Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 583
Filtrar
1.
Int J Nanomedicine ; 19: 6603-6618, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38979533

RESUMEN

Objective: Ovarian cancer cells are prone to acquire tolerance to chemotherapeutic agents, which seriously affects clinical outcomes. The development of novel strategies to enhance the targeting of chemotherapeutic agents to overcome drug resistance and minimize side effects is significant for improving the clinical outcomes of ovarian cancer patients. Methods: We employed folic acid (FA)-modified ZIF-90 nanomaterials (FA-ZIF-90) to deliver the chemotherapeutic drug, cisplatin (DDP), via dual targeting to improve its targeting to circumvent cisplatin resistance in ovarian cancer cells, especially by targeting mitochondria. FA-ZIF-90/DDP could rapidly release DDP in response to dual stimulation of acidity and ATP in tumor cells. Results: FA-ZIF-90/DDP showed good blood compatibility. It was efficiently taken up by human ovarian cancer cisplatin-resistant cells A2780/DDP and aggregated in the mitochondrial region. FA-ZIF-90/DDP significantly inhibited the mitochondrial activity and metastatic ability of A2780/DDP cells. In addition, it effectively induced apoptosis in A2780/DDP cells and overcame cisplatin resistance. In vivo experiments showed that FA-ZIF-90/DDP increased the accumulation of DDP in tumor tissues and significantly inhibited tumor growth. Conclusion: FA-modified ZIF-90 nanocarriers can improve the tumor targeting and anti-tumor effects of chemotherapeutic drugs, reduce toxic side effects, and are expected to be a novel therapeutic strategy to reverse drug resistance in ovarian cancer.


Asunto(s)
Antineoplásicos , Apoptosis , Cisplatino , Resistencia a Antineoplásicos , Ácido Fólico , Imidazoles , Neoplasias Ováricas , Zeolitas , Femenino , Cisplatino/farmacología , Cisplatino/química , Cisplatino/farmacocinética , Neoplasias Ováricas/tratamiento farmacológico , Neoplasias Ováricas/patología , Humanos , Resistencia a Antineoplásicos/efectos de los fármacos , Animales , Zeolitas/química , Línea Celular Tumoral , Antineoplásicos/farmacología , Antineoplásicos/química , Antineoplásicos/administración & dosificación , Ácido Fólico/química , Ácido Fólico/farmacología , Imidazoles/química , Imidazoles/farmacología , Imidazoles/administración & dosificación , Apoptosis/efectos de los fármacos , Sistemas de Liberación de Medicamentos/métodos , Mitocondrias/efectos de los fármacos , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Portadores de Fármacos/química , Estructuras Metalorgánicas/química , Estructuras Metalorgánicas/farmacología , Ensayos Antitumor por Modelo de Xenoinjerto
2.
J Nanobiotechnology ; 22(1): 387, 2024 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-38951841

RESUMEN

Metal-organic frameworks (MOFs) are metal-organic skeleton compounds composed of self-assembled metal ions or clusters and organic ligands. MOF materials often have porous structures, high specific surface areas, uniform and adjustable pores, high surface activity and easy modification and have a wide range of prospects for application. MOFs have been widely used. In recent years, with the continuous expansion of MOF materials, they have also achieved remarkable results in the field of antimicrobial agents. In this review, the structural composition and synthetic modification of MOF materials are introduced in detail, and the antimicrobial mechanisms and applications of these materials in the healing of infected wounds are described. Moreover, the opportunities and challenges encountered in the development of MOF materials are presented, and we expect that additional MOF materials with high biosafety and efficient antimicrobial capacity will be developed in the future.


Asunto(s)
Estructuras Metalorgánicas , Cicatrización de Heridas , Estructuras Metalorgánicas/química , Estructuras Metalorgánicas/farmacología , Cicatrización de Heridas/efectos de los fármacos , Humanos , Animales , Antiinfecciosos/farmacología , Antiinfecciosos/química , Antibacterianos/farmacología , Antibacterianos/química , Porosidad , Infección de Heridas/tratamiento farmacológico
3.
ACS Appl Mater Interfaces ; 16(24): 30728-30741, 2024 Jun 19.
Artículo en Inglés | MEDLINE | ID: mdl-38847598

RESUMEN

The prevalence of pathogenic bacterial infections with high morbidity and mortality poses a widespread challenge to the healthcare system. Therefore, it is imperative to develop nanoformulations capable of adaptively releasing antimicrobial factors and demonstrating multimodal synergistic antimicrobial activity. Herein, an NIR-activated multifunctional synergistic antimicrobial nanospray MXene/ZIF-90@ICG was prepared by incorporating ZIF-90@ICG nanoparticles onto MXene-NH2 nanosheets. MXene/ZIF-90@ICG can on-demand release the antimicrobial factors MXenes, ICG, and Zn2+ in response to variations in pH and ATP levels within the bacterial infection microenvironment. Under NIR radiation, the combination of MXenes, Zn2+, and ICG generated a significant amount of ROS and elevated heat, thereby enhancing the antimicrobial efficacy of PDT and PTT. Meanwhile, NIR excitation could accelerate the further release of ICG and Zn2+, realizing the multimodal synergistic antibacterial effect of PDT/PTT/Zn2+. Notably, introducing MXenes improved the dispersion of the synthesized antimicrobial nanoparticles in aqueous solution, rendering MXene/ZIF-90@ICG a candidate for application as a nanospray. Importantly, MXene/ZIF-90@ICG demonstrated antimicrobial activity and accelerated wound healing in the constructed in vivo subcutaneous Staphylococcus aureus infection model with NIR activation, maintaining a favorable biosafety level. Therefore, MXene/ZIF-90@ICG holds promise as an innovative nanospray for adaptive multimodal synergistic and efficient antibacterial applications with NIR activation.


Asunto(s)
Adenosina Trifosfato , Antibacterianos , Verde de Indocianina , Rayos Infrarrojos , Staphylococcus aureus , Cicatrización de Heridas , Antibacterianos/farmacología , Antibacterianos/química , Animales , Cicatrización de Heridas/efectos de los fármacos , Concentración de Iones de Hidrógeno , Staphylococcus aureus/efectos de los fármacos , Adenosina Trifosfato/metabolismo , Adenosina Trifosfato/química , Ratones , Verde de Indocianina/química , Verde de Indocianina/farmacología , Nanopartículas/química , Pruebas de Sensibilidad Microbiana , Estructuras Metalorgánicas/química , Estructuras Metalorgánicas/farmacología , Escherichia coli/efectos de los fármacos , Humanos , Fotoquimioterapia
4.
Inorg Chem ; 63(26): 12377-12384, 2024 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-38902911

RESUMEN

Pathogenic bacteria have consistently posed a formidable challenge to human health, creating the critical need for effective antibacterial solutions. In response, enzyme-metal-organic framework (MOF) composites have emerged as a promising class of antibacterial agents. This study focuses on the development of an enzyme-MOF composite based on HZIF-8, incorporating the advantages of simple synthesis, ZIF-8 antibacterial properties, lysozyme hydrolysis, and high biological safety. Through a one-pot method, core-shell nanoparticles (HZIF-8) were synthesized. This structure enables efficient immobilization of lysozyme and lactoferrin within the HZIF-8, resulting in the formation of the lysozyme-lactoferrin@HZIF-8 (LYZ-LF@HZIF-8) composite. Upon exposure to light irradiation, HZIF-8 itself possessed antibacterial properties. Lysozyme initiated the degradation of bacterial peptidoglycan and lactoferrin synergistically enhanced the antibacterial effect of lysozyme. All of the above ultimately contributed to comprehensive antibacterial activity. Antibacterial assessments demonstrated the efficacy of the LYZ-LF@HZIF-8 composite, effectively eradicating Staphylococcus aureus at a cell density of 1.5 × 106 CFU/mL with a low dosage of 200 µg/mL and completely inactivating Escherichia coli at 400 µg/mL with the same cell density. The enzyme-MOF composite exhibited significant and durable antibacterial efficacy, with no apparent cytotoxicity in vitro, thereby unveiling expansive prospects for applications in the medical and food industries.


Asunto(s)
Antibacterianos , Escherichia coli , Lactoferrina , Estructuras Metalorgánicas , Pruebas de Sensibilidad Microbiana , Muramidasa , Staphylococcus aureus , Zeolitas , Muramidasa/farmacología , Muramidasa/química , Muramidasa/metabolismo , Lactoferrina/química , Lactoferrina/farmacología , Estructuras Metalorgánicas/química , Estructuras Metalorgánicas/farmacología , Estructuras Metalorgánicas/síntesis química , Antibacterianos/farmacología , Antibacterianos/química , Antibacterianos/síntesis química , Staphylococcus aureus/efectos de los fármacos , Escherichia coli/efectos de los fármacos , Zeolitas/química , Zeolitas/farmacología , Imidazoles/química , Imidazoles/farmacología , Imidazoles/síntesis química , Porosidad , Propiedades de Superficie , Tamaño de la Partícula , Enzimas Inmovilizadas/química , Enzimas Inmovilizadas/farmacología
5.
ACS Appl Mater Interfaces ; 16(25): 32118-32127, 2024 Jun 26.
Artículo en Inglés | MEDLINE | ID: mdl-38862123

RESUMEN

The SARS-CoV-2 (COVID-19) pandemic outbreak led to enormous social and economic repercussions worldwide, felt even to this date, making the design of new therapies to combat fast-spreading viruses an imperative task. In the face of this, diverse cutting-edge nanotechnologies have risen as promising tools to treat infectious diseases such as COVID-19, as well as challenging illnesses such as cancer and diabetes. Aside from these applications, nanoscale metal-organic frameworks (nanoMOFs) have attracted much attention as novel efficient drug delivery systems for diverse pathologies. However, their potential as anti-COVID-19 therapeutic agents has not been investigated. Herein, we propose a pioneering anti-COVID MOF approach by studying their potential as safe and intrinsically antiviral agents through screening various nanoMOF. The iron(III)-trimesate MIL-100 showed a noteworthy antiviral effect against SARS-CoV-2 at the micromolar range, ensuring a high biocompatibility profile (90% of viability) in a real infected human cellular scenario. This research effectively paves the way toward novel antiviral therapies based on nanoMOFs, not only against SARS-CoV-2 but also against other challenging infectious and/or pulmonary diseases.


Asunto(s)
Antivirales , Tratamiento Farmacológico de COVID-19 , COVID-19 , Estructuras Metalorgánicas , SARS-CoV-2 , Estructuras Metalorgánicas/química , Estructuras Metalorgánicas/farmacología , Humanos , SARS-CoV-2/efectos de los fármacos , Antivirales/química , Antivirales/farmacología , Antivirales/uso terapéutico , COVID-19/virología , Chlorocebus aethiops , Células Vero , Supervivencia Celular/efectos de los fármacos
6.
Dalton Trans ; 53(26): 10928-10937, 2024 Jul 02.
Artículo en Inglés | MEDLINE | ID: mdl-38888155

RESUMEN

Multifunctional materials that combine antimicrobial properties with the ability to stimulate bone formation are needed to overcome the problem of infected bone defects. As a novel approach, a new composite based on bioactive glass nanoparticles in a simple system of SiO2-CaO (BG) coated with NH4[Cu3(µ3-OH)(µ3-4-carboxypyrazolato)3] (Cu-MOF) with additionally anchored silver nanoparticles (AgNPs) was proposed. Ag@Cu-MOF@BG obtained by the spin coating approach in the form of a disc was characterized using PXRD, ATR-FTIR, XPS, ICP-OES, and TEM. Importantly, the material retained its bioactivity, although ion exchange in the bioactive glass administered as a disc is limited. Hydroxyapatite (HA) formation was identified in TEM images after 7 days of immersion of the composite in a physiological-like buffer (pH 7.4, 37 °C). The Cu and Ag contents of Ag@Cu-MOF@BG were as low as 0.013 and 0.018 wt% respectively, but the slow release of the AgNPs ensured its antibacterial nature. Ag@Cu-MOF@BG exhibited antibacterial activity against all tested bacteria (E. coli, S. aureus, P. aeruginosa, and K. pneumoniae) with the diameter of the inhibition zones of their growth between 8 and 10 mm and the reduction index determined to be ≥3. Moreover, the biocompatibility of the new composite has been demonstrated, as shown by cell culture assays with human dermal fibroblasts (HDFs). The results from the migration test also proved that the HDF cell's phenotypic properties were not changed, and the cell adhesion and migration ability were the same as in control indirect assays.


Asunto(s)
Antibacterianos , Materiales Biocompatibles , Vidrio , Nanopartículas del Metal , Estructuras Metalorgánicas , Pruebas de Sensibilidad Microbiana , Plata , Antibacterianos/farmacología , Antibacterianos/química , Antibacterianos/síntesis química , Plata/química , Plata/farmacología , Nanopartículas del Metal/química , Estructuras Metalorgánicas/química , Estructuras Metalorgánicas/farmacología , Vidrio/química , Humanos , Materiales Biocompatibles/química , Materiales Biocompatibles/farmacología , Materiales Biocompatibles/síntesis química , Cobre/química , Cobre/farmacología , Staphylococcus aureus/efectos de los fármacos , Escherichia coli/efectos de los fármacos , Fibroblastos/efectos de los fármacos , Pseudomonas aeruginosa/efectos de los fármacos , Klebsiella pneumoniae/efectos de los fármacos
7.
ACS Appl Mater Interfaces ; 16(26): 33093-33105, 2024 Jul 03.
Artículo en Inglés | MEDLINE | ID: mdl-38884171

RESUMEN

The morphological features of materials significantly influence their interactions with cells, consequently affecting the cellular uptake of these materials. In this study, we examine the cellular uptake behavior of spherical metal-organic frameworks (MOFs) and petaloid MOFs, both possessing similar sizes and compositions. In comparison to spherical MOFs, dendritic cells (DCs) and macrophages exhibit superior phagocytic uptake of petaloid MOFs. Next, the results demonstrate that R848@petaloid MOFs more effectively promote the repolarization of tumor-associated macrophages (TAMs) from the M2 to M1 phenotype and the maturation of DCs. More importantly, the R848-loaded petaloid MOFs are found to significantly enhance the therapeutic effects of radiotherapy (RT) by eliciting antitumor responses. Furthermore, R848@petaloid MOFs combined with RT and αPD-L1 elicit a potent abscopal effect, effectively suppressing tumor metastasis. Therefore, this work proposes a new strategy to enhance the uptake of immunomodulators by immune cells through modulating the morphology of drug delivery carriers.


Asunto(s)
Imidazoles , Estructuras Metalorgánicas , Estructuras Metalorgánicas/química , Estructuras Metalorgánicas/farmacología , Ratones , Animales , Imidazoles/química , Imidazoles/farmacología , Células Dendríticas/efectos de los fármacos , Células Dendríticas/inmunología , Portadores de Fármacos/química , Antineoplásicos/química , Antineoplásicos/farmacología , Células RAW 264.7 , Línea Celular Tumoral , Ratones Endogámicos C57BL , Macrófagos/efectos de los fármacos , Macrófagos/metabolismo , Macrófagos/inmunología , Femenino , Antígeno B7-H1/metabolismo , Humanos , Neoplasias/tratamiento farmacológico , Neoplasias/patología , Neoplasias/inmunología
8.
ACS Appl Mater Interfaces ; 16(26): 33070-33080, 2024 Jul 03.
Artículo en Inglés | MEDLINE | ID: mdl-38904394

RESUMEN

Nanomaterials have been extensively exploited in tumor treatment, leading to numerous innovative strategies for cancer therapy. While nanomedicines present immense potential, their application in cancer therapy is characterized by significant complexity and unpredictability, especially regarding biocompatibility and anticancer efficiency. These considerations underscore the essential need for the development of ex vivo research models, which provide invaluable insights and understanding into the biosafety and efficacy of nanomedicines in oncology. Fortunately, the emergence of organoid technology offers a novel approach to the preclinical evaluation of the anticancer efficacy of nanomedicines in vitro. Hence, in this study, we constructed intestine and hepatocyte organoid models (Intestine-orgs and Hep-orgs) for assessing intestinal and hepatic toxicity at the microtissue level. We utilized three typical metal-organic frameworks (MOFs), ZIF-8, ZIF-67, and MIL-125, as nanomedicines to further detect their interactions with organoids. Subsequently, the MIL-125 with biocompatibility loaded methotrexate (MTX), forming the nanomedicine (MIL-125-PEG-MTX), indicated a high loading efficiency (82%) and a well-release capability in an acid microenvironment. More importantly, the anticancer effect of the nanomedicine was investigated using an in vitro patient-derived organoids (PDOs) model, achieving inhibition rates of 48% and 78% for PDO-1 and PDO-2, respectively, demonstrating that PDOs could predict clinical response and facilitate prospective therapeutic selection. These achievements presented great potential for organoid-based ex vivo models for nano theragnostic evaluation in biosafety and function.


Asunto(s)
Estructuras Metalorgánicas , Nanomedicina , Organoides , Estructuras Metalorgánicas/química , Estructuras Metalorgánicas/farmacología , Humanos , Organoides/efectos de los fármacos , Organoides/metabolismo , Nanomedicina/métodos , Metotrexato/farmacología , Metotrexato/química , Metotrexato/uso terapéutico , Neoplasias/tratamiento farmacológico , Neoplasias/patología , Hepatocitos/efectos de los fármacos , Hepatocitos/metabolismo , Antineoplásicos/química , Antineoplásicos/farmacología , Antineoplásicos/uso terapéutico , Intestinos/efectos de los fármacos , Intestinos/patología , Animales
9.
Carbohydr Polym ; 340: 122200, 2024 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-38857994

RESUMEN

Cyclodextrin metal-organic framework (CD-MOF) is an edible and porous material that can serve as a template for synthesizing small-sized metal nanoparticles. However, its highly hydrophilic nature has limited its wider application. Herein, ultra-small gold nanoparticles (U-AuNPs) were loaded into CD-MOF to produce a composite material Au@CD-MOF. The CD-MOF was utilized as a template to control the size of the AuNPs. The synthesized Au@CD-MOF was easily dispersible in aqueous medium and its released U-AuNPs exhibited effective water dispersion stability within 120 days. Additionally, compared to gold nanoparticles prepared using traditional methods (T-AuNPs), the U-AuNPs exhibited superior antibacterial properties. Furthermore, hydrophilic Au@CD-MOF was incorporated into a hydrophobic polydimethylsiloxane (PDMS) matrix (Au@CD-MOF/PDMS) to achieve a humidity-responsive antibacterial function. The composite membrane exhibited remarkable responsiveness to humidity, showing almost no release of U-AuNPs at 0 % humidity. However, it exhibited approximately 89 % release within 1 h, and complete release of U-AuNPs was observed within 4 h under 100 % humidity. These findings highlight the successful preparation of a humidity-responsive antibacterial composite membrane, which has great potential applications in various scenarios, particularly in the field of antibacterial food packaging.


Asunto(s)
Antibacterianos , Ciclodextrinas , Oro , Humedad , Nanopartículas del Metal , Estructuras Metalorgánicas , Oro/química , Estructuras Metalorgánicas/química , Estructuras Metalorgánicas/farmacología , Antibacterianos/farmacología , Antibacterianos/química , Nanopartículas del Metal/química , Ciclodextrinas/química , Tamaño de la Partícula , Pruebas de Sensibilidad Microbiana , Interacciones Hidrofóbicas e Hidrofílicas , Escherichia coli/efectos de los fármacos , Staphylococcus aureus/efectos de los fármacos
10.
J Mater Chem B ; 12(25): 6242-6256, 2024 Jun 27.
Artículo en Inglés | MEDLINE | ID: mdl-38842217

RESUMEN

Designing artificial nano-enzymes for scavenging reactive oxygen species (ROS) in chondrocytes (CHOs) is considered the most feasible pathway for the treatment of osteoarthritis (OA). However, the accumulation of ROS due to the amount of nano-enzymatic catalytic site exposure and insufficient oxygen supply seriously threatens the clinical application of this therapy. Although metal-organic framework (MOF) immobilization of artificial nano-enzymes to enhance active site exposure has been extensively studied, artificial nano-enzymes/MOFs for ROS scavenging in OA treatment are still lacking. In this study, a biocompatible lubricating hydrogel-loaded iron-doped zeolitic imidazolate framework-8 (Fe/ZIF-8/Gel) centrase was engineered to scavenge endogenous overexpressed ROS synergistically generating dissolved oxygen and enhancing sustained lubrication for CHOs as a ternary artificial nano-enzyme. This property enabled the nano-enzymatic hydrogels to mitigate OA hypoxia and inhibit oxidative stress damage successfully. Ternary strategy-based therapies show excellent cartilage repair in vivo. The experimental results suggest that nano-enzyme-enhanced lubricating hydrogels are a potentially effective OA treatment and a novel strategy.


Asunto(s)
Condrocitos , Hidrogeles , Especies Reactivas de Oxígeno , Hidrogeles/química , Hidrogeles/farmacología , Animales , Condrocitos/metabolismo , Condrocitos/efectos de los fármacos , Condrocitos/citología , Especies Reactivas de Oxígeno/metabolismo , Estructuras Metalorgánicas/química , Estructuras Metalorgánicas/farmacología , Osteoartritis/tratamiento farmacológico , Materiales Biocompatibles/química , Materiales Biocompatibles/farmacología , Cartílago/efectos de los fármacos , Cartílago/metabolismo , Tamaño de la Partícula , Humanos , Zeolitas/química
11.
Chem Commun (Camb) ; 60(51): 6476-6487, 2024 Jun 20.
Artículo en Inglés | MEDLINE | ID: mdl-38853690

RESUMEN

Pyroptosis is regarded as a promising strategy to modulate tumor immune microenvironments for anticancer therapy. Although pyroptosis inducers have been extensively explored in the biomedical field, their drug resistance, off-targeting capacity, and adverse effects do not fulfill the growing demands of therapy. Nowadays, metal-organic frameworks (MOFs) with unique structures and facile synthesis/functionalization characteristics have shown great potential in anticancer therapy. The flexible choices of metal ions and ligands endow MOFs with inherent anti-cancer efficiency, whereas the porous structures in MOFs make them ideal vehicles for delivering various chemodrug-based pyroptosis inducers. In this review, we provide the latest advances in MOF-based materials to evoke pyroptosis and give a brief but comprehensive review of the different types of MOFs for pyroptosis-mediated cancer therapy. Finally, we also discuss the current challenges of MOF-based pyroptosis inducers and their future prospects in this field.


Asunto(s)
Antineoplásicos , Estructuras Metalorgánicas , Neoplasias , Piroptosis , Estructuras Metalorgánicas/química , Estructuras Metalorgánicas/farmacología , Piroptosis/efectos de los fármacos , Humanos , Antineoplásicos/farmacología , Antineoplásicos/química , Neoplasias/tratamiento farmacológico , Neoplasias/patología , Animales
12.
ACS Appl Mater Interfaces ; 16(25): 32058-32077, 2024 Jun 26.
Artículo en Inglés | MEDLINE | ID: mdl-38872401

RESUMEN

The development of growth factor-free biomaterials for bone tissue regeneration with anti-infection and anti-inflammatory activities remains challenging. Black phosphorus nanosheets (BPNs), with distinctive attributes, including photothermal conversion and calcium ion chelation, offer potential for use in bone tissue engineering and infection prevention. However, BPNs are prone to oxidation and degradation in aqueous environments, and methods to stabilize BPNs for long-term bone repair remain insufficient. Herein, zeolitic imidazolate framework-8 (ZIF-8) was used to stabilize BPNs via in situ crystallization onto the surface of BPNs (BP@ZIF-8 nanocomposite). A novel injectable dual-component hydrogel comprising gelatin methacryloyl (GelMA) and methacrylate-modified hyaluronic acid (HAMA) was used as a BP@ZIF-8 nanocomposite carrier (GelMA/HAMA/BP@ZIF-8). The BP@ZIF-8 nanocomposite could effectively protect internal BPNs from oxidation and enhance the long-term photothermal performance of the hydrogel in both in vitro and in vivo settings. The GelMA/HAMA/BP@ZIF-8 hydrogel was injectable and exhibited outstanding performance for photothermal conversion, mechanical strength, and biodegradability, as well as excellent photothermal antibacterial activity against Staphylococcus aureus and Escherichia coli in vitro and in an in vivo rat model. The GelMA/HAMA/BP@ZIF-8 hydrogel also provided a microenvironment conducive to osteogenic differentiation, promoting the transformation of M2 macrophages and inhibiting inflammatory responses. Furthermore, the hydrogel promoted bone regeneration and had a synergistic effect with near-infrared irradiation in a rat skull-defect model. Transcriptome sequencing analysis revealed that the PI3K-AKT- and calcium-signaling pathways may be involved in promoting osteogenic differentiation induced by the GH-BZ hydrogel. This study presents an innovative, multifaceted solution to the challenges of bone tissue regeneration with antibacterial and anti-inflammatory effects, providing insights into the design of smart biomaterials with dual therapeutic capabilities.


Asunto(s)
Antibacterianos , Escherichia coli , Hidrogeles , Osteogénesis , Fósforo , Staphylococcus aureus , Zeolitas , Antibacterianos/química , Antibacterianos/farmacología , Hidrogeles/química , Hidrogeles/farmacología , Animales , Osteogénesis/efectos de los fármacos , Fósforo/química , Escherichia coli/efectos de los fármacos , Staphylococcus aureus/efectos de los fármacos , Ratas , Zeolitas/química , Zeolitas/farmacología , Gelatina/química , Gelatina/farmacología , Ratones , Ácido Hialurónico/química , Ácido Hialurónico/farmacología , Ratas Sprague-Dawley , Metacrilatos/química , Metacrilatos/farmacología , Pruebas de Sensibilidad Microbiana , Estructuras Metalorgánicas/química , Estructuras Metalorgánicas/farmacología , Nanocompuestos/química , Células RAW 264.7 , Regeneración Ósea/efectos de los fármacos , Nanoestructuras/química
13.
J Hazard Mater ; 474: 134807, 2024 Aug 05.
Artículo en Inglés | MEDLINE | ID: mdl-38850939

RESUMEN

Nanocrop protectants have attracted much attention as sustainable platforms for controlling pests and diseases and improving crop nutrition. Here, we reported the fungicidal activity and disease inhibition potential of pectin-coated metal-iron organic framework nanoparticles (Fe-MOF-PT NPs) against rice stripe blight (RSB). An in vitro bacterial inhibition assay showed that Fe-MOF-PT NPs (80 mg/L) significantly inhibited mycelial growth and nucleus formation. The Fe-MOF-PT NPs adsorbed to the surface of mycelia and induced toxicity by disrupting cell membranes, mitochondria, and DNA. The results of a nontargeted metabolomics analysis showed that the metabolites of amino acids and their metabolites, heterocyclic compounds, fatty acids, and nucleotides and their metabolites were significantly downregulated after treatment with 80 mg/L NPs. The difference in metabolite abundance between the CK and Fe-MOF-PT NPs (80 mg/L) treatment groups was mainly related to nucleotide metabolism, pyrimidine metabolism, purine metabolism, fatty acid metabolism, and amino acid metabolism. The results of the greenhouse experiment showed that Fe-MOF-PT NPs improved rice resistance to R. solani by inhibiting mycelial invasion, enhancing antioxidant enzyme activities, activating the jasmonic acid signaling pathway, and enhancing photosynthesis. These findings indicate the great potential of Fe-MOF-PT NPs as a new RSB disease management strategy and provide new insights into plant fungal disease management.


Asunto(s)
Hierro , Estructuras Metalorgánicas , Oryza , Pectinas , Enfermedades de las Plantas , Rhizoctonia , Oryza/metabolismo , Oryza/efectos de los fármacos , Oryza/microbiología , Rhizoctonia/efectos de los fármacos , Enfermedades de las Plantas/prevención & control , Enfermedades de las Plantas/microbiología , Hierro/química , Hierro/metabolismo , Pectinas/química , Pectinas/farmacología , Estructuras Metalorgánicas/química , Estructuras Metalorgánicas/farmacología , Fungicidas Industriales/farmacología , Fungicidas Industriales/química , Fungicidas Industriales/toxicidad , Nanopartículas del Metal/química , Nanopartículas del Metal/toxicidad , Resistencia a la Enfermedad/efectos de los fármacos
15.
ACS Nano ; 18(27): 17852-17868, 2024 Jul 09.
Artículo en Inglés | MEDLINE | ID: mdl-38939981

RESUMEN

The discovery of cuproptosis, a copper-dependent mechanism of programmed cell death, has provided a way for cancer treatment. However, cuproptosis has inherent limitations, including potential cellular harm, the lack of targeting, and insufficient efficacy as a standalone treatment. Therefore, exogenously controlled combination treatments have emerged as key strategies for cuproptosis-based oncotherapy. In this study, a Cu2-xSe@cMOF nanoplatform was constructed for combined sonodynamic/cuproptosis/gas therapy. This platform enabled precise cancer cotreatment, with external control allowing the selective induction of cuproptosis in cancer cells. This approach effectively prevented cancer metastasis and recurrence. Furthermore, Cu2-xSe@cMOF was combined with the antiprogrammed cell death protein ligand-1 antibody (aPD-L1), and this combination maximized the advantages of cuproptosis and immune checkpoint therapy. Additionally, under ultrasound irradiation, the H2Se gas generated from Cu2-xSe@cMOF induced cytotoxicity in cancer cells. Further, it generated reactive oxygen species, which hindered cell survival and proliferation. This study reports an externally controlled system for cuproptosis induction that combines a carbonized metal-organic framework with aPD-L1 to enhance cancer treatment. This precision and reinforced cuproptosis cancer therapy platform could be valuable as an effective therapeutic agent to reduce cancer mortality and morbidity in the future.


Asunto(s)
Cobre , Inhibidores de Puntos de Control Inmunológico , Estructuras Metalorgánicas , Estructuras Metalorgánicas/química , Estructuras Metalorgánicas/farmacología , Humanos , Inhibidores de Puntos de Control Inmunológico/farmacología , Inhibidores de Puntos de Control Inmunológico/química , Ratones , Animales , Cobre/química , Cobre/farmacología , Supervivencia Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Especies Reactivas de Oxígeno/metabolismo , Antineoplásicos/farmacología , Antineoplásicos/química , Ensayos de Selección de Medicamentos Antitumorales , Línea Celular Tumoral , Neoplasias/tratamiento farmacológico , Neoplasias/patología , Neoplasias/terapia , Femenino , Carbono/química , Carbono/farmacología , Ratones Endogámicos BALB C
16.
Lab Chip ; 24(14): 3521-3527, 2024 Jul 10.
Artículo en Inglés | MEDLINE | ID: mdl-38939907

