Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 84
Filtrer
Plus de filtres











Base de données
Gamme d'année
1.
Small ; : e2407365, 2024 Oct 04.
Article de Anglais | MEDLINE | ID: mdl-39363827

RÉSUMÉ

Atopic dermatitis (AD) is one of the most common allergic skin disorders affecting over 230 million people worldwide, while safe and efficient therapeutic options for AD are currently rarely available. Reactive oxygen species (ROS) accumulation plays a key role in AD's disease progression. Therefore, a novel single-atom catalyst is designed with isolated Cu1-N4 sites anchored on carbon support (Cu1-N4 ISAC), featuring triple antioxidant enzyme-mimicking activities, for efficient AD cascade catalytic therapy (CCT). The excellent superoxide dismutase (SOD)-, glutathione peroxidase (GPx)-, and ascorbate peroxidase (APx)-like activities of Cu1-N4 ISACs enable the sequential conversion of O2•- to H2O2 and then to harmless H2O, thereby protecting keratinocytes from oxidative stress damage. Notably, two novel experimental methods are developed to directly prove the SOD-GPx and SOD-APx cascade catalytic activities for the first time. In vivo experiments show that Cu1-N4 ISACs are more potent than a recommended typical medicine (halcinonide solution). Additionally, RNA sequencing and bioinformatic analysis reveal that Cu1-N4 ISACs reduce inflammation and inhibit ROS production by activating PPAR signaling, which is aberrantly reduced in AD. Therefore, the synthesized catalytic medicine offers an alternative to alleviate AD and has the potential to serve as PPAR agonists for treating similar diseases.

2.
J Coll Physicians Surg Pak ; 34(9): 1112-1116, 2024 Sep.
Article de Anglais | MEDLINE | ID: mdl-39262015

RÉSUMÉ

OBJECTIVE: To assess the clinical efficacy of combined microsurgery and postoperative radiotherapy for the treatment of intramedullary spinal gliomas and its impact on neurological function. STUDY DESIGN: An observational study. Place and Duration of the Study: Department of Neurosurgery, Baoding No.1 Central Hospital, Hebei, China, between January 2020 and 2023. METHODOLOGY: Sixty patients diagnosed with spinal cord intramedullary gliomas were divided equally into an experimental and control group. The control group received microsurgical treatment, and the experimental group received microsurgical treatment combined with postoperative radiotherapy. The treatment effectiveness, neurological function, and follow-up results of the two groups were compared. RESULTS: After treatment, the clinical efficacy of the experimental group treatment was significantly better than that of the control group (p <0.05). The National Institutes of Health Stroke Scale (NIHSS) scores were significantly lower, and the European Organisation for Research and Treatment of Cancer Quality of Life Questionnaire-30 (EORTC QLQ-C30) scores were significantly higher in the experimental group than in the control group (p <0.05). The 1-3-year survival rate and median survival time of the experimental group were significantly higher than those of the control group (p <0.05). The incidence of complications was 3.33% in the experimental group and 6.67% in the control group, but the difference was not statistically significant (p >0.05). The postoperative recurrence rate was significantly lower in the experimental (0%) than in the control group (13.33%, p <0.05). CONCLUSION: Combined microsurgery and postoperative radiotherapy was found to be more effective than microsurgery alone. It was also more conducive to the recovery of neurological function and improved the patient's quality of life. KEY WORDS: Intramedullary spinal cord glioma, Microsurgery, Neurological function, Radiotherapy.


Sujet(s)
Gliome , Microchirurgie , Qualité de vie , Tumeurs de la moelle épinière , Humains , Tumeurs de la moelle épinière/chirurgie , Tumeurs de la moelle épinière/radiothérapie , Microchirurgie/méthodes , Mâle , Femelle , Adulte d'âge moyen , Gliome/chirurgie , Gliome/radiothérapie , Adulte , Résultat thérapeutique , Radiothérapie adjuvante , Chine/épidémiologie
3.
Pak J Med Sci ; 40(8): 1675-1681, 2024 Sep.
Article de Anglais | MEDLINE | ID: mdl-39281213

RÉSUMÉ

Objective: To compare and analyze the clinical effectiveness of conventional puncture hematoma drainage and stereotactic robot-guided puncture hematoma drainage in managing intracerebral hemorrhage. Methods: This is clinical comparative research. One hundred and twenty patients with the intracerebral hemorrhage who underwent puncture hematoma drainage in Baoding No.1 Central Hospital from March 2020 to May 2023 were included and were assigned into the control groups(n=60) and experimental groups(n=60) according to different treatment methods. The experimental group underwent stereotactic robot-guided puncture hematoma drainage, while the control group underwent conventional puncture hematoma drainage treatment. The duration and situation of surgery, levels of inflammatory factors, as well as preoperative and 1-week postoperative GCS scores and NIHSS scores were compared and analyzed between the two groups. Results: In comparison with the control group, the experimental group exhibited considerably less surgical duration(p=0.00), higher amount of intraoperative blood drainage and hematoma clearance rate(p=0.00). The experimental group possessed a substantially more reduced incidence of complications(10%) in comparison with the control group(25%), with a statistically substantial distinction(p=0.03). After therapy, CRP, TNF-a, and IL-6 degrees were considerably more decreased (p=0.00) in the experimental group in comparison with the control group, while GCS grades were considerably more prominent and NIHSS grades were considerably more reduced (p=0.00). Conclusion: Stereotactic robot-guided puncture hematoma drainage is a dependable and safe operative method to treat patients who had intracerebral hemorrhage, resulting in various benefits such as short length of operation, less injury, less inflammatory reaction, high hematoma clear efficiency and satisfactory recovery of neurological function.

4.
ACS Nano ; 18(32): 21221-21235, 2024 Aug 13.
Article de Anglais | MEDLINE | ID: mdl-39079080

RÉSUMÉ

The mechanical properties of nanoparticles play a crucial role in regulating nanobiointeractions, influencing processes such as blood circulation, tumor accumulation/penetration, and internalization into cancer cells. Consequently, they have a significant impact on drug delivery and therapeutic efficacy. However, it remains unclear whether and how macrophages alter their biological function in response to nanoparticle elasticity. Here, we report on the nano-mechanical biological effects resulting from the interactions between elastic silica nanoparticles (SNs) and macrophages. The SNs with variational elasticity Young's moduli ranging from 81 to 837 MPa were synthesized, and it was demonstrated that M2 [tumor-associated macrophages (TAMs)] could be repolarized to M1 by the soft SNs. Additionally, our findings revealed that cell endocytosis, membrane tension, the curvature protein Baiap2, and the cytoskeleton were all influenced by the elasticity of SNs. Moreover, the mechanically sensitive protein Piezo1 on the cell membrane was activated, leading to calcium ion influx, activation of the NF-κB pathway, and the initiation of an inflammatory response. In vivo experiments demonstrated that the softest 81 MPa SNs enhanced tumor penetration and accumulation and repolarized TAMs in intratumoral hypoxic regions, ultimately resulting in a significant inhibition of tumor growth. Taken together, this study has established a cellular feedback mechanism in response to nanoparticle elasticity, which induces plasma membrane deformation and subsequent activation of mechanosensitive signals. This provides a distinctive "nano-mechanical immunoengineering" strategy for reprogramming TAMs to enhance cancer immunotherapy.


Sujet(s)
Canaux ioniques , Nanoparticules , Macrophages associés aux tumeurs , Animaux , Nanoparticules/composition chimique , Souris , Canaux ioniques/métabolisme , Canaux ioniques/composition chimique , Macrophages associés aux tumeurs/métabolisme , Macrophages associés aux tumeurs/immunologie , Silice/composition chimique , Élasticité , Cellules RAW 264.7 , Humains , Souris de lignée BALB C
5.
Small ; 20(27): e2308565, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38339770

RÉSUMÉ

Cuproptosis is an emerging programmed cell death, displaying great potential in cancer treatment. However, intracellular copper content to induce cuproptosis is unmet, which mainly ascribes to the intracellular pumping out equilibrium mechanism by copper exporter ATP7A and ATP7B. Therefore, it is necessary to break such export balance mechanisms for desired cuproptosis. Mediated by diethyldithiocarbamate (DTC) coordination, herein a strategy to efficiently assemble copper ions into polydopamine nanostructure (PDA-DTC/Cu) for reprogramming copper metabolism of tumor is developed. The deposited Cu2+ can effectively trigger the aggregation of lipoylated proteins to induce cuproptosis of tumor cells. Beyond elevating intracellular copper accumulation, PDA-DTC/Cu enables to break the balance of copper metabolism by disrupting mitochondrial function and restricting the adenosine triphosphate (ATP) energy supply, thus catalytically inhibiting the expressions of ATP7A and ATP7B of tumor cells to enhance cuproptosis. Meanwhile, the killed tumor cells can induce immunogenic cell death (ICD) to stimulate the immune response. Besides, PDA-DTC/Cu NPs can promote the repolarization of tumor-associated macrophages (TAMs ) to relieve the tumor immunosuppressive microenvironment (TIME). Collectively, PDA-DTC/Cu presented a promising "one stone two birds" strategy to realize copper accumulation and inhibit copper export simultaneously to enhance cuproptosis for 4T1 murine breast cancer immunotherapy.


Sujet(s)
Cuivre , Immunothérapie , Indoles , Nanostructures , Polymères , Cuivre/composition chimique , Polymères/composition chimique , Animaux , Immunothérapie/méthodes , Indoles/composition chimique , Indoles/pharmacologie , Souris , Nanostructures/composition chimique , Lignée cellulaire tumorale , Humains , Catalyse , Femelle , Tumeurs/thérapie , Tumeurs/traitement médicamenteux , Tumeurs/métabolisme
6.
ACS Appl Mater Interfaces ; 16(8): 9640-9655, 2024 Feb 28.
Article de Anglais | MEDLINE | ID: mdl-38364050

RÉSUMÉ

The successful treatment of diabetic wounds requires strategies that promote anti-inflammation, angiogenesis, and re-epithelialization of the wound. Excessive oxidative stress in diabetic ulcers (DUs) inhibits cell proliferation and hinders timely vascular formation and macrophage polarization from pro-inflammatory M1 to anti-inflammatory M2, resulting in a persistent inflammatory environment and a nonhealing wound. We designed arginine-nanoenzyme (FTA) with mimic-catalase and arginine-loading. 2,3,4-trihydroxy benzaldehyde and arginine (Arg) were connected by a Schiff base bond, and the nanoassembly of Arg to FTA was driven by the coordination force between a ferric ion and polyphenol and noncovalent bond force such as a hydrogen bond. FTA could remove excess reactive oxygen species at the wound site in situ and convert it to oxygen to improve hypoxia. Meanwhile, Arg was released and catalytically metabolized by NO synthase in M1 to promote vascular repair in the early phase. In the late phase, the metabolite of Arg catalyzed by arginase in M2 was mainly ornithine, which played a vital role in promoting tissue repair, which implemented angiogenesis timely and prevented hypertrophic scars. Mechanistically, FTA activated the cAMP signaling pathway combined with reducing inflammation and ameliorating angiogenesis, which resulted in excellent therapeutic effects on a DU mice model.


Sujet(s)
Arginine , Diabète expérimental , Souris , Animaux , Arginine/pharmacologie , Arginine/usage thérapeutique , Angiogenesis , Diabète expérimental/traitement médicamenteux , Cicatrisation de plaie , Réépithélialisation
7.
J Am Chem Soc ; 146(5): 3186-3199, 2024 02 07.
Article de Anglais | MEDLINE | ID: mdl-38266487

RÉSUMÉ

Atopic dermatitis (AD) is a prevalent chronic inflammatory skin disease that carries a significant global economic burden. Elevated levels of reactive oxygen species (ROS) have been recognized as contributing to AD exacerbation, making them a potential therapeutic target for AD treatment. Here, we introduce a dual-site biomimetic copper/zinc metal-organic framework (Cu/Zn-MOF) featuring four types of enzyme-like activities for AD treatment via suppressing the Fcγ receptor (FcγR)-mediated phagocytosis signal by mimicking the bimetallic sites of natural copper-zinc superoxide dismutase (CuZn-SOD). Interestingly, the neighboring Cu and Zn sites in both Cu/Zn-MOF and CuZn-SOD are at similar distances of ∼5.98 and ∼6.3 Šfrom each other, respectively, and additionally, both Cu and Zn sites are coordinated to nitrogen atoms in both structures, and the coordinating ligands to Cu and Zn are both imidazole rings. Cu/Zn-MOF exhibits remarkable SOD-like activity as well as its glutathione peroxidase (GPx)-, thiol peroxidase (TPx)-, and ascorbate peroxidase (APx)-like activities to continuously consume ROS and mitigate oxidative stress in keratinocytes. Animal experiments show that Cu/Zn-MOF outperforms halcinonide solution (a potent steroid medication) in terms of preventing mechanical injuries, reducing cutaneous water loss, and inhibiting inflammatory responses while presenting favorable biosafety. Mechanistically, Cu/Zn-MOF functions through an FcγR-mediated phagocytosis signal pathway, decreasing the continuous accumulation of ROS in AD and ultimately suppressing disease progression. These findings will provide an effective paradigm for AD therapy and contribute to the development of two-site bionics (TSB).


Sujet(s)
Eczéma atopique , Réseaux organométalliques , Humains , Animaux , Superoxide dismutase/métabolisme , Cuivre , Récepteurs du fragment Fc des IgG , Zinc/métabolisme , Espèces réactives de l'oxygène/métabolisme , Biomimétique , Glutathione peroxidase/métabolisme
8.
Biomaterials ; 306: 122474, 2024 Apr.
Article de Anglais | MEDLINE | ID: mdl-38271788

RÉSUMÉ

Repolarizing the tumor-associated macrophages (TAMs) towards the antitumoral M1-like phenotype has been a promising approach for cancer immunotherapy. However, the anti-cancer immune response is severely limited mainly by the repolarized M1-like macrophages belatedly returning to the M2-like phenotype (i.e., negative feedback). Inspired by nitric oxide (NO) effectively preventing repolarization of inflammatory macrophages in inflammatory diseases, herein, we develop an arginine assembly, as NO nano-donor for NO generation to prevent the negative feedback of the macrophage repolarization. The strategy is to first apply reversible tagging of hydrophobic terephthalaldehyde to create an arginine nano-assembly, and then load a toll-like receptor 7/8 agonist resiquimod (R848) (R848@Arg). Through this strategy, a high loading efficiency of 40 % for the arginine and repolarization characteristics for TAMs can be achieved. Upon the macrophage repolarization by R848, NO can be intracellularly generated from the released arginine by the upregulated inducible nitric oxide synthase. Mechanistically, NO effectively prevented the negative feedback of the repolarized macrophage by mitochondrial dysfunction via blocking oxidative phosphorylation. Notably, R848@Arg significantly increased the tumor inhibition ratio by 3.13-fold as compared to the free R848 by maintaining the M1-like phenotype infiltrating into tumor. The Arg-assembly as NO nano-donor provides a promising method for effective repolarization of macrophages.


Sujet(s)
Maladies mitochondriales , Tumeurs , Humains , Donneur d'oxyde nitrique , Rétroaction , Macrophages , Tumeurs/anatomopathologie , Adjuvants immunologiques/pharmacologie , Monoxyde d'azote/pharmacologie , Immunothérapie/méthodes , Maladies mitochondriales/anatomopathologie , Microenvironnement tumoral
9.
Bioresour Technol ; 395: 130367, 2024 Mar.
Article de Anglais | MEDLINE | ID: mdl-38266788

RÉSUMÉ

The impact and mechanism of fluoranthene (Flr), a typical polycyclic aromatic hydrocarbon highly detected in sludge, on alkaline fermentation for volatile fatty acids (VFAs) recovery and antibiotic resistance genes (ARGs) transfer were studied. The results demonstrated that VFAs production increased from 2189 to 4272 mg COD/L with a simultaneous reduction of ARGs with Flr. The hydrolytic enzymes and genes related to glucose and amino acid metabolism were provoked. Also, Flr benefited for the enrichment of hydrolytic-acidifying consortia (i.e., Parabacteroides and Alkalibaculum) while reduced VFAs consumers (i.e., Rubrivivax) and ARGs potential hosts (i.e., Rubrivivax and Pseudomonas). Metagenomic analysis indicated that the genes related to cell wall synthesis, biofilm formation and substrate transporters to maintain high VFAs-producer activities were upregulated. Moreover, cell functions of efflux pump and Type IV secretion system were suppressed to inhibit ARGs proliferation. This study provided intrinsic mechanisms of Flr-induced VFAs promotion and ARGs reduction during alkaline fermentation.


Sujet(s)
Antibactériens , Fluorènes , Eaux d'égout , Fermentation , Eaux d'égout/composition chimique , Consortiums microbiens , Acides gras volatils , Résistance microbienne aux médicaments , Concentration en ions d'hydrogène
10.
Adv Healthc Mater ; : e2302387, 2023 Nov 17.
Article de Anglais | MEDLINE | ID: mdl-37975271

RÉSUMÉ

Macrophages, capable of both direct killing and antigen presentation, are crucial for the interplay between innate and adaptive immunity. However, strategies mainly focus on polarizing tumor-associated macrophages (TAMs) to M1 phenotype, while overlooking the inefficient antigen cross-presentation due to hyperactive hydrolytic protease within lysosomes which leads to antigen degradation. In light of the significant influence of reactive oxygen species (ROS) on TAMs' polarization and the inhibition of phagosomal proteolysis, a novel nanosystem termed OVA-Fe-GA (OFG) is engineered, drawing inspiration from the NOX2 enzyme's role. OFG integrates ovalbumin (OVA) and a network composed of Fe-gallic acid (GA), emulating the NOX2 enzyme's sequential ROS generation process ("O2 to O2 •- to H2 O2 /•OH"). Furthermore, it elucidates a biological mechanism that augments antigen cross-presentation by suppressing the expression of cysteine proteases. OFG restores the innate anti-tumor functionality of TAMs and significantly amplifies their antigen cross-presentation (4.5-fold compared to the PBS control group) in B16-OVA tumor-bearing mice. Notably, the infiltration and activity of intratumoral CD8+ T cells are enhanced, indicating an adaptive immune response. Moreover, OFG exhibits excellent photothermal properties, thereby fostering a system antitumor immune response. This study provides a promising strategy for initiating both innate and adaptive immunity via TAMs activation. This article is protected by copyright. All rights reserved.

11.
Nat Commun ; 14(1): 6767, 2023 10 25.
Article de Anglais | MEDLINE | ID: mdl-37880231

RÉSUMÉ

Psoriasis is a common inflammatory disease of especially high recurrence rate (90%) which is suffered by approximately 3% of the world population. The overexpression of reactive oxygen species (ROS) plays a critical role in psoriasis progress. Here we show that biomimetic iron single-atom catalysts (FeN4O2-SACs) with broad-spectrum ROS scavenging capability can be used for psoriasis treatment and relapse prevention via related gene restoration. FeN4O2-SACs demonstrate attractive multiple enzyme-mimicking activities based on atomically dispersed Fe active structures, which are analogous to those of natural antioxidant enzymes, iron superoxide dismutase, human erythrocyte catalase, and ascorbate peroxidase. Further, in vitro and in vivo experiments show that FeN4O2-SACs can effectively ameliorate psoriasis-like symptoms and prevent the relapse with augmented efficacy compared with the clinical drug calcipotriol. Mechanistically, estrogen receptor 1 (ESR1) is identified as the core protein upregulated in psoriasis treatment through RNA sequencing and bioinformatic analysis. Together, this study provides a proof of concept of psoriasis catalytic therapy (PCT) and multienzyme-inspired bionics (MIB).


Sujet(s)
Récepteur alpha des oestrogènes , Psoriasis , Humains , Espèces réactives de l'oxygène/métabolisme , Prévention secondaire , Superoxide dismutase/métabolisme , Psoriasis/traitement médicamenteux , Psoriasis/métabolisme
12.
J Nanobiotechnology ; 21(1): 227, 2023 Jul 17.
Article de Anglais | MEDLINE | ID: mdl-37461079

RÉSUMÉ

Myocardial infarction (MI) resulting from coronary artery occlusion is the leading global cause of cardiovascular disability and mortality. Anti-inflammatory treatment plays an important role in MI treatment. Triptolide (TPL), as a Chinese medicine monomer, has a variety of biological functions, including anti-inflammatory, anti-tumor, and immunoregulation. However, it has been proved that TPL is poorly water soluble, and has clear hepatotoxicity and nephrotoxicity, which seriously limits its clinical application. Herein, we designed a long-acting hydrogel platform (TPL@PLGA@F127) for MI treatment by intramyocardial injection. First, we found that the inflammatory response and immune regulation might be the main mechanisms of TPL against MI by network pharmacology. Subsequently, we prepared the hydrogel platform (TPL@PLGA@F127) and tested its effects and toxicity on normal organs in the early stage of MI (3 days after MI-operation). The results showed that TPL@PLGA@F127 could not only promote "repair" macrophages polarization (to M2 macrophage) by day 3 after MI, but also has a long-lasting anti-inflammatory effect in the later stage of MI (28 days after MI-operation). Additionally, we proved that TPL@PLGA@F127 could attenuate the toxicity of TPL by releasing it more slowly and stably. Finally, we observed the long-term effects of TPL@PLGA@F127 on MI and found that it could improve cardiac function, depress the myocardial fibrosis and protect the cardiomyocytes. In summary, this study indicated that TPL@PLGA@F127 could not only enhance the therapeutic effects of TPL on MI, but also attenuate the hepatotoxicity and nephrotoxicity, which established a strong foundation for the clinical application of TPL for MI.


Sujet(s)
Lésions hépatiques dues aux substances , Infarctus du myocarde , Humains , Hydrogels/pharmacologie , Infarctus du myocarde/traitement médicamenteux , Infarctus du myocarde/anatomopathologie , Myocytes cardiaques
14.
J Nanobiotechnology ; 21(1): 120, 2023 Apr 07.
Article de Anglais | MEDLINE | ID: mdl-37024939

RÉSUMÉ

Antigen self-assembly nanovaccines advance the minimalist design of therapeutic cancer vaccines, but the issue of inefficient cross-presentation has not yet been fully addressed. Herein, we report a unique approach by combining the concepts of "antigen multi-copy display" and "calcium carbonate (CaCO3) biomineralization" to increase cross-presentation. Based on this strategy, we successfully construct sub-100 nm biomineralized antigen nanosponges (BANSs) with high CaCO3 loading (38.13 wt%) and antigen density (61.87%). BANSs can be effectively uptaken by immature antigen-presenting cells (APCs) in the lymph node upon subcutaneous injection. Achieving efficient spatiotemporal coordination of antigen cross-presentation and immune effects, BANSs induce the production of CD4+ T helper cells and cytotoxic T lymphocytes, resulting in effective tumor growth inhibition. BANSs combined with anti-PD-1 antibodies synergistically enhance anti-tumor immunity and reverse the tumor immunosuppressive microenvironment. Overall, this CaCO3 powder-mediated biomineralization of antigen nanosponges offer a robust and safe strategy for cancer immunotherapy.


Sujet(s)
Vaccins anticancéreux , Tumeurs , Humains , Poudres , Lymphocytes T CD8+ , Biominéralisation , Cellules présentatrices d'antigène , Tumeurs/traitement médicamenteux , Vaccins anticancéreux/usage thérapeutique , Immunothérapie/méthodes , Microenvironnement tumoral
15.
Adv Healthc Mater ; 12(11): e2202695, 2023 04.
Article de Anglais | MEDLINE | ID: mdl-36622285

RÉSUMÉ

Methionine metabolism has a significant impact on T cells' survival and activation even in comparison to arginine, a well-documented amino acid in metabolic therapy. However, hydrophilic methionine is hardly delivered into TME due to difficult loading and rapid diffusion. Herein, the labeling assembly of methionine into nanoparticle is developed to overcome high hydrophilicity for mild-heat mediated immunometabolic therapy. The strategy is to first label methionine with protocatechualdehyde (as the tag) via reversible Schiff-base bond, and then drive nanoassembly of methionine (MPC@Fe) mediated by iron ions. In this fashion, a loading efficiency of 40% and assembly induced photothermal characteristics can be achieved. MPC@Fe can accumulate persistently in tumor up to 36 h due to tumor-selective aggregation in acidic TME. A mild heat of 43 °C on tumor by light irradiation stimulated the immunogenic cell death and effectively generated CD8+ T cells. Notably, MPC@Fe assisted by mild heat promoted 4.2-fold of tumor-infiltrating INF-γ+ CD8+ T cells, leading to an inhibition ratio of 27.3-fold versus the free methionine. Such labeling assembly provides a promising methionine delivery platform to realize mild heat mediated immunometabolic therapy, and is potentially extensible to other amino acids.


Sujet(s)
Nanoparticules , Tumeurs , Humains , Méthionine , Température élevée , Lymphocytes T CD8+ , Nanoparticules/composition chimique , Racéméthionine , Acides aminés , Lignée cellulaire tumorale
16.
J Mater Chem B ; 11(6): 1171-1190, 2023 02 08.
Article de Anglais | MEDLINE | ID: mdl-36650960

RÉSUMÉ

Apoptosis-based treatment plays an important role in regulating the death of tumor cells (e.g., chemotherapy, radiotherapy, and immunotherapy). Nevertheless, cancer cells can escape surveillance from apoptosis-associated signaling by bypassing other biological pathways and thus result in considerable resistance to therapies. Significantly, ferroptosis, a newly identified type of regulated cell death that is characterized by iron-dependent and lipid peroxidation accumulation, has aroused great research interest in cancer therapy. Increasing approaches have been developed to induce ferroptosis of tumor cells, including using clinically approved drugs, experimentally used compounds, and nanomedicine formulations. More importantly, the emerging nanomedicine-based strategy has made great advances in tumor treatment because of the promising targeting efficacy and enhanced therapeutic effects. In this review, we mainly overview state-of-the-art research on nanomedicine-mediated ferroptosis targeting strategies for synergistic cancer therapies, such as immunotherapy, chemotherapy, radiotherapy, and photothermal therapy. The potential targeting mechanism of nanomedicine for ferroptosis induction was also included. Finally, the future development of nanomedicine in the field of ferroptosis-based cell death in tumor treatment will be envisioned, aiming to provide new insight for tumor treatment in the clinic.


Sujet(s)
Ferroptose , Tumeurs , Nanomédecine , Immunothérapie , Apoptose , Mort cellulaire , Tumeurs/traitement médicamenteux
17.
Adv Sci (Weinh) ; 10(2): e2203308, 2023 01.
Article de Anglais | MEDLINE | ID: mdl-36424137

RÉSUMÉ

Diabetic foot ulcers (DFU), one of the most serious complications of diabetes, are essentially chronic, nonhealing wounds caused by diabetic neuropathy, vascular disease, and bacterial infection. Given its pathogenesis, the DFU microenvironment is rather complicated and characterized by hyperglycemia, ischemia, hypoxia, hyperinflammation, and persistent infection. However, the current clinical therapies for DFU are dissatisfactory, which drives researchers to turn attention to advanced nanotechnology to address DFU therapeutic bottlenecks. In the last decade, a large number of multifunctional nanosystems based on the microenvironment of DFU have been developed with positive effects in DFU therapy, forming a novel concept of "DFU nanomedicine". However, a systematic overview of DFU nanomedicine is still unavailable in the literature. This review summarizes the microenvironmental characteristics of DFU, presents the main progress of wound healing, and summaries the state-of-the-art therapeutic strategies for DFU. Furthermore, the main challenges and future perspectives in this field are discussed and prospected, aiming to fuel and foster the development of DFU nanomedicines successfully.


Sujet(s)
Microenvironnement cellulaire , Pied diabétique , Nanomédecine , Humains , Diabète , Pied diabétique/traitement médicamenteux , Pied diabétique/physiopathologie , Hyperglycémie , Cicatrisation de plaie , Microenvironnement cellulaire/physiologie
18.
Biomaterials ; 292: 121938, 2023 01.
Article de Anglais | MEDLINE | ID: mdl-36493715

RÉSUMÉ

L-arginine metabolism is essential for the activation, survival, and effector function of the T lymphocytes and critical in eliminating tumors via T-cell-mediated immunotherapy, such as immune checkpoint blockade (ICB). Unfortunately, efficient delivery of hydrophilic L-arginine to the tumor microenvironment (TME) has met tremendous difficulties because of the limited loading efficacy and rapid diffusion. Inspired by the small-molecule prodrug nanoassemblies with ultrahigh drug-loading, we screen out aromatic aldehydes compounds to be used as dynamic tags to decorate L-arginine (reversible imine). Nano-Arginine (ArgNP, 104 nm) was created based on dynamic tag-mediated self-assembly. Molecular dynamics simulations indicate that the driving force of this self-assembly process is intermolecular hydrogen bonds, π-π stacking, and cation-π interactions. Notably, ArgNP metabolic synergy with anti-PD-L1 antibody (aPDL1) can promote tumor-infiltrating T cells (3.3-fold than aPDL1), resulting in a tumor inhibition ratio of 2.6-fold than aPDL1. Besides, such a strategy efficiently reduces the myeloid-derived suppressor cells, increases the M1-macrophages against the tumor, and induces the production of memory T cells. Furthermore, this synergistic therapy effectively restrains lung metastasis and prolongs mouse survival (60% survival ratio). The study highlights the dynamic tags strategy with facility and advance to deliver L-arginine that can metabolically promote ICB therapy.


Sujet(s)
Inhibiteurs de points de contrôle immunitaires , Tumeurs , Souris , Animaux , Inhibiteurs de points de contrôle immunitaires/pharmacologie , Inhibiteurs de points de contrôle immunitaires/usage thérapeutique , Arginine , Microenvironnement tumoral , Immunothérapie , Tumeurs/thérapie , Lignée cellulaire tumorale
19.
J Nanobiotechnology ; 20(1): 544, 2022 Dec 28.
Article de Anglais | MEDLINE | ID: mdl-36577992

RÉSUMÉ

Hypoxia and high accumulation of lactic acid in the tumor microenvironment provide fertile soil for tumor development, maintenance and metastasis. Herein, we developed a calcium peroxide (CaO2)-loaded nanostructure that can play a role of "one stone kill two birds", i.e., acidic and hypoxic tumor microenvironment can be simultaneously regulated by CaO2 loaded nanostructure. Specifically, CaO2-loaded mesoporous polydopamine nanoparticles modified with sodium hyaluronate (denoted as CaO2@mPDA-SH) can gradually accumulate in a tumor site. CaO2 exposed in acidic microenvironment can succeed in consuming the lactic acid with oxygen generation simultaneously, which could remodel the acid and hypoxia tumor microenvironment. More importantly, the relief of hypoxia could further reduce lactate production from the source by down-regulating the hypoxia inducible factor-1α (HIF-1α), which further down-regulated the glycolysis associated enzymes including glycolysis-related glucose transporter 1 (GLUT1) and lactate dehydrogenase A (LDHA). As a result, CaO2@mPDA-SH alone without the employment of other therapeutics can dually regulate the tumor hypoxia and lactic acid metabolism, which efficiently represses tumor progression in promoting immune activation, antitumor metastasis, and anti-angiogenesis.


Sujet(s)
Nanoparticules , Microenvironnement tumoral , Humains , Lignée cellulaire tumorale , Hypoxie , Nanoparticules/composition chimique , Acide lactique/métabolisme
20.
Pak J Med Sci ; 38(7): 1796-1801, 2022.
Article de Anglais | MEDLINE | ID: mdl-36246717

RÉSUMÉ

Objectives: To compare the clinical efficacy of robot of stereotactic assistant (ROSA) and frame-assisted stereotactic drilling and drainage for intracerebral hematoma in hypertensive intracerebral hemorrhage (HICH). Methods: A total of 142 patients with HICH treated in Baoding First Central Hospital from January 2018 to January 2020 were selected and divided into two groups using a random number table. The ROSA group was treated with a robot of stereotactic assistant, while the frame group underwent frame-assisted stereotactic drilling and drainage for intracerebral hematoma. Surgical duration, postoperative extubation time and complications were compared between the two groups. Venous blood (5 mL) was collected before and three days after surgery. The levels of inflammatory factors [tumor necrosis factor-α (TNF-α), high-sensitivity C-reactive protein (hs-CRP) and interleukin-6 (IL-6)], as well as neurological function indexes [neuron-specific enolase (NSE), nerve growth factor (NGF) and brain-derived neurotrophic factor (BDNF)] were detected by enzyme-linked immunosorbent assay. Results: The surgical duration, postoperative extubation time, and incidences of infection and postoperative rehemorrhage in the ROSA group were lower than those in the frame group (P < 0.05). In the ROSA group, postoperative TNF-α, hs-CRP, IL-6 and NSE levels were significantly lower while NGF and BDNF levels were higher than those in the frame group (all P < 0.05). Conclusion: Compared with frame-assisted stereotactic drilling and drainage for intracerebral hematoma, ROSA in HICH treatment shortens the surgical duration and postoperative extubation time, reduces the risks of infection and rehemorrhage and decreases inflammatory level, which is helpful for the recovery of neurological function.

SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE