Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 9 de 9
Filtrer
Plus de filtres










Base de données
Gamme d'année
1.
Nat Commun ; 15(1): 4241, 2024 May 18.
Article de Anglais | MEDLINE | ID: mdl-38762500

RÉSUMÉ

Glioblastoma multiforme (GBM) is a highly aggressive brain tumor characterized by invasive behavior and a compromised immune response, presenting treatment challenges. Surgical debulking of GBM fails to address its highly infiltrative nature, leaving neoplastic satellites in an environment characterized by impaired immune surveillance, ultimately paving the way for tumor recurrence. Tracking and eradicating residual GBM cells by boosting antitumor immunity is critical for preventing postoperative relapse, but effective immunotherapeutic strategies remain elusive. Here, we report a cavity-injectable bacterium-hydrogel superstructure that targets GBM satellites around the cavity, triggers GBM pyroptosis, and initiates innate and adaptive immune responses, which prevent postoperative GBM relapse in male mice. The immunostimulatory Salmonella delivery vehicles (SDVs) engineered from attenuated Salmonella typhimurium (VNP20009) seek and attack GBM cells. Salmonella lysis-inducing nanocapsules (SLINs), designed to trigger autolysis, are tethered to the SDVs, eliciting antitumor immune response through the intracellular release of bacterial components. Furthermore, SDVs and SLINs administration via intracavitary injection of the ATP-responsive hydrogel can recruit phagocytes and promote antigen presentation, initiating an adaptive immune response. Therefore, our work offers a local bacteriotherapy for stimulating anti-GBM immunity, with potential applicability for patients facing malignancies at a high risk of recurrence.


Sujet(s)
Tumeurs du cerveau , Glioblastome , Récidive tumorale locale , Salmonella typhimurium , Glioblastome/thérapie , Glioblastome/immunologie , Animaux , Souris , Salmonella typhimurium/immunologie , Mâle , Récidive tumorale locale/prévention et contrôle , Récidive tumorale locale/immunologie , Tumeurs du cerveau/immunologie , Tumeurs du cerveau/thérapie , Humains , Lignée cellulaire tumorale , Souris de lignée C57BL , Pyroptose , Immunité acquise , Immunité innée , Hydrogels/composition chimique , Immunothérapie/méthodes
3.
ACS Nano ; 18(13): 9511-9524, 2024 Apr 02.
Article de Anglais | MEDLINE | ID: mdl-38499440

RÉSUMÉ

Glioblastoma multiforme (GBM) is the most aggressive and lethal form of human brain tumors. Dismantling the suppressed immune microenvironment is an effective therapeutic strategy against GBM; however, GBM does not respond to exogenous immunotherapeutic agents due to low immunogenicity. Manipulating the mitochondrial electron transport chain (ETC) elevates the immunogenicity of GBM, rendering previously immune-evasive tumors highly susceptible to immune surveillance, thereby enhancing tumor immune responsiveness and subsequently activating both innate and adaptive immunity. Here, we report a nanomedicine-based immunotherapeutic approach that targets the mitochondria in GBM cells by utilizing a Trojan-inspired nanovector (ABBPN) that can cross the blood-brain barrier. We propose that the synthetic photosensitizer IrPS can alter mitochondrial electron flow and concurrently interfere with mitochondrial antioxidative mechanisms by delivering si-OGG1 to GBM cells. Our synthesized ABBPN coloaded with IrPS and si-OGG1 (ISA) disrupts mitochondrial electron flow, which inhibits ATP production and induces mitochondrial DNA oxidation, thereby recruiting immune cells and endogenously activating intracranial antitumor immune responses. The results of our study indicate that strategies targeting the mitochondrial ETC have the potential to treat tumors with limited immunogenicity.


Sujet(s)
Tumeurs du cerveau , Glioblastome , Humains , Glioblastome/anatomopathologie , Barrière hémato-encéphalique/anatomopathologie , Électrons , Transport biologique , Tumeurs du cerveau/génétique , Mitochondries , Lignée cellulaire tumorale , Microenvironnement tumoral
4.
J Headache Pain ; 25(1): 2, 2024 Jan 05.
Article de Anglais | MEDLINE | ID: mdl-38177986

RÉSUMÉ

BACKGROUND: The pathogenesis of pediatric migraine remains unclear and presents challenges in diagnosis. Recently, growing evidence has indicated that the gut microbiota can exert modulatory functions at the gut-brain axis by directly or indirectly regulating tryptophan metabolism. Consequently, we aimed to elucidate the potential association among gut microbiota, tryptophan metabolism, and pediatric migraine while also identifying diagnostic biomarkers for pediatric migraine. METHODS: The gut microbiota composition of 33 migraine children and 42 healthy children, aged less than ten years, from the GMrepo database, was analyzed using the Shannon index, Simpson index, principal coordinates analysis, and Wilcoxon rank-sum test. Microbial diagnostic biomarkers were identified using linear discriminant analysis effect size, ridge regression, and random forest. Plasma concentrations of tryptophan metabolites investigated by enzyme-linked immunosorbent assay were compared between 51 migraine children and 120 healthy children, aged less than eighteen years, using t tests and analysis of variance. The receiver operating characteristic curve was performed to evaluate the diagnostic value of microbial and metabolite biomarkers in pediatric migraine. RESULTS: Differences in the composition of gut microbiota, notably the genera that regulate tryptophan metabolism, were observed in pediatric migraine children. Further investigations revealed a significant decrease in plasma kynurenic acid levels (p < 0.001) among migraine children, along with a significant increase in serotonin (p < 0.05) and quinolinic acid (p < 0.001). Subsequently, we established the normal reference intervals for plasma concentrations of tryptophan metabolites in children. More importantly, the ratio of kynurenic acid to quinolinic acid (AUC: 0.871, sensitivity: 86.3%, specificity: 83.3%) exhibited excellent diagnostic efficacy for pediatric migraine. CONCLUSION: Our study suggests that the gut microbiota may play an important role in the development of pediatric migraine by regulating tryptophan metabolism. We believe that microbial and metabolite biomarkers are sensitive diagnostic tests for pediatric migraine. TRIAL REGISTRATION: The study was registered at ClinicalTrials.gov (NCT05969990).


Sujet(s)
Microbiome gastro-intestinal , Migraines , Humains , Enfant , Microbiome gastro-intestinal/physiologie , Tryptophane/métabolisme , Acide kynurénique , Acide quinolinique , Migraines/diagnostic , Marqueurs biologiques
5.
Biomaterials ; 278: 121163, 2021 11.
Article de Anglais | MEDLINE | ID: mdl-34601197

RÉSUMÉ

Glioblastoma multiforme (GBM) is a common malignancy of the central nervous system, but conventional treatments yield unsatisfactory results. Although innovative therapeutic approaches have been developed, they prolong survival by only approximately 5 months. The heterogeneity of GBM renders growth inhibition with a single drug difficult, and exploring combination approaches with multiple targets for the comprehensive treatment of GBM is expected to overcome this limitation. In this study, we designed a biocompatible cRGD/Pt + DOX@GFNPs (RPDGs) nanoformulation to disrupt redox homeostasis in GBM cells and promote the simultaneous occurrence of efficient apoptosis and ferroptosis. Taking advantage of the highly stable Fenton reaction catalytic activity of gallic acid (GA)/Fe2+ nanoparticles in physiological environments, the ability of Pt (IV) to deplete glutathione (GSH) and increase reactive oxygen species (ROS) levels, and the efficient photothermal conversion efficiency of GA/Fe2+ nanoparticles, our synthesized multifunctional and multitargeted RPDGs significantly increased intracellular ROS levels and thus induced ferroptosis. Furthermore, the RPDGs displayed superior photothermal responsiveness and magnetic resonance imaging (MRI) capabilities. These results indicate that RPDGs can not only directly inhibit the growth of tumors but also effectively improve the efficient translocation of conventional chemotherapeutic drugs across the blood-brain barrier, thereby providing a new approach for the comprehensive treatment of GBM.


Sujet(s)
Glioblastome , Nanoparticules métalliques , Nanoparticules , Apoptose , Lignée cellulaire tumorale , Association thérapeutique , Glioblastome/imagerie diagnostique , Glioblastome/traitement médicamenteux , Humains , Spectroscopie par résonance magnétique
6.
Oncogene ; 40(8): 1425-1439, 2021 02.
Article de Anglais | MEDLINE | ID: mdl-33420375

RÉSUMÉ

Dysregulated iron metabolism is a hallmark of many cancers, including glioblastoma (GBM). However, its role in tumor progression remains unclear. Herein, we identified coatomer protein complex subunit zeta 1 (COPZ1) as a therapeutic target candidate which significantly dysregulated iron metabolism in GBM cells. Overexpression of COPZ1 was associated with increasing tumor grade and poor prognosis in glioma patients based on analysis of expression data from the publicly available database The Cancer Genome Atlas (P < 0.001). Protein levels of COPZ1 were significantly increased in GBM compared to non-neoplastic brain tissue samples in immunohistochemistry and western blot analysis. SiRNA knockdown of COPZ1 suppressed proliferation of U87MG, U251 and P3#GBM in vitro. Stable expression of a COPZ1 shRNA construct in U87MG inhibited tumor growth in vivo by ~60% relative to controls at day 21 after implantation (P < 0.001). Kaplan-Meier analysis of the survival data demonstrated that the overall survival of tumor bearing animals increased from 20.8 days (control) to 27.8 days (knockdown, P < 0.05). COPZ1 knockdown also led to the increase in nuclear receptor coactivator 4 (NCOA4), resulting in the degradation of ferritin, and a subsequent increase in the intracellular levels of ferrous iron and ultimately ferroptosis. These data demonstrate that COPZ1 is a critical mediator in iron metabolism. The COPZ1/NCOA4/FTH1 axis is therefore a novel therapeutic target for the treatment of human GBM.


Sujet(s)
Protéine du coatomère/génétique , Ferritines/génétique , Glioblastome/génétique , Coactivateurs de récepteurs nucléaires/génétique , Oxidoreductases/génétique , Apoptose/génétique , Autophagie/génétique , Lignée cellulaire tumorale , Prolifération cellulaire/génétique , Femelle , Ferroptose/génétique , Humains , Estimation de Kaplan-Meier , Mâle , Adulte d'âge moyen , Petit ARN interférent/génétique
7.
ACS Appl Mater Interfaces ; 12(39): 43408-43421, 2020 Sep 30.
Article de Anglais | MEDLINE | ID: mdl-32885649

RÉSUMÉ

Glioblastoma (GBM) is the most common and lethal type of malignant brain tumor in adults. Currently, interventions are lacking, the median overall survival of patients with GBM is less than 15 months, and the postoperative recurrence rate is greater than 60%. We proposed an innovative local chemotherapy involving the construction of gene therapy-based iron oxide nanoparticles (IONPs) as a treatment for patients with glioblastoma after surgery that targeted ferroptosis and apoptosis to address these problems. The porous structure of IONPs with attached carboxyl groups was modified for the codelivery of small interfering RNA (siRNA) targeting glutathione peroxidase 4 (si-GPX4) and cisplatin (Pt) with high drug loading efficiencies. The synthesized folate (FA)/Pt-si-GPX4@IONPs exerted substantial effects on glioblastoma in U87MG and P3#GBM cells, but limited effects on normal human astrocytes (NHAs). During intracellular degradation, IONPs significantly increased iron (Fe2+ and Fe3+) levels, while Pt destroyed nuclear DNA and mitochondrial DNA, leading to apoptosis. Furthermore, IONPs increased H2O2 levels by activating reduced nicotinamide adenine dinucleotide phosphate (NADPH) oxidase (NOX). The Fenton reaction between Fe2+, Fe3+, and intracellular H2O2 generated potent reactive oxygen species (ROS) to initiate ferroptosis, while the co-released si-GPX4 inhibited GPX4 expression and synergistically improved the therapeutic efficacy through a mechanism related to ferroptosis. As a result, superior therapeutic effects with low systemic toxicity were achieved both in vitro and in vivo, indicating that our nanoformulations might represent safe and efficient ferroptosis and apoptosis inducers for use in combinatorial glioblastoma therapy.


Sujet(s)
Antinéoplasiques/pharmacologie , Tumeurs du cerveau/traitement médicamenteux , Cisplatine/pharmacologie , Glioblastome/traitement médicamenteux , Nanoparticules/composition chimique , Phospholipid hydroperoxide glutathione peroxidase/antagonistes et inhibiteurs , Petit ARN interférent/pharmacologie , Animaux , Antinéoplasiques/composition chimique , Apoptose/effets des médicaments et des substances chimiques , Tumeurs du cerveau/métabolisme , Tumeurs du cerveau/chirurgie , Prolifération cellulaire/effets des médicaments et des substances chimiques , Survie cellulaire/effets des médicaments et des substances chimiques , Cellules cultivées , Cisplatine/composition chimique , Tests de criblage d'agents antitumoraux , Femelle , Composés du fer III/composition chimique , Glioblastome/métabolisme , Glioblastome/chirurgie , Humains , Souris , Souris nude , Tumeurs expérimentales/traitement médicamenteux , Tumeurs expérimentales/métabolisme , Tumeurs expérimentales/anatomopathologie , Taille de particule , Phospholipid hydroperoxide glutathione peroxidase/métabolisme , Porosité , Petit ARN interférent/composition chimique , Propriétés de surface
8.
Neurosci Lett ; 732: 135050, 2020 07 27.
Article de Anglais | MEDLINE | ID: mdl-32450188

RÉSUMÉ

Current studies have illustrated that circular RNAs (circRNAs) are a vital part of non-coding RNA (ncRNAs) species and highly abundant and dynamically expressed in brain. However, the exact mechanisms by which circRNAs modulate methamphetamine (METH)-induced neuronal damage still remain largely unexplored. Consistent with our previous study, the expression of circHomer1 was significantly up-regulated after METH treatment in HT-22 cells. We confirmed its loop structure by detection of its back-splice junction with qRT-PCR product via sequence. Moreover, circHomer1 was resistant against RNase R digestion compared with its linear mRNA Homer1. Inhibition of circHomer1 expression indeed alleviated METH-induced neurotoxicity, with lower apoptosis rate via flow cytometry and cleaved Caspase3 protein level. Furthermore, we speculated that Bbc3 functioned as a target of circHomer1 based on computational algorithm, and knockdown of circHomer1 actually reduced Bbc3 expression at the mRNA and protein level. Besides, suppression of Bbc3 decreased the reactive oxygen species (ROS) level and radio of PI-positive cells. Furthermore, we analyzed the correlation in pairs among circHomer1, Bbc3 and behaviors in well-developed METH-addicted models using Pearson's correlation coefficient, which implied an important role of circHomer1 and Bbc3 in addictive behaviors. In all, we for the first time identified a novel circRNA, circHomer1 and our results suggested that circHomer1 regulated METH-induced lethal process by suppressing the Bbc3 expression.


Sujet(s)
Protéines régulatrices de l'apoptose/métabolisme , Métamfétamine/pharmacologie , Neurones/effets des médicaments et des substances chimiques , ARN circulaire/génétique , ARN circulaire/métabolisme , Protéines suppresseurs de tumeurs/métabolisme , Animaux , Apoptose/effets des médicaments et des substances chimiques , Caspase-3/métabolisme , Lignée cellulaire , Extinction de l'expression des gènes , Protéines d'échafaudage Homer/métabolisme , Souris , Souris de lignée C57BL , Neurones/métabolisme , ARN messager/métabolisme , Espèces réactives de l'oxygène/métabolisme
9.
Neurosci Lett ; 701: 146-153, 2019 05 14.
Article de Anglais | MEDLINE | ID: mdl-30797870

RÉSUMÉ

Methamphetamine (METH) has been a worldwide health threat for years. Recent studies have reported that circular RNA (circRNA) are highly abundant and dynamically expressed in brain. However, connections between circRNA and METH-induced neurotoxicity remains indefinite. In the present study, primary cortical neurons were treated with METH in vitro. We profiled circRNA via high-throughput RNA sequencing and identified 2458 circRNAs. Bioinformatics analysis was performed to predict potential functions of these circRNAs which revealed several relevant pathways including 'morphine addiction' that may contribute to the pathogenesis of neuronal damage by METH. Especially, a METH-addicted mouse model was established with conditional place preference paradigm for validation of screened circRNAs. At last, we established co-expression networks of circRNAs with miRNAs and mRNAs to exhibit potential association among them. In conclusion, we firstly unveiled a role of circRNAs in METH-induced neuronal damage and METH addiction.


Sujet(s)
Métamfétamine/pharmacologie , Neurones/effets des médicaments et des substances chimiques , ARN circulaire/effets des médicaments et des substances chimiques , ARN circulaire/génétique , Animaux , Survie cellulaire , Analyse de profil d'expression de gènes , Réseaux de régulation génique , Séquençage nucléotidique à haut débit , Mâle , Métamfétamine/toxicité , Souris , Souris de lignée C57BL , microARN , Culture de cellules primaires , ARN , ARN messager
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE
...