RESUMEN

Zeolitic imidazolate framework-8 (ZIF-8) encapsulating enzymatically active biomolecules has emerged as a novel biocompatible nanozyme and offers significant implications for bioanalysis of various biomarkers towards early diagnosis of severe diseases such as cancers. However, the rapid, continuous and scalable synthesis of these nanozymes still remains challenging. In this work, we proposed a novel microfluidic approach for rapid and continuous synthesis of hemin@ZIF-8 nanozyme. By employing a distinctive combination of zigzag-shaped channel and spiral channel with sudden expansion structures, we have enhanced the mixing efficiency within the chip and achieved effective encapsulation of hemin in ZIF-8. The resulting hemin@ZIF-8 nanoparticles exhibit peroxidase-like activity and are capable of detecting free H2O2 with a limit of detection (LOD) as low as 45 nM, as well as H2O2 secreted by viable cells with a detection threshold of approximately 10 cells per mL. By leveraging this method, we achieved successful detection of cancer cells and effective screening of anticancer drugs that induce oxidative stress injury in cancer cells. This innovative microfluidic strategy offers a new avenue for synthesizing functional nanocomposites to facilitate the development of next-generation diagnostic tools for early disease detection and personalized medicine.


Asunto(s)
Antineoplásicos , Hemina , Estructuras Metalorgánicas , Especies Reactivas de Oxígeno , Hemina/química , Hemina/metabolismo , Humanos , Antineoplásicos/farmacología , Antineoplásicos/química , Especies Reactivas de Oxígeno/metabolismo , Especies Reactivas de Oxígeno/análisis , Estructuras Metalorgánicas/química , Estructuras Metalorgánicas/síntesis química , Estructuras Metalorgánicas/farmacología , Peróxido de Hidrógeno/análisis , Peróxido de Hidrógeno/química , Técnicas Analíticas Microfluídicas/instrumentación , Ensayos de Selección de Medicamentos Antitumorales , Dispositivos Laboratorio en un Chip , Zeolitas/química , Límite de Detección , Imidazoles
17.
J Colloid Interface Sci ; 672: 589-599, 2024 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-38852359

RESUMEN

Failure of articular cartilage lubrication and inflammation are the main causes of osteoarthritis (OA), and integrated treatment realizing joint lubrication and anti-inflammation is becoming the most effective treat model. Inspired by low friction of human synovial fluid and adhesive chemical effect of mussels, our work reports a biomimetic lubricating system that realizes long-time lubrication, photothermal responsiveness and anti-inflammation property. To build the system, a dopamine-mediated strategy is developed to controllably graft hyaluronic acid on the surface of metal organic framework. The design constructs a biomimetic core-shell structure that has good dispersity and stability in water with a high drug loading ratio of 99%. Temperature of the solution rapidly increases to 55 °C under near-infrared light, and the hard-soft lubricating system well adheres to wear surfaces, and greatly reduces frictional coefficient by 75% for more than 7200 times without failure. Cell experiments show that the nanosystem enters cells by endocytosis, and releases medication in a sustained manner. The anti-inflammatory outcomes validate that the nanosystem prevents the progression of OA by down-regulating catabolic proteases and pain-related genes and up-regulating genes that are anabolic in cartilage. The study provides a bioinspired strategy to employ metal organic framework with controlled surface and structure for friction reduction and anti-inflammation, and develops a new concept of OA synergistic therapy model for practical applications.


Asunto(s)
Materiales Biomiméticos , Ácido Hialurónico , Osteoartritis , Osteoartritis/tratamiento farmacológico , Osteoartritis/patología , Osteoartritis/metabolismo , Humanos , Materiales Biomiméticos/química , Materiales Biomiméticos/farmacología , Ácido Hialurónico/química , Ácido Hialurónico/farmacología , Animales , Estructuras Metalorgánicas/química , Estructuras Metalorgánicas/farmacología , Propiedades de Superficie , Lubrificación , Antiinflamatorios/química , Antiinflamatorios/farmacología , Tamaño de la Partícula , Dopamina/química , Dopamina/farmacología , Liberación de Fármacos
18.
J Colloid Interface Sci ; 672: 724-735, 2024 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-38870763

RESUMEN

The integration of functional nanomaterials with tissue engineering scaffolds has emerged as a promising solution for simultaneously treating malignant bone tumors and repairing resected bone defects. However, achieving a uniform bioactive interface on 3D-printing polymer scaffolds with minimized microstructural heterogeneity remains a challenge. In this study, we report a facile metal-coordination self-assembly strategy for the surface engineering of 3D-printed polycaprolactone (PCL) scaffolds with nanostructured two-dimensional conjugated metal-organic frameworks (cMOFs) consisting of Cu ions and 2,3,6,7,10,11-hexahydroxytriphenylene (HHTP). A tunable thickness of Cu-HHTP cMOF on PCL scaffolds was achieved via the alternative deposition of metal ions and HHTP. The resulting composite PCL@Cu-HHTP scaffolds not only demonstrated potent photothermal conversion capability for efficient OS ablation but also promoted the bone repair process by virtue of their cell-friendly hydrophilic interfaces. Therefore, the cMOF-engineered dual-functional 3D-printing scaffolds show promising potential for treating bone tumors by offering sequential anti-tumor effects and bone regeneration capabilities. This work also presents a new avenue for the interface engineering of bioactive scaffolds to meet multifaceted demands in osteosarcoma-related bone defects.


Asunto(s)
Neoplasias Óseas , Regeneración Ósea , Osteosarcoma , Poliésteres , Impresión Tridimensional , Andamios del Tejido , Osteosarcoma/patología , Osteosarcoma/tratamiento farmacológico , Osteosarcoma/terapia , Regeneración Ósea/efectos de los fármacos , Andamios del Tejido/química , Neoplasias Óseas/patología , Neoplasias Óseas/tratamiento farmacológico , Neoplasias Óseas/terapia , Poliésteres/química , Humanos , Estructuras Metalorgánicas/química , Estructuras Metalorgánicas/farmacología , Estructuras Metalorgánicas/síntesis química , Propiedades de Superficie , Cobre/química , Cobre/farmacología , Hipertermia Inducida , Ingeniería de Tejidos , Tamaño de la Partícula , Catálisis , Animales , Antineoplásicos/farmacología , Antineoplásicos/química , Ratones , Supervivencia Celular/efectos de los fármacos , Nanoestructuras/química , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos
19.
ACS Biomater Sci Eng ; 10(7): 4227-4236, 2024 Jul 08.
Artículo en Inglés | MEDLINE | ID: mdl-38848308

RESUMEN

Anticancer drugs are often associated with limitations such as poor stability in aqueous solutions, limited cell membrane permeability, nonspecific targeting, and irregular drug release when taken orally. One possible solution to these problems is the use of nanocarriers of drug molecules, particularly those with targeting ability, stimuli-responsive properties, and high drug loading capacity. These nanocarriers can improve drug stability, increase cellular uptake, allow specific targeting of cancer cells, and provide controlled drug release. While improving the therapeutic efficacy of cancer drugs, contemporary researchers also aim to reduce their associated side effects, such that cancer patients are offered with a more effective and targeted treatment strategy. Herein, a set of nine porous covalent organic frameworks (COFs) were tested as drug delivery nanocarriers. Among these, paclitaxel loaded in COF-3 was most effective against the proliferation of ovarian cancer cells. This study highlights the emerging potential of COFs in the field of therapeutic drug delivery. Due to their biocompatibility, these porous COFs provide a viable substrate for controlled drug release, making them attractive candidates for improving drug delivery systems. This work also demonstrates the potential of COFs as efficient drug delivery agents, thereby opening up new opportunities in the field of sarcoma therapy.


Asunto(s)
Antineoplásicos , Portadores de Fármacos , Estructuras Metalorgánicas , Neoplasias Ováricas , Paclitaxel , Humanos , Neoplasias Ováricas/tratamiento farmacológico , Neoplasias Ováricas/metabolismo , Portadores de Fármacos/química , Femenino , Antineoplásicos/uso terapéutico , Antineoplásicos/química , Antineoplásicos/farmacología , Paclitaxel/uso terapéutico , Paclitaxel/química , Paclitaxel/farmacología , Estructuras Metalorgánicas/química , Estructuras Metalorgánicas/uso terapéutico , Estructuras Metalorgánicas/farmacología , Línea Celular Tumoral , Porosidad , Liberación de Fármacos
20.
ACS Appl Mater Interfaces ; 16(27): 34591-34606, 2024 Jul 10.
Artículo en Inglés | MEDLINE | ID: mdl-38917296

RESUMEN

Immunotherapy has emerged as a revolutionizing therapeutic modality for cancer. However, its efficacy has been largely limited by a weak immune response and an immunosuppressive tumor microenvironment. Herein, we report a metal-organic framework (MOF)-derived titanium oxide nanoparticle (MCTx NP) as an immune booster that can greatly improve the immunotherapy efficacy by inducing "immunogenic cell death" (ICD) and remodeling the tumor microenvironment. The NPs, inheriting the characteristic structure of MIL-125 and enriched with oxygen vacancies (OVs), demonstrate both high photothermal conversion efficiency and a reactive oxygen species (ROS) generation yield upon near-infrared (NIR) activation. Moreover, the NPs can release O2 and reduce glutathione (GSH) in the tumor environment, showcasing their potential to reverse the immunosuppressive microenvironment. In vitro/vivo results demonstrate that MCTx NPs directly kill tumor cells and effectively eliminate primary tumors by exerting dual photodynamic/photothermal therapy under a single NIR irritation. At the same time, MCTx NPs augment the PD-L1 blockade efficacy by potently inducing ICDs and reversing the immunosuppressive tumor microenvironment, including promoting dendritic cell (DC) maturation, decreasing regulatory T cells (Tregs)' infiltration, and increasing cytotoxic T lymphocytes (CTLs) and helper T cells (Ths), resulting in effective distant tumor suppression. This work highlights MCTx NP-mediated photodynamic- and photothermal-enhanced immunotherapy as an effective strategy for tumor treatment.


Asunto(s)
Inmunoterapia , Estructuras Metalorgánicas , Oxígeno , Fotoquimioterapia , Titanio , Microambiente Tumoral , Titanio/química , Titanio/farmacología , Estructuras Metalorgánicas/química , Estructuras Metalorgánicas/farmacología , Animales , Ratones , Humanos , Microambiente Tumoral/efectos de los fármacos , Oxígeno/química , Terapia Fototérmica , Nanopartículas/química , Línea Celular Tumoral , Especies Reactivas de Oxígeno/metabolismo , Neoplasias/terapia , Neoplasias/inmunología , Neoplasias/patología , Antineoplásicos/farmacología , Antineoplásicos/química
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA