Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 4 de 4
Filtrar
Más filtros

Banco de datos
Tipo del documento
País de afiliación
Intervalo de año de publicación
1.
Nano Lett ; 19(1): 124-134, 2019 01 09.
Artículo en Inglés | MEDLINE | ID: mdl-30521345

RESUMEN

The targeted delivery of therapeutics to sites of rheumatoid arthritis (RA) has been a long-standing challenge. Inspired by the intrinsic inflammation-targeting capacity of macrophages, a macrophage-derived microvesicle (MMV)-coated nanoparticle (MNP) was developed for targeting RA. The MMV was efficiently produced through a novel method. Cytochalasin B (CB) was applied to relax the interaction between the cytoskeleton and membrane of macrophages, thus stimulating MMV secretion. The proteomic profile of the MMV was analyzed by iTRAQ (isobaric tags for relative and absolute quantitation). The MMV membrane proteins were similar to those of macrophages, indicating that the MMV could exhibit bioactivity similar to that of RA-targeting macrophages. A poly(lactic- co-glycolic acid) (PLGA) nanoparticle was subsequently coated with MMV, and the inflammation-mediated targeting capacity of the MNP was evaluated both in vitro and in vivo. The in vitro binding of MNP to inflamed HUVECs was significantly stronger than that of the red blood cell membrane-coated nanoparticle (RNP). Compared with bare NP and RNP, MNP showed a significantly enhanced targeting effect in vivo in a collagen-induced arthritis (CIA) mouse model. The targeting mechanism was subsequently revealed according to the proteomic analysis, indicating that Mac-1 and CD44 contributed to the outstanding targeting effect of the MNP. A model drug, tacrolimus, was encapsulated in MNP (T-RNP) and significantly suppressed the progression of RA in mice. The present study demonstrates MMV as a promising and rich material, with which to mimic macrophages, and demonstrates that MNP is an efficient biomimetic vehicle for RA targeting and treatment.


Asunto(s)
Artritis Experimental/tratamiento farmacológico , Artritis Reumatoide/tratamiento farmacológico , Nanopartículas/administración & dosificación , Proteómica , Animales , Artritis Reumatoide/patología , Citocalasina B/química , Modelos Animales de Enfermedad , Eritrocitos/química , Eritrocitos/efectos de los fármacos , Regulación de la Expresión Génica/efectos de los fármacos , Células Endoteliales de la Vena Umbilical Humana , Humanos , Receptores de Hialuranos/genética , Antígeno de Macrófago-1/genética , Macrófagos/química , Ratones , Nanopartículas/química , Poliésteres/química , Copolímero de Ácido Poliláctico-Ácido Poliglicólico/química , Tacrolimus
2.
Int J Pharm ; 612: 121356, 2022 Jan 25.
Artículo en Inglés | MEDLINE | ID: mdl-34919996

RESUMEN

PURPOSE: Drug-resistant gram-negative bacteria have emerged as a global crisis. Therefore, novel antibiotics and novel anti-infection strategies are urgently needed. Current antibiotics remain unsatisfactory due to poor targeting efficiency and poor drug penetration through the bacterial cell wall. Thus, targeted delivery of antibiotics into gram-negative bacteria should be a promising approach. Moreover, gram-negative bacteria can release lipopolysaccharide (LPS) to induce inflammatory response and septic shock, further increasing the disease burden. Hence, it is also promising to neutralize LPS while delivering antibiotics. This study aims to develop a multifunctional bacteria-targeting liposome that could enhance the delivery of antibiotics and adsorb LPS. METHODS: A polymyxin B (PMB)-modified liposomal system (P-Lipo) was developed as novel carrier of cinnamaldehyde (CA) by using a thin-film evaporation method. Liposome morphology, size, zeta potential, stability, entrapment efficiency, and in vitro release were systematically evaluated. The bacteria-targeting effect and LPS-neutralizing capacity of P-Lipo were evaluated both in vitro and in vivo. The antibacterial effect of CA-loaded P-Lipo was assessed in Escherichia coli (E. coli) O157:H7 and Pseudomonas aeruginosa (P. aeruginosa). Ultimately, the therapeutic effect of P-CA-Lipo was investigated in E. coli O157:H7-infected mice. RESULTS: P-Lipo was successfully synthesized and encapsulated with CA, which was well characterized. Both in vivo and in vitro experiments demonstrated that P-Lipo could efficiently target the E. coli after modification with PMB. Compared with free CA, CA-Lipo, and P-Lipo, P-CA-Lipo exhibited a significantly enhanced inhibitory effect on E. coli and P. aeruginosa. Further analysis demonstrated that P-CA-Lipo improved the bacterial uptake of CA and enhanced its antibacterial effect. It was also confirmed that P-Lipo could neutralize the LPS to avoid the inflammatory responses and inhibit the release of proinflammatory cytokines in both macrophages and mice. Finally, P-CA-Lipo inhibited E. coli-induced skin damage and death in mice and showed good biocompatibility. CONCLUSION: The P-Lipo could target E. coli by binding with LPS and enhancing the delivery and internalization of CA. In addition, P-Lipo could adsorb free LPS synergistically, thus promoting the infection management. We believe that this strategy can provide innovative insights into antibacterial agent delivery for the treatment of persistent and severe bacterial infections.


Asunto(s)
Escherichia coli , Liposomas , Acroleína/análogos & derivados , Animales , Bacterias , Ratones
3.
ACS Nano ; 15(3): 4173-4185, 2021 03 23.
Artículo en Inglés | MEDLINE | ID: mdl-33606516

RESUMEN

Antivirulence therapy by cell membrane coated nanoparticles has shown promise against bacterial infections. However, current approaches remain unsatisfactory when facing Escherichia coli (E. coli) infections, since the E. coli secretes multiple bacterial toxins including endotoxins and exotoxins that are challenging to eliminate simultaneously. What is worse, the absorptive scavengers normally rely on random contact of the diffuse toxins, which is not efficient. For the current cell membrane coated platform, the single type of cell membrane cannot fully meet the detoxing requirement facing multiple toxins. To address these problems, a polymyxin B (PMB)-modified, red blood cell (RBC)-mimetic hybrid liposome (P-RL) was developed. The P-RL was fabricated succinctly through fusion of PMB-modified lipids and the RBC membranes. By the strong interaction between PMB and the E. coli membrane, P-RL could attach and anchor to the E. coli; attributed to the fused RBC membrane and modified PMB, the P-RL could then efficiently neutralize both endotoxins and exotoxins from the toxin fountainhead. In vitro and in vivo results demonstrated the P-RL had a significant anchoring effect to E. coli. Moreover, compared with the existing RBC vesicles or PMB-modified liposomes, P-RL exhibited a superior therapeutic effect against RBC hemolysis, macrophage activation, and a mixed-toxin infection in mice. Potently, P-RL could inhibit E. coli O157:H7-induced skin damage, intestinal infection, and mouse death. Overall, the P-RL could potentially improve the detoxing efficiency and markedly expand the detoxification spectrum of current antivirulence systems, which provides different insights into drug-resistant E. coli treatment.


Asunto(s)
Infecciones por Escherichia coli , Escherichia coli O157 , Animales , Endotoxinas , Membrana Eritrocítica , Infecciones por Escherichia coli/tratamiento farmacológico , Liposomas , Ratones
4.
ACS Nano ; 13(4): 4148-4159, 2019 04 23.
Artículo en Inglés | MEDLINE | ID: mdl-30855941

RESUMEN

Pore-forming toxins (PFTs) are the most common bacterial virulence proteins and play a significant role in the pathogenesis of bacterial infections; thus, PFTs are an attractive therapeutic target in bacterial infections. Inspired by the pore-forming process and mechanism of PFTs, we designed an integrated hybrid nanovesicle-the erythroliposome (called the RM-PL)-for PFT detoxification by fusing natural red blood cell (RBC) membranes with artificial lipid membranes. The lipid and RBC membranes were mutually beneficial when integrated into a hybrid nanovesicle structure. The RBC membrane endowed RM-PLs with the capacity for detoxification, while the PEGylated lipid membrane stabilized the RM-PLs and greatly improved the detoxification capacity of the RBC membrane. With α-hemolysin (Hlα) as a model PFT, we demonstrated that RM-PLs could not only significantly reduce the toxicity of Hlα to erythrocytes in vitro but also effectively sponge Hlα in vivo and rescue mice from Hlα-induced damage. Moreover, the high detoxification capacity of RM-PLs was shown to be partly related to the expression of the Hlα receptor protein, a disintegrin and metalloproteinase domain-containing protein 10 on the RBC membrane. Consequently, as a component integrating natural and artificial materials, the erythroliposome nanoplatform inspires potential strategies for antivirulence therapy.


Asunto(s)
Membrana Eritrocítica/metabolismo , Proteínas Hemolisinas/aislamiento & purificación , Liposomas/uso terapéutico , Infecciones Estafilocócicas/terapia , Staphylococcus aureus/fisiología , Animales , Proteínas Hemolisinas/metabolismo , Liposomas/metabolismo , Lípidos de la Membrana/metabolismo , Lípidos de la Membrana/uso terapéutico , Membranas Artificiales , Ratones Endogámicos BALB C , Ratones Endogámicos ICR , Ratones Desnudos , Infecciones Estafilocócicas/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA