Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 31
Filtrar
1.
Chembiochem ; 24(9): e202200698, 2023 05 02.
Artículo en Inglés | MEDLINE | ID: mdl-36793188

RESUMEN

Cannabidiol (CBD) is a non-intoxicating cannabinoid from cannabis sativa that has demonstrated efficacious against inflammation, which can be considered as a potential drug for arthritis treatment. However, the poor solubility and low bioavailability limit its clinical application. Here, we report an effective strategy to fabricate Cannabidiol-loaded poly(lactic-co-glycolic acid) copolymer (CBD-PLGA) nanoparticles (NPs), with a spherical morphology and an average diameter of 238 nm. CBD was sustained release from CBD-PLGA-NPs, which improved the bioavailability of CBD. The CBD-PLGA-NPs effectively protect the damage of LPS to cell viability. We observed that CBD-PLGA-NPs significantly suppressed LPS-induced primary rat chondrocyte expression of inflammatory cytokines, including interleukin 1ß (IL-1ß), interleukin 6 (IL-6), tumor necrosis factor-α (TNF-α) and matrix metalloproteinase 13 (MMP-13). Remarkably, CBD-PLGA-NPs also showed better therapeutic effects of inhibiting the degradation of the extracellular matrix of chondrocytes than equivalent CBD solution. In general, the fabrication CBD-PLGA-NPs showed good protection of primary chondrocytes in vitro and is a promising system for osteoarthritis treatment.


Asunto(s)
Cannabidiol , Nanopartículas , Osteoartritis , Ratas , Animales , Cannabidiol/farmacología , Cannabidiol/uso terapéutico , Glicoles , Disponibilidad Biológica , Lipopolisacáridos , Copolímero de Ácido Poliláctico-Ácido Poliglicólico , Osteoartritis/tratamiento farmacológico , Portadores de Fármacos
2.
J Nanobiotechnology ; 19(1): 197, 2021 Jul 03.
Artículo en Inglés | MEDLINE | ID: mdl-34217311

RESUMEN

Intra-articular (IA) injection is an efficient treatment for osteoarthritis, which will minimize systemic side effects. However, the joint experiences rapid clearance of therapeutics after intra-articular injection. Delivering system modified through active targeting strategies to facilitate localization within specific joint tissues such as cartilage is hopeful to increase the therapeutic effects. In this study, we designed a nanoscaled amphiphilic and cartilage-targeting polymer-drug delivery system by using formononetin (FMN)-poly(ethylene glycol) (PEG) (denoted as PCFMN), which was prepared by PEGylation of FMN followed by coupling with cartilage-targeting peptide (CollBP). Our results showed that PCFMN was approximately regular spherical with an average diameter about 218 nm. The in vitro test using IL-1ß stimulated chondrocytes indicated that PCFMN was biocompatible and upregulated anabolic genes while simultaneously downregulated catabolic genes of the articular cartilage. The therapeutic effects in vivo indicated that PCFMN could effectively attenuate the progression of OA as evidenced by immunohistochemical staining and histological analysis. In addition, PCFMN showed higher intention time in joints and better anti-inflammatory effects than FMN, indicating the efficacy of cartilage targeting nanodrug on OA. This study may provide a reference for clinical OA therapy.


Asunto(s)
Isoflavonas/química , Isoflavonas/farmacología , Osteoartritis/tratamiento farmacológico , Polietilenglicoles/química , Polietilenglicoles/farmacología , Animales , Antiinflamatorios , Cartílago Articular/efectos de los fármacos , Cartílago Articular/metabolismo , Cartílago Articular/patología , Condrocitos/efectos de los fármacos , Condrocitos/metabolismo , Condrocitos/patología , Modelos Animales de Enfermedad , Sistemas de Liberación de Medicamentos , Interleucina-1beta/metabolismo , Masculino , Nanopartículas , Osteoartritis/metabolismo , Osteoartritis/patología , Péptidos , Ratas Sprague-Dawley
3.
J Nanobiotechnology ; 19(1): 395, 2021 Nov 27.
Artículo en Inglés | MEDLINE | ID: mdl-34838028

RESUMEN

Stimulus-responsive therapy that allows precise imaging-guided therapy is limited for osteoarthritis (OA) therapy due to the selection of proper physiological markers as stimulus. Based on that the over-production of Reactive Oxygen Species (ROS) is associated with the progression in OA, we selected ROS as markers and designed a cartilage targeting and ROS-responsive theranostic nanoprobe that can be used for effective bioimaging and therapy of OA. This nanoprobe was fabricated by using PEG micelles modified with ROS-sensitive thioketal linkers (TK) and cartilage-targeting peptide, termed TKCP, which was then encapsulated with Dexamethasone (DEX) to form TKCP@DEX nanoparticles. Results showed that the nanoprobe can smartly "turn on" in response to excessive ROS and "turn off" in the normal joint. By applying different doses of ROS inducer and ROS inhibitor, this nanoprobe can emit ROS-dependent fluorescence according to the degree of OA severity, helpful to precise disease classification in clinic. Specifically targeting cartilage, TKCP@DEX could effectively respond to ROS and sustained release DEX to remarkably reduce cartilage damage in the OA joints. This smart, sensitive and endogenously activated ROS-responsive nanoprobe is promising for OA theranostics.


Asunto(s)
Nanopartículas/química , Osteoartritis , Especies Reactivas de Oxígeno , Nanomedicina Teranóstica/métodos , Animales , Masculino , Ratones , Ratones Endogámicos C57BL , Sondas Moleculares/química , Sondas Moleculares/metabolismo , Osteoartritis/diagnóstico por imagen , Osteoartritis/tratamiento farmacológico , Especies Reactivas de Oxígeno/química , Especies Reactivas de Oxígeno/metabolismo
4.
J Nanobiotechnology ; 18(1): 139, 2020 Sep 29.
Artículo en Inglés | MEDLINE | ID: mdl-32993662

RESUMEN

Drug therapy of osteoarthritis (OA) is limited by the short retention and lacking of stimulus-responsiveness after intra-articular (IA) injection. The weak acid microenvironment in joint provides a potential trigger for controlled drug release systems in the treatment of OA. Herein, we developed an pH-responsive metal - organic frameworks (MOFs) system modified by hyaluronic acid (HA) and loaded with an anti-inflammatory protocatechuic acid (PCA), designated as MOF@HA@PCA, for the therapy of OA. Results demonstrated that MOF@HA@PCA could smartly respond to acidic conditions in OA microenvironment and gradually release PCA, which could remarkably reduce synovial inflammation in both IL-1ß induced chondrocytes and the OA joints. MOF@HA@PCA also down-regulated the expression of inflammatory markers of OA and promoted the expression of cartilage-specific makers. This work may provide a new insight for the design of efficient nanoprobes for precision theranostics of OA .


Asunto(s)
Ácido Hialurónico/química , Ácido Hialurónico/farmacología , Estructuras Metalorgánicas/química , Estructuras Metalorgánicas/farmacología , Osteoartritis/tratamiento farmacológico , Animales , Antiinflamatorios/uso terapéutico , Biomarcadores , Supervivencia Celular/efectos de los fármacos , Condrocitos/metabolismo , Concentración de Iones de Hidrógeno , Hidroxibenzoatos , Inflamación/tratamiento farmacológico , Inyecciones Intraarticulares , Interleucina-1beta , Masculino , Osteoartritis/patología , Ratas , Ratas Sprague-Dawley , Especies Reactivas de Oxígeno
5.
J Nanobiotechnology ; 18(1): 117, 2020 Aug 27.
Artículo en Inglés | MEDLINE | ID: mdl-32854712

RESUMEN

Stimulus-responsive therapy permits precise control of therapeutic effect only at lesion of interest, which determines it a promising method for diagnosis and imaging-guided precision therapy. The acid environment and overexpressed matrix metalloproteinases-13 (MMP-13) are typical markers in osteoarthritis (OA), which enables the development of stimulus-responsive drug delivery system with high specificity for OA. We herein demonstrate a nano-micelle based stimuli-responsive theranostic strategy with reporting and drug release controlled by acidic pH and MMP-13 for OA therapy. Such nanoplatform is incorporated with a motif specifically targeting on cartilage, a motif responsive to matrix metalloproteinases-13 to specifically report OA condition and biodynamics of nano-micelles, an anti-inflammatory drug (e.g., psoralidin (PSO)) from traditional Chinese medicine, and a biocompatible polymeric skeleton for sustainable drug release in response to the acidic OA condition. The high effectiveness of this targeted precision therapy is demonstrated comprehensively by both in vitro and vivo evidences.


Asunto(s)
Metaloproteinasa 13 de la Matriz/metabolismo , Osteoartritis/metabolismo , Nanomedicina Teranóstica/métodos , Animales , Benzofuranos , Células Cultivadas , Condrocitos/metabolismo , Cumarinas , Concentración de Iones de Hidrógeno , Ratones , Ratones Endogámicos C57BL
6.
Nanomedicine ; 28: 102210, 2020 08.
Artículo en Inglés | MEDLINE | ID: mdl-32334102

RESUMEN

Intra-articular (IA) drug delivery to treat osteoarthritis (OA) is limited by the short retention time of drugs in the joints due to poor specific targeting and non-responsiveness under acidic environment. A cartilage-targeting peptide was engineered to the surface of ferritin nanocages (CT-Fn) and loaded with an anti-inflammatory drug, metformin (Met), via the self-assembling nature of Fn nanocages. It demonstrated that the CT-Fn/Met could specifically bind to type II collagen, leading to the downregulation of catabolic markers of OA and promotion of cartilage-specific makers in IL-1ß-induced chondrocytes. IA delivery of CT-Fn/Met prolonged the retention time for 3 weeks and remarkably reduced inflammation. Moreover, better release under acidic conditions which enabling longer retention time of Met after IA delivery in OA joints for one more week. CT-Fn/Met could target and efficiently enter chondrocytes, further inducing prolonged IA accumulation and achieving enhanced therapeutic efficacy for OA treatment.


Asunto(s)
Antiinflamatorios/uso terapéutico , Sistemas de Liberación de Medicamentos/métodos , Ferritinas/química , Osteoartritis/tratamiento farmacológico , Animales , Supervivencia Celular/efectos de los fármacos , Colágeno Tipo II/metabolismo , Técnica del Anticuerpo Fluorescente , Inmunohistoquímica , Masculino , Metformina/uso terapéutico , Ratones , Ratones Endogámicos C57BL , Reacción en Cadena en Tiempo Real de la Polimerasa
7.
Angew Chem Int Ed Engl ; 58(1): 269-272, 2019 01 02.
Artículo en Inglés | MEDLINE | ID: mdl-30421488

RESUMEN

We report the rational design of coordination-driven self-assembly metal-organic nanostructures for multifunctional nanotheranostics. Zinc(II) coordination-based nano-formulations capable of loading indocyanine green (ICG) and therapeutic genes were prepared to achieve a fluorescence/photoacoustic imaging-guided combination photo/gene therapy strategy. We showed the enhanced theranostic capability of zinc(II)-dipicolylamine-assisted assembly of ICG, as well as simultaneous targeted gene delivery in an experimental mouse model of cancer. Such a co-assembly strategy provides a facile way to achieve combined therapeutic functions for personalized nanomedicine.


Asunto(s)
Terapia Genética/métodos , Nanomedicina/métodos , Fototerapia/métodos , Zinc/química , Humanos
8.
J Am Chem Soc ; 140(44): 14980-14989, 2018 11 07.
Artículo en Inglés | MEDLINE | ID: mdl-30359020

RESUMEN

Tumor microenvironment-induced ultrasmall nanodrug generation (TMIUSNG) is an unprecedented approach to overcome the drug penetration barriers across complex biological systems, poor circulation stability and limited drug loading efficiency (DLE). Herein, a novel strategy was designed to synthesize metal-organic nanodrug complexes (MONCs) through supramolecular coassembly of photosensitizer sinoporphyrin sodium, chemotherapeutic drug doxorubicin and ferric ions. Compared with the free photosensitizer, MONCs produced 3-fold more reactive oxygen species (ROS) through the energy transfer-mediated fluorescence quenching. Remarkably, the self-delivering supramolecular MONCs with high DLE acted as a potent ultrasmall-nanodrug generator in response to the mild acidic tumor microenvironment to release ultrasmall nanodrugs (5-10 nm in diameter) from larger parental nanoparticles (140 nm in diameter), which in turn enhanced the intratumor permeability and therapeutic efficacy. The key mechanism of MONC synthesis was proposed, and we, for the first time, validated the generation of supramolecular scaffold intermediates between MONCs' assembly/disassembly states, as well as their involvement in multidrug ligands interactions. This proof-of-concept TMIUSNG strategy provides a foundation for the rational design of analogous carrier-free nanotheranostics through the combination of multiple therapeutic agents and metal ions with imaging functions.


Asunto(s)
Antineoplásicos/farmacología , Doxorrubicina/farmacología , Sistemas de Liberación de Medicamentos , Estructuras Metalorgánicas/farmacología , Nanopartículas/química , Fármacos Fotosensibilizantes/farmacología , Porfirinas/farmacología , Animales , Antineoplásicos/síntesis química , Antineoplásicos/química , Proliferación Celular/efectos de los fármacos , Doxorrubicina/química , Ensayos de Selección de Medicamentos Antitumorales , Humanos , Células MCF-7 , Estructuras Metalorgánicas/síntesis química , Estructuras Metalorgánicas/química , Ratones , Imagen Óptica , Tamaño de la Partícula , Fármacos Fotosensibilizantes/síntesis química , Fármacos Fotosensibilizantes/química , Porfirinas/química , Especies Reactivas de Oxígeno/metabolismo , Propiedades de Superficie , Células Tumorales Cultivadas , Microambiente Tumoral/efectos de los fármacos
9.
Int J Pharm ; 661: 124397, 2024 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-38945463

RESUMEN

Rhein, a natural anthraquinone compound derived from traditional Chinese medicine, exhibits potent anti-inflammatory properties via modulating the level of Reactive oxygen or nitrogen species (RONS). Nevertheless, its limited solubility in water, brief duration of plasma presence, as well as its significant systemic toxicity, pose obstacles to its in vivo usage, necessitating the creation of a reliable drug delivery platform to circumvent these difficulties. In this study, an esterase-responsive and mitochondria-targeted nano-prodrug was synthesized by conjugating Rhein with the polyethylene glycol (PEG)-modified triphenyl phosphonium (TPP) molecule, forming TPP-PEG-RH, which could spontaneously self-assemble into RPT NPs when dispersed in aqueous media. The TPP outer layer of these nanoparticles enhances their pharmacokinetic profile, facilitates efficient delivery to mitochondria, and promotes cellular uptake, thereby enabling enhanced accumulation in mitochondria and improved therapeutic effects in vitro. The decline in RONS was observed in IL-1ß-stimulated chondrocyte after RPT NPs treating. RPT NPs also exert excellent anti-inflammatory (IL-1ß, TNF-α, IL-6 and MMP-13) and antioxidative effects (Cat and Sod) via the Nrf2 signalling pathway, upregulation of cartilage related genes (Col2a1 and Acan). Moreover, RPT NPs shows protection of mitochondrial membrane potential and inhibition of chondrocyte apoptosis. Moreover, These findings suggest that the mitochondria-targeted polymer-Rhein conjugate may offer a therapeutic solution for patients suffering from chronic joint disorders, by attenuating the progression of osteoarthritis (OA).


Asunto(s)
Antraquinonas , Antiinflamatorios , Mitocondrias , Nanopartículas , Osteoartritis , Profármacos , Antraquinonas/administración & dosificación , Antraquinonas/farmacología , Antraquinonas/farmacocinética , Antraquinonas/química , Mitocondrias/efectos de los fármacos , Mitocondrias/metabolismo , Osteoartritis/tratamiento farmacológico , Animales , Antiinflamatorios/administración & dosificación , Antiinflamatorios/farmacología , Antiinflamatorios/química , Antiinflamatorios/farmacocinética , Profármacos/administración & dosificación , Profármacos/química , Nanopartículas/química , Condrocitos/efectos de los fármacos , Polietilenglicoles/química , Antioxidantes/administración & dosificación , Antioxidantes/farmacología , Antioxidantes/química , Sistemas de Liberación de Medicamentos/métodos , Especies Reactivas de Oxígeno/metabolismo , Humanos , Apoptosis/efectos de los fármacos , Compuestos Organofosforados/química , Compuestos Organofosforados/administración & dosificación , Ratas
10.
Front Bioeng Biotechnol ; 12: 1339530, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38361795

RESUMEN

Articular cartilage regeneration is still a difficult task due to the cartilage's weak capacity for self-healing and the effectiveness of the available therapies. The engineering of cartilage tissue has seen widespread use of stem cell-based therapies. However, efficient orientation of line-specific bone marrow mesenchymal stem cells (BMSCs) to chondrogenesis and maintenance of chondrogenic differentiation challenged stem cell-based therapy. Herein, we developed a Fe-based metal-organic framework (MOF) loaded with hematoporphyrin monomethyl ether (HMME) and cartilage-targeting arginine-aspartate-glycine (RGD) peptide to form MOF-HMME-RGD sonosensitizer to regulate BMSCs chondrogenic differentiation for cartilage regeneration via the modulation of reactive oxygen species (ROS). By using sonodynamic therapy (SDT), the MOF-HMME-RGD demonstrated favorable biocompatibility, could generate a modest amount of ROS, and enhanced BMSCs chondrogenic differentiation through increased accumulation of glycosaminoglycan, an ECM component specific to cartilage, and upregulated expression of key chondrogenic genes (ACAN, SOX9, and Col2a1). Further, transplanted BMSCs loading MOF-HMME-RGD combined with SDT enhanced cartilage regeneration for cartilage defect repair after 8 weeks into treatment. This synergistic strategy based on MOF nanoparticles provides an instructive approach to developing alternative sonosensitizers for cartilage regeneration combined with SDT.

11.
Research (Wash D C) ; 7: 0310, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38410279

RESUMEN

The activation of pro-inflammatory M1-type macrophages by overexpression of reactive oxygen species (ROS) and reactive nitrogen species (RONS) in synovial membranes contributes to osteoarthritis (OA) progression and cartilage matrix degradation. Here, combing Pt and Se with potent catalytic activities, we developed a hybrid Pt-Se nanozymes as ROS and RONS scavengers to exert synergistic effects for OA therapy. As a result, Pt-Se nanozymes exhibited efficient scavenging effect on ROS and RONS levels, leading to repolarization of M1-type macrophages. Furthermore, the polarization of synovial macrophages to the M2 phenotype inhibited the expression of pro-inflammatory factors and salvaged mitochondrial function in arthritic chondrocytes. In vivo results also suggest that Pt-Se nanozymes effectively suppress the early progression of OA with an Osteoarthritis Research International Association score reduction of 68.21% and 82.66% for 4 and 8 weeks, respectively. In conclusion, this study provides a promising strategy to regulate inflammatory responses by macrophage repolarization processes for OA therapeutic.

12.
Biomed Pharmacother ; 161: 114366, 2023 May.
Artículo en Inglés | MEDLINE | ID: mdl-36857913

RESUMEN

The activation of M1-type macrophages are dominant cells secreting proinflammatory present within the inflamed synovium in the progression of osteoarthritis (OA). Increased oxidative stress, such as redundant ROS and hydrogen peroxide (H2O2), are important factors in driving macrophages to polarize into M1 type. In this study, metal-polyphenol nanoformulations (Cu-Epigallocatechin-3-gallate (Cu-EGCG) nanosheets) were synthesized through the coordination interaction between EGCG and copper ions, which possessed the antioxidant effect of EGCG and anti-inflammatory of Cu2+. Results showed that Cu-EGCG nanosheets were biocompatible and the Cu2+ could be sustained released from the nanoparticles. Cu-EGCG nanosheets with multienzyme-like antioxidative activity could effectively scavenge the excessive intracellular ROS, leading to significantly decreased expression of the pro-inflammatory cytokines, which could reduce the expression of M1-type macrophages and exhibit excellent promotion on shifting macrophages to M2 phenotypes. Moreover, the secreted factor from the cell supernatant of Cu-EGCG treated macrophages exhibited anti-inflammatory potential in chondrocytes of inflamed synovial joints. This study suggests a novel strategy for OA therapy by using metal-polyphenol nanoformulations targeting macrophages.


Asunto(s)
Antiinflamatorios , Osteoartritis , Humanos , Antiinflamatorios/farmacología , Antiinflamatorios/uso terapéutico , Antioxidantes/farmacología , Antioxidantes/uso terapéutico , Catequina/uso terapéutico , Condrocitos/metabolismo , Peróxido de Hidrógeno/farmacología , Inflamación/tratamiento farmacológico , Inflamación/metabolismo , Macrófagos , Osteoartritis/tratamiento farmacológico , Osteoartritis/metabolismo , Polifenoles/farmacología , Especies Reactivas de Oxígeno/metabolismo
13.
Int J Nanomedicine ; 18: 1-16, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-36632237

RESUMEN

Introduction: Combination therapy is a promising approach to promote the efficacy and reduce the systemic toxicity of cancer therapy. Herein, we examined the potency of a combined chemo-phototherapy approach by constructing a hyaluronidase- and reactive oxygen species-responsive hyaluronic acid nanoparticle carrying a chemotherapy drug and a photosensitizer in a tumor-bearing mouse model. We hypothesized that following decomposition, the drugs inside the nanocomplex will be released in the tumors to provide effective tumor treatment. We aimed to design a smart drug delivery system that can improve traditional chemotherapy drug delivery and enhance the therapeutic efficacy in combination with photodynamic therapy. Methods: Hydrophilic hyaluronic acid (HA) was covalently modified with a hydrophobic 5ß-cholanic acid (CA) via an ROS-cleavable thioketal (tk) linker for a targeted co-deliver of 10-Hydroxy camptothecin (HCPT) and Chlorin e6 (Ce6) into tumors to improve the efficiency of combined chemo-photodynamic therapy. Results: The obtained HA-tk-CA nanoparticle carrying HCPT and Ce6, named HTCC, accumulated in the tumor through the enhanced permeable response (EPR) effect and HA-mediated CD44 targeting after intravenous administration. Upon laser irradiation and hyaluronidase degradation, HTCC was disrupted to release HCPT and Ce6 into the tumors. Compared to the monotherapy approach, HTCC demonstrated enhanced tumor growth inhibition and minimized systemic toxicity in a tumor-bearing mouse model. Conclusion: Our results suggested that controlled dual-drug release not only improved tumor drug delivery efficacy, but also reduced systemic side effects. In addition to HCPT and Ce6 delivery, the HA-tk-CA nanocomplex can be used to deliver other drugs in synergistic cancer therapy. Since most current combined therapy uses free drugs with distinct spatiotemporal distributions, the simultaneous co-delivery of dual drugs with a remote on-demand drug delivery nanosystem provides an alternative strategy for drug delivery design.


Asunto(s)
Sistemas de Liberación de Medicamentos , Nanopartículas , Neoplasias , Fotoquimioterapia , Fármacos Fotosensibilizantes , Porfirinas , Animales , Ratones , Camptotecina/química , Línea Celular Tumoral , Sistemas de Liberación de Medicamentos/métodos , Ácido Hialurónico/química , Hialuronoglucosaminidasa , Nanopartículas/química , Neoplasias/tratamiento farmacológico , Fotoquimioterapia/métodos , Fármacos Fotosensibilizantes/administración & dosificación , Porfirinas/química , Especies Reactivas de Oxígeno
14.
Research (Wash D C) ; 6: 0068, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-36930778

RESUMEN

Osteoarthritis (OA) is always characterized as excessive reactive oxygen species (ROS) inside articular cavity. Mimicking natural metalloenzymes with metal ions as the active centers, stable metal organic framework (MOF) formed by natural polyphenols and metal ions shows great potential in alleviating inflammatory diseases. Herein, a series of novel copper-morin-based MOF (CuMHs) with different molar ratios of Cu2+ and MH were employed to serve as ROS scavengers for OA therapy. As a result, CuMHs exhibited enhanced dispersion in aqueous solution, improved biocompatibility, and efficient ROS-scavenging ability compared to MH. On the basis of H2O2-stimulated chondrocytes, intracellular ROS levels were efficiently declined and cell death was prevented after treated by Cu6MH (Cu2+ and MH molar ratio of 6:1). Meanwhile, Cu6MH also exhibited efficient antioxidant and anti-inflammation function by down-regulating the expression of IL6, MMP13, and MMP3, and up-regulating cartilage specific gene expression as well. Importantly, Cu6MH could repair mitochondrial function by increasing mitochondrial membrane potential, reducing the accumulation of calcium ions, as well as promoting ATP content production. In OA joint model, intra-articular (IA) injected Cu6MH suppressed the progression of OA. It endowed that Cu6MH might be promising nanoenzymes for the prevention and treatment of various inflammatory diseases.

15.
NPJ Regen Med ; 7(1): 66, 2022 Nov 03.
Artículo en Inglés | MEDLINE | ID: mdl-36323709

RESUMEN

Excessive reactive oxygen species (ROS) are one of the leading mechanisms in the initiation and development of osteoarthritis (OA). However, conventional injection of ROS-responsive drug delivery systems (DDSs) such as nanoparticles and hydrogels usually cannot provide effective treatment due to rapid clearance and degradation or low bioavailability. In this study, a ROS-responsive nanofiber membrane named PLA/PEGDA-EDT@rGO-Fucoxanthin (PPGF) is fabricated by electrospinning, wherein PEGDA-EDT served as the ROS-responsive motif, reduced graphene oxide (rGO) as the drug carrier and fucoxanthin (Fx) as the antioxidative and anti-inflammatory agent. The results demonstrated that the PPGF nanofiber membrane exhibited sustained and long-term Fx release behavior (at least 66 days) in response to hydrogen peroxide (H2O2) in vitro. With low cytotoxicity and smart ROS responsiveness, PPGF showed excellent anti-inflammatory and antioxidative effects on IL-1ß-induced chondrocytes by potent ROS scavenging potential and upregulation of antioxidative enzymes. It also demonstrated the attenuation of OA progression with the reduced Osteoarthritis Research Society International (OARSI) score by 93.17% in 8 weeks. The smart ROS-responsive, biodegradable and biocompatible nanofiber membranes possess great potential for OA therapy under arthroscopy.

16.
Biomater Sci ; 10(22): 6549-6557, 2022 Nov 08.
Artículo en Inglés | MEDLINE | ID: mdl-36205771

RESUMEN

Three-dimensional (3D) bioink with favorable printability, strength, and biocompatibility challenged the 3D bioprinting technology in cartilage tissue engineering. Herein, we innovatively fabricated photo-crosslinkable methacrylated konjac glucomannan (KGMMA) as a novel biomaterial ink for 3D extrusion bioprinting in an attempt to construct precisely patterned tissues. Specifically, konjac glucomannan (KGM) was modified by methacrylic anhydride, which is a kind of photoreactive group, to form KGMMA. After UV crosslinking, the printed KGMMA hydrogel formed a covalent crosslinking network with high strength, desired shearing, and swelling and degradation characteristics. The properties of the KGMMA hydrogel could be modulated by changing the contents of MA. The shear-thinning property of the KGMMA biomaterial ink enables excellent printability, which can print different shapes including lattices, hexagons, and flowers. Furthermore, the bioinks support cell growth after being printed with chondrocytes for a culture. Therefore, the biodegradable, injectable, and photo-crosslinkable KGMMA biomaterial ink holds a great promise for cartilage tissue engineering.


Asunto(s)
Bioimpresión , Bioimpresión/métodos , Hidrogeles , Tinta , Impresión Tridimensional , Reología , Ingeniería de Tejidos/métodos , Materiales Biocompatibles , Andamios del Tejido
17.
Adv Healthc Mater ; 11(8): e2101715, 2022 04.
Artículo en Inglés | MEDLINE | ID: mdl-34997700

RESUMEN

Methods that leverage bone marrow mesenchymal stem cells (BMSCs) and stimulating factor kartogenin (KGN) for chondrocyte differentiation have paved the way for cartilage repair. However, the scarce carriers for efficiently bridging the two components significantly impede their further application. Therefore, one kind of bifunctional ferritin has designed and synthesized: RC-Fn, a genetically engineered ferritin nanocage with RGD peptide and WYRGRL peptide on the surface. The RGD can target the integrin αvß3 of BMSCs and promote proliferation, and the WYRGRL peptide has an inherent affinity for the cartilage matrix component of collagen II protein. RC-Fn nanocages have an ideal size for penetrating the proteoglycan network of cartilage. Thus, intra-articularly injected RC-Fn with KGN loading can convert the articular cavity from a barrier into a reservoir to prevent rapid release and clearance of KGN and exogenous BMSCs, which results in efficient and persistent chondrogenesis in cartilage regeneration.


Asunto(s)
Condrogénesis , Células Madre Mesenquimatosas , Anilidas , Cartílago/metabolismo , Diferenciación Celular , Ferritinas/metabolismo , Ácidos Ftálicos
18.
Regen Biomater ; 8(4): rbab020, 2021 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-34221446

RESUMEN

Andrographolide (AG) has favorable anti-inflammatory and antioxidative capacity. However, it has low bioavailability due to high lipophilicity and can be easily cleared by the synovial fluid after intra-articular injection, leading to low therapeutic efficiency in osteoarthritis (OA). Herein, we designed a nano-sized pH-responsive drug delivery system (DDS) for OA treatment by using modified mesoporous silica nanoparticles (MSNs) with pH-responsive polyacrylic acid (PAA) for loading of AG to form AG@MSNs-PAA nanoplatform. The nanoparticles have uniform size (∼120 nm), high drug loading efficiency (22.38 ± 0.71%) and pH-responsive properties, beneficial to sustained release in OA environment. Compared with AG, AG@MSNs-PAA showed enhanced antiarthritic efficacy and chondro-protective capacity based on IL-1ß-stimulated chondrocytes and anterior cruciate ligament transection-induced rat OA model, as demonstrated by lower expression of inflammatory factors and better prevention of proteoglycan loss. Therefore, the AG@MSNs-PAA nanoplatform may be developed as a promising OA-specific and on-demand DDS.

19.
Adv Sci (Weinh) ; 7(6): 2000588, 2020 03.
Artículo en Inglés | MEDLINE | ID: mdl-32195106

RESUMEN

[This corrects the article DOI: 10.1002/advs.201900099.].

20.
Adv Sci (Weinh) ; 6(16): 1900099, 2019 Aug 21.
Artículo en Inglés | MEDLINE | ID: mdl-31453055

RESUMEN

Directed differentiation of bone-marrow-derived stem cells (BMSCs) toward chondrogenesis has served as a predominant method for cartilage repair but suffers from poor oriented differentiation tendency and low differentiation efficiency. To overcome these two obstacles, an injectable composite hydrogel that consists of collagen hydrogels serving as the scaffold support to accommodate BMSCs and cadmium selenide (CdSe) quantum dots (QDs) is constructed. The introduction of CdSe QDs considerably strengthens the stiffness of the collagen hydrogels via mutual crosslinking using a natural crosslinker (i.e., genipin), which simultaneously triggers photodynamic provocation (PDP) to produce reactive oxygen species (ROS). Experimental results demonstrate that the intensified stiffness and augmented ROS production can synergistically promote the proliferation of BMSCs, induce cartilage-specific gene expression and increase secretion of glycosaminoglycan. As a result, this approach can facilitate the directed differentiation of BMSCs toward chondrogenesis and accelerate cartilage regeneration in cartilage defect repair, which routes through activation of the TGF-ß/SMAD and mTOR signaling pathways, respectively. Thus, this synergistic strategy based on increased stiffness and PDP-mediated ROS production provides a general and instructive approach for developing alternative materials applicable for cartilage repair.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA