Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 6 de 6
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
2.
Acta Biomater ; 144: 168-182, 2022 05.
Artigo em Inglês | MEDLINE | ID: mdl-35358735

RESUMO

Osteoarthritis (OA), a widespread degenerative disease characterized by cartilage destruction, has emerged as a public health challenge in the current aging society. In addition to applied steroids and surgery, near-infrared (NIR) sensitive nano-enzyme for the treatment of osteoarthritis through mitochondrial repair and cartilage protection is attractive and promising. In this study, a NIR sensitive multifunctional heterostructure (EGCG (Epigallocatechin gallate) decorated Au-Ag nano-jars (E@Au-Ag)) was introduced as an enzyme-sensitive active nanoplatform for the treatment of osteoarthritis. Molecular biology results indicated that E@Au-Ag possesses intrinsic properties of anti-oxidative stress and was able to reduce the apoptosis rate of chondrocytes by 83.3%. The area of the intra-articular joint cavity injected with E@Au-Ag can be elevated to 46.6 °C under NIR to promote the release of EGCG further to induce cartilage regeneration. X-ray radiography and section staining showed that E@Au-Ag reduced cartilage damage and decreased OARSI scores by approximately 52% after 8 weeks of treatment in a surgically induced OA model. The results demonstrated that this multifunctional enzyme-like nanoplatform with a synergistic NIR sensitive property to facilitate cartilage migration and regeneration repair provides a promising OA treatment strategy. STATEMENT OF SIGNIFICANCE: 1. NIR-sensitive nano-enzyme is gaining much attention in the field of biomedical materials. 2. EGCG decorated Au-Ag nano-heterostructures were utilized as NIR-sensitive nano-enzymes for the treatment of osteoarthritis through mitochondrial repair and cartilage protection. 3. The obtained multifunctional Au-Ag nano-heterostructures are promising for osteoarthritis treatment.


Assuntos
Cartilagem Articular , Catequina , Osteoartrite , Catequina/análogos & derivados , Catequina/farmacologia , Condrócitos , Humanos , Osteoartrite/tratamento farmacológico
3.
Acta Biomater ; 137: 199-217, 2022 01 01.
Artigo em Inglês | MEDLINE | ID: mdl-34644613

RESUMO

Multidrug-resistant (MDR) bacteria-induced infection is becoming a huge challenge for clinical treatment, especially for non-healing diabetic wound infections, which increase patient mortality. MRSA infections and delayed wound healing (methicillin-resistant Staphylococcus aureus) accounted for a higher proportion. Although surgical debridement and continuous use of antibiotics are still the main clinical treatments, new multifunctional therapeutic nanoplatform are attractive for MIDW. Thus, in the present study, black phosphorus quantum dots (BPQDs) encapsulated in hydrogel (BPQDs@NH) were utilized as nanoplatforms for MIDW treatment to achieve the multifunctional properties of NIR (near infrared) responsiveness, ROS (reactive oxygen species) generation and antibacterial activity. Upon NIR irradiation, the temperature of the BPQDs@NH-treated MIDW area rapidly increased up to 55 °C for sterilization. In vitro experiments showed that BPQDs@NH exerted a synergistic effect on inhibiting MRSA by producing of ROS, lipid peroxidation, glutathione, adenosine triphosphate accumulation and bacterial membrane destruction upon NIR irradiation. The resulting BPQDs@NH achieved an effective sterilization rate of approximately 90% for MRSA. Furthermore, animal experiments revealed that BPQDs@NH achieved an effective closure rate of 95% for MIDW after 12 days by reducing the inflammatory response and regulating the expression of vascular endothelial growth factor (VEGF) and basic fibroblast growth factor (bFGF). Meanwhile, intravenous circulation experiments showed good biocompatibility of BPQDs, and no obvious damage to rat major organs was observed. The obtained results indicated that BPQDs@NH achieved the synergistic functions of NIR-responsiveness, ROS generation, and antibacterial activity and promoted wound healing, suggesting that they are promising multifunctional nanoplatforms for MIDW healing. STATEMENT OF SIGNIFICANCE: 1. NIR-triggered ROS-generating and antibacterial nanoplatforms are attractive in the wound healing field. 2. In this work, black phosphorus quantum dots encapsulated in a hydrogel were used as a nanoplatform for treating MRSA infected wounds. 3. The obtained materials have achieved an effective sterilization rate for MRSA and effective wound closure rate.


Assuntos
Diabetes Mellitus Experimental , Staphylococcus aureus Resistente à Meticilina , Pontos Quânticos , Animais , Antibacterianos/farmacologia , Diabetes Mellitus Experimental/tratamento farmacológico , Humanos , Fósforo , Ratos , Espécies Reativas de Oxigênio , Fator A de Crescimento do Endotélio Vascular , Cicatrização
4.
Regen Biomater ; 8(6): rbab067, 2021 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-34858635

RESUMO

Arthritis is a kind of chronic inflammatory autoimmune disease, which can destroy joint cartilage and bone, leading to joint pain, joint swelling, and limited mobility. Traditional therapies have many side effects or focus too much on anti-inflammation while neglecting joint repair. In this experiment, we combined Epigallocatechin gallate (EGCG) with extracellular vesicles derived from macrophages to treat rheumatoid arthritis. Sustained-release resulted in a significant decrease in chondrocyte expression of hypoxia-inducible factor 1-alpha, a decrease in apoptosis-related proteins Cytochrome C, Caspase-3, Caspase-9, and Bax. Molecular biological analysis showed that extracellular vesicles-encapsulated EGCG (EVs-EGCG) more significantly upregulated type II collagen expression by about 1.8-fold than EGCG alone, which was more beneficial for arthritis repair. Animal experiments revealed that these EGCG-coated extracellular vesicles significantly reduced swelling, decreased synovial hyperplasia, repaired cartilage, and attenuated arthritis-related pathology scores in arthritic rats. Measurement data showed that EVs-EGCG treatment reduced joint swelling by approximately 39.5% in rheumatoid rats. In vitro studies have shown that this EVs-EGCG can increase the expression of cartilage type II collagen and reduce apoptosis of chondrocytes. Moreover, it was demonstrated in vivo experiments to reduce cartilage destruction in rheumatoid arthritis rats, providing a solution for the treatment of rheumatoid arthritis.

5.
J Nanobiotechnology ; 19(1): 362, 2021 Nov 10.
Artigo em Inglês | MEDLINE | ID: mdl-34758829

RESUMO

BACKGROUND: Healing of MRSA (methicillin-resistant Staphylococcus aureus) infected deep burn wounds (MIDBW) in diabetic patients remains an obstacle but is a cutting-edge research problem in clinical science. Surgical debridement and continuous antibiotic use remain the primary clinical treatment for MIDBW. However, suboptimal pharmacokinetics and high doses of antibiotics often cause serious side effects such as fatal complications of drug-resistant bacterial infections. MRSA, which causes wound infection, is currently a bacterium of concern in diabetic wound healing. In more severe cases, it can even lead to amputation of the patient's limb. The development of bioactive nanomaterials that can promote infected wound healing is significant. RESULTS: The present work proposed a strategy of using EGCG (Epigallocatechin gallate) modified black phosphorus quantum dots (BPQDs) as therapeutic nanoplatforms for MIDBW to achieve the synergistic functions of NIR (near-infrared)-response, ROS-generation, sterilization, and promoting wound healing. The electron spin resonance results revealed that EGCG-BPQDs@H had a more vital photocatalytic ability to produce singlet oxygen than BPQDs@H. The inhibition results indicated an effective bactericidal rate of 88.6% against MRSA. Molecular biology analysis demonstrated that EGCG-BPQDs significantly upregulated CD31 nearly fourfold and basic fibroblast growth factor (bFGF) nearly twofold, which were beneficial for promoting the proliferation of vascular endothelial cells and skin epidermal cells. Under NIR irradiation, EGCG-BPQDs hydrogel (EGCG-BPQDs@H) treated MIDBW area could rapidly raise temperature up to 55 °C for sterilization. The MIBDW closure rate of rats after 21 days of treatment was 92.4%, much better than that of 61.1% of the control group. The engineered EGCG-BPQDs@H were found to promote MIDBW healing by triggering the PI3K/AKT and ERK1/2 signaling pathways, which could enhance cell proliferation and differentiation. In addition, intravenous circulation experiment showed good biocompatibility of EGCG-BPQDs@H. No significant damage to major organs was observed in rats. CONCLUSIONS: The obtained results demonstrated that EGCG-BPQDs@H achieved the synergistic functions of photocatalytic property, photothermal effects and promoted wound healing, and are promising multifunctional nanoplatforms for MIDBW healing in diabetics.


Assuntos
Fósforo , Polifenóis/farmacologia , Pontos Quânticos/química , Espécies Reativas de Oxigênio/metabolismo , Chá/química , Animais , Queimaduras/metabolismo , Células Cultivadas , Diabetes Mellitus Experimental/metabolismo , Células Endoteliais da Veia Umbilical Humana , Humanos , Masculino , Staphylococcus aureus Resistente à Meticilina/efeitos dos fármacos , Fósforo/química , Fósforo/farmacologia , Processos Fotoquímicos , Ratos , Ratos Sprague-Dawley , Cicatrização/efeitos dos fármacos
6.
Mater Sci Eng C Mater Biol Appl ; 125: 112098, 2021 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-33965108

RESUMO

Photothermal responsive nanoplatforms are attracting for photothermal therapy (PTT) of cancer. Herein, we propose a strategy to prepare IR-780 modified hydroxyapatite (HAP) nanorods as photothermic agents (HAP@IR-780). The results demonstrated that the obtained HAP@IR-780 was photothermal responsive under near-infrared laser irradiation the photothermal conversion efficiency was 69.3%, and it remained photostability after 4 cycles of irradiation. This advantage overcomes the optical instability of IR780. MTT and cellular uptake research proved that HAP@IR-780 was biocompatible in appropriate concentration range (0-20 µg/mL) without laser irradiation. Concentration-dependent internalization and reactive oxygen species (ROS) related apoptosis of HAP@IR-780 for MCF-7 cells were observed. Animal experiments showed that the gathered HAP@IR-780 at the tumor site reached a photothermal responsive temperature up to 57.9 °C, which could almost ablate the tumor with volumes as large as 1500 mm3. In general, our photothermal material has good photothermal conversion characteristics, and may have the least safety problems while showing excellent therapeutic effects. Therefore, HAP@IR-780 has a brilliant prospect in the field of tumor photothermal therapy.


Assuntos
Hipertermia Induzida , Nanotubos , Animais , Linhagem Celular Tumoral , Durapatita , Humanos , Fototerapia , Espécies Reativas de Oxigênio
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA