma/pathology , Glioblastoma/metabolism
19.
Commun Biol ; 7(1): 677, 2024 Jun 03.
Article En | MEDLINE | ID: mdl-38830977

We present a quantitative sandwich immunoassay for CD63 Extracellular Vesicles (EVs) and a constituent surface cargo, EGFR and its activity state, that provides a sensitive, selective, fluorophore-free and rapid alternative to current EV-based diagnostic methods. Our sensing design utilizes a charge-gating strategy, with a hydrophilic anion exchange membrane functionalized with capture antibodies and a charged silica nanoparticle reporter functionalized with detection antibodies. With sensitivity and robustness enhancement by the ion-depletion action of the membrane, this hydrophilic design with charged reporters minimizes interference from dispersed proteins, thus enabling direct plasma analysis without the need for EV isolation or sensor blocking. With a LOD of 30 EVs/µL and a high relative sensitivity of 0.01% for targeted proteomic subfractions, our assay enables accurate quantification of the EV marker, CD63, with colocalized EGFR by an operator/sample insensitive universal normalized calibration. We analysed untreated clinical samples of Glioblastoma to demonstrate this new platform. Notably, we target both total and "active" EGFR on EVs; with a monoclonal antibody mAb806 that recognizes a normally hidden epitope on overexpressed or mutant variant III EGFR. Analysis of samples yielded an area-under-the-curve (AUC) value of 0.99 and a low p-value of 0.000033, surpassing the performance of existing assays and markers.


ErbB Receptors , Extracellular Vesicles , Glioblastoma , Tetraspanin 30 , Humans , Glioblastoma/blood , Glioblastoma/diagnosis , Glioblastoma/metabolism , Tetraspanin 30/metabolism , ErbB Receptors/metabolism , Extracellular Vesicles/metabolism , Immunoassay/methods , Biomarkers, Tumor/blood , Biomarkers, Tumor/metabolism , Brain Neoplasms/blood , Brain Neoplasms/metabolism , Brain Neoplasms/diagnosis
20.
J Med Chem ; 67(11): 9662-9685, 2024 Jun 13.
Article En | MEDLINE | ID: mdl-38831692

The new ligand L2Ad, obtained by conjugating the bifunctional species bis(3,5-dimethylpyrazol-1-yl)-acetate and the drug amantadine, was used as a chelator for the synthesis of new Cu complexes 1-5. Their structures were investigated by synchrotron radiation-induced X-ray photoelectron spectroscopy (SR-XPS), near-edge X-ray absorption fine structure (NEXAFS) spectroscopy, and by combining X-ray absorption fine structure (XAFS) spectroscopy techniques and DFT modeling. The structure of complex 3 was determined by single-crystal X-ray diffraction analysis. Tested on U87, T98, and U251 glioma cells, Cu(II) complex 3 and Cu(I) complex 5 decreased cell viability with IC50 values significantly lower than cisplatin, affecting cell growth, proliferation, and death. Their effects were prevented by treatment with the Cu chelator tetrathiomolybdate, suggesting the involvement of copper in their cytotoxic activity. Both complexes were able to increase ROS production, leading to DNA damage and death. Interestingly, nontoxic doses of 3 or 5 enhanced the chemosensitivity to Temozolomide.


Adamantane , Antineoplastic Agents , Coordination Complexes , Copper , Glioblastoma , Humans , Copper/chemistry , Antineoplastic Agents/pharmacology , Antineoplastic Agents/chemistry , Antineoplastic Agents/chemical synthesis , Coordination Complexes/pharmacology , Coordination Complexes/chemistry , Coordination Complexes/chemical synthesis , Glioblastoma/drug therapy , Glioblastoma/pathology , Glioblastoma/metabolism , Ligands , Adamantane/pharmacology , Adamantane/chemistry , Adamantane/chemical synthesis , Adamantane/analogs & derivatives , Cell Line, Tumor , Cell Proliferation/drug effects , Pyrazoles/chemistry , Pyrazoles/pharmacology , Pyrazoles/chemical synthesis , Cell Survival/drug effects , Density Functional Theory , Drug Screening Assays, Antitumor , Reactive Oxygen Species/metabolism , Molecular Structure , Chelating Agents/chemistry , Chelating Agents/pharmacology , Chelating Agents/chemical synthesis , Structure-Activity Relationship , Acetates/chemistry , Acetates/pharmacology , Acetates/chemical synthesis
...