Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 3.485
Filtrar
1.
Biomaterials ; 312: 122720, 2025 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-39084098

RESUMEN

Mesenchymal stem cells (MSCs) are expected to be useful therapeutics in osteoarthritis (OA), the most common joint disorder characterized by cartilage degradation. However, evidence is limited with regard to cartilage repair in clinical trials because of the uncontrolled differentiation and weak cartilage-targeting ability of MSCs after injection. To overcome these drawbacks, here we synthesized CuO@MSN nanoparticles (NPs) to deliver Sox9 plasmid DNA (favoring chondrogenesis) and recombinant protein Bmp7 (inhibiting hypertrophy). After taking up CuO@MSN/Sox9/Bmp7 (CSB NPs), the expressions of chondrogenic markers were enhanced while hypertrophic markers were decreased in response to these CSB-engineered MSCs. Moreover, a cartilage-targeted peptide (designated as peptide W) was conjugated onto the surface of MSCs via a click chemistry reaction, thereby prolonging the residence time of MSCs in both the knee joint cavity of mice and human-derived cartilage. In a surgery-induced OA mouse model, the NP and peptide dual-modified W-CSB-MSCs showed an enhancing therapeutic effect on cartilage repair in knee joints compared with other engineered MSCs after intra-articular injection. Most importantly, W-CSB-MSCs accelerated cartilage regeneration in damaged cartilage explants derived from OA patients. Thus, this new peptide and NPs dual engineering strategy shows potential for clinical applications to boost cartilage repair in OA using MSC therapy.


Asunto(s)
Diferenciación Celular , Trasplante de Células Madre Mesenquimatosas , Células Madre Mesenquimatosas , Nanopartículas , Osteoartritis , Péptidos , Células Madre Mesenquimatosas/citología , Células Madre Mesenquimatosas/metabolismo , Animales , Osteoartritis/terapia , Osteoartritis/patología , Nanopartículas/química , Humanos , Diferenciación Celular/efectos de los fármacos , Péptidos/química , Trasplante de Células Madre Mesenquimatosas/métodos , Condrogénesis/efectos de los fármacos , Ratones , Factor de Transcripción SOX9/metabolismo , Factor de Transcripción SOX9/genética , Cartílago Articular/patología , Cartílago Articular/efectos de los fármacos , Proteína Morfogenética Ósea 7/química , Proteína Morfogenética Ósea 7/farmacología , Ingeniería de Tejidos/métodos , Regeneración/efectos de los fármacos
2.
Nutrients ; 16(17)2024 Sep 04.
Artículo en Inglés | MEDLINE | ID: mdl-39275306

RESUMEN

The extracellular matrix of cartilage primarily constitutes of collagen and aggrecan. Cartilage degradation starts with aggrecan loss in osteoarthritis (OA). Vitamin D (VD) plays an essential role in several inflammation-related diseases and can protect the collagen in cartilage during OA. The present study focused on the role of VD in aggrecan turnover of human articular chondrocytes treated with tumor necrosis factor α (TNF-α) and the possible mechanism. Treatment with different doses of VD and different periods of intervention with TNF-α and TGF-ß1 receptor (TGFßR1) inhibitor SB525334 were investigated. The viability of human chondrocytes and extracellular secretion of TGF-ß1 were measured. The expression of intracellular TGFßR1 and VD receptor was examined. Transcriptional and translational levels of aggrecan and the related metabolic factors were analyzed. The results showed that TNF-α markedly reduced the viability, TGFßR1 expressions and aggrecan levels of human chondrocytes, and increased disintegrin and metalloproteinase with thrombospondin motifs. The alterations were partially inhibited by VD treatment. Furthermore, the effects of VD were blocked by the TGFßR1 inhibitor SB525334 in TNF-α-treated cells. VD may prevent proteoglycan loss due to TNF-α via TGF-ß1 signaling in human chondrocytes.


Asunto(s)
Agrecanos , Cartílago Articular , Condrocitos , Proteoglicanos , Transducción de Señal , Factor de Crecimiento Transformador beta1 , Factor de Necrosis Tumoral alfa , Vitamina D , Humanos , Condrocitos/metabolismo , Condrocitos/efectos de los fármacos , Transducción de Señal/efectos de los fármacos , Factor de Crecimiento Transformador beta1/metabolismo , Agrecanos/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo , Vitamina D/farmacología , Proteoglicanos/metabolismo , Proteoglicanos/farmacología , Cartílago Articular/metabolismo , Cartílago Articular/efectos de los fármacos , Células Cultivadas , Supervivencia Celular/efectos de los fármacos , Osteoartritis/metabolismo , Receptor Tipo I de Factor de Crecimiento Transformador beta/metabolismo , Receptores de Calcitriol/metabolismo
3.
Int J Mol Sci ; 25(17)2024 Sep 04.
Artículo en Inglés | MEDLINE | ID: mdl-39273553

RESUMEN

Osteoarthritis (OA) is an age-related disease characterized by inflammation, pain, articular cartilage damage, synovitis, and irreversible disability. Gynostemma pentaphyllum (Thunb.) Makino (GP), a herbal medicine traditionally used in East Asia for its anti-inflammatory properties, was investigated for its potential to modulate OA pathology and symptoms. This study evaluated GP's efficacy in inhibiting pain, functional decline, and cartilage destruction in monosodium iodoacetate-induced OA and acetic acid-induced writhing models. Additionally, the effects of GP on OA-related inflammatory targets were assessed via mRNA and protein expression in rat knee cartilage and lipopolysaccharide-induced RAW 264.7 cells. The GP group demonstrated significant pain relief, functional improvement, and cartilage protection. Notably, GP inhibited key inflammatory mediators, including interleukin (IL)-1ß, IL-6, matrix metalloproteinases (MMP)-3 and MMP-13, cyclooxygenase-2, and prostaglandin E receptor 2, surpassing the effects of active controls. These findings suggest that GP is a promising candidate for disease-modifying OA drugs and warrants further comprehensive studies.


Asunto(s)
Analgésicos , Antiinflamatorios , Gynostemma , Osteoartritis , Extractos Vegetales , Animales , Gynostemma/química , Ratones , Osteoartritis/tratamiento farmacológico , Osteoartritis/patología , Osteoartritis/inducido químicamente , Osteoartritis/metabolismo , Antiinflamatorios/farmacología , Antiinflamatorios/uso terapéutico , Células RAW 264.7 , Ratas , Extractos Vegetales/farmacología , Extractos Vegetales/uso terapéutico , Analgésicos/farmacología , Analgésicos/uso terapéutico , Masculino , Cartílago Articular/efectos de los fármacos , Cartílago Articular/patología , Cartílago Articular/metabolismo , Modelos Animales de Enfermedad , Ratas Sprague-Dawley , Dolor/tratamiento farmacológico
4.
Nat Commun ; 15(1): 7712, 2024 Sep 04.
Artículo en Inglés | MEDLINE | ID: mdl-39231947

RESUMEN

Osteoarthritis is a degenerative joint disease with joint pain as the main symptom, caused by fibrosis and loss of articular cartilage. Due to the complexity and heterogeneity of osteoarthritis, there is a lack of effective individualized disease-modifying osteoarthritis drugs in clinical practice. Chondrocyte senescence is reported to participate in occurrence and progression of osteoarthritis. Here we show that small molecule 10-hydroxy-2-decenoic acid suppresses cartilage degeneration and relieves pain in the chondrocytes, cartilage explants from osteoarthritis patients, surgery-induced medial meniscus destabilization or naturally aged male mice. We further confirm that 10-hydroxy-2-decenoic acid exerts a protective effect by targeting the glycosylation site in the Asp_Arg_Hydrox domain of aspartyl ß-hydroxylase. Mechanistically, 10-hydroxy-2-decenoic acid alleviate cellular senescence through the ERK/p53/p21 and GSK3ß/p16 pathways in the chondrocytes. Our study uncovers that 10-hydroxy-2-decenoic acid modulate cartilage metabolism by targeting aspartyl ß-hydroxylase to inhibit chondrocyte senescence in osteoarthritis. 10-hydroxy-2-decenoic acid may be a promising therapeutic drug against osteoarthritis.


Asunto(s)
Cartílago Articular , Senescencia Celular , Condrocitos , Ácidos Grasos Monoinsaturados , Osteoartritis , Animales , Condrocitos/efectos de los fármacos , Condrocitos/metabolismo , Condrocitos/patología , Masculino , Osteoartritis/metabolismo , Osteoartritis/patología , Osteoartritis/tratamiento farmacológico , Osteoartritis/prevención & control , Ratones , Senescencia Celular/efectos de los fármacos , Humanos , Ácidos Grasos Monoinsaturados/farmacología , Cartílago Articular/efectos de los fármacos , Cartílago Articular/metabolismo , Cartílago Articular/patología , Ratones Endogámicos C57BL , Modelos Animales de Enfermedad , Femenino
5.
Physiol Res ; 73(4): 633-642, 2024 Aug 31.
Artículo en Inglés | MEDLINE | ID: mdl-39264083

RESUMEN

The study aims to elucidate the therapeutic mechanism of Baicalin (BAI) in alleviating cartilage injury in osteoarthritic (OA) rat models, concentrating on its regulation of the miR-766-3p/AIFM1 axis. An OA rat model was developed with unilateral anterior cruciate ligament transection (ACLT). Interventions comprised of BAI treatment and intra-articular administration of miR-766-3p inhibitor. For evaluation, histopathological staining was conducted to investigate the pathological severity of knee cartilage injury. The levels of oxidative stress (OS) indicators including MDA, SOD, and GSH-Px, were quantified using colorimetric assays. Inflammatory factors (IFs; TNF-?, IL-1?, and IL-6) in knee joint lavage fluids were assessed using ELISA, while RT-PCR was employed to quantify miR-766-3p expression. TUNEL apoptosis staining was utilized to detect chondrocyte apoptosis, and western blotting examined autophagy-related markers (LC3, Beclin, p62), extracellular matrix (ECM) synthesis-associated indices (COL2A, ACAN, MMP13), and apoptosis-inducing factor mitochondrion-associated 1 (AIFM1). Histological examination revealed a marked amelioration of cartilage injury in the BAI-treated OA rat models compared to controls. BAI treatment significantly reduced inflammation and OS of knee joint fluid, activated autophagy, and decreased chondrocyte apoptosis and ECM degradation. Interestingly, the inhibitory effects of BAI on these pathological markers were significantly decreased by the miR-766-3p inhibitor. Further assessment revealed that BAI efficiently promoted miR-766-3p expression while inhibiting AIFM1 protein expression. BAI potentially mitigates articular cartilage injury in OA rats, likely through modulation of miR-766-3p/AIFM1 axis. Keywords: Baicalin, microRNA, AIFM1, Osteoarthritisv, Rat.


Asunto(s)
Flavonoides , MicroARNs , Ratas Sprague-Dawley , Animales , Flavonoides/farmacología , Flavonoides/uso terapéutico , MicroARNs/metabolismo , MicroARNs/genética , MicroARNs/biosíntesis , Ratas , Masculino , Osteoartritis/tratamiento farmacológico , Osteoartritis/metabolismo , Osteoartritis/patología , Cartílago Articular/efectos de los fármacos , Cartílago Articular/metabolismo , Cartílago Articular/patología , Factor Inductor de la Apoptosis/metabolismo , Estrés Oxidativo/efectos de los fármacos , Apoptosis/efectos de los fármacos
6.
Int Immunopharmacol ; 140: 112858, 2024 Oct 25.
Artículo en Inglés | MEDLINE | ID: mdl-39111145

RESUMEN

OBJECTIVE: The aim of this study was to investigate whether ASA VI controls osteoarthritis (OA) by regulating mitochondrial function. METHODS: Primary chondrocytes were isolated and cultured from rat knee joints. The chondrocytes were treated with ASA VI and interleukin-1ß (IL-1ß) to simulate the inflammatory environment of OA. Cell viability, apoptosis, inflammatory cytokine levels, and extracellular matrix (ECM) component levels were assessed. Mitochondrial function, including ATP levels, mitochondrial membrane potential, reactive oxygen species (ROS) levels, and mitochondrial DNA content, was evaluated. The expression of Sirtuin 3 (Sirt3), a key regulator of mitochondrial homeostasis, was examined. Additionally, a rat OA model was established by destabilizing the medial meniscus, and the effects of ASA VI on cartilage degeneration were assessed. RESULTS: ASA VI treatment improved cell viability, reduced apoptosis, and decreased IL-6 and TNF-α levels in IL-1ß-induced chondrocytes. ASA VI also upregulated Collagen II and Aggrecan expression, while downregulating ADAMTS5 and MMP-13 expression. Furthermore, ASA VI mitigated IL-1ß-induced mitochondrial dysfunction by increasing ATP levels, restoring mitochondrial membrane potential, reducing ROS production, and preserving mitochondrial DNA content. These effects were accompanied by the activation of Sirt3. In the rat OA model, ASA VI treatment increased Sirt3 expression and alleviated cartilage degeneration. CONCLUSION: ASA VI exerts chondroprotective and anti-inflammatory effects on IL-1ß-induced chondrocytes by improving mitochondrial function through Sirt3 activation. ASA VI also attenuates cartilage degeneration in a rat OA model. These findings suggest that ASA VI may be a potential therapeutic agent for the treatment of osteoarthritis by targeting mitochondrial dysfunction.


Asunto(s)
Condrocitos , Homeostasis , Mitocondrias , Osteoartritis , Saponinas , Sirtuina 3 , Animales , Masculino , Ratones , Ratas , Antiinflamatorios/farmacología , Antiinflamatorios/uso terapéutico , Apoptosis/efectos de los fármacos , Cartílago Articular/patología , Cartílago Articular/metabolismo , Cartílago Articular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Células Cultivadas , Condrocitos/efectos de los fármacos , Condrocitos/metabolismo , Citocinas/metabolismo , Modelos Animales de Enfermedad , Interleucina-1beta/metabolismo , Potencial de la Membrana Mitocondrial/efectos de los fármacos , Mitocondrias/metabolismo , Mitocondrias/efectos de los fármacos , Osteoartritis/tratamiento farmacológico , Osteoartritis/metabolismo , Osteoartritis/patología , Ratas Sprague-Dawley , Especies Reactivas de Oxígeno/metabolismo , Sirtuina 3/metabolismo , Sirtuina 3/genética , Saponinas/farmacología
7.
Biomater Sci ; 12(18): 4747-4758, 2024 Sep 10.
Artículo en Inglés | MEDLINE | ID: mdl-39118400

RESUMEN

Synovial fluid lubricates articular joints by forming a hydrated layer between the cartilage surfaces. In degenerative joint diseases like osteoarthritis (OA), the synovial fluid is compromised, which leads to less effective innate lubrication and exacerbated cartilage degeneration. Studies over the years have led to the development of partially or fully synthetic biolubricants to reduce the coefficient of friction with cartilage in knee joints. Cartilage-adhering, hydrated lubricants are particularly important to provide cartilage lubrication and chondroprotection under high normal load and slow speed. Here, we report the development of a hyaluronic acid (HA)-based lubricant functionalized with cationic branched poly-L-lysine (BPL) molecules that bind to cartilage via electrostatic interactions. We surmised that the electrostatic interactions between the BPL-modified HA molecules (HA-BPL) and the cartilage facilitate localization of the HA molecules to the cartilage surface. The number of BPL molecules on the HA backbone was varied to determine the optimal grafting density for cartilage binding and HA localization. Collectively, our results show that our HA-BPL molecules adhered readily to cartilage and were effective as a lubricant in cartilage-on-cartilage shear measurements where the modified HA molecules significantly reduce the coefficient of friction compared to phosphate-buffered saline or HA alone. This proof-of-concept study shows how the incorporation of cartilage adhering moieties, such as cationic molecules, can be used to enhance cartilage binding and lubrication properties of HA.


Asunto(s)
Cartílago Articular , Cationes , Ácido Hialurónico , Lubrificación , Polilisina , Ácido Hialurónico/química , Ácido Hialurónico/farmacología , Adsorción , Cartílago Articular/efectos de los fármacos , Cartílago Articular/metabolismo , Cationes/química , Animales , Polilisina/química , Polilisina/farmacología , Bovinos , Lubricantes/química , Lubricantes/farmacología , Fricción/efectos de los fármacos , Líquido Sinovial/metabolismo , Líquido Sinovial/química , Líquido Sinovial/efectos de los fármacos
8.
Phytomedicine ; 133: 155922, 2024 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-39126921

RESUMEN

BACKGROUND: Cartilage metabolism dysregulation is a crucial driver in knee osteoarthritis (KOA). Modulating the homeostasis can mitigate the cartilage degeneration in KOA. Curcumenol, derived from traditional Chinese medicine Curcuma Longa L., has demonstrated potential in enhancing chondrocyte proliferation and reducing apoptosis. However, the specific mechanism of Curcumenol in treating KOA remains unclear. This study aimed to demonstrate the molecular mechanism of Curcumenol in treating KOA based on the transcriptomics and metabolomics, and both in vivo and in vitro experimental validations. MATERIALS AND METHODS: In this study, a destabilization medial meniscus (DMM)-induced KOA mouse model was established. And the mice were intraperitoneally injected with Curcumenol at 4 and 8 mg/kg concentrations. The effects of Curcumenol on KOA cartilage and subchondral was evaluated using micro-CT, histopathology, and immunohistochemistry (IHC). In vitro, OA chondrocytes were induced with 10 µg/mL lipopolysaccharide (LPS) and treated with Curcumenol to evaluate the proliferation, apoptosis, and extracellular matrix (ECM) metabolism through CCK8 assay, flow cytometry, and chondrocyte staining. Furthermore, transcriptomics and metabolomics were utilized to identify differentially expressed genes (DEGs) and metabolites. Finally, integrating multi-omics analysis, virtual molecular docking (VMD), and molecular dynamics simulation (MDS), IHC, immunofluorescence (IF), PCR, and Western blot (WB) validation were conducted to elucidate the mechanism by which Curcumenol ameliorates KOA cartilage degeneration. RESULTS: Curcumenol ameliorated cartilage destruction and subchondral bone loss in KOA mice, promoted cartilage repair, upregulated the expression of COL2 while downregulated MMP3, and improved ECM synthesis metabolism. Additionally, Curcumenol also alleviated the damage of LPS on the proliferation activity and suppressed apoptosis, promoted ECM synthesis. Transcriptomic analysis combined with weighted gene co-expression network analysis (WGCNA) identified a significant downregulation of 19 key genes in KOA. Metabolomic profiling showed that Curcumenol downregulates the expression of d-Alanyl-d-alanine, 17a-Estradiol, Glutathione, and Succinic acid, while upregulating Sterculic acid and Azelaic acid. The integrated multi-omics analysis suggested that Curcumenol targeted KDM6B to regulate downstream protein H3K27me3 expression, which inhibited methylation at the histone H3K27, consequently reducing Succinic acid levels and improving KOA cartilage metabolism homeostasis. Finally, both in vivo and in vitro findings indicated that Curcumenol upregulated KDM6B, suppressed H3K27me3 expression, and stimulated collagen II expression and ECM synthesis, thus maintaining cartilage metabolism homeostasis and alleviating KOA cartilage degeneration. CONCLUSION: Curcumenol promotes cartilage repair and ameliorates cartilage degeneration in KOA by upregulating KDM6B expression, thereby reducing H3K27 methylation and downregulating Succinic Acid, restoring metabolic stability and ECM synthesis.


Asunto(s)
Condrocitos , Curcuma , Modelos Animales de Enfermedad , Ratones Endogámicos C57BL , Osteoartritis de la Rodilla , Ácido Succínico , Animales , Condrocitos/efectos de los fármacos , Condrocitos/metabolismo , Ratones , Masculino , Curcuma/química , Osteoartritis de la Rodilla/tratamiento farmacológico , Osteoartritis de la Rodilla/metabolismo , Ácido Succínico/metabolismo , Histona Demetilasas con Dominio de Jumonji/metabolismo , Proliferación Celular/efectos de los fármacos , Apoptosis/efectos de los fármacos , Sesquiterpenos/farmacología , Simulación del Acoplamiento Molecular , Cartílago Articular/efectos de los fármacos , Cartílago Articular/metabolismo , Matriz Extracelular/metabolismo , Matriz Extracelular/efectos de los fármacos , Humanos
9.
Arch Dermatol Res ; 316(8): 512, 2024 Aug 12.
Artículo en Inglés | MEDLINE | ID: mdl-39133320

RESUMEN

Vitamin A derivatives have inhibitory effects on cartilage tissue, such as decreasing chondrocyte proliferation and collagen synthesis, and increasing the loss of glycosaminoglycans and proteoglycans. Therefore, isotretinoin (a vitamin A derivative) may play a role in the pathogenesis of cartilage-related diseases like osteoarthritis by affecting the balance of cartilage tissue. The aim of this study was to evaluate the distal femoral cartilage thickness in acne patients under the systemic isotretinoin therapy and to determine whether it constitutes a risk factor for the development of osteoarthritis. The study included 52 patients (42 female, 10 male, mean age 23.31 ± 3.89 years) who were prescribed systemic isotretinoin for acne and completed at least 3 months of treatment, along with 45 healthy controls ((35 female, 10 male, mean age 23.85 ± 4.77 years). Bilateral distal femoral cartilage thickness was measured by ultrasonography before isotretinoin treatment and after the completion of the third month of treatment. After treatment, a statistically significant increase was found in the thickness of the right medial, right lateral, left medial, left lateral, and left intercondylar cartilage (p = 0.014, 0.012, 0.019, 0.027, 0.002, respectively). There was also an increase in the right intercondylar cartilage thickness, but this was not statistically significant (p = 0.1). Systemic isotretinoin seems to make cartilage thicker. The increase in femoral cartilage thickness observed after short-term isotretinoin treatment might be an indicator of very early-stage osteoarthritis. Extended follow-up studies with larger participant pools are necessary to substantiate this result.


Asunto(s)
Acné Vulgar , Cartílago Articular , Fémur , Isotretinoína , Humanos , Isotretinoína/efectos adversos , Isotretinoína/uso terapéutico , Isotretinoína/administración & dosificación , Femenino , Masculino , Acné Vulgar/tratamiento farmacológico , Acné Vulgar/patología , Acné Vulgar/diagnóstico , Adulto , Adulto Joven , Cartílago Articular/patología , Cartílago Articular/efectos de los fármacos , Cartílago Articular/diagnóstico por imagen , Fémur/diagnóstico por imagen , Fémur/efectos de los fármacos , Fémur/patología , Ultrasonografía , Fármacos Dermatológicos/efectos adversos , Fármacos Dermatológicos/uso terapéutico , Fármacos Dermatológicos/administración & dosificación , Osteoartritis/tratamiento farmacológico , Osteoartritis/patología , Osteoartritis/diagnóstico por imagen , Factores de Riesgo , Estudios de Casos y Controles
10.
Biomed Mater Eng ; 35(5): 475-485, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-39150826

RESUMEN

BACKGROUND: Osteoarthritis (OA) is a chronic and degenerative joint disease that remains a great challenge in treatment due to the lack of effective therapies. 4-octyl itaconate (4-OI) is a novel and potent modulator of inflammation for the treatment of inflammatory disease. However, the clinical usage of 4-OI is limited due to its poor solubility and low bioavailability. As a promising drug delivery strategy, injectable hydrogels offers an effective approach to address these limitations of 4-OI. OBJECTIVE: The aim of the study was to verify that the composite 4-OI/SA hydrogels could achieve a controlled release of 4-OI and reduce damage to articular cartilage in the group of osteoarthritic rats treated with the system. METHODS: In this study, an injectable composite hydrogel containing sodium alginate (SA) and 4-octyl itaconate (4-OI) has been developed for continuous intra-articular administration in the treatment of OA. RESULTS: After intra-articular injection in arthritic rats, the as-prepared 4-OI/SA hydrogel containing of 62.5 µM 4-OI effectively significantly reduced the expression of TNF-α, IL-1ß, IL-6 and MMP3 in the ankle fluid. Most importantly, the as-prepared 4-OI/SA hydrogel system restored the morphological parameters of the ankle joints close to normal. CONCLUSION: 4-OI/SA hydrogel shows a good anti-inflammatory activity and reverse cartilage disruption, which provide a new strategy for the clinical treatment of OA.


Asunto(s)
Alginatos , Antiinflamatorios , Preparaciones de Acción Retardada , Hidrogeles , Osteoartritis , Ratas Sprague-Dawley , Succinatos , Animales , Hidrogeles/química , Alginatos/química , Succinatos/química , Succinatos/farmacología , Antiinflamatorios/administración & dosificación , Antiinflamatorios/química , Antiinflamatorios/farmacología , Antiinflamatorios/farmacocinética , Preparaciones de Acción Retardada/química , Osteoartritis/tratamiento farmacológico , Osteoartritis/patología , Ratas , Masculino , Inyecciones Intraarticulares , Cartílago Articular/efectos de los fármacos , Cartílago Articular/patología , Cartílago Articular/metabolismo
11.
Proc Natl Acad Sci U S A ; 121(33): e2405454121, 2024 Aug 13.
Artículo en Inglés | MEDLINE | ID: mdl-39106310

RESUMEN

Regeneration of hyaline cartilage in human-sized joints remains a clinical challenge, and it is a critical unmet need that would contribute to longer healthspans. Injectable scaffolds for cartilage repair that integrate both bioactivity and sufficiently robust physical properties to withstand joint stresses offer a promising strategy. We report here on a hybrid biomaterial that combines a bioactive peptide amphiphile supramolecular polymer that specifically binds the chondrogenic cytokine transforming growth factor ß-1 (TGFß-1) and crosslinked hyaluronic acid microgels that drive formation of filament bundles, a hierarchical motif common in natural musculoskeletal tissues. The scaffold is an injectable slurry that generates a porous rubbery material when exposed to calcium ions once placed in cartilage defects. The hybrid material was found to support in vitro chondrogenic differentiation of encapsulated stem cells in response to sustained delivery of TGFß-1. Using a sheep model, we implanted the scaffold in shallow osteochondral defects and found it can remain localized in mechanically active joints. Evaluation of resected joints showed significantly improved repair of hyaline cartilage in osteochondral defects injected with the scaffold relative to defects injected with the growth factor alone, including implantation in the load-bearing femoral condyle. These results demonstrate the potential of the hybrid biomimetic scaffold as a niche to favor cartilage repair in mechanically active joints using a clinically relevant large-animal model.


Asunto(s)
Condrogénesis , Andamios del Tejido , Factor de Crecimiento Transformador beta1 , Animales , Andamios del Tejido/química , Ovinos , Factor de Crecimiento Transformador beta1/metabolismo , Condrogénesis/efectos de los fármacos , Polímeros/química , Ácido Hialurónico/química , Ácido Hialurónico/farmacología , Cartílago Articular/efectos de los fármacos , Regeneración/efectos de los fármacos , Diferenciación Celular/efectos de los fármacos , Ingeniería de Tejidos/métodos , Humanos , Materiales Biocompatibles/química , Condrocitos/efectos de los fármacos , Cartílago Hialino/metabolismo
12.
Int Immunopharmacol ; 141: 112863, 2024 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-39146779

RESUMEN

Rheumatoid arthritis (RA) is an autoimmune disease that causes persistent inflammation involving the joints, cartilage, and synovium. In individuals with RA, alterations in the composition of intestinal bacteria suggest the vital role of gut microbiota in immune dysfunction. Multiple therapies commonly used to treat RA can also alter the diversity of gut microbiota, further suggesting the modulation of gut microbiota as a prevention or treatment for RA. Therefore, a better understanding of the changes in the gut microbiota that accompany RA should facilitate the development of novel therapeutic approaches. In this study, B. coagulans BACO-17 not only significantly reduced paw swelling, arthritis scores, and hind paw and forepaw thicknesses but also protected articular cartilage and the synovium against RA degeneration, with a corresponding downregulation of TNF-α expression. The inhibition or even reversing of RA progression highlights B. coagulans BACO-17 as a novel therapeutic for RA worth investigating.


Asunto(s)
Artritis Reumatoide , Bacillus coagulans , Progresión de la Enfermedad , Microbioma Gastrointestinal , Animales , Artritis Reumatoide/tratamiento farmacológico , Artritis Reumatoide/inmunología , Microbioma Gastrointestinal/efectos de los fármacos , Masculino , Artritis Experimental/tratamiento farmacológico , Artritis Experimental/inmunología , Factor de Necrosis Tumoral alfa/metabolismo , Membrana Sinovial/inmunología , Membrana Sinovial/patología , Membrana Sinovial/efectos de los fármacos , Cartílago Articular/patología , Cartílago Articular/efectos de los fármacos , Probióticos/uso terapéutico , Humanos , Ratones , Ratas , Condrocitos/efectos de los fármacos , Condrocitos/inmunología
13.
ACS Biomater Sci Eng ; 10(9): 5796-5806, 2024 Sep 09.
Artículo en Inglés | MEDLINE | ID: mdl-39155687

RESUMEN

Cartilage defects caused by joint diseases are difficult to treat clinically. Tissue engineering materials provide a new means to promote the repair of cartilage defects. The purpose of this study is to design a novel scaffold of porous magnesium alloy loaded with icariin and sustained release in order to explore the effect and possible mechanism of this scaffold in repairing SD rat knee articular cartilage defect. We constructed a novel type of icariin/porous magnesium alloy scaffold, observed the structure of the scaffold by electron microscope, detected the drug release of icariin in the scaffold and the biological safety, and established an animal model of cartilage defect in the femoral intercondylar fossa of the knee joint in rats; the scaffold was placed in the defect. After 12 weeks of repair, the rat knee articular cartilage repair was evaluated by gross specimens and micro-CT, HE, safranin O-fast green, and toluidine blue staining combined with the modified Mankin's score. The protein expressions of the Wnt/ß-catenin signaling pathway-related factors (ß-catenin, Wnt5a, Wnt1, sFRP1) and chondrogenic differentiation-related factors (Sox9, Aggrecan, Col2α1) were detected by immunohistochemical staining. We found that the novel scaffold of icariin/porous magnesium alloy can release icariin slowly and has biosafety in rats. Compared with other groups, icariin/porous magnesium alloy can significantly promote the repair of cartilage defects and the expressions of ß-catenin, Wnt5a, Wnt1, Sox9, Aggrecan, and Col2α1 (P < 0.05). This novel scaffold can promote the repair of rat knee cartilage defects, and this process may be achieved by activating the Wnt/ß-catenin signaling pathway.


Asunto(s)
Aleaciones , Cartílago Articular , Flavonoides , Magnesio , Andamios del Tejido , Vía de Señalización Wnt , Animales , Masculino , Ratas , Aleaciones/química , Aleaciones/farmacología , beta Catenina/metabolismo , Cartílago Articular/efectos de los fármacos , Cartílago Articular/metabolismo , Cartílago Articular/patología , Flavonoides/farmacología , Flavonoides/química , Articulación de la Rodilla/patología , Articulación de la Rodilla/efectos de los fármacos , Magnesio/química , Magnesio/farmacología , Porosidad , Ratas Sprague-Dawley , Andamios del Tejido/química , Vía de Señalización Wnt/efectos de los fármacos
14.
Am J Sports Med ; 52(11): 2882-2892, 2024 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-39214071

RESUMEN

BACKGROUND: Posttraumatic osteoarthritis (PTOA) arises secondarily to joint trauma and is driven by catabolic inflammatory pathways. Alpha-2-macroglobulin (α2M) is a naturally occurring proteinase inhibitor found in human serum and synovial fluid that binds proteases as well as proinflammatory cytokines involved in the pathogenesis of PTOA. PURPOSE: (1) To investigate the therapeutic potential of intra-articular α2M injections during the acute stages of PTOA by inhibiting inflammatory pathways driven by the cytokines expressed by the synovium in a large preclinical Yucatan minipig model and (2) to determine if 3 intra-articular α2M injections have greater chondroprotective effects compared with 1 intra-articular injection. STUDY DESIGN: Controlled laboratory study. METHODS: A total of 48 Yucatan minipigs were randomized into 4 groups (n = 12 each): (1) modified intra-articular drilling (mIAD) and saline (mIAD + saline), (2) mIAD and 1 intra-articular α2M injection (mIAD +α2M-1), (3) mIAD and 3 α2M injections (mIAD +α2M-3), and (4) sham control. Surgical hindlimbs were harvested at 15 weeks after surgery. Cartilage degeneration, synovial changes, inflammatory gene expression, and matrix metalloproteinase levels were evaluated. Gait asymmetry was measured before and after surgery using a pressure-sensing walkway system. RESULTS: Macroscopic lesion areas and microscopic cartilage degeneration scores were lower in the mIAD +α2M-1 and mIAD +α2M-3 groups compared with the mIAD + saline group (P < .05) and similar to those in the sham group (P > .05). Synovial membrane scores of the mIAD +α2M-1 and mIAD +α2M-3 groups were lower than that of the mIAD + saline group (P < .05) and higher than that of the sham group (P < .05). Interleukin-1 beta, nuclear factor kappa B, and tumor necrosis factor alpha mRNA expression in the synovium and matrix metalloproteinase-1 levels in synovial fluid were significantly lower in the mIAD +α2M-1 and mIAD +α2M-3 groups compared with the mIAD + saline group (P < .05). No significant differences were observed between the mIAD +α2M-1 and mIAD +α2M-3 groups for all measured outcomes. There were early changes in gait (P < .05) between preoperative and postoperative time points for the mIAD + saline, mIAD +α2M-1, and mIAD +α2M-3 groups that normalized by 15 weeks. CONCLUSION: Animals receiving early α2M treatment exhibited less cartilage damage, milder synovitis, and lower inflammation compared with animals with no α2M treatment. These results exemplify the early anti-inflammatory effects of α2M and provide evidence that intra-articular α2M injections may slow the progression of PTOA. CLINICAL RELEVANCE: In patients presenting with an acute joint injury, an early intervention with α2M may have the potential to reduce cartilage degeneration from catabolic pathways and delay the development of PTOA.


Asunto(s)
Cartílago Articular , Modelos Animales de Enfermedad , Porcinos Enanos , Animales , Porcinos , Inyecciones Intraarticulares , Cartílago Articular/efectos de los fármacos , alfa-Macroglobulinas/metabolismo , Osteoartritis , Membrana Sinovial/efectos de los fármacos , Citocinas/metabolismo , alfa 2-Macroglobulinas Asociadas al Embarazo , Femenino , Inflamación , Osteoartritis de la Rodilla , Distribución Aleatoria
15.
J Mater Chem B ; 12(35): 8672-8687, 2024 Sep 11.
Artículo en Inglés | MEDLINE | ID: mdl-39115288

RESUMEN

Articular cartilage damage represents a prevalent clinical disease in orthopedics, with its regeneration and repair constituting a central focus in ongoing research endeavors. While hydrogel technology has achieved notable progress in the field of cartilage regeneration, addressing the repair of larger cartilage defects remains a significant and formidable challenge. In pursuit of achieving the repair of extensive cartilage defects, this study designed a polydopamine-modified chondroitin sulfate hydrogel loaded with SDF-1α (P-SCMA). This hydrogel, capable of directly providing glycosaminoglycans (GAGs), served as a platform for carrying growth factors and attracting mesenchymal stem cells for the in situ reconstruction of extensive cartilage defects. The results indicate that the P-SCMA hydrogel is capable of not only directly providing GAGs but also sustainably releasing SDF-1α. In the early stages, it promotes cell adhesion and proliferation and induces cell homing, while in the later stages, it further induces chondrogenesis by inhibiting the Wnt/ß-catenin pathway. This bioactive hydrogel, which possesses the functions of providing GAGs, promoting cell proliferation, inducing cell homing and chondrogenesis, is capable of promoting cartilage repair in multiple ways, providing new perspectives for the repair of extensive cartilage defects.


Asunto(s)
Quimiocina CXCL12 , Condrogénesis , Sulfatos de Condroitina , Hidrogeles , Sulfatos de Condroitina/química , Sulfatos de Condroitina/farmacología , Condrogénesis/efectos de los fármacos , Quimiocina CXCL12/metabolismo , Quimiocina CXCL12/farmacología , Animales , Hidrogeles/química , Hidrogeles/farmacología , Células Madre Mesenquimatosas/efectos de los fármacos , Células Madre Mesenquimatosas/citología , Proliferación Celular/efectos de los fármacos , Cartílago Articular/efectos de los fármacos , Cartílago Articular/metabolismo , Humanos , Preparaciones de Acción Retardada/química , Células Cultivadas
16.
J Cell Mol Med ; 28(16): e70019, 2024 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-39164798

RESUMEN

Knee osteoarthritis (KOA) is a chronic joint disease that significantly affects the health of the elderly. As an herbal remedy, Gubi decoction (GBD) has been traditionally used for the treatment of osteoarthritis-related syndromes. However, the anti-KOA efficacy and mechanism of GBD remain unclear. This study aimed to experimentally investigate the anti-KOA efficacy and the underlying mechanism of GBD. The medial meniscus (DMM) mice model and IL-1ß-stimulated chondrocytes were, respectively, constructed as in vivo and in vitro models of KOA to evaluate the osteoprotective effect and molecular mechanism of GBD. The UPLC-MS/MS analysis showed that GBD mainly contained pinoresinol diglucoside, rehmannioside D, hesperidin, liquiritin, baohuoside I, glycyrrhizic acid, kaempferol and tangeretin. Animal experiment showed that GBD could alleviate articular cartilage destruction and recover histopathological alterations in DMM mice. In addition, GBD inhibited chondrocyte apoptosis and restored DMM-induced dysregulated autophagy evidenced by the upregulation of ATG7 and LC3 II/LC3 I but decreased P62 level. Mechanistically, METTL3-mediated m6A modification decreased the expression of ATG7 in DMM mice, as it could be significantly attenuated by GBD. METTL3 overexpression significantly counteracted the protective effect of GBD on chondrocyte autophagy. Further research showed that GBD promoted proteasome-mediated ubiquitination degradation of METLL3. Our findings suggest that GBD could act as a protective agent against KOA. The protective effect of GBD may result from its promotion on chondrocyte autophagy by suppressing METTL3-dependent ATG7 m6A methylation.


Asunto(s)
Proteína 7 Relacionada con la Autofagia , Autofagia , Condrocitos , Metiltransferasas , Osteoartritis de la Rodilla , Animales , Condrocitos/metabolismo , Condrocitos/efectos de los fármacos , Autofagia/efectos de los fármacos , Osteoartritis de la Rodilla/metabolismo , Osteoartritis de la Rodilla/patología , Osteoartritis de la Rodilla/tratamiento farmacológico , Ratones , Proteína 7 Relacionada con la Autofagia/metabolismo , Proteína 7 Relacionada con la Autofagia/genética , Metiltransferasas/metabolismo , Metilación/efectos de los fármacos , Masculino , Medicamentos Herbarios Chinos/farmacología , Modelos Animales de Enfermedad , Apoptosis/efectos de los fármacos , Ratones Endogámicos C57BL , Adenosina/análogos & derivados , Adenosina/farmacología , Adenosina/metabolismo , Humanos , Cartílago Articular/metabolismo , Cartílago Articular/efectos de los fármacos , Cartílago Articular/patología
17.
Sichuan Da Xue Xue Bao Yi Xue Ban ; 55(4): 878-885, 2024 Jul 20.
Artículo en Chino | MEDLINE | ID: mdl-39170014

RESUMEN

Objective: To investigate the ameliorative effect of tanshinone ⅡA (Tan) on osteoarticular degeneration in ovariectomized rats (a postmenopausal estrogen deficiency model) and the mechanisms involved. Methods: Eight-week-old female Sprague Dawley (SD) rats were randomly allocated to 5 groups (n=10 each), including a Sham operation group (Sham), an ovariectomy group (OVX), and low, medium, and high-dose Tan groups. Eight weeks after bilateral ovariectomy, the rats in the low, medium, and high-dose Tan groups were treated with Tan at the doses of 5, 10, and 20 mg/kg for a duration of 28 days. Evaluation of the rat articular cartilage was performed using X-ray imaging, anatomical observation, hematoxylin and eosin (H&E) staining, and toluidine blue staining. Immunohistochemistry was performed to assess the expression levels of transforming growth factor ß1 (TGF-ß1), phosphorylated-smad2 (p-Smad2), type Ⅱ collagen (CⅡ), matrix metalloproteinase 9 (MMP-9), and MMP-13 in the cartilage tissue. Results: The knee joints of the OVX rats exhibited narrowed joint spaces, osteophyte formation, cartilage erosion or even localized cartilage cracks, faded methylene blue staining on the cartilage surface, disordered arrangement of chondrocytes, unclear or interrupted tidal line, and increased Kellgren-Lawrence grading, Pelletier grading, Mankin grading, and OARSI scores compared to those of the Sham group (P<0.01), as revealed by X-ray imaging, anatomical observation, and histological examination results. Tan ameliorated the degenerative changes in the knee joint caused by OVX in a dose-dependent manner while improving Kellgren-Lawrence grading, Pelletier grading, Mankin grading, and OARSI scores. Immunohistochemistry findings showed that TGF-ß1, p-Smad2, and CⅡ expression levels were significantly increased (P<0.01), while MMP-9 and MMP-13 expression levels were significantly decreased (P<0.01) in the articular cartilage of the Tan group compared to those of the OVX group, with all these effects being dose-dependent. Conclusion: Tan mitigates articular cartilage degeneration in ovariectomized rats, which may be related to the regulation of TGF-ß1/Smad2/MMPs signaling pathway.


Asunto(s)
Abietanos , Cartílago Articular , Ovariectomía , Ratas Sprague-Dawley , Transducción de Señal , Proteína Smad2 , Factor de Crecimiento Transformador beta1 , Animales , Femenino , Factor de Crecimiento Transformador beta1/metabolismo , Ratas , Abietanos/farmacología , Abietanos/uso terapéutico , Transducción de Señal/efectos de los fármacos , Proteína Smad2/metabolismo , Cartílago Articular/metabolismo , Cartílago Articular/efectos de los fármacos , Cartílago Articular/patología , Metaloproteinasa 9 de la Matriz/metabolismo , Metaloproteinasa 13 de la Matriz/metabolismo , Colágeno Tipo II/metabolismo
18.
Int J Rheum Dis ; 27(8): e15297, 2024 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-39175261

RESUMEN

BACKGROUND: Ferroptosis is caused by iron-dependent peroxidation of membrane phospholipids and chondrocyte ferroptosis contributes to osteoarthritis (OA) progression. Glutathione peroxidase 4 (GPX4) plays a master role in blocking ferroptosis. N6-methyladenosine (m6A) is an epigenetic modification among mRNA post-transcriptional modifications. This study investigated the effect of methyltransferase-like 14 (METTL14), the key component of the m6A methyltransferase, on chondrocyte ferroptosis via m6A modification. METHODS: An OA rat model was established through an intra-articular injection of monosodium iodoacetate in the right knee. OA cartilages in rat models were used for gene expression analysis. Primary mouse chondrocytes or ADTC5 cells were stimulated with IL-1ß or erastin. The m6A RNA methylation quantification kit was used to measure m6A level. The effect of METTL14 and GPX4 on ECM degradation and ferroptosis was investigated through western blotting, fluorescence immunostaining, propidium iodide staining, and commercially available kits. The mechanism of METTL14 action was explored through MeRIP-qPCR assays. RESULTS: METTL14 and m6A expression was upregulated in osteoarthritic cartilages and IL-1ß-induced chondrocytes. METTL14 depletion repressed the IL-1ß or erastin-stimulated ECM degradation and ferroptosis in mouse chondrocytes. METTL14 inhibited GPX4 gene through m6A methylation modification. GPX4 knockdown reversed the si-METTL14-mediated protection in IL-1ß-induced chondrocytes. CONCLUSION: METTL14 depletion inhibits ferroptosis and ECM degradation by suppressing GPX4 mRNA m6A modification in injured chondrocytes.


Asunto(s)
Condrocitos , Ferroptosis , Metiltransferasas , Fosfolípido Hidroperóxido Glutatión Peroxidasa , Animales , Condrocitos/efectos de los fármacos , Condrocitos/patología , Condrocitos/metabolismo , Condrocitos/enzimología , Ferroptosis/efectos de los fármacos , Fosfolípido Hidroperóxido Glutatión Peroxidasa/metabolismo , Fosfolípido Hidroperóxido Glutatión Peroxidasa/genética , Metiltransferasas/metabolismo , Metiltransferasas/genética , Ratones , Masculino , Adenosina/análogos & derivados , Adenosina/metabolismo , Adenosina/farmacología , Osteoartritis/patología , Osteoartritis/metabolismo , Osteoartritis/enzimología , Osteoartritis/genética , Osteoartritis/inducido químicamente , Cartílago Articular/patología , Cartílago Articular/metabolismo , Cartílago Articular/efectos de los fármacos , Células Cultivadas , Modelos Animales de Enfermedad , Ratas , Humanos , Ratas Sprague-Dawley
19.
Carbohydr Polym ; 343: 122424, 2024 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-39174114

RESUMEN

Articular cartilage and subchondral bone defects have always been problematic because the osteochondral tissue plays a crucial role in the movement of the body and does not recover spontaneously. Here, an injectable hydrogel composed of oxidized sodium alginate/gelatin/chondroitin sulfate (OSAGC) was designed for the minimally invasive treatment and promotion of osteochondral regeneration. The OSAGC hydrogel had a double network based on dynamic covalent bonds, demonstrating commendable injectability and self-healing properties. Chondroitin sulfate was organically bound to the hydrogel network, retaining its own activity and gradually releasing during the degradation process as well as improving mechanical properties. The compressive strength could be increased up to 3 MPa by regulating the concentration of chondroitin sulphate and the oxidation level, and this mechanical stimulation could help repair injured tissue. The OSAGC hydrogel had a favourable affinity to articular cartilage and was able to release active ingredients in a sustained manner over 3 months. The OSAGC showed no cytotoxic effects. Results from animal studies demonstrated its capacity to regenerate new bone tissue in four weeks and new cartilage tissue in twelve weeks. The OSAGC hydrogel represented a promising approach to simplify bone surgery and repair damaged osteochondral tissue.


Asunto(s)
Alginatos , Cartílago Articular , Sulfatos de Condroitina , Hidrogeles , Alginatos/química , Alginatos/farmacología , Animales , Sulfatos de Condroitina/química , Sulfatos de Condroitina/farmacología , Cartílago Articular/efectos de los fármacos , Hidrogeles/química , Hidrogeles/farmacología , Regeneración Ósea/efectos de los fármacos , Gelatina/química , Conejos , Fuerza Compresiva , Ingeniería de Tejidos/métodos , Inyecciones , Condrocitos/efectos de los fármacos , Condrocitos/citología , Andamios del Tejido/química , Regeneración/efectos de los fármacos
20.
J Nanobiotechnology ; 22(1): 466, 2024 Aug 02.
Artículo en Inglés | MEDLINE | ID: mdl-39095867

RESUMEN

BACKGROUND: Osteoarthritis (OA) is a degenerative joint disease characterized by the progressive degeneration of articular cartilage, leading to pain, stiffness, and loss of joint function. The pathogenesis of OA involves multiple factors, including increased intracellular reactive oxygen species (ROS), enhanced chondrocyte apoptosis, and disturbances in cartilage matrix metabolism. These processes contribute to the breakdown of the extracellular matrix (ECM) and the loss of cartilage integrity, ultimately resulting in joint damage and dysfunction. RNA interference (RNAi) therapy has emerged as a promising approach for the treatment of various diseases, including hATTR and acute hepatic porphyria. By harnessing the natural cellular machinery for gene silencing, RNAi allows for the specific inhibition of target genes involved in disease pathogenesis. In the context of OA, targeting key molecules such as matrix metalloproteinase-13 (MMP13), which plays a critical role in cartilage degradation, holds great therapeutic potential. RESULTS: In this study, we developed an innovative therapeutic approach for OA using a combination of liposome-encapsulated siMMP13 and NG-Monomethyl-L-arginine Acetate (L-NMMA) to form an injectable hydrogel. The hydrogel served as a delivery vehicle for the siMMP13, allowing for sustained release and targeted delivery to the affected joint. Experiments conducted on destabilization of the medial meniscus (DMM) model mice demonstrated the therapeutic efficacy of this composite hydrogel. Treatment with the hydrogel significantly inhibited the degradation of cartilage matrix, as evidenced by histological analysis showing preserved cartilage structure and reduced loss of proteoglycans. Moreover, the hydrogel effectively suppressed intracellular ROS accumulation in chondrocytes, indicating its anti-oxidative properties. Furthermore, it attenuated chondrocyte apoptosis, as demonstrated by decreased levels of apoptotic markers. CONCLUSION: In summary, the injectable hydrogel containing siMMP13, endowed with anti-ROS and anti-apoptotic properties, may represent an effective therapeutic strategy for osteoarthritis in the future.


Asunto(s)
Apoptosis , Condrocitos , Hidrogeles , Metaloproteinasa 13 de la Matriz , Osteoartritis , Especies Reactivas de Oxígeno , Animales , Osteoartritis/tratamiento farmacológico , Osteoartritis/metabolismo , Osteoartritis/patología , Especies Reactivas de Oxígeno/metabolismo , Apoptosis/efectos de los fármacos , Hidrogeles/química , Metaloproteinasa 13 de la Matriz/metabolismo , Ratones , Condrocitos/metabolismo , Condrocitos/efectos de los fármacos , Ratones Endogámicos C57BL , Masculino , Cartílago Articular/metabolismo , Cartílago Articular/efectos de los fármacos , Cartílago Articular/patología , Liposomas/química , Humanos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA