Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 5 de 5
Filtrar
Más filtros

Bases de datos
Tipo del documento
País de afiliación
Intervalo de año de publicación
1.
Heliyon ; 9(9): e19812, 2023 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-37809453

RESUMEN

Tenghuang Jiangu Capsule (THJGC) is a Chinese herbal formula used for the treatment of osteoporosis and osteoarthritis in China, but its mechanism for treating osteoporosis is not clear. The aim of this study was to investigate the therapeutic effect of THJGC on osteoporosis and its intrinsic mechanism through network pharmacology and experimental validation. Drugs and potential targets were obtained from several reliable databases through network pharmacology, and these targets were integrated and analyzed using bioinformatics and molecular docking strategies. Quercetin, lignans and kaempferol were identified as key components, and the key targets included Akt1, MAPKs, and CASP3. Subsequently, UPLC-MS/MS analysis confirmed the presence of components in THJGC for the treatment of osteoporosis. In addition, using ex vivo and in vivo models, it was confirmed that THJGC inhibited H2O2-induced ROS generation and apoptosis, and reduced OVX-induced bone loss in a mouse model of osteoporosis. Our data suggest that THJGC has antioxidant, bone formation-promoting, bone resorption-inhibiting, and MC3T3-E1 apoptosis-reducing effects, and thus has anti-osteoporotic properties. In conclusion, it may be a promising pharmacologic adjuvant treatment for osteoporosis.

2.
J Cell Biochem ; 124(8): 1155-1172, 2023 08.
Artículo en Inglés | MEDLINE | ID: mdl-37357411

RESUMEN

This study aimed to explore the effect of myricitrin on osteoblast differentiation in mice immortalised bone marrow mesenchymal stem cells (imBMSCs). Additionally, ovariectomy (OVX) mice were employed to examine the effect of myricitrin on bone trabecular loss in vivo. The effect of myricitrin on the proliferation of imBMSCs was evaluated using a cell counting kit-8 assay. Alizarin red staining, alkaline phosphatase staining were performed to elucidate osteogenesis. Furthermore, qRT-PCR and western blot determined the expression of osteo-specific genes and proteins. To screen for candidate targets, mRNA transcriptome genes were sequenced using bioinformatics analyses. Western blot and molecular docking analysis were used to examine target signalling markers. Moreover, rescue experiments were used to confirm the effect of myricitrin on the osteogenic differentiation of imBMSCs. OVX mice were also used to estimate the delay capability of myricitrin on bone trabecular loss in vivo using western blot, micro-CT, tartaric acid phosphatase (Trap) staining, haematoxylin and eosin staining, Masson staining and immunochemistry. In vitro, myricitrin significantly enhanced osteo-specific genes and protein expression and calcium deposition. Moreover, mRNA transcriptome gene sequencing and molecular docking analysis revealed that this enhancement was accompanied by an upregulation of the PI3K/AKT signalling pathway. Furthermore, copanlisib, a PI3K inhibitor, partially reversed the osteogenesis promotion induced by myricitrin. In vivo, western blot, micro-CT, hematoxylin and eosin staining, Masson staining, Trap staining and immunochemistry revealed that bone trabecular loss rate was significantly alleviated in the myricitrin low- and high-dose groups, with an increased expression of osteopontin, osteoprotegerin, p-PI3K and p-AKT compared to the OVX group. Myricitrin enhances imBMSC osteoblast differentiation and attenuate bone mass loss partly through the upregulation of the PI3K/AKT signalling pathway. Thus, myricitrin has therapeutic potential as an antiosteoporosis drug.


Asunto(s)
Enfermedades Óseas Metabólicas , Osteogénesis , Animales , Femenino , Ratones , Diferenciación Celular , Células Cultivadas , Eosina Amarillenta-(YS)/farmacología , Simulación del Acoplamiento Molecular , Osteogénesis/genética , Ovariectomía , Fosfatidilinositol 3-Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-akt/genética , Proteínas Proto-Oncogénicas c-akt/metabolismo , ARN Mensajero
3.
Biomed Pharmacother ; 157: 113915, 2023 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-36379122

RESUMEN

BACKGROUND: Iron homeostasis plays a positive role in articular cartilage health. Excessive iron or iron overload can induce oxidative stress damage in chondrocytes and ferroptosis cell death, advancing knee osteoarthritis (KOA). However, up to date, few effective agents treat iron overload-induced KOA (IOKOA). Chinese herbal medicine (CHM) provides abundant resources for drug selection to manage bone metabolic conditions, including osteoporosis. Biochanin A (BCA) is a novel bioactive multifunctional natural compound isolated from Huangqi, which has protective effects on bone loss. Nevertheless, the function and mechanism of BCA in treating IOKOA are still elusive. PURPOSE: This study seeks to uncover the potential therapeutic targets and mechanisms of BCA in the management of KOA with iron accumulation. METHODS: Iron dextrin (500 mg/kg) was intraperitoneally injected into mice to establish the iron overloaded mice model. OA was induced through surgery, and the progression was evaluated eight weeks following surgery. OA severity was evaluated with micro-CT and Safranin-O/Fast green staining in vivo. Iron deposition in the knee joint and synovium was assessed using Perl's Prussian blue staining. Ferric ammonium citrate (FAC) was then administered to primary chondrocytes to evaluate iron regulators mediated iron homeostasis. Toluidine blue staining was utilized to identify chondrocytes in vitro. The vitality of the cells was assessed using the CCK-8 test. The apoptosis rate of cells was measured using Annexin V-FITC/PI assay. The intracellular iron level was detected utilizing the calcein-AM test. Reactive oxygen species (ROS), lipid-ROS, and mitochondrial membrane potentiality were reflected via fluorescence density. Utilizing RT-qPCR and western blotting, the expression level was determined. RESULTS: Micro-CT and histological staining of knee joints showed greater cartilage degradation and higher iron buildup detected in iron-overloaded mice. BCA can reduce iron deposition and the severity of KOA. Toluidine blue staining and the CCK-8 assay indicated that BCA could rescue chondrocytes killed by iron. Cell apoptosis rates were increased due to iron overload but improved by BCA. Further, the intracellular content of iron, ROS, and lipid-ROS was increased with ferric ammonium citrate (FAC) treatment but restored after treatment with different concentrations of BCA. JC-1 staining revealed that BCA could reduce mitochondrial damage induced by iron overload. CONCLUSION: Iron overload was shown to promote chondrocyte ferroptosis in vivo and in vitro. Moreover, iron overload suppressed the expression of collagen II and induced MMP expression by catalyzing ROS generation with mitochondrial dysfunction. Our results showed that BCA could directly reduce intracellular iron concentration by inhibiting TfR1 and promoting FPN but also target the Nrf2/system xc-/GPX4 signaling pathway to scavenge free radicals and prevent lipid peroxidation. The results of this research indicate that BCA regulates iron homeostasis during the progression of osteoarthritis, which can open a new field of treatment for KOA.


Asunto(s)
Sobrecarga de Hierro , Osteoartritis de la Rodilla , Animales , Ratones , Condrocitos/metabolismo , Hierro/metabolismo , Sobrecarga de Hierro/complicaciones , Sobrecarga de Hierro/tratamiento farmacológico , Sobrecarga de Hierro/metabolismo , Lípidos/farmacología , Osteoartritis de la Rodilla/patología , Especies Reactivas de Oxígeno/metabolismo , Cloruro de Tolonio/metabolismo , Cloruro de Tolonio/farmacología
4.
Phytomedicine ; 105: 154330, 2022 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-35905566

RESUMEN

BACKGROUND: The traditional Chinese medicine Gusuibu, the rhizome of Rhizoma Drynariae, is used to treat rheumatism and fractures. Naringenin (NAR) is an active ingredient in Gusuibu and has significant anti-inflammatory and antioxidant effects. However, the role of naringenin in iron overload-induced osteoarthritis (IOOA) is unknown. HYPOTHESIS: NAR reduces cartilage damage in IOOA. METHODS: The effects of NAR on the viability of IOOA chondrocytes and the synthesis ability of type II collagen were evaluated using cell counting kit (CCK8) and toluidine blue assays. To determine the mechanism of action and characteristics of NAR, the intracellular iron ion content, apoptosis rate, and mitochondrial membrane potential (MMP) change, and malondialdehyde (MDA) levels, as well as the degree of reactive oxygen species (ROS) and lipid hydroperoxide (LPO) accumulation in the cells were detected in vitro and verified using western blotting and quantitative real-time PCR (qRT-PCR). To verify the role of NAR in vivo, IOOA mice were established using iron dextran and surgery-induced destabilised medial meniscus. Changes in the articular cartilage and subchondral bone were examined using Safranin O-fast Green staining (S-O), haematoxylin-eosin staining (H&E), and microcomputed tomography (µCT). RESULTS: In vitro, NAR attenuated the impairment of cell viability, apoptosis, and MMP caused by ferric ammonium citrate and interleukin-1ß co-culture, increased the levels of MDA, reduced the expression of matrix metallopeptidase (MMP)3, MMP13, and Bax, and restored the expression of type II collagen (Col II). NAR showed a slight iron accumulation-reducing effect. NAR alleviated the accumulation of ROS and LPO in IOOA chondrocytes and upregulated antioxidant genes nuclear factor E2-related factor 2 (NRF2) and haem oxygenase 1 (HO-1). When ML385, a specific NRF-2 inhibitor, was added, the protective effect of NAR was significantly inhibited. In vivo, NAR reduced synovitis and attenuated cartilage damage and subchondral bone proliferation in IOOA mice. CONCLUSIONS: NAR can reduce oxidative stress through the NRF2-HO-1 pathway, alleviate cartilage damage under iron overload, and has the potential to treat IOOA.


Asunto(s)
Sobrecarga de Hierro , Osteoartritis , Animales , Antioxidantes , Apoptosis , Colágeno Tipo II , Flavanonas , Hierro , Ratones , Factor 2 Relacionado con NF-E2 , Estrés Oxidativo , Especies Reactivas de Oxígeno , Transducción de Señal , Microtomografía por Rayos X
5.
J Ethnopharmacol ; 294: 115292, 2022 Aug 10.
Artículo en Inglés | MEDLINE | ID: mdl-35447200

RESUMEN

ETHNOPHARMACOLOGICAL RELEVANCE: XianLing GuBao Capsule (XLGB) is often used to treat osteoarthritis (OA), osteoporosis, fractures, and other musculoskeleton disorders. However, the molecular mechanism of XLGB for treating OA is still unclear. AIM OF THE STUDY: This study set out to uncover the molecular mechanism underlying the treatment of osteoarthritis with XLGB. MATERIALS AND METHODS: Disease genes were obtained from CTD, DisGeNET, and GeneCards databases, and XLGB drug targets were obtained from ETCM and target genes predicted by XLGB metabolic components reported in the literature. Then we used the Venn diagram viewer to extract disease and drug intersection genes as potential therapeutic genes for Protein-protein interaction (PPI), GO terminology, and KEGG pathway analysis. Subsequently, we performed qRT-PCR, Western blot and histological analysis to validate the therapeutic effect of XLGB against OA and its molecular mechanism. RESULTS: A total of 1039 OA genes and 949 XLGB target genes were collected, and finally 188 potential therapeutic target genes were obtained. PPI network analysis indicated that the main target genes for XLGB to treat OA include Akt1, Mapk3, Il-6, Il-1ß, Ptgs2, Mmp9, etc. The results of KEGG and GO enrichment analysis suggested that XLGB may treat OA by anti-inflammatory and reducing extracellular matrix degradation. In vitro, XLGB down-regulated the expressions of Mmp3, Mmp9, Mmp12, Mmp13, Cox-2, Il-6, increased the expression of Collagen II and Sox9. Mechanistically, XLGB inhibits the activation of PI3K/AKT/NF-κB and MAPK pathways. Moreover, the results of animal experiments indicated that XLGB reduced cartilage destruction, bone resorption, and synovitis in osteoarthritic rats. CONCLUSIONS: XLGB has a protective effect against OA by suppressing PI3K/AKT/NF-κB and MAPK signaling. Our study provides a theoretical basis for XLGB in the treatment of osteoarthritis.


Asunto(s)
Osteoartritis , Proteínas Proto-Oncogénicas c-akt , Animales , Condrocitos , Biología Computacional , Interleucina-6 , Metaloproteinasa 9 de la Matriz , FN-kappa B/metabolismo , Osteoartritis/tratamiento farmacológico , Osteoartritis/genética , Fosfatidilinositol 3-Quinasas , Ratas
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA