Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 8 de 8
Filtrar
Mais filtros

Bases de dados
Tipo de documento
Intervalo de ano de publicação
1.
J Nanobiotechnology ; 20(1): 205, 2022 Apr 27.
Artigo em Inglês | MEDLINE | ID: mdl-35477452

RESUMO

BACKGROUND: The reactive oxygen species (ROS) and inflammation, a critical contributor to tissue damage, is well-known to be associated with various disease. The kidney is susceptible to hypoxia and vulnerable to ROS. Thus, the vicious cycle between oxidative stress and renal hypoxia critically contributes to the progression of chronic kidney disease and finally, end-stage renal disease. Thus, delivering therapeutic agents to the ROS-rich inflammation site and releasing the therapeutic agents is a feasible solution. RESULTS: We developed a longer-circulating, inflammation-sensing, ROS-scavenging versatile nanoplatform by stably loading catalase-mimicking 1-dodecanethiol stabilized Mn3O4 (dMn3O4) nanoparticles inside ROS-sensitive nanomicelles (PTC), resulting in an ROS-sensitive nanozyme (PTC-M). Hydrophobic dMn3O4 nanoparticles were loaded inside PTC micelles to prevent premature release during circulation and act as a therapeutic agent by ROS-responsive release of loaded dMn3O4 once it reached the inflammation site. CONCLUSIONS: The findings of our study demonstrated the successful attenuation of inflammation and apoptosis in the IRI mice kidneys, suggesting that PTC-M nanozyme could possess promising potential in AKI therapy. This study paves the way for high-performance ROS depletion in treating various inflammation-related diseases.


Assuntos
Injúria Renal Aguda , Injúria Renal Aguda/tratamento farmacológico , Animais , Catalase , Feminino , Humanos , Hipóxia , Inflamação/tratamento farmacológico , Masculino , Camundongos , Estresse Oxidativo , Espécies Reativas de Oxigênio
2.
J Nanobiotechnology ; 19(1): 109, 2021 Apr 17.
Artigo em Inglês | MEDLINE | ID: mdl-33865397

RESUMO

BACKGROUND: Recently, we developed hydrophobically modified glycol chitosan (HGC) nanomicelles loaded with tacrolimus (TAC) (HGC-TAC) for the targeted renal delivery of TAC. Herein, we determined whether the administration of the HGC-TAC nanomicelles decreases kidney injury in a model of lupus nephritis. Lupus-prone female MRL/lpr mice were randomly assigned into three groups that received intravenous administration of either vehicle control, an equivalent dose of TAC, or HGC-TAC (0.5 mg/kg TAC) weekly for 8 weeks. Age-matched MRL/MpJ mice without Faslpr mutation were also treated with HGC vehicle and used as healthy controls. RESULTS: Weekly intravenous treatment with HGC-TAC significantly reduced genetically attributable lupus activity in lupus nephritis-positive mice. In addition, HGC-TAC treatment mitigated renal dysfunction, proteinuria, and histological injury, including glomerular proliferative lesions and tubulointerstitial infiltration. Furthermore, HGC-TAC treatment reduced renal inflammation and inflammatory gene expression and ameliorated increased apoptosis and glomerular fibrosis. Moreover, HGC-TAC administration regulated renal injury via the TGF-ß1/MAPK/NF-κB signaling pathway. These renoprotective effects of HGC-TAC treatment were more potent in lupus mice compared to those of TAC treatment alone. CONCLUSION: Our study indicates that weekly treatment with the HGC-TAC nanomicelles reduces kidney injury resulting from lupus nephritis by preventing inflammation, fibrosis, and apoptosis. This advantage of a new therapeutic modality using kidney-targeted HGC-TAC nanocarriers may improve drug adherence and provide treatment efficacy in lupus nephritis mice.


Assuntos
Quitosana/farmacologia , Quitosana/uso terapêutico , Nefrite Lúpica/tratamento farmacológico , Micelas , Tacrolimo/farmacologia , Tacrolimo/uso terapêutico , Animais , Apoptose , Quitosana/química , Feminino , Fibrose/patologia , Expressão Gênica , Interações Hidrofóbicas e Hidrofílicas , Inflamação , Rim/lesões , Rim/patologia , Nefrite Lúpica/patologia , Camundongos , Camundongos Endogâmicos MRL lpr , NF-kappa B/metabolismo , Transdução de Sinais
3.
Biomater Sci ; 2024 Jul 23.
Artigo em Inglês | MEDLINE | ID: mdl-39041781

RESUMO

Reactive oxygen species (ROS) drive processes in various pathological conditions serving as an attractive target for therapeutic strategies. This review highlights the development and use of ROS-dependent prodrug-based nanoscale carriers that has transformed many biomedical applications. Incorporating prodrugs into nanoscale carriers not only improves their stability and solubility but also enables site-specific drug delivery ultimately enhancing the therapeutic effectiveness of the nanoscale carriers. We critically examine recent advances in ROS-responsive nanoparticulate platforms, encompassing liposomes, polymeric nanoparticles, and inorganic nanocarriers. These platforms facilitate precise control over drug release upon encountering elevated ROS levels at disease sites, thereby minimizing off-target effects and maximizing therapeutic efficiency. Furthermore, we investigate the potential of combination therapies in which ROS-activated prodrugs are combined with other therapeutic agents and underscore their synergistic potential for treating multifaceted diseases. This comprehensive review highlights the immense potential of ROS-dependent prodrug-based nanoparticulate systems in revolutionizing biomedical applications; such nanoparticulate systems can facilitate selective and controlled drug delivery, reduce toxicity, and improve therapeutic outcomes for ROS-associated diseases.

4.
Adv Healthc Mater ; 13(17): e2304093, 2024 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-38409920

RESUMO

Combinatorial immuno-cancer therapy is recognized as a promising approach for efficiently treating malignant tumors. Yet, the development of multifunctional nanomedicine capable of precise tumor targeting, remote activation, and immune-regulating drug delivery remains a significant challenge. In this study, nanoparticles loaded with an immune checkpoint inhibitor (JQ-1) using polypyrrole/hyaluronic acid (PPyHA/JQ-1) are developed. These nanoparticles offer active tumor targeting, photothermal tumor ablation using near-infrared light, and laser-controlled JQ-1 release for efficient breast cancer treatment. When the molecular weight of HA varies (from 6.8 kDa to 3 MDa) in the PPyHA nanoparticles, it is found that the nanoparticles synthesized using 1 MDa HA, referred to as PPyHA (1 m), show the most suitable properties, including small hydrodynamic size, high surface HA contents, and colloidal stability. Upon 808 nm laser irradiation, PPyHA/JQ-1 elevates the temperature above 55 °C, which is sufficient for thermal ablation and active release of JQ-1 in the tumor microenvironment (TME). Notably, the controlled release of JQ-1 substantially inhibits the expression of cancer-promoting genes. Furthermore, PPyHA/JQ-1 effectively suppresses the expression of programmed cell death ligand 1 (PD-L1) and prolongs dendritic cell maturation and CD8+ T cell activation against the tumor both in vitro and in vivo. PPyHA/JQ-1 treatment simultaneously provides a significant tumor regression through photothermal therapy and immune checkpoint blockade, leading to a durable antitumor-immune response. Overall, "Three-in-one" immunotherapeutic photo-activable nanoparticles have the potential to be beneficial for a targeted combinatorial treatment approach for TNBC.


Assuntos
Antígeno B7-H1 , Imunoterapia , Nanopartículas , Fatores de Transcrição , Animais , Antígeno B7-H1/metabolismo , Nanopartículas/química , Camundongos , Imunoterapia/métodos , Humanos , Fatores de Transcrição/metabolismo , Feminino , Proteínas de Ciclo Celular/metabolismo , Linhagem Celular Tumoral , Proteínas Proto-Oncogênicas c-myc/metabolismo , Fototerapia/métodos , Ácido Hialurônico/química , Ácido Hialurônico/farmacologia , Inibidores de Checkpoint Imunológico/química , Inibidores de Checkpoint Imunológico/farmacologia , Camundongos Endogâmicos BALB C , Polímeros/química , Polímeros/farmacologia , Microambiente Tumoral/efeitos dos fármacos , Proteínas que Contêm Bromodomínio , Azepinas , Triazóis
5.
ACS Appl Bio Mater ; 2023 Aug 16.
Artigo em Inglês | MEDLINE | ID: mdl-37586084

RESUMO

Antiretroviral drugs are limited in their ability to target latent retroviral reservoirs in CD4+ T cells, highlighting the need for a T cell-targeted drug delivery system that activates the transcription of inactivated viral DNA in infected cells. Histone deacetylase inhibitors (HDACi) disrupt chromatin-mediated silencing of the viral genome and are explored in HIV latency reversal. But single drug formulations of HDACi are insufficient to elicit therapeutic efficacy, warranting combination therapy. Furthermore, protein kinase C activators (PKC) have shown latency reversal activity in HIV by activating the NF-κB signaling pathway. Combining HDACi (SAHA) with PKC (PMA) activators enhances HIV reservoir activation by promoting chromatin decondensation and subsequent transcriptional activation. In this study, we developed a mixed nanomicelle (PD-CR4) drug delivery system for simultaneous targeting of HIV-infected CD4+ T cells with two drugs, suberoylanilide hydroxamic acid (SAHA) and phorbol 12-myristate 13-acetate (PMA). SAHA is a HDACi that promotes chromatin decondensation, while PMA is a PKC agonist that enhances transcriptional activation. The physicochemical properties of the formulated PD-CR4 nanoparticles were characterized by NMR, CMC, DLS, and TEM analyses. Further, we investigated in vitro safety profiles, targeting efficacy, and transcriptional activation of inactivated HIV reservoir cells. Our results suggest that we successfully prepared a targeted PD system with dual drug loading. We have compared latency reversal efficacy of a single drug nanoformulation and combination drug nanoformulation. Final PD-SP-CR4 successfully activated infected CD4+ T cell reservoirs and showed enhanced antigen release from HIV reservoir T cells, compared with the single drug treatment group as expected. To summarize, our data shows PD-SP-CR4 has potential T cell targeting efficiency and efficiently activated dormant CD4+ T cells. Our data indicate that a dual drug-loaded particle has better therapeutic efficacy than a single loaded particle as expected. Hence, PD-CR4 can be further explored for HIV therapeutic drug delivery studies.

6.
Biomater Sci ; 11(21): 7188-7202, 2023 Oct 24.
Artigo em Inglês | MEDLINE | ID: mdl-37750339

RESUMO

Magnetic hyperthermia has attracted considerable attention for efficient cancer therapy because of its noninvasive nature, deep tissue penetration, and minimal damage to healthy tissues. Herein, we have fused cancer cell membrane fragments with lipids and cloaked them on magnetic nanorings to form targeted Fe nanorings (TF) for tumor-targeted magnetic hyperthermia-induced tumor ablation. In our approach, cell membrane fragments from cancer cells were fused with lipids to form vesicles, which could efficiently encapsulate magnetic nanorings, thereby forming TF. We observed that TF have high tumor uptake via homotypic targeting, where cancer cells take up TF through membrane fusion. Under an external alternating magnetic field (AMF), TF accumulated in the tumors are heated, driving magnetic-hyperthermia-induced tumor cell death. Our in vitro studies show that self-targeting TF efficiently localized in cancer cells and induced cell death with an AMF, which was shown by a live/dead assay. Our findings demonstrate the potential of TF in tumor ablation, thereby making them promising and efficient nanosystems for tumor-targeted theranostics.


Assuntos
Hipertermia Induzida , Nanopartículas de Magnetita , Linhagem Celular Tumoral , Membrana Celular , Fenômenos Magnéticos , Lipídeos , Campos Magnéticos
7.
Biomater Sci ; 10(5): 1248-1256, 2022 Mar 02.
Artigo em Inglês | MEDLINE | ID: mdl-35079755

RESUMO

Excessive inflammatory response during sepsis causes irreversible damage to healthy tissues and results in multi-organ failure. During infection, bacterial endotoxin-triggered inflammatory responses in macrophages facilitate the recruitment of circulating leukocytes, including neutrophils and monocytes. A key component that aggravates the systemic inflammatory response is the generation of stable reactive oxygen species such as hydrogen peroxide (H2O2). In this study, we present a versatile strategy to reduce the activation of tissue-resident macrophages and prevent leukocyte infiltration in an LPS-induced endotoxemia model. We designed and synthesized hyaluronic acid-stabilized Prussian blue (HAPB) nanoparticles and validated their activity in the dismutation of H2O2 in LPS-induced tissue-resident macrophages. Hyaluronic acid provided stability and enhanced the intracellular uptake of insoluble Prussian blue via the CD44 receptor on LPS-activated macrophages. Following HAPB administration to an LPS-induced peritonitis murine model, the level of M1 inflammatory macrophage population decreased, and the infiltration of neutrophils along with monocytes was suppressed. Overall, we have developed biocompatible Prussian blue nanoparticles to ameliorate inflammatory stress in LPS-induced endotoxemia by scavenging the intracellular peroxide thereby inhibiting inflammatory cascade in tissue-resident macrophages. Therefore, HAPB nanoparticles may potentially be used as novel nano-stress relievers in sepsis. The nanomaterials may have clinical application in sepsis and in other inflammatory diseases involving peroxides as key inflammatory agents.


Assuntos
Nanopartículas , Peritonite , Animais , Ferrocianetos , Ácido Hialurônico , Peróxido de Hidrogênio , Lipopolissacarídeos , Macrófagos , Camundongos , Estresse Oxidativo , Peritonite/induzido quimicamente , Peritonite/tratamento farmacológico
8.
Int J Pharm ; 600: 120497, 2021 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-33753165

RESUMO

ACE inhibitors or angiotensin II receptor blockers (ACEi/ARBs) have been a cornerstone of the management in kidney disease, but their use is often limited by undesired systemic effects, such as symptomatic hypotension. To minimize the extra-renal effects of ACEi/ARBs, we formulated hydrophobically modified glycol chitosan (HGC) nanomicelles releasing olmesartan (HGC-Olm) that specifically accumulated in the kidney, and investigated whether kidney-specific delivery of olmesartan by HGC nanomicelles could ameliorate organ damage in Col4a3-/- mouse, a murine model of progressive chronic kidney disease mimicking human Alport syndrome. Ex vivo tracing demonstrated that intravenously injected HGC-Olm nanomicelles were specifically delivered to the kidney, with sustained release of olmesartan for more than 48 h. Contrary to the conventional delivery of olmesartan via oral route, injection of HGC-Olm nanomicelles did not alter blood pressure in Col4a3-/- mice. Immunohistochemistry revealed that HGC nanomicelles were diffusely distributed from the cortex and glomeruli to the outer medulla, sparing the inner medulla. Phenotypic analysis showed that the attenuation of kidney fibrosis in the kidney of Col4a3-/- mice by HGC-Olm nanomicelles was comparable to that noted with conventionally delivered olmesartan. Therefore, our results suggest that HGC-Olm nanomicelles could be a safe and effective alternative drug delivery system for kidney diseases.


Assuntos
Nefrite Hereditária , Antagonistas de Receptores de Angiotensina , Inibidores da Enzima Conversora de Angiotensina , Animais , Modelos Animais de Doenças , Imidazóis , Rim , Camundongos , Nefrite Hereditária/genética , Tetrazóis
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA