Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
Add more filters










Publication year range
1.
Heliyon ; 10(1): e23510, 2024 Jan 15.
Article in English | MEDLINE | ID: mdl-38170113

ABSTRACT

Esophageal cancer (EC) is a common and devastating tumor of the upper digestive tract. Unfortunately, by the time any symptoms have manifested, the disease has often progressed to an advanced stage and is accompanied by macro- and micrometastases, including in the bones. The treatment of esophageal cancer with bone metastases remains clinically challenging, given the poor prognosis associated with this condition. Effective prognostic biomarkers can help medical staff choose the appropriate operation and treatment plan, that is for most beneficial for making patients. Current treatments for esophageal cancer with bone metastases include pain-relieving drugs, surgical therapy, radiotherapy (RT), chemotherapy (CT, including molecular-targeted drug therapy), endocrine therapy (ET), bisphosphonates (BPs) and interventional therapy. Of these robust measures, radiotherapy has emerged as a particularly promising therapy for bone metastases from esophageal cancer. Substantial progress has been made in radiation therapy techniques since the discovery of X-rays by Roentgen in 1895. In its palliative capacity, the key goals of radiotherapy are to relieve the patients' bone pain and debilitate effects, including relieving spinal cord compression, correcting the spinal deformity and restoring spinal stability. However, it is worth mentioning that RT for esophageal cancer has various side effects. Currently, the available studies focused exclusively on radiotherapy for ECBM are too small to draw any definitive conclusions, and each of these studies has significant limitations. In this review, in addition to the epidemiology described at the beginning, we will explore the current prognostic biomarkers and radiotherapy for esophageal cancer, with a particular focus on those with bone metastases.

2.
J Immunol Res ; 2023: 2061071, 2023.
Article in English | MEDLINE | ID: mdl-37425490

ABSTRACT

Dysregulation of miRNAs in chondrocytes has been confirmed to participate in osteoarthritis (OA) progression. Previous study has screen out several key miRNAs may play crucial role in OA based on bioinformatic analysis. Herein, we identified the downregulation of miR-1 in OA samples and inflamed chondrocytes. The further experiments revealed that miR-1 played an essential role in maintaining chondrocytes proliferation, migration, antiapoptosis, and anabolism. Connexin 43 (CX43) was further predicted and confirmed to be the target of miR-1, and mediated the promotion effects of miR-1 in regulating chondrocyte functions. Mechanistically, miR-1 maintained the expression of GPX4 and SLC7A11 by targeting CX43, attenuated the accumulation of intracellular ROS, lipid ROS, MDA, and Fe2+ in chondrocytes, thereby inhibiting the ferroptosis of chondrocytes. Finally, experimental OA model was constructed by anterior cruciate ligament transection surgery, and Agomir-1 was injected into the joint cavity of mice to assess the protective effect of miR-1 in OA progression. Histological staining, immunofluorescence staining and Osteoarthritis Research Society International score revealed that miR-1 could alleviate the OA progression. Therefore, our study elucidated the mechanism of miR-1 in OA in detail and provided a new insight for the treatment of OA.


Subject(s)
Ferroptosis , MicroRNAs , Osteoarthritis , Mice , Animals , Chondrocytes/metabolism , Connexin 43/genetics , Connexin 43/metabolism , Reactive Oxygen Species/metabolism , Osteoarthritis/genetics , Osteoarthritis/metabolism , MicroRNAs/genetics , MicroRNAs/metabolism , Apoptosis/genetics
3.
Biomed Res Int ; 2023: 1150768, 2023.
Article in English | MEDLINE | ID: mdl-36817861

ABSTRACT

Background: Osteosarcoma (OS) is the most common primary aggressive sarcoma of bone, with massive aberrant expression of oncogenes related to the development of OS. RALA, a kind of small Ras-like guanosine triphosphatases, has been identified as a potential therapeutic target in several types of tumor, but its role in OS remains largely unknown. Methods: Abnormal expression of RALA was proven in the Cancer Genome Atlas (TCGA), Genotype-Tissue Expression (GTEx), Therapeutically Applicable Research to Generate Effective Treatments (TARGET), and RNA-sequence of samples and cell lines. The role of RALA in OS was analyzed in terms of DNA methylation, immune cell infiltration, and patient survival. The cancer-promoting effect of RALA was demonstrated in cell lines and xenograft osteosarcoma models. A prognostic scoring model incorporating RALA as an indicator was established with the clinical samples that we collected. Results: The results showed that RALA was highly expressed in human OS tissues and cell lines. Survival analysis demonstrated that RALA was the sole independent risk factor for poor overall survival and disease-free survival in OS patients and impacted the proportion of infiltrating immune cells and DNA methylation in the OS tumor microenvironment. By gene-gene interaction analysis, we found that the expression of RALA was highly correlated to the expression of ABCE1. Similar to RALA, upregulated ABCE1 is correlated with poor survival outcome of OS patients. In addition, the functional experiment demonstrated that higher expression of RALA promoted the proliferation, migration, and invasion of OS cells. In vivo results were similar with the in vitro results. We examined m6a methylation-related genes and found that m6A methylation is responsible for the abnormal expression of RALA. Finally, the prognostic prediction model of RALA could be used to predict the long-term outcome of OS patients. Conclusions: We identified RALA as an oncogene in OS, and RALA upregulation in a concerted manner with ABCE1 was significantly associated with worse outcomes of OS patients. Targeting RALA may prove to be a novel target for OS immunotherapy in future clinical practice.


Subject(s)
Bone Neoplasms , Osteosarcoma , Humans , Prognosis , Bone Neoplasms/pathology , Cell Movement/genetics , Bone and Bones/metabolism , Osteosarcoma/pathology , Tumor Microenvironment , ral GTP-Binding Proteins/metabolism
4.
Mater Sci Eng C Mater Biol Appl ; 118: 111388, 2021 Jan.
Article in English | MEDLINE | ID: mdl-33254994

ABSTRACT

As cartilage tissue lacks the innate ability to mount an adequate regeneration response, damage to it is detrimental to the quality of life of the subject. The emergence of three-dimensional bioprinting (3DBP) technology presents an opportunity to repair articular cartilage defects. However, widespread adoption of this technique has been impeded by difficulty in preparing a suitable bioink and the toxicity inherent in the chemical crosslinking process of most bioinks. Our objective was to develop a crosslinker-free bioink with the same biological activity as the original cartilage extracellular matrix (ECM) and good mechanical strength. We prepared bioinks containing different concentrations of silk fibroin and decellularized extracellular matrix (SF-dECM bioinks) mixed with bone marrow mesenchymal stem cells (BMSCs) for 3D bioprinting. SF and dECM interconnect with each other through physical crosslinking and entanglement. A porous structure was formed by removing the polyethylene glycol from the SF-dECM bioink. The results showed the SF-dECM construct had a suitable mechanical strength and degradation rate, and the expression of chondrogenesis-specific genes was found to be higher than that of the SF control construct group. Finally, we confirmed that a SF-dECM construct that was designed to release TGF-ß3 had the ability to promote chondrogenic differentiation of BMSCs and provided a good cartilage repair environment, suggesting it is an ideal scaffold for cartilage tissue engineering.


Subject(s)
Bioprinting , Extracellular Matrix , Porosity , Printing, Three-Dimensional , Quality of Life , Silk , Tissue Engineering , Tissue Scaffolds
5.
Jt Dis Relat Surg ; 31(3): 411-418, 2020.
Article in English | MEDLINE | ID: mdl-32962569

ABSTRACT

OBJECTIVES: This study aims to explore the mechanism by which osteoblast autophagy participated in glucocorticoid-induced femoral head necrosis (FHN). MATERIALS AND METHODS: Thirty male specific-pathogen-free C57 mice (age, one month; weighing 20-25 g) were randomly divided into blank control, dexamethasone and rapamycin-dexamethasone groups (n=10). After six weeks of intervention, right femoral head was obtained to observe morphology and to calculate percentage of empty lacunae. MC3T3-E1 cells were randomly divided into normal, dexamethasone, rapamycin and dexamethasone-rapamycin groups, and cultured for 24 h. Microtubule-associated protein 1 light chain 3 (LC3)-I, LC3-II, mammalian target of rapamycin (mTOR) and Beclin-1 protein expressions were detected by Western blot. RESULTS: In rapamycin-dexamethasone group, some bone trabeculae in medullary cavity ruptured and atrophied, and subchondral bone underwent local necrosis. The total apoptosis rates of dexamethasone and rapamycin-dexamethasone groups surpassed that of blank control group, and the former two groups had significantly different rates (p<0.001). LC3-II/LC3-I of dexamethasone group was lower than those of rapamycin and dexamethasone-rapamycin groups (p<0.001), and the ratio of rapamycin group surpassed that of dexamethasone-rapamycin group (p<0.001). Dexamethasone group had higher mTOR protein expression than those of rapamycin and dexamethasone- rapamycin groups (p<0.001), and the expression of rapamycin group was lower than that of dexamethasone-rapamycin group (p<0.001). The Beclin-1 protein expression of dexamethasone group was lower than those of rapamycin and dexamethasone- rapamycin groups (p<0.001), and the expression of rapamycin group exceeded that of dexamethasone-rapamycin group (p<0.05). CONCLUSION: Osteoblast autophagy may play a crucial protective role in dexamethasone-induced FHN. The attenuation of autophagy may be related to the affected expressions of key autophagy regulators mTOR and Beclin-1.


Subject(s)
Autophagy/drug effects , Femur Head Necrosis , Glucocorticoids/pharmacology , Osteoblasts , Sirolimus/pharmacology , Animals , Apoptosis/drug effects , Beclin-1/metabolism , Femur Head Necrosis/chemically induced , Femur Head Necrosis/metabolism , Femur Head Necrosis/pathology , Femur Head Necrosis/prevention & control , Immunosuppressive Agents/pharmacology , Male , Mice , Microtubule-Associated Proteins/metabolism , Osteoblasts/drug effects , Osteoblasts/metabolism , Osteoblasts/pathology , TOR Serine-Threonine Kinases/metabolism , Treatment Outcome
6.
Regen Med ; 14(7): 663-680, 2019 07.
Article in English | MEDLINE | ID: mdl-31313645

ABSTRACT

Aim: To investigate the effect of cartilage extracellular matrix (ECM) particle size on the chondrogenic differentiation of bone marrow-derived mesenchymal stem cells (BMSCs). Materials & methods: BMSCs were seeded into the scaffolds fabricated by small particle ECM materials and large particle ECM materials. For the positive control, chondrogenically induced BMSCs were seeded into commercial poly-lactic-glycolic acid scaffolds. Macroscopic observation, histological and immunohistochemical staining, mechanical testing and biochemical analysis were performed to the cell-scaffold constructs. Results: BMSCs in small particle ECM materials and poly-lactic-glycolic acid scaffolds were induced to differentiate into chondrocytes, while BMSCs in the large particle ECM materials scaffold did not differentiate into chondrocytes. Conclusion: The small ECM particle materials improved the induction ability of the cartilage ECM-derived scaffold.


Subject(s)
Bone Marrow Cells , Cartilage/chemistry , Cell Differentiation , Cells, Immobilized , Chondrogenesis , Extracellular Matrix/chemistry , Mesenchymal Stem Cell Transplantation , Mesenchymal Stem Cells , Animals , Bone Marrow Cells/cytology , Bone Marrow Cells/metabolism , Cells, Immobilized/cytology , Cells, Immobilized/metabolism , Cells, Immobilized/transplantation , Female , Goats , Heterografts , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/metabolism , Mice , Mice, Nude , Particle Size , Rats
7.
Tissue Eng Part C Methods ; 24(11): 645-658, 2018 11.
Article in English | MEDLINE | ID: mdl-30351193

ABSTRACT

IMPACT STATEMENT: This study presented a new method to fabricate SF-ECM scaffolds that potentially promote chondrogenesis of BMSCs, and open up new possibilities for using SF-ECM scaffolds as an off-the-shelf strategy for joint cartilage regeneration. It is worthy of further investigation in knee joints of animals, and beyond knee cartilage, this scaffold may also serve as an ideal biomaterial for the regeneration of other joint cartilages.


Subject(s)
Cartilage, Articular/cytology , Chondrogenesis , Extracellular Matrix/chemistry , Mesenchymal Stem Cells/cytology , Silk/chemistry , Tissue Engineering/methods , Tissue Scaffolds , Animals , Bombyx , Cell Differentiation , Cells, Cultured , Extracellular Matrix/metabolism , Female , Goats , Mice , Mice, Nude , Rats , Rats, Sprague-Dawley , Regeneration
8.
Tissue Eng Part A ; 24(21-22): 1680-1692, 2018 11.
Article in English | MEDLINE | ID: mdl-29779446

ABSTRACT

Repair of articular cartilage defects using bilayered scaffolds is problematic because tissue-engineered cartilage is prone to overgrowth toward subchondral bone, resulting in structural abnormalities of cartilage and subchondral bone. A "twice freeze-drying" technique was used to construct a dense isolation layer between the cartilage and subchondral bone layers in an integrated bilayered scaffold to prevent cartilage from excessive downgrowth. Briefly, beta-tricalcium phosphate was used for the subchondral bone layer of the scaffold, high-concentration chitosan/gelatin solution for the dense isolation layer, and low-concentration chitosan/gelatin solution for the cartilage layer. As controls, cell-free trilayered scaffolds, autologous osteochondral transplantation, and the bilayered scaffolds were used for repair of osteochondral defects. After 6 months, two of the eight goats in the bilayered scaffold group showed conspicuous cartilage downgrowth, whereas no excessive downgrowth of cartilage was observed in the trilayered scaffold group. Moreover, there was no difference in the repair efficacy between the trilayered scaffold and mosaicplasty group. The results confirmed that the trilayered scaffold effectively prevented cartilage downgrowth with better cartilage repair.


Subject(s)
Biomimetic Materials , Cartilage, Articular , Femur , Tissue Engineering , Tissue Scaffolds/chemistry , Animals , Biomimetic Materials/chemistry , Biomimetic Materials/pharmacology , Cartilage, Articular/injuries , Cartilage, Articular/metabolism , Cartilage, Articular/pathology , Chitosan/chemistry , Chitosan/pharmacology , Chondrocytes/metabolism , Chondrocytes/pathology , Chondrocytes/transplantation , Femur/injuries , Femur/metabolism , Femur/pathology , Goats , Male
9.
J Orthop Res ; 2018 Feb 21.
Article in English | MEDLINE | ID: mdl-29464749

ABSTRACT

To evaluate the effects of intraarticular injections of autologous platelet-rich plasma (PRP) or bone marrow concentrate (BMC) on osteoarthritis (OA), 24 adult goats were equally divided into control (Ctrl), saline (NS), PRP, and BMC groups, and OA was induced by surgery in NS, PRP, and BMC groups. Autologous PRP and BMC were obtained from whole blood and bone marrow aspirates, respectively. The data revealed, platelets were increased in BMC by 1.8-fold, monocytes by 5.6-fold, TGF-ß1 by 7.7-fold, and IGF-1 by 3.6-fold (p < 0.05), and platelets were increased in PRP by 2.9-fold, and TGF-ß1 by 3.3-fold (p < 0.05). From the sixth week post-operation, saline, PRP, and BMC were administered by intraarticular injection once every 4 weeks, three consecutive times. After the animals were sacrificed, inflammatory cytokines in the synovial fluid was measured, and bone and cartilage degeneration progression was observed by macroscopy, histology, and immunohistochemistry. Compared with the NS group, the level of inflammatory cytokines was reduced in the PRP and BMC groups (p < 0.05). Histologically, delayed cartilage degeneration and higher levels of extracellular matrix (ECM) were observed in both PRP and BMC treated groups (p < 0.05). Furthermore, the BMC group showed greater cartilage protection and less ECM loss than the PRP group (p < 0.05). In summary, this study showed that intraarticular injection of autologous PRP and BMC has therapeutic efficacy in a goat osteoarthritis model, with the greater benefit in terms of cartilage protection being observed in the BMC-treated group than PRP. © 2018 Orthopaedic Research Society. Published by Wiley Periodicals, Inc. J Orthop Res.

10.
Cell Physiol Biochem ; 44(4): 1509-1525, 2017.
Article in English | MEDLINE | ID: mdl-29197863

ABSTRACT

BACKGROUND/AIMS: Periodic mechanical stress could significantly promote chondrocyte proliferation and matrix synthesis. However, the mechanisms underlying the ability of chondrocyte detecting and responding to periodic mechanical stimuli have not been well delineated. METHODS: Quantitative proteomic analysis was performed to construct the differently expressed proteome profiles of chondrocyte under pressure. Then a combination of Western blot, quantitative real-time PCR, lentiviral vector and histological methods were used to confirm the proteomic results and investigate the mechanoseing mechanism. RESULTS: Growth factor receptor-bound protein 2 (Grb2), a component of integrin adhesome, was found a 1.49-fold increase in dynamic stress group. This process was mediated through integrin ß1, leading to increased phosphorylation of focal adhesion kinase (FAK) and extracellular signal-regulated kinase 1/2 (ERK1/2) respectively and then produce the corresponding biological effects. CONCLUSION: This was the first time to demonstrate Grb2 has such an important role in periodic mechanotransduction, and the proteomic data could facilitate the further investigation of chondrocytes mechanosensing.


Subject(s)
GRB2 Adaptor Protein/metabolism , Proteomics , Stress, Mechanical , Aggrecans/genetics , Aggrecans/metabolism , Animals , Cells, Cultured , Chondrocytes/cytology , Chondrocytes/metabolism , Chromatography, High Pressure Liquid , Collagen Type II/genetics , Collagen Type II/metabolism , Focal Adhesion Protein-Tyrosine Kinases/genetics , Focal Adhesion Protein-Tyrosine Kinases/metabolism , GRB2 Adaptor Protein/antagonists & inhibitors , GRB2 Adaptor Protein/genetics , Immunohistochemistry , Mitogen-Activated Protein Kinase 1/genetics , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/genetics , Mitogen-Activated Protein Kinase 3/metabolism , Phosphorylation , RNA Interference , RNA, Small Interfering/metabolism , Rats , Rats, Sprague-Dawley , Real-Time Polymerase Chain Reaction , Spectrometry, Mass, Electrospray Ionization , Tissue Engineering , Up-Regulation
11.
Int Immunopharmacol ; 43: 1-6, 2017 Feb.
Article in English | MEDLINE | ID: mdl-27936459

ABSTRACT

The inflammatory cytokine interleukin 17 (IL-17) is an important contributor of rheumatoid arthritis (RA) chronicity. Although several microRNAs (miRNAs) have been shown to regulate RA pathogenesis, the function of miRNAs in articular chondrocytes during rheumatoid arthritis pathogenesis is unclear. Here we showed that miR-23a was downregulated in articular cartilage tissues from rheumatoid arthritis patients. MiR-23a suppressed IL-17 inflammatory cytokine-induced NF-κB activation and several proinflammatory mediators expression, such as cytokine IL-6, chemokine MCP-1, and matrix metalloproteinase MMP-3 in articular chondrocytes. Furthermore, we found that the miR-23a expression was inversely correlated with IKKα expression in articular cartilage tissues from rheumatoid arthritis patients. We identified that IKKα was the direct target of miR-23a and miR-23a inhibited IL-17-mediated proinflammatory mediators expression via targeting the IKKα in primary articular chondrocytes. Together, our study provides the first evidence of a role for miR-23a regulated IL-17-mediated proinflammatory mediators expression in rheumatoid arthritis by directly targeting IKKα. Our findings provide novel evidence that may be useful for future studies exploring therapeutic approaches for rheumatoid arthritis by targeting miR-23a. Thus, miR-23a may be a common therapeutic target for rheumatoid arthritis.


Subject(s)
Arthritis, Rheumatoid/genetics , Cartilage, Articular/pathology , Chondrocytes/physiology , I-kappa B Kinase/genetics , Interleukin-17/immunology , MicroRNAs/genetics , Arthritis, Rheumatoid/immunology , Arthritis, Rheumatoid/therapy , Cells, Cultured , Chemokine CCL2/genetics , Chemokine CCL2/metabolism , Gene Expression Regulation/genetics , Humans , I-kappa B Kinase/metabolism , Inflammation Mediators/metabolism , Interleukin-6/genetics , Interleukin-6/metabolism , Matrix Metalloproteinase 3/genetics , Matrix Metalloproteinase 3/metabolism , Molecular Targeted Therapy
12.
Cell Physiol Biochem ; 37(4): 1301-14, 2015.
Article in English | MEDLINE | ID: mdl-26473622

ABSTRACT

BACKGROUND/AIMS: Periodic mechanical stress activates integrin ß1-initiated signal pathways to promote chondrocyte proliferation and matrix synthesis. Integrin ß1 overexpression has been demonstrated to play important roles in improving the activities and functions of several non-chondrocytic cell types. Therefore, in the current study, we evaluated the effects of integrin ß1 up-regulation on periodic mechanical stress-induced chondrocyte proliferation, matrix synthesis and ERK1/2 phosphorylation in chondrocyte monolayer culture, and evaluated the quality of tissue-engineered cartilage constructed in vitro under periodic mechanical stress combined with integrin ß1 up-regulation. METHODS AND RESULTS: Our results revealed that under periodic mechanical stress, pre-treatment with integrin ß1-wild type vector significantly enhanced chondrocyte proliferation and matrix synthesis and promoted ERK1/2 phosphorylation in comparison to mock transfectants. Furthermore, when chondrocytes were seeded in PLGA scaffolds, more accumulated GAG and type II collagen tissue were detected after Lv-integrin ß1 transfection compared with sham controls exposed to periodic mechanical stress. In contrast, in the Lv-shRNA-integrin ß1 group, the opposite results were observed. CONCLUSION: Our findings collectively suggest that in addition to periodic mechanical stress, integrin ß1 up-regulation in chondrocytes could further improve the quality of tissue-engineered cartilage.


Subject(s)
Cartilage, Articular/pathology , Integrin beta1/metabolism , Stress, Mechanical , Tissue Engineering , Aggrecans/genetics , Aggrecans/metabolism , Animals , Cartilage, Articular/metabolism , Cell Proliferation , Cells, Cultured , Chondrocytes/cytology , Chondrocytes/metabolism , Collagen Type II/genetics , Collagen Type II/metabolism , Female , Genetic Therapy , Integrin beta1/chemistry , Integrin beta1/genetics , Lactic Acid/chemistry , Male , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Polyglycolic Acid/chemistry , Polylactic Acid-Polyglycolic Acid Copolymer , RNA Interference , Rats , Rats, Sprague-Dawley , Real-Time Polymerase Chain Reaction , Tissue Scaffolds , Up-Regulation
13.
Mol Med Rep ; 12(2): 1753-60, 2015 Aug.
Article in English | MEDLINE | ID: mdl-25892273

ABSTRACT

In the present study, the effects and mechanisms of mesenchymal stem cells (MSCs) on interleukin (IL)-1ß-stimulated rat chondrocytes, as well as cartilage from a rat model of osteoarthritis (OA) induced by anterior cruciate ligament transection and medial meniscectomy were investigated. Confluent rat chondrocytes were treated with IL-1ß (10 ng/ml), then cultured indirectly with or without MSCs at a ratio of 2:1. Total RNA and protein were collected at various time-points, and western blot and reverse transcription-quantitative polymerase chain reaction analyses were used to investigate the expression of type II collagen (Col2), aggrecan, matrix metalloproteinase-13 (MMP-13) and cyclooxygenase-2 (COX-2). The activation of extracellular signal-regulated kinases 1/2 (ERK1/2), c-Jun N-terminal kinase (JNK), p38 mitogen-activated protein kinase (MAPK), nuclear factor-κB (NF-κB) p65 and inhibitory-κ-B-α (IκBα) were also assessed by western blotting. In addition, the in vivo effects of MSCs in a rat OA model were assessed by histology and western blot analysis. The results indicated that in vitro, IL-1ß markedly upregulated the expression of MMP-13, COX-2, phosphorylated ERK1/2, JNK, p38 MAPK and NF-κB p65, and inhibited the expression of Col2, aggrecan and IκBα. Conversely, MSCs enhanced the expression of Col2, aggrecan and IκBα, and inhibited the expression of MMP-13 and NF-κB p65 in IL-1ß-stimulated rat chondrocytes. In vivo histological and western blot analyses revealed analogous results to the in vitro findings. The results of the present study demonstrated that MSCs suppressed the inflammatory response and extracellular matrix degradation in IL-1ß­induced rat chondrocytes, as well as cartilage in a osteoarthritic rat model, in part via the NF-κB signaling pathway.


Subject(s)
Cartilage/cytology , Chondrocytes/drug effects , Interleukin-1beta/pharmacology , Mesenchymal Stem Cells/metabolism , Aggrecans/genetics , Aggrecans/metabolism , Animals , Bone Marrow Cells/cytology , Cartilage/pathology , Cells, Cultured , Chondrocytes/cytology , Chondrocytes/metabolism , Coculture Techniques , Collagen Type II/genetics , Collagen Type II/metabolism , Cyclooxygenase 2/genetics , Cyclooxygenase 2/metabolism , Disease Models, Animal , JNK Mitogen-Activated Protein Kinases/genetics , JNK Mitogen-Activated Protein Kinases/metabolism , Male , Matrix Metalloproteinase 13/genetics , Matrix Metalloproteinase 13/metabolism , Mesenchymal Stem Cells/cytology , Osteoarthritis/metabolism , Osteoarthritis/pathology , Rats , Rats, Sprague-Dawley , Transcriptome/drug effects , p38 Mitogen-Activated Protein Kinases/genetics , p38 Mitogen-Activated Protein Kinases/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...