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











Base de dados
Intervalo de ano de publicação
1.
J Mater Chem B ; 12(32): 7814-7825, 2024 Aug 14.
Artigo em Inglês | MEDLINE | ID: mdl-38895823

RESUMO

In recent decades, orthopedic implants have been widely used as materials to replace human bone tissue functions. Among these, metal implants play a crucial role. Metals with better chemical stability, such as stainless steel, titanium alloys, and cobalt-chromium-molybdenum (CoCrMo) alloy, are commonly used for long-term applications. However, good chemical stability can result in poor tissue integration between the tissue and the implant, leading to potential inflammation risks. This study creates hydrogenated CoCrMo (H-CoCrMo) surfaces, which have shown promise as anti-inflammatory orthopedic implants. Using the electrochemical cathodic hydrogen-charging method, the surface of the CoCrMo alloy was hydrogenated, resulting in improved biocompatibility, reduced free radicals, and an anti-inflammatory response. Hydrogen diffusion to a depth of approximately 106 ± 27 nm on the surface facilitated these effects. This hydrogen-rich surface demonstrated a reduction of 85.2% in free radicals, enhanced hydrophilicity as evidenced by a decrease in a contact angle from 83.5 ± 1.9° to 52.4 ± 2.2°, and an increase of 11.4% in hydroxyapatite deposition surface coverage. The cell study results revealed a suppression of osteosarcoma cell activity to 50.8 ± 2.9%. Finally, the in vivo test suggested the promotion of new bone formation and a reduced inflammatory response. These findings suggest that electrochemical hydrogen charging can effectively modify CoCrMo surfaces, offering a potential solution for improving orthopedic implant outcomes through anti-inflammatory mechanisms.


Assuntos
Materiais Biocompatíveis , Hidrogênio , Inflamação , Vitálio , Inflamação/tratamento farmacológico , Inflamação/induzido quimicamente , Humanos , Hidrogênio/química , Vitálio/química , Vitálio/farmacologia , Materiais Biocompatíveis/química , Materiais Biocompatíveis/farmacologia , Propriedades de Superfície , Próteses e Implantes , Animais , Ligas/química , Ligas/farmacologia , Cobalto/química , Teste de Materiais , Camundongos , Tamanho da Partícula
2.
ACS Appl Mater Interfaces ; 16(20): 25622-25636, 2024 May 22.
Artigo em Inglês | MEDLINE | ID: mdl-38739745

RESUMO

Breast cancer is a malignant tumor with a high mortality rate among women. Therefore, it is necessary to develop novel therapies to effectively treat this disease. In this study, iron selenide nanorods (FeSe2 NRs) were designed for use in magnetic hyperthermic, photothermal, and chemodynamic therapy (MHT/PTT/CDT) for breast cancer. To illustrate their efficacy, FeSe2 NRs were modified with the chemotherapeutic agent methotrexate (MTX). MTX-modified FeSe2 (FeSe2-MTX) exhibited excellent controlled drug release properties. Fe2+ released from FeSe2 NRs induced the release of •OH from H2O2 via a Fenton/Fenton-like reaction, enhancing the efficacy of CDT. Under alternating magnetic field (AMF) stimulation and 808 nm laser irradiation, FeSe2-MTX exerted potent hyperthermic and photothermal effects by suppressing tumor growth in a breast cancer nude mouse model. In addition, FeSe2 NRs can be used for magnetic resonance imaging in vivo by incorporating their superparamagnetic characteristics into a single nanomaterial. Overall, we presented a novel technique for the precise delivery of functional nanosystems to tumors that can enhance the efficacy of breast cancer treatment.


Assuntos
Neoplasias da Mama , Hipertermia Induzida , Metotrexato , Camundongos Nus , Nanotubos , Metotrexato/química , Metotrexato/farmacologia , Animais , Nanotubos/química , Camundongos , Feminino , Humanos , Neoplasias da Mama/tratamento farmacológico , Neoplasias da Mama/patologia , Neoplasias da Mama/terapia , Camundongos Endogâmicos BALB C , Terapia Fototérmica , Ferro/química , Compostos de Selênio/química , Compostos de Selênio/farmacologia , Compostos de Selênio/efeitos da radiação , Linhagem Celular Tumoral , Raios Infravermelhos
3.
J Mater Chem B ; 12(15): 3569-3593, 2024 Apr 17.
Artigo em Inglês | MEDLINE | ID: mdl-38494982

RESUMO

In recent years, inorganic nanoparticles (NPs) have attracted increasing attention as potential theranostic agents in the field of oncology. Photothermal therapy (PTT) is a minimally invasive technique that uses nanoparticles to produce heat from light to kill cancer cells. PTT requires two essential elements: a photothermal agent (PTA) and near-infrared (NIR) radiation. The role of PTAs is to absorb NIR, which subsequently triggers hyperthermia within cancer cells. By raising the temperature in the tumor microenvironment (TME), PTT causes damage to the cancer cells. Nanoparticles (NPs) are instrumental in PTT given that they facilitate the passive and active targeting of the PTA to the TME, making them crucial for the effectiveness of the treatment. In addition, specific targeting can be achieved through their enhanced permeation and retention effect. Thus, owing to their significant advantages, such as altering the morphology and surface characteristics of nanocarriers comprised of PTA, NPs have been exploited to facilitate tumor regression significantly. This review highlights the properties of PTAs, the mechanism of PTT, and the results obtained from the improved curative efficacy of PTT by utilizing NPs platforms.


Assuntos
Hipertermia Induzida , Nanopartículas , Neoplasias , Humanos , Fototerapia/métodos , Hipertermia Induzida/métodos , Neoplasias/tratamento farmacológico , Neoplasias/patologia , Nanomedicina Teranóstica/métodos , Microambiente Tumoral
4.
Mater Sci Eng C Mater Biol Appl ; 126: 112187, 2021 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-34082986

RESUMO

A composite coating of polyelectrolyte multilayers (PEMs) consisting of collagen, a chitosan barrier, and poly-γ-glutamic acid was fabricated using a spin coating technique to investigate and overcome the limited osseointegration capacity of 316 L stainless steel (316 L SS). To further enhance the biocompatibility, bone morphogenetic protein 2 (BMP-2) and basic fibroblast growth factor-2 (FGF-2) were loaded separately as dual growth factors, allowing for progressive drug release following the natural process of bone regeneration. The first burst release of FGF-2 triggered the proliferation of surrounding cells, and the subsequent release of BMP-2 stimulated their differentiation. The microstructure, surface potential, hardness, reduced Young's modulus, and wettability were assessed using scanning electron microscopy, nanoindentation, and water contact angle. The formation of apatite layers after immersion in simulated body fluid confirmed the bioactivity of this PEM. PEMs loaded with BMP-2 and FGF-2 showed a long sustained release of growth factors for up to 48 days. The biological properties were studied in vitro with rat bone mesenchymal stem cells (rBMSCs) and in vivo using a rat critical-sized calvarial defect model. PEMs loaded with growth factors further stimulated the proliferation and osteogenic differentiation of rBMSCs and the histology results indicated that new bone tissues could directly grow onto the PEMs. These findings suggest that PEM composite coating possesses significant potential for surface modification and long-term drug release of metallic implants to assist with bone restoration.


Assuntos
Osteogênese , Aço Inoxidável , Animais , Proteína Morfogenética Óssea 2 , Regeneração Óssea , Preparações de Ação Retardada/farmacologia , Polieletrólitos , Ratos
5.
Pharmaceutics ; 12(11)2020 Nov 16.
Artigo em Inglês | MEDLINE | ID: mdl-33207577

RESUMO

The lack of optimal methods employing nanoparticles to administer local anesthesia often results in posing severe risks such as non-biocompatibility, in vivo cytotoxicity, and drug overdose to patients. Here, we employed magnetic field-induced hyperthermia to achieve localized anesthesia. We synthesized iron-gold alloy nanoparticles (FeAu Nps), conjugated an anesthetic drug, Lidocaine, and coated the product with gelatin to increase the biocompatibility, resulting in a FeAu@Gelatin-Lidocaine nano-complex formation. The biocompatibility of this drug-nanoparticle conjugate was evaluated in vitro, and its ability to trigger local anesthesia was also evaluated in vivo. Upon exposure to high-frequency induction waves (HFIW), 7.2 ± 2.8 nm sized superparamagnetic nanoparticles generated heat, which dissociated the gelatin coating, thereby triggering Lidocaine release. MTT assay revealed that 82% of cells were viable at 5 mg/mL concentration of Lidocaine, indicating that no significant cytotoxicity was induced. In vivo experiments revealed that unless stimulated with HFIW, Lidocaine was not released from the FeAu@Gelatin-Lidocaine complex. In a proof-of-concept experiment, an intramuscular injection of FeAu@Gelatin-Lidocaine complex was administered to the rat posterior leg, which upon HFIW stimulation triggered an anesthetic effect to the injected muscle. Based on our findings, the FeAu@Gelatin-Lidocaine complex can deliver hyperthermia-induced controlled anesthetic drug release and serve as an ideal candidate for site-specific anesthesia administration.

6.
Int J Nanomedicine ; 13: 5499-5509, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30271143

RESUMO

INTRODUCTION: The efficacy of a chemotherapy drug in cancer therapy is highly determined by the ability to control the rate and extent of its release in vivo. However, the lack of techniques to accurately control drug release drastically limits the potency of a chemotherapy drug. MATERIALS AND METHODS: Here, we present a novel strategy to precisely monitor drug release under magnetic stimulation. Methotrexate (MTX), an anticancer drug, was covalently functionalized onto iron-gold alloy magnetic nanoparticles (Fe-Au alloy nanoparticles or NFAs) through 2-aminoethanethiol grafting and the ability of this drug-nanoparticle conjugate (NFA-MTX) in limiting HepG2 (liver carcinoma) cell growth was studied. Well-dispersed NFAs were prepared through pyrolysis. RESULTS AND DISCUSSION: Transmission electron microscopy revealed the average nanoparticle size to be 7.22±2.6 nm, while X-ray diffraction showed distinct 2θ peaks for iron and gold, confirming the presence of iron and gold nanoparticles. Inductively coupled plasma mass spectrometry revealed that the amount of NFA-MTX conjugate ingested by HepG2 cancer cells was 1.5 times higher than that ingested by L929 fibroblasts, thereby proving a higher selective ingestion by cancer cells compared to normal cells. Fourier-transform infrared spectroscopy revealed the breakage of Au-S bonds by the heat generated under magnetic field stimulation to release MTX from the NFA-MTX conjugate, triggering a 95% decrease in cellular viability at 100 µg/mL. CONCLUSION: The ability of NFA-MTX to dissociate under the influence of an applied magnetic field provides a new strategy to induce cancer cell death via hyperthermia. Applications in drug delivery, drug development, and cancer research are expected.


Assuntos
Preparações de Ação Retardada/uso terapêutico , Ligas de Ouro/química , Ouro/química , Hipertermia Induzida , Ferro/química , Nanopartículas Metálicas/química , Neoplasias/tratamento farmacológico , Animais , Antineoplásicos/química , Antineoplásicos/farmacologia , Antineoplásicos/uso terapêutico , Apoptose/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Preparações de Ação Retardada/farmacologia , Liberação Controlada de Fármacos , Células Hep G2 , Humanos , Campos Magnéticos , Nanopartículas Metálicas/ultraestrutura , Metotrexato/química , Metotrexato/uso terapêutico , Camundongos , Neoplasias/patologia , Espectroscopia de Infravermelho com Transformada de Fourier , Difração de Raios X
7.
J Biomed Nanotechnol ; 14(4): 725-735, 2018 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-31352946

RESUMO

Bacterial infection in wounds or implants can cause osteomyelitis, eventually leading to orthopedic implant failure. In this study, polyelectrolyte multilayer (PEM) coating comprising collagen as the cationic layer, chitosan as the barrier layer and γ-poly-glutamic acid as the anionic layer were fabricated onto a 316L stainless steel substrate by spin coating technique. Tetracycline-loaded 57S mesoporous bioactive glass nanoparticles (57S MBG, SiO2:CaO:P2O5 = 57:33:10 by wt%) were introduced into the γ-poly-glutamic acid layers. Herein, 57S MBG nanoparticles were successfully incorporated into the PEMs with a total thickness of ∼53 µm on 316L stainless steel (SS-PEMs-57S), which exhibited good hydrophilicity with a contact angle of 18.71°. The hardness of SS-PEMs-57S was 0.66 GPa while the Young's modulus was 11.5 GPa; these values are similar to those for the cortical bone. The surface roughness of MBG nanoparticle-incorporated PEMs increased from 231 to 384 nm. Controlled release of tetracycline loaded in MBG nanoparticles resulted in sustained antibacterial effect for up to 7 days, with higher release efficacy at low pH, which may be induced by inflammation or infection. Tetracycline loaded in SS-PEMs-57S showed good bacterial inhibition and maintained good cell viability in rat bone marrow mesenchymal stem cells (BMSCs) in the MTT assay. Moreover, SS-PEMs-57S also promoted mineralization of BMSCs. Therefore, this surface modification technology has great potential for endowing orthopedic implants with antibacterial and osteoconductive properties.


Assuntos
Nanopartículas , Polieletrólitos , Animais , Antibacterianos , Ratos , Dióxido de Silício , Aço Inoxidável , Propriedades de Superfície
8.
Biomed Mater ; 9(4): 045013, 2014 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-25065552

RESUMO

Reconstruction of critical size defects in the load-bearing area has long been a challenge in orthopaedics. In the past, we have demonstrated the feasibility of using a biodegradable load-sharing scaffold fabricated from poly(propylene fumarate)/tricalcium phosphate (PPF/TCP) loaded with bone morphogenetic protein-2 (BMP-2) to successfully induce healing in those defects. However, there is limited osteoconduction observed with the PPF/TCP scaffold itself. For this reason, 13-93 bioactive glass scaffolds with local BMP-2 delivery were investigated in this study for inducing segmental defect repairs in a load-bearing region. Furthermore, a recent review on BMP-2 revealed greater risks in radiculitis, ectopic bone formation, osteolysis and poor global outcome in association with the use of BMP-2 for spinal fusion. We also evaluated the potential side effects of locally delivered BMP-2 on the structures of adjacent bones. Therefore, cylindrical 13-93 glass scaffolds were fabricated by indirect selective laser sintering with side holes on the cylinder filled with dicalcium phosphate dehydrate as a BMP-2 carrier. The scaffolds were implanted into critical size defects created in rat femurs with and without 10 µg of BMP-2. The x-ray and micro-CT results showed that a bridging callus was found as soon as three weeks and progressed gradually in the BMP group while minimal bone formation was observed in the control group. Degradation of the scaffolds was noted in both groups. Stiffness, peak load and energy to break of the BMP group were all higher than the control group. There was no statistical difference in bone mineral density, bone area and bone mineral content in the tibiae and contralateral femurs of the control and BMP groups. In conclusion, a 13-93 bioactive glass scaffold with local BMP-2 delivery has been demonstrated for its potential application in treating large bone defects.


Assuntos
Cimentos Ósseos/química , Proteína Morfogenética Óssea 2/farmacologia , Fosfatos de Cálcio/química , Fêmur/efeitos dos fármacos , Fumaratos/química , Vidro/química , Polipropilenos/química , Alicerces Teciduais/química , Animais , Fenômenos Biomecânicos , Desenvolvimento Ósseo , Fixação de Fratura/instrumentação , Masculino , Microscopia Eletrônica de Varredura , Ratos , Ratos Long-Evans , Espectroscopia de Infravermelho com Transformada de Fourier , Propriedades de Superfície , Difração de Raios X , Microtomografia por Raio-X
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA