Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 400
Filter
1.
Aging (Albany NY) ; 16(8): 7043-7059, 2024 Apr 17.
Article in English | MEDLINE | ID: mdl-38637111

ABSTRACT

Osteoarthritis (OA) is a prevalent degenerative condition commonly observed in the elderly, leading to consequential disability. Despite notable advancements made in clinical strategies for OA, its pathogenesis remains uncertain. The intricate association between OA and metabolic processes has yet to receive comprehensive exploration. In our investigation, we leveraged public databases and applied machine learning algorithms, including WGCNA, LASSO, RF, immune infiltration analysis, and pathway enrichment analysis, to scrutinize the role of lipid metabolism-associated genes (LAGs) in the OA. Our findings identified three distinct biomarkers, and evaluated their expression to assess their diagnostic value in the OA patients. The exploration of immune infiltration in these patients revealed an intricate relationship between immune cells and the identified biomarkers. In addition, in vitro experiments, including qRT-PCR, Western blot, chondrocyte lipid droplets detection and mitochondrial fatty acid oxidation measurement, further verified abnormal expressions of selected LAGs in OA cartilage and confirmed the correlation between lipid metabolism and OA.


Subject(s)
Biomarkers , Lipid Metabolism , Machine Learning , Osteoarthritis , Humans , Lipid Metabolism/genetics , Osteoarthritis/genetics , Osteoarthritis/immunology , Osteoarthritis/metabolism , Biomarkers/metabolism , Algorithms , Chondrocytes/metabolism , Chondrocytes/immunology
2.
Exp Cell Res ; 410(1): 112945, 2022 01 01.
Article in English | MEDLINE | ID: mdl-34838812

ABSTRACT

OBJECTIVE: Mesenchymal stem cells (MSCs), especially genetically modified MSCs, have become a promising therapeutic approach for the treatment of rheumatoid arthritis (RA) through modulating immune responses. However, most MSCs used in the treatment of RA are modified based on a single gene. In this study, we evaluated the therapeutic effects of human BMSCs (hBMSCs) with COX-2 silence and TGF-ß3 overexpression in the treatment of RA in a rabbit model. MATERIALS AND METHODS: hBMSCs were cotransfected with shCOX-2 and TGF-ß3 through lentiviral vector delivery. After SPIO-Molday ION Rhodamine-B™ (MIRB) labeling, lenti-shCOX2-TGF-ß3 hBMSCs, lenti-shCOX2 hBMSCs, lenti-TGF-ß3 hBMSCs, hBMSCs without genetic modification, or phosphate-buffered saline (PBS) were injected into the knee joint of rabbits with antigen-induced arthritis (AIA). The diameter of the knee joint and soft-tissue swelling score (STS) were recorded, and the levels of inflammatory mediators, including interleukin-1ß (IL-1ß), tumor necrosis factor alpha (TNF-α), interleukin-6 (IL-6), and prostaglandin E2 (PGE2) were evaluated by ELISA. Clinical 3.0T MR imaging (MRI) was used to track the distribution and dynamic migration of hBMSCs in the joint. Histopathological and immunohistochemical assays were conducted to localize labeled hBMSCs and assess the alteration of synovial hyperplasia, inflammatory cell infiltration, and cartilage damage. RESULTS: COX-2 silencing and TGF-ß3 overexpression in hBMSCs were confirmed through real-time PCR and Western blot analyses. Reduced joint diameter, soft-tissue swelling (STS) score, and PGE2, IL-1ß, and TNF-α expression were detected 4 weeks after injection of MIRB-labeled lenti-shCOX2-TGF-ß3 hBMSCs into the joint in rabbits with AIA. Eight weeks after hBMSC injection, reduced inflammatory cell infiltration, improved hyperplasia of the synovial lining, recovered cartilage damage, and increased matrix staining were observed in joints injected with lenti-shCOX2-TGF-ß3 hBMSCs and lenti-shCOX2 hBMSCs. Slight synovial hyperplasia, no surface fibrillation, and strong positive expression of collagen II staining in chondrocytes and cartilage matrix were detected in the joints 12 weeks after injection of lenti-shCOX2-TGF-ß3 hBMSCs. In addition, hBMSCs were detected by MRI imaging throughout the process of hBMSC treatment. CONCLUSION: Intra-articular injection of hBMSCs with COX-2 silence and TGFß3 overexpression not only significantly inhibited joint inflammation and synovium hyperplasia, but also protected articular cartilage at the early stage. In addition, intra-articular injection of hBMSCs with COX-2 silence and TGFß3 overexpression promoted chondrocyte and matrix proliferation. This study provides an alternative therapeutic strategy for the treatment of RA using genetically modified hBMSCs.


Subject(s)
Arthritis, Rheumatoid/genetics , Cyclooxygenase 2/genetics , Inflammation/genetics , Transforming Growth Factor beta3/genetics , Animals , Antigens/pharmacology , Arthritis, Rheumatoid/etiology , Arthritis, Rheumatoid/immunology , Arthritis, Rheumatoid/pathology , Cell Differentiation/genetics , Cell Differentiation/immunology , Chondrocytes/immunology , Chondrocytes/metabolism , Cyclooxygenase 2/drug effects , Cyclooxygenase 2 Inhibitors/pharmacology , Disease Models, Animal , Humans , Immunity/genetics , Inflammation/etiology , Inflammation/immunology , Inflammation/pathology , Mesenchymal Stem Cell Transplantation , Mesenchymal Stem Cells , Rabbits
3.
Int Immunopharmacol ; 101(Pt A): 108179, 2021 Dec.
Article in English | MEDLINE | ID: mdl-34601329

ABSTRACT

The term "osteoimmunology" was coined to denote the bridge between the immune system and the skeletal system. Osteoimmunology is interdisciplinary, and a full understanding and development of this "bridge" will provide an in-depth understanding of the switch between body health and disease development. B lymphocytes can promote the maturation and differentiation of osteoclasts, and osteoclasts have a negative feedback effect on B lymphocytes. Different subtypes of T lymphocytes regulate osteoclasts in different directions. T lymphocytes have a two-way regulatory effect on osteoblasts, while B lymphocytes have minimal regulatory effects on osteoblasts. In contrast, osteoblasts can promote the differentiation and maturation of T lymphocytes and B lymphocytes. Different immune cells have different effects on chondrocytes; some cooperate with each other, while some antagonize each other. In a healthy adult body, bone resorption and bone formation are in a dynamic balance under the action of multiple mechanisms. In this review, we summarize the interactions and key signaling molecular mechanisms between each type of cell in the immune system and the skeletal system.


Subject(s)
Cell Communication/immunology , Osteoarthritis/immunology , Animals , B-Lymphocytes/immunology , B-Lymphocytes/metabolism , Cell Differentiation/immunology , Chondrocytes/immunology , Chondrocytes/pathology , Disease Models, Animal , Humans , Mesenchymal Stem Cells/physiology , Osteoarthritis/pathology , Osteoblasts/immunology , Osteoblasts/metabolism , Osteoclasts/immunology , Osteoclasts/metabolism , T-Lymphocytes/immunology , T-Lymphocytes/metabolism
4.
Aging (Albany NY) ; 13(18): 22134-22147, 2021 09 22.
Article in English | MEDLINE | ID: mdl-34550907

ABSTRACT

BACKGROUND: Osteoarthritis (OA) is degenerative joint disorder mainly characterized by long-term pain with limited activity of joints, the disease has no effective preventative therapy. Rutin (RUT) is a flavonoid compound, present naturally. The flavonoid shows range of biological activities such as anti-inflammatory and anti-cancer effect. We screened RUT for its activity against osteoarthritis with in vivo and in vitro models of osteoarthritis. METHODS: Animal model of OA was developed using C57BL/6 mice by surgical destabilization of medial meniscus. For in vitro studies the human articular cartilage tissues were used which were collected from osteoarthritis patients and were processed to isolate chondrocytes. The chondrocytes were submitted to advanced glycation end products (AGEs) for inducing osteoarthritis in vitro. Cell viability was done by CCK-8 assay, ELISA analysis for MMP13, collage II, PGE2, IL-6, TNF-α, ADAMTS-5 and MMP-13. Western blot analysis was done for expression of proteins and in silico analysis was done by docking studies. RESULTS: Pretreatment of RT showed no cytotoxic effect and also ameliorated the AGE mediated inflammatory reaction on human chondrocytes in vitro. Treatment of RT inhibited the levels of COX-2 and iNOS in AGE exposed chondrocytes. RT decreased the AGE mediated up-regulation of IL-6, NO, TNF-α and PGE-2 in a dose dependent manner. Pretreatment of RT decreased the extracellular matrix degradation, inhibited expression of TRAF-6 and BCL-2 the NF-κB/MAPK pathway proteins. The treatment of RT in mice prevented the calcification of cartilage tissues, loss of proteoglycans and also halted the narrowing of joint space is mice subjected to osteoarthritis. The in-silico analysis suggested potential binding affinity of RT with TRAF-6 and BCL-2. CONCLUSION: In brief RT inhibited AGE-induced inflammatory reaction and also degradation of ECM via targeting the NF-κB/MAPK pathway proteins BCL-2 and TRAF-6. RT can be a potential molecule in treating OA.


Subject(s)
Anti-Inflammatory Agents/administration & dosage , Extracellular Matrix/immunology , Glycation End Products, Advanced/immunology , Osteoarthritis/drug therapy , Osteoarthritis/immunology , Proto-Oncogene Proteins c-bcl-2/immunology , Rutin/administration & dosage , TNF Receptor-Associated Factor 6/immunology , Animals , Cartilage, Articular/drug effects , Cartilage, Articular/immunology , Chondrocytes/drug effects , Chondrocytes/immunology , Cyclooxygenase 2/genetics , Cyclooxygenase 2/immunology , Dinoprostone/immunology , Disease Models, Animal , Extracellular Matrix/drug effects , Extracellular Matrix/genetics , Humans , Male , Mice, Inbred C57BL , NF-kappa B/genetics , NF-kappa B/immunology , Osteoarthritis/genetics , Proto-Oncogene Proteins c-bcl-2/genetics , TNF Receptor-Associated Factor 6/genetics
5.
Inflamm Res ; 70(10-12): 1129-1139, 2021 Dec.
Article in English | MEDLINE | ID: mdl-34562102

ABSTRACT

OBJECTIVE: This study aimed to investigate the biochemical effects of osteoarthritic infrapatellar fat pad (IPFP) on cartilage and the underlying mechanisms. METHODS: Human IPFP and articular cartilage were collected from end-stage osteoarthritis (OA) patients during total knee arthroplasty. IPFP-derived fat-conditioned medium (FCM) was used to stimulate human primary chondrocytes and cartilage explants. Functional effect of osteoarthritic IPFP was explored in human primary chondrocytes and articular cartilage in vitro and ex vivo. Activation of relative pathways and its effects on chondrocytes were assessed through immunoblotting and inhibition experiments, respectively. Neutralization test was performed to identify the main factors and their associated pathways responsible for the effects of IPFP. RESULTS: Osteoarthritic IPFP-derived FCM significantly induced extracellular matrix (ECM) degradation in both human primary chondrocytes and cartilage explants. Several pathways, such as NF-κB, mTORC1, p38MAPK, JNK, and ERK1/2 signaling, were significantly activated in human chondrocytes with osteoarthritic IPFP-derived FCM stimulation. Interestingly, inhibition of p38MAPK and ERK1/2 signaling pathway could alleviate the detrimental effects of FCM on chondrocytes, while inhibition of other signaling pathways had no similar results. In addition, IL-1ß and TNF-α instead of IL-6 in osteoarthritic IPFP-derived FCM played key roles in cartilage degradation via activating p38MAPK rather than ERK1/2 signaling pathway. CONCLUSION: Osteoarthritic IPFP induces the degradation and inflammation of cartilage via activation of p38MAPK and ERK1/2 pathways, in which IL-1ß and TNF-α act as the key factors. Our study suggests that modulating the effects of IPFP on cartilage may be a promising strategy for knee OA intervention.


Subject(s)
Adipose Tissue/immunology , Cartilage, Articular/immunology , Osteoarthritis, Knee/immunology , Patella/immunology , Cells, Cultured , Chondrocytes/immunology , Cytokines/immunology , Humans , MAP Kinase Signaling System , p38 Mitogen-Activated Protein Kinases/immunology
6.
Int J Mol Sci ; 22(17)2021 Aug 31.
Article in English | MEDLINE | ID: mdl-34502384

ABSTRACT

Chronic low-grade inflammation plays a central role in the pathogenesis of osteoarthritis (OA), and several pro- and anti-inflammatory cytokines have been implicated to mediate and regulate this process. Out of these cytokines, particularly IFNγ, IL-1ß, IL-4 and IL-17 are associated with different phenotypes of T helper (TH) cells and macrophages, both examples of cells known for great phenotypic and functional heterogeneity. Chondrocytes also display various phenotypic changes during the course of arthritis. We set out to study the hypothesis of whether chondrocytes might adopt polarized phenotypes analogous to TH cells and macrophages. We studied the effects of IFNγ, IL-1ß, IL-4 and IL-17 on gene expression in OA chondrocytes with RNA-Seq. Chondrocytes were harvested from the cartilage of OA patients undergoing knee replacement surgery and then cultured with or without the cytokines for 24 h. Total RNA was isolated and sequenced, and GO (Gene Ontology) functional analysis was performed. We also separately investigated genes linked to OA in recent genome wide expression analysis (GWEA) studies. The expression of more than 2800 genes was significantly altered in chondrocytes treated with IL-1ß [in the C(IL-1ß) phenotype] with a fold change (FC) > 2.5 in either direction. These included a large number of genes associated with inflammation, cartilage degradation and attenuation of metabolic signaling. The profile of genes differentially affected by IFNγ (the C(IFNγ) phenotype) was relatively distinct from that of the C(IL-1ß) phenotype and included several genes associated with antigen processing and presentation. The IL-17-induced C(IL-17) phenotype was characterized by the induction of a more limited set of proinflammatory factors compared to C(IL-1ß) cells. The C(IL-4) phenotype induced by IL-4 displayed a differential expression of a rather small set of genes compared with control, primarily those associated with TGFß signaling and the regulation of inflammation. In conclusion, our results show that OA chondrocytes can adopt diverse phenotypes partly analogously to TH cells and macrophages. This phenotypic plasticity may play a role in the pathogenesis of arthritis and open new therapeutic avenues for the development of disease-modifying treatments for (osteo)arthritis.


Subject(s)
Chondrocytes/immunology , Chondrocytes/metabolism , Osteoarthritis/metabolism , Cells, Cultured , Cytokines/immunology , Cytokines/metabolism , Humans , Inflammation/metabolism , Osteoarthritis/immunology , Phenotype , Sequence Analysis, RNA/methods , Signal Transduction/immunology , Th1 Cells/immunology , Th1 Cells/metabolism , Th17 Cells/immunology , Th17 Cells/metabolism , Th2 Cells/immunology , Th2 Cells/metabolism
7.
Mol Pharm ; 18(9): 3235-3246, 2021 09 06.
Article in English | MEDLINE | ID: mdl-34387081

ABSTRACT

Nanoparticles (NPs) have wide potential applications in the biomedical field. To promote targeted and controlled delivery of encapsulated drugs, it is fundamentally important to understand the factors regulating NP uptake by different cells. Thus, the goal of the present study is to assess the internalization rates of different NPs under normal and proinflammatory states in primary human articular chondrocytes (hACs), human umbilical vein endothelial cells (EA), and human monocytes (THP-1). Here, we compared chitosan-hyaluronic acid (Ch-HA) polymeric NPs, methoxypolyethylene glycol amine-glutathione-palmitic acid (mPEG-GSHn-PA) micelles, and cholesterol/l-α-phosphatidylcholine/DSPE-PEG-Mal (Chol/EPC/DSPE-PEG-Mal) unilamellar liposomes (LUVs). Our results reveal the importance of surface charge and chemistry in determining the levels of NP internalization. Under normal conditions, the cellular uptake was ≈30% for Ch-HA NPs and ≈100% for mPEG-GSHn-PA micelles and Chol/EPC/DSPE-PEG-Mal LUVs. A proinflammatory cell state promoted a higher uptake of the Ch-HA NPs by EA cells (93% after 24 h). Since the therapeutic efficacy of the NP-loaded cargo is dependent on trafficking routes after cellular internalization, we tested their internalization pathways. Accordingly, caveolae-mediated endocytosis or energy-independent non-endocytic pathways, which circumvent lysosomal degradation, were accomplished in hACs and EA by LUVs and in M1 polarized macrophages by micelles. The present outcomes highlight the importance of considering cellular uptake and internalization pathways by the target cell when designing functional NPs for therapeutic applications.


Subject(s)
Anti-Inflammatory Agents/administration & dosage , Arthritis/drug therapy , Nanoparticle Drug Delivery System/pharmacokinetics , Arthritis/immunology , Chondrocytes/immunology , Chondrocytes/metabolism , Endocytosis , Human Umbilical Vein Endothelial Cells/immunology , Human Umbilical Vein Endothelial Cells/metabolism , Humans , Macrophages/immunology , Macrophages/metabolism , Micelles , Polymers/chemistry , Primary Cell Culture , THP-1 Cells
8.
Aging (Albany NY) ; 13(16): 20774-20792, 2021 08 29.
Article in English | MEDLINE | ID: mdl-34455406

ABSTRACT

MicroRNAs (miRNAs) contribute to osteoarthritis (OA) development. Nevertheless, the function and mechanism of miR-30b-5p in OA are unclear. In the present article, we gauged the miR-30b-5p level in OA patients and analyzed its correlation with OA stages. Then, we conducted in-vivo and in-vitro gain-of-function assays to determine the function of miR-30b-5p, silent information regulator 2 homolog 1 (SIRT1) and Fox. Cell counting Kit-8 (CCK-8) assay, BrdU assay and flow cytometry were utilized to gauge cell viability and apoptosis of human chondrocyte (HC-A). The targeting association between miR-30b-5p and SIRT1 was validated through the dual-luciferase reporter assay and RNA immunoprecipitation (RIP) experiment. The results signified that miR-30b-5p was up-regulated in OA patients, OA rats and interleukin-1ß (IL-1ß)-induced chondrocytes. The higher miR-30b-5p expression brought about progressive stages of OA patients and enhanced levels of pro-inflammatory mediators in the synovial fluid. Functionally, overexpressing miR-30b-5p hampered cell viability, aggravated chondrocyte apoptosis and NLRP3 inflammasome activation induced by IL-1ß, while down-regulating miR-30b-5p exerted the reverse effects. The in-vivo experiment exhibited that down-regulating miR-30b-5p improved joint pain and loss of articular cartilage in the rats with restrained inflammation and NLRP3 inflammasome activation. Mechanistically, miR-30b-5p targeted the 3'-non-translated region (3'UTR) of SIRT1, and miR-30b-5p was inducible with NF-κB phosphorylation enhancement. Overexpressing SIRT1 or inhibiting NF-κB relieved miR-30b-5p-induced apoptosis and NLRP3 inflammasome activation by promoting FoxO3a, while down-regulating SIRT1 or FoxO3a reversed miR-30b-5p-in-induced anti-inflammatory and apoptosis-suppressive effects. Collectively, NF-κB-induced miR-30b-5p modulates chondrocyte apoptosis and OA progression by regulating the SIRT1-FoxO3a-mediated NLRP3 inflammasome.


Subject(s)
Cartilage, Articular/immunology , Forkhead Box Protein O3/immunology , Inflammasomes/immunology , NF-kappa B/immunology , NLR Family, Pyrin Domain-Containing 3 Protein/immunology , Osteoarthritis/immunology , Sirtuin 1/immunology , Aged , Animals , Arthralgia/genetics , Arthralgia/immunology , Chondrocytes/immunology , Female , Forkhead Box Protein O3/genetics , Humans , Inflammasomes/genetics , Interleukin-1beta/genetics , Interleukin-1beta/immunology , Male , MicroRNAs/genetics , MicroRNAs/immunology , Middle Aged , NF-kappa B/genetics , NLR Family, Pyrin Domain-Containing 3 Protein/genetics , Osteoarthritis/genetics , Rats , Sirtuin 1/genetics
9.
Aging (Albany NY) ; 13(16): 20651-20660, 2021 08 26.
Article in English | MEDLINE | ID: mdl-34438368

ABSTRACT

NOD-like receptor family caspase recruitment domain family domain containing 5 (NLRC5) has been found to be a critical mediator of inflammatory response. However, the role of NLRC5 in osteoarthritis (OA) has not been reported. Our results showed that NLRC5 was down-regulated by IL-1ß induction in chondrocytes. Overexpression of NLRC5 in chondrocytes significantly suppressed IL-1ß-induced inflammatory response through inhibiting the production of multiple inflammatory mediators including inducible nitric oxide synthases (iNOS), and cyclooxygenase-2 (COX-2), prostaglandin E2 (PGE2), NO, TNF-α and IL-6, as well matrix metalloproteinase 3 (MMP-3) and MMP-13. Consistently, NLRC5 knockdown exhibited opposite effects on the production of these inflammatory mediators in IL-1ß-induced chondrocytes. Furthermore, overexpression of NLRC5 increased the IĸBα expression, while decreased the p-p65 expression, indicating that NLRC5 inhibited the activation of NF-κB signaling. Additionally, inhibition of NF-κB by PDTC mitigated the si-NLRC5-mediated promotion of IL-1ß-induced inflammatory injury in chondrocytes. Finally, NLRC5 treatment ameliorated cartilage degeneration in an OA model in rats. Taken together, these findings revealed that NLRC5 attenuated IL-1ß-induced inflammatory injury in chondrocytes through regulating the NF-κB signaling.


Subject(s)
Chondrocytes/immunology , Interleukin-1beta/immunology , Intracellular Signaling Peptides and Proteins/immunology , NF-kappa B/immunology , Osteoarthritis/immunology , Animals , Cyclooxygenase 2/genetics , Cyclooxygenase 2/immunology , Dinoprostone/immunology , Humans , Interleukin-1beta/genetics , Intracellular Signaling Peptides and Proteins/genetics , Male , Matrix Metalloproteinase 13/genetics , Matrix Metalloproteinase 13/immunology , NF-kappa B/genetics , Nitric Oxide Synthase Type II/genetics , Nitric Oxide Synthase Type II/immunology , Osteoarthritis/genetics , Rats , Rats, Sprague-Dawley , Signal Transduction
10.
Int J Mol Sci ; 22(13)2021 Jul 05.
Article in English | MEDLINE | ID: mdl-34281278

ABSTRACT

Osteoarthritis (OA) is hallmarked by a progressive degradation of articular cartilage. One major driver of OA is inflammation, in which cytokines such as IL-6, TNF-α and IL-1ß are secreted by activated chondrocytes, as well as synovial cells-including macrophages. Intra-articular injection of blood products-such as citrate-anticoagulated plasma (CPRP), hyperacute serum (hypACT), and extracellular vesicles (EVs) isolated from blood products-is gaining increasing importance in regenerative medicine for the treatment of OA. A co-culture system of primary OA chondrocytes and activated M1 macrophages was developed to model an OA joint in order to observe the effects of EVs in modulating the inflammatory environment. Primary OA chondrocytes were obtained from patients undergoing total knee replacement. Primary monocytes obtained from voluntary healthy donors and the monocytic cell line THP-1 were differentiated and activated into proinflammatory M1 macrophages. EVs were isolated by ultracentrifugation and characterized by nanoparticle tracking analysis and Western blot. Gene expression analysis of chondrocytes by RT-qPCR revealed increased type II collagen expression, while cytokine profiling via ELISA showed lower TNF-α and IL-1ß levels associated with EV treatment. In conclusion, the inflammation model provides an accessible tool to investigate the effects of blood products and EVs in the inflammatory context of OA.


Subject(s)
Chondrocytes/immunology , Extracellular Vesicles/immunology , Osteoarthritis/therapy , Chondrocytes/metabolism , Coculture Techniques , Female , Gene Expression Profiling , Humans , Inflammation/immunology , Inflammation/therapy , Injections, Intra-Articular , Interleukin-1beta/metabolism , Male , Models, Biological , Monocytes/immunology , Osteoarthritis/genetics , Osteoarthritis/immunology , Regenerative Medicine/methods , Tumor Necrosis Factor-alpha/metabolism
11.
Int Immunopharmacol ; 98: 107903, 2021 Sep.
Article in English | MEDLINE | ID: mdl-34192661

ABSTRACT

BACKGROUND: Long non-coding RNA (lncRNA) HOX transcript antisense RNA (HOTAIR) contributes to cartilage damages including osteoarthritis (OA). While, its role and mechanism in chondrocytes is incompletely clear. METHODS: HOTAIR, microRNA (miR)-222-3p and ADAM metalloproteinase-like domain 10 (ADAM10) expressions were detected by real-time quantitative PCR and western blotting. The interaction between miR-222-3p and HOTAIR or ADAM10 was confirmed by dual-luciferase reporter assay. Cell injury was measured by MTS method, flow cytometry, western blotting, enzyme-linked immunosorbent assay for collagen Type II, type X, sex determining region Y-box 9 (SOX9), matrix metalloproteinase (MMP)-13, interleukin (IL)-6, IL-10, and tumor necrosis factor (TNF)-α, and special assay kits for malondialdehyde (MDA), reactive oxygen species (ROS) and superoxide dismutase (SOD). RESULTS: HOTAIR was highly expressed in human OA cartilages and IL-1ß-induced OA model in immortalized chondrocytes (C-28/I2). Under IL-1ß stress, blocking HOTAIR was responsible to high mitochondrial activity and low early apoptosis rate, accompanied with increased B cell lymphoma (Bcl)-2 and LC3B-II/I proteins, boosted IL-10 and SOD productions, suppressed cleaved caspase-3 and p62 proteins, and decreased MDA and ROS levels, as well as elevated secretions of Type II collagen, Type X collagen, SOX9, MMP-13, IL-6, and TNF-α. Moreover, miR-222-3p was a target of HOTAIR, and its overexpression and knockdown could suppress and aggravate IL-1ß-induced chondrocytes injury. Furthermore, restoring ADAM10, a target gene of miR-222-3p, counteracted the protective role of miR-222-3p upregulation. CONCLUSION: HOTAIR might contribute to IL-1ß-induced chondrocytes death, inflammation, extracellular matrix degradation, and oxidative stress in OA via miR-222-3p/ADAM10 axis.


Subject(s)
ADAM10 Protein/genetics , Amyloid Precursor Protein Secretases/genetics , Membrane Proteins/genetics , MicroRNAs/metabolism , Osteoarthritis, Knee/immunology , RNA, Long Noncoding/metabolism , Apoptosis/genetics , Apoptosis/immunology , Cell Line , Chondrocytes/immunology , Chondrocytes/metabolism , Chondrocytes/pathology , Cohort Studies , Extracellular Matrix/pathology , Gene Expression Regulation/immunology , Humans , Inflammation/genetics , Inflammation/immunology , Inflammation/pathology , Interleukin-1beta/metabolism , Osteoarthritis, Knee/genetics , Osteoarthritis, Knee/pathology , Oxidative Stress/genetics , Oxidative Stress/immunology , RNA, Long Noncoding/antagonists & inhibitors , RNA, Long Noncoding/genetics , Recombinant Proteins/metabolism , Signal Transduction/genetics
12.
Front Immunol ; 12: 676173, 2021.
Article in English | MEDLINE | ID: mdl-34054865

ABSTRACT

Increased interleukin (IL)-17A has been identified in joints affected by osteoarthritis (OA), but it is unclear how IL-17A, and its family members IL-17AF and IL-17F, can contribute to human OA pathophysiology. Therefore, we aimed to evaluate the gene expression and signalling pathway activation effects of the different IL-17 family members in chondrocytes and synovial fibroblasts derived from cartilage and synovium of patients with end-stage knee OA. Immunohistochemistry staining confirmed that IL-17 receptor A (IL-17RA) and IL-17RC are expressed in end-stage OA-derived cartilage and synovium. Chondrocytes and synovial fibroblasts derived from end-stage OA patients were treated with IL-17A, IL-17AF, or IL-17F, and gene expression was assessed with bulk RNA-Seq. Hallmark pathway analysis showed that IL-17 cytokines regulated several OA pathophysiology-related pathways including immune-, angiogenesis-, and complement-pathways in both chondrocytes and synovial fibroblasts derived from end-stage OA patients. While overall IL-17A induced the strongest transcriptional response, followed by IL-17AF and IL-17F, not all genes followed this pattern. Disease-Gene Network analysis revealed that IL-17A-related changes in gene expression in these cells are associated with experimental arthritis, knee arthritis, and musculoskeletal disease gene-sets. Western blot analysis confirmed that IL-17A significantly activates p38 and p65 NF-κB. Incubation of chondrocytes and synovial fibroblasts with anti-IL-17A monoclonal antibody secukinumab significantly inhibited IL-17A-induced gene expression. In conclusion, the association of IL-17-induced transcriptional changes with arthritic gene-sets supports a role for IL-17A in OA pathophysiology. Future studies should further investigate the role of IL-17A in the OA joint to establish whether anti-IL-17 treatment could be a potential therapeutic option in OA patients with an inflammatory phenotype.


Subject(s)
Chondrocytes/immunology , Interleukin-17/physiology , Osteoarthritis, Knee/etiology , Synovial Membrane/immunology , Antibodies, Monoclonal, Humanized/pharmacology , Cells, Cultured , Chondrocytes/drug effects , Fibroblasts/drug effects , Fibroblasts/immunology , Humans , Interleukin-17/pharmacology , NF-kappa B/physiology , Osteoarthritis, Knee/immunology , Receptors, Interleukin-17/analysis , Signal Transduction/drug effects , Synovial Membrane/drug effects , Transcription, Genetic/drug effects , p38 Mitogen-Activated Protein Kinases/physiology
13.
Aging (Albany NY) ; 13(8): 11646-11664, 2021 04 20.
Article in English | MEDLINE | ID: mdl-33879632

ABSTRACT

OBJECTIVE: To investigate the heterogeneous responses of in vitro expanded chondrocytes, which were cultured in an interleukin (IL)-1ß -induced inflammatory environment. METHOD: Human articular chondrocytes were expanded, in vitro, for 13 days and treated with IL-1ß for 0, 24, and 48 h. Cells were collected and subjected to single-cell RNA sequencing. Multiple bioinformatics tools were used to determine the signatures that define chondrocyