Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 11 de 11
Filtrer
Plus de filtres










Base de données
Gamme d'année
1.
Sci Rep ; 14(1): 10646, 2024 05 09.
Article de Anglais | MEDLINE | ID: mdl-38724530

RÉSUMÉ

Individual theranostic agents with dual-mode MRI responses and therapeutic efficacy have attracted extensive interest due to the real-time monitor and high effective treatment, which endow the providential treatment and avoid the repeated medication with side effects. However, it is difficult to achieve the integrated strategy of MRI and therapeutic drug due to complicated synthesis route, low efficiency and potential biosafety issues. In this study, novel self-assembled ultrasmall Fe3O4 nanoclusters were developed for tumor-targeted dual-mode T1/T2-weighted magnetic resonance imaging (MRI) guided synergetic chemodynamic therapy (CDT) and chemotherapy. The self-assembled ultrasmall Fe3O4 nanoclusters synthesized by facilely modifying ultrasmall Fe3O4 nanoparticles with 2,3-dimercaptosuccinic acid (DMSA) molecule possess long-term stability and mass production ability. The proposed ultrasmall Fe3O4 nanoclusters shows excellent dual-mode T1 and T2 MRI capacities as well as favorable CDT ability due to the appropriate size effect and the abundant Fe ion on the surface of ultrasmall Fe3O4 nanoclusters. After conjugation with the tumor targeting ligand Arg-Gly-Asp (RGD) and chemotherapy drug doxorubicin (Dox), the functionalized Fe3O4 nanoclusters achieve enhanced tumor accumulation and retention effects and synergetic CDT and chemotherapy function, which serve as a powerful integrated theranostic platform for cancer treatment.


Sujet(s)
Imagerie par résonance magnétique , Nanomédecine théranostique , Imagerie par résonance magnétique/méthodes , Nanomédecine théranostique/méthodes , Animaux , Souris , Humains , Doxorubicine/composition chimique , Doxorubicine/administration et posologie , Doxorubicine/pharmacologie , Doxorubicine/usage thérapeutique , Lignée cellulaire tumorale , Tumeurs/imagerie diagnostique , Tumeurs/traitement médicamenteux , Tumeurs/thérapie , Nanoparticules de magnétite/composition chimique , Nanoparticules de magnétite/usage thérapeutique , Succimer/composition chimique , Antinéoplasiques/usage thérapeutique , Antinéoplasiques/composition chimique , Antinéoplasiques/administration et posologie , Antinéoplasiques/pharmacologie
2.
J Mater Chem B ; 12(16): 3959-3969, 2024 Apr 24.
Article de Anglais | MEDLINE | ID: mdl-38477096

RÉSUMÉ

The Fenton reaction-based chemodynamic therapy is a form of cancer therapy, and its efficacy can be significantly improved by promoting catalytic reactions involving iron ions. A system with high catalytic capacity and low biological toxicity that effectively inhibits tumor progression is required for optimal treatment. In this study, iron-loaded carbonaceous nanoparticles (CNPs@Fe) with Fenton catalytic activity were fabricated and applied for the chemodynamic therapy of cancer. The carbonaceous nanoparticles derived from glucose via a caramelization reaction demonstrated high biocompatibility. Besides, aromatic structures in the carbonaceous nanoparticles helped accelerate electron transfer to enhance the catalytic decomposition of H2O2, resulting in the formation of highly reactive hydroxyl radicals (˙OH). At pH 6.0 (representing weak acidity in the tumor microenvironment), the Fenton catalytic activity of CNPs@Fe in the decomposition of H2O2 was 15.3 times higher than that of Fe2+ and 28.3 times higher than that of Fe3O4via a chromogenic reaction. The reasons for the enhancement were revealed by analyzing the chemical composition of carbonaceous nanoparticles using high-resolution mass spectra. The developed Fenton agent also demonstrated significant therapeutic effectiveness and minimal side effects in in vitro and in vivo anticancer studies. This work proposes a novel approach to promote the generation of reactive oxygen species (ROS) for the chemodynamic therapy of cancer.


Sujet(s)
Carbone , Peroxyde d'hydrogène , Fer , Nanoparticules , Peroxyde d'hydrogène/composition chimique , Concentration en ions d'hydrogène , Fer/composition chimique , Humains , Animaux , Nanoparticules/composition chimique , Souris , Carbone/composition chimique , Antinéoplasiques/composition chimique , Antinéoplasiques/pharmacologie , Prolifération cellulaire/effets des médicaments et des substances chimiques , Souris de lignée BALB C , Survie cellulaire/effets des médicaments et des substances chimiques , Taille de particule , Femelle , Tests de criblage d'agents antitumoraux
3.
Adv Healthc Mater ; 13(11): e2303963, 2024 04.
Article de Anglais | MEDLINE | ID: mdl-38296248

RÉSUMÉ

Adoptively transferred cells usually suffer from exhaustion, limited expansion, and poor infiltration, partially attributing to the complicated immunosuppressive microenvironment of solid tumors. Therefore, it is necessary to explore more effective strategies to improve the poor tumor microenvironment (TME) to efficaciously deliver and support extrinsic effector cells in vivo. Herein, an intelligent biodegradable hollow manganese dioxide nanoparticle (MnOX) that possesses peroxidase activity to catalyze excess H2O2 in the TME to produce oxygen and relieve the hypoxia of solid tumors is developed. MnOX nanoenzymes modified with CD56 antibody could specifically bind CAR-NK (chimeric antigen receptor modified natural killer) cells. It is demonstrated that CAR-NK cells incorporated with MnOX nanoenzymes effectively infiltrate into tumor tissues with an improved TME, which results in superior antitumor activity in solid tumor-bearing mice. The antibody connection between MnOX nanoenzymes and CAR-NK endows the lowest efficient dosage of MnOX. This study features a smart synergistic immunotherapy approach for solid tumors using MnOX nanoenzyme-armed CAR-NK cells, which would provide a valuable tool for immunocyte therapy in solid tumors.


Sujet(s)
Cellules tueuses naturelles , Composés du manganèse , Nanoparticules , Oxydes , Microenvironnement tumoral , Animaux , Composés du manganèse/composition chimique , Souris , Microenvironnement tumoral/effets des médicaments et des substances chimiques , Oxydes/composition chimique , Nanoparticules/composition chimique , Humains , Cellules tueuses naturelles/immunologie , Lignée cellulaire tumorale , Tumeurs/thérapie , Tumeurs/métabolisme , Tumeurs/anatomopathologie , Récepteurs chimériques pour l'antigène/métabolisme , Récepteurs chimériques pour l'antigène/immunologie , Peroxyde d'hydrogène/composition chimique , Peroxyde d'hydrogène/métabolisme
4.
Front Mol Neurosci ; 15: 822129, 2022.
Article de Anglais | MEDLINE | ID: mdl-35557557

RÉSUMÉ

Mutations in the SCN8A gene encoding the voltage-gated sodium channel α-subunit Nav1. 6 have been reported in individuals with epilepsy, intellectual disability and features of autism spectrum disorder. SCN8A is widely expressed in the central nervous system, including the cerebellum. Cerebellar dysfunction has been implicated in autism spectrum disorder. We investigated conditional Scn8a knockout mice under C57BL/6J strain background that specifically lack Scn8a expression in cerebellar Purkinje cells (Scn8a flox/flox , L7Cre + mice). Cerebellar morphology was analyzed by immunohistochemistry and MR imaging. Mice were subjected to a battery of behavioral tests including the accelerating rotarod, open field, elevated plus maze, light-dark transition box, three chambers, male-female interaction, social olfaction, and water T-maze tests. Patch clamp recordings were used to evaluate evoked action potentials in Purkinje cells. Behavioral phenotyping demonstrated that Scn8a flox/flox , L7Cre + mice have impaired social interaction, motor learning and reversal learning as well as increased repetitive behavior and anxiety-like behaviors. By 5 months of age, Scn8a flox/flox , L7Cre + mice began to exhibit cerebellar Purkinje cell loss and reduced molecular thickness. At 9 months of age, Scn8a flox/flox , L7Cre + mice exhibited decreased cerebellar size and a reduced number of cerebellar Purkinje cells more profoundly, with evidence of additional neurodegeneration in the molecular layer and deep cerebellar nuclei. Purkinje cells in Scn8a flox/flox , L7Cre + mice exhibited reduced repetitive firing. Taken together, our experiments indicated that loss of Scn8a expression in cerebellar Purkinje cells leads to cerebellar degeneration and several ASD-related behaviors. Our study demonstrated the specific contribution of loss of Scn8a in cerebellar Purkinje cells to behavioral deficits characteristic of ASD. However, it should be noted that our observed effects reported here are specific to the C57BL/6 genome type.

5.
Ann N Y Acad Sci ; 1513(1): 140-152, 2022 07.
Article de Anglais | MEDLINE | ID: mdl-35419858

RÉSUMÉ

Aseptic loosening is a major complication of prosthetic joint surgery and is associated with impaired osteoblast homeostasis. Cortistatin (CST) is a neuropeptide that protects against inflammatory conditions. In this study, we found that expression of CST was diminished in patients with prosthetic joint loosening and in titanium (Ti) particle-induced animal models. A Ti particle-induced calvarial osteolysis model was established in wild-type and CST gene knockout mice; CST deficiency enhanced, while exogenously added CST attenuated, the severity of Ti particle-mediated osteolysis. CST protected against inflammation as well as apoptosis and maintained the osteogenic function of MC3T3-E1 osteoblasts upon stimulation with Ti particles. Furthermore, CST antagonized reactive oxygen species production and suppressed caspase-3-associated apoptosis mediated by Ti particles in osteoblasts. Additionally, CST protects against Ti particle-induced osteolysis through tumor necrosis factor receptor 1. Taken together, CST might provide a therapeutic strategy for wear debris-induced inflammatory osteolysis.


Sujet(s)
Neuropeptides , Ostéolyse , Animaux , Caspase-3/génétique , Caspase-3/métabolisme , Souris , Souris de lignée C57BL , Neuropeptides/génétique , Neuropeptides/métabolisme , Ostéoblastes/métabolisme , Ostéoclastes , Ostéolyse/induit chimiquement , Ostéolyse/prévention et contrôle , Espèces réactives de l'oxygène/métabolisme , Récepteur au facteur de nécrose tumorale de type I/métabolisme , Titane/effets indésirables
6.
Acta Biomater ; 142: 284-297, 2022 04 01.
Article de Anglais | MEDLINE | ID: mdl-35151925

RÉSUMÉ

Pancreatic adenocarcinoma (PDAC) is one of the deadliest cancers, and it is resistant to most conventional antineoplastic therapies. To address this challenge, gemcitabine (Gem)-loaded carbonaceous nanoparticles (MFC-Gem) as nanozymes and a theranostic platform were fabricated and used for MR-guided ferroptosis-chemo synergetic therapy of PDAC. As a biocompatible carrier, MFC-Gem nanoparticles are regarded as peroxidase-like and glutathione peroxidase-like nanozymes that promote ferroptosis therapy by effectively generating ROS and consuming GSH. Meanwhile, the combination of MnFe2O4 and Gem can markedly enhance synergetic therapy by both ferroptosis and Gem chemotherapy. MFC-Gem has higher magnetic susceptibility and was used for simultaneous magnetic resonance imaging (MRI) monitoring of the PDAC treatment. In conclusion, these salient features unequivocally indicate that this biocompatible nanotheranostic system has cooperative and enhancing chemotherapy effects for anti-PDAC therapy with simultaneous MRI monitoring. STATEMENT OF SIGNIFICANCE: Pancreatic adenocarcinoma (PDAC) is one of the deadliest cancers, and it is resistant to most conventional antineoplastic therapies. To address this challenge, gemcitabine (Gem)-loaded carbonaceous nanoparticles (MFC-Gem) as nanozymes and a theranostic platform were fabricated and used for MR-guided ferroptosis-chemo synergetic therapy of PDAC. i) MFC nanoparticles are regarded as peroxidase-like and glutathione peroxidase-like nanozymes that enhance ferroptosis therapy by effectively generating ROS and consuming GSH. ii) The combination of MnFe2O4 and Gem can markedly enhance synergetic therapy by both ferroptosis and Gem chemotherapy. iii) MFC-Gem has higher magnetic susceptibility and was used for simultaneous magnetic resonance imaging (MRI) monitoring of the PDAC treatment.


Sujet(s)
Adénocarcinome , Antinéoplasiques , Ferroptose , Tumeurs du pancréas , Antinéoplasiques/usage thérapeutique , Lignée cellulaire tumorale , Désoxycytidine/analogues et dérivés , Glutathione peroxidase/usage thérapeutique , Humains , Imagerie par résonance magnétique , Tumeurs du pancréas/imagerie diagnostique , Tumeurs du pancréas/traitement médicamenteux , Tumeurs du pancréas/anatomopathologie , Myeloperoxidase , Espèces réactives de l'oxygène , , Tumeurs du pancréas
7.
Front Aging Neurosci ; 13: 656430, 2021.
Article de Anglais | MEDLINE | ID: mdl-33935688

RÉSUMÉ

OBJECTIVE: Low-intensity transcranial ultrasound stimulation (TUS) is a non-invasive neuromodulation technique with high spatial resolution and feasible penetration depth. To date, the mechanisms of TUS modulated neural oscillations are not fully understood. This study designed a very low acoustic intensity (AI) TUS system that produces considerably reduced AI Ultrasound pulses (I SPTA < 0.5 W/cm2) when compared to previous methods used to measure regional neural oscillation patterns under different TUS parameters. METHODS: We recorded the local field potential (LFP) of five brain nuclei under TUS with three groups of simulating parameters. Spectrum estimation, time-frequency analysis (TFA), and relative power analysis methods have been applied to investigate neural oscillation patterns under different stimulation parameters. RESULTS: Under PRF, 500 Hz and 1 kHz TUS, high-amplitude LFP activity with the auto-rhythmic pattern appeared in selected nuclei when I SPTA exceeded 12 mW/cm2. With TFA, high-frequency energy (slow gamma and high gamma) was significantly increased during the auto-rhythmic patterns. We observed an initial plateau in nuclei response when I SPTA reached 16.4 mW/cm2 for RPF 500 Hz and 20.8 mW/cm2 for RPF 1 kHz. The number of responding nuclei started decreasing while I SPTA continued increasing. Under 1.5 kHz TUS, no auto-rhythmic patterns have been observed, but slow frequency power was increased during TUS. TUS inhibited most of the frequency band and generated obvious slow waves (theta and delta band) when stimulated at RPF = 1.5 kHz, I SPTA = 8.8 mW/cm2. CONCLUSION: These results demonstrate that very low intensity Transcranial Ultrasound Stimulation (VLTUS) exerts significant neuromodulator effects under specific parameters in rat models and may be a valid tool to study neuronal physiology.

8.
J Mater Chem B ; 8(48): 11021-11032, 2020 12 23.
Article de Anglais | MEDLINE | ID: mdl-33191409

RÉSUMÉ

Metastasis and spread are currently the main factors leading to high mortality of cancer, so developing a synergetic antitumor strategy with high specificity and hypotoxicity is in urgent demand. Based on the design concept of "nanocatalytic medicine", multifunctional nanotherapeutic agent FePt@COP-FA nanocomposites (FPCF NCs) are developed for cancer treatment. Specifically, in the tumor microenvironment (TME), FePt could catalyze intracellular over-expressed H2O2 to generate highly active hydroxyl radicals (˙OH), which could not only induce the apoptosis of tumor cells, but also activate the "ferroptosis" pathway resulting in the lipid peroxide accumulation and ferroptotic cell death. Moreover, owing to the excellent photothermal effect, the FPCF NCs could effectively ablate primary tumors under near-infrared (NIR) laser irradiation and produce numerous tumor-associated antigens in situ. With the assistance of a checkpoint blockade inhibitor, anti-CTLA4 antibody, the body's specific immune response would be initiated to inhibit the growth of metastatic tumors. In particular, such synergistic therapeutics could produce an effective immunological memory effect, which could prevent tumor metastasis and recurrence again. In summary, the FPCF NC is an effective multifunctional antitumor therapeutic agent for nanocatalytic/photothermal/checkpoint blockade combination therapy, which exhibits great potential in nanocatalytic anticancer therapeutic applications.


Sujet(s)
Antinéoplasiques/synthèse chimique , Composés du fer III/synthèse chimique , Nanocomposites/composition chimique , Platine/composition chimique , Polymères/synthèse chimique , Microenvironnement tumoral/effets des médicaments et des substances chimiques , Animaux , Antinéoplasiques/administration et posologie , Catalyse/effets des médicaments et des substances chimiques , Lignée cellulaire tumorale , Survie cellulaire/effets des médicaments et des substances chimiques , Survie cellulaire/physiologie , Relation dose-effet des médicaments , Composés du fer III/administration et posologie , Humains , Cellules MCF-7 , Souris , Nanocomposites/administration et posologie , Tumeurs/traitement médicamenteux , Tumeurs/anatomopathologie , Thérapie photothermique/méthodes , Platine/administration et posologie , Polymères/administration et posologie , Microenvironnement tumoral/physiologie , Tests d'activité antitumorale sur modèle de xénogreffe/méthodes
9.
Adv Healthc Mater ; 9(21): e1901634, 2020 11.
Article de Anglais | MEDLINE | ID: mdl-32959536

RÉSUMÉ

Multimodal imaging-guided synergistic anticancer strategies have attracted increasing attention for efficient diagnosis and therapy of cancer. Herein, a multifunctional nanotheranostic agent FePtMn-Ce6/FA (FPMCF NPs) is constructed by covalently anchoring photosensitizer chlorin e6 (Ce6) and targeting molecule folic acid (FA) on ultrasmall homogeneous ternary FePtMn nanocrystals. Response to tumor microenvironment (TME), FPMCF NPs can release Fe2+ to catalyze H2 O2 into •OH by Fenton reaction and simultaneously catalyze hydrogen peroxide (H2 O2 ) into O2 to overcome the tumor hypoxia barrier. Released O2 is further catalyzed into 1 O2 under 660 nm laser irradiation with Ce6. Thus, the FPMCF NPs exhibit superior dual-ROS oxidization capability including ferroptosis chemodynamic oxidization and 1 O2 -based photodynamic oxidization. Interestingly, FPMCF NPs reveal strong photothermal conversion efficiency exposed to an 808 nm laser, which can assist dual-ROS oxidization to suppress solid tumor remarkably. Additionally, Mn2+ can be released from FPMCF NPs to enhance longitudinal relaxivity (T1 -weighted magnetic resonance (MR) imaging) and Fe-synergistic transverse relaxivity (T2 -weighted MR imaging), which is convenient for diagnosis of solid tumors. Meanwhile, the fluorescent/photothermal (FL/PT) imaging function of FPMCF NPs can also accurately monitor tumor location. Therefore, FPMCF NPs with multimodal MR/FL/PT imaging-guided synergistic chemodynamic/photodynamic/photothermal cancer therapy capability have potential bioapplication in bionanomedicine field.


Sujet(s)
Nanoparticules , Tumeurs , Photothérapie dynamique , Humains , Hypoxie , Tumeurs/imagerie diagnostique , Tumeurs/traitement médicamenteux , Photosensibilisants/usage thérapeutique , Microenvironnement tumoral
10.
J Mater Chem B ; 8(31): 6866-6876, 2020 08 12.
Article de Anglais | MEDLINE | ID: mdl-32249886

RÉSUMÉ

Engineering nanoparticles (NPs) with multifunctionality has become a promising strategy for cancer theranostics. Herein, theranostic polymer NPs are fabricated via the assembly of amphiphilic paramagnetic block copolymers (PCL-b-PIEtMn), in which IR-780 and doxorubicin (DOX) were co-encapsulated, for magnetic resonance (MR) and near infrared fluorescence (NIRF) imaging as well as for photo thermal therapy (PTT)-enhanced chemotherapy. The synthesized amphiphilic paramagnetic block copolymers demonstrated high relaxivity (r1 = 7.05 mM-1 s-1). The encapsulated DOX could be released with the trigger of near infrared (NIR) light. In vivo imaging confirmed that the paramagnetic NPs could be accumulated effectively at the tumor sites. Upon the NIR laser irradiation, tumor growth was inhibited by PTT-enhanced chemotherapy. The advantages of the reported system lie in the one-step convergence of multiple functions (i.e., imaging and therapy agents) into a one delivery vehicle and the dual mode imaging-guided synergistic PTT and chemotherapy. This study represents a new drug delivery vehicle of paramagnetic NPs for visualized theranostics.


Sujet(s)
Interactions hydrophobes et hydrophiles , Aimants/composition chimique , Polymères/composition chimique , Polymères/usage thérapeutique , Doxorubicine/pharmacologie , Synergie des médicaments , Humains , Cellules MCF-7 , Nanoparticules/composition chimique , Imagerie optique , Photothérapie
11.
ACS Appl Mater Interfaces ; 11(42): 38395-38404, 2019 Oct 23.
Article de Anglais | MEDLINE | ID: mdl-31554396

RÉSUMÉ

Reactive oxygen species (ROS)-based anticancer therapy methods were heavily dependent on specific tumor microenvironments such as acidity and excess hydrogen peroxide (H2O2). In this work, an acidity-sensitive nanotheranostic agent (FePt@MnO)@DSPE-PEG5000-FA (FMDF NPs)  was successfully constructed for MR imaging guided ferroptosis chemodynamic therapy (FCDT) of cancer. The FMDF NPs could specifically target folic acid (FA) receptor-positive tumor cells (HeLa etc.) and induce ferroptosis efficiently by rapidly releasing active Fe2+ to catalyze intracellular H2O2 into ROS based on Fenton reaction. On the other hand, the Mn2+ could also be released due to acidity  and further coordinate with GSH to enhance the longitudinal-transverse relaxivity (T1/T2-weighted MR imaging), which could obviously strengthen the contrast distinction between solid tumors and the surrounding tissue to accurately real-time monitor the tumor location. Furthermore, the in vivo anticancer study revealed that the growth of solid tumor models could be suppressed remarkably after treating with FMDF NPs and no obvious damage to other major organs. Therefore, the FMDF NPs were competent simultaneously as an enhanced imaging diagnosis contrast agent and efficient therapy agent for promoting more precise and effective treatment in the bionanomedicine field.


Sujet(s)
Ferroptose , Fer/composition chimique , Composés du manganèse/composition chimique , Nanoparticules/composition chimique , Oxydes/composition chimique , Platine/composition chimique , Animaux , Lignée cellulaire tumorale , Survie cellulaire/effets des médicaments et des substances chimiques , Produits de contraste/composition chimique , Ferroptose/effets des médicaments et des substances chimiques , Récepteurs des folates à ancre GPI/composition chimique , Récepteurs des folates à ancre GPI/métabolisme , Acide folique/composition chimique , Acide folique/métabolisme , Humains , Peroxyde d'hydrogène/composition chimique , Peroxyde d'hydrogène/métabolisme , Concentration en ions d'hydrogène , Imagerie par résonance magnétique , Souris , Souris de lignée BALB C , Nanoparticules/usage thérapeutique , Nanoparticules/toxicité , Tumeurs/imagerie diagnostique , Tumeurs/traitement médicamenteux , Tumeurs/anatomopathologie , Polyéthylène glycols/composition chimique , Distribution tissulaire
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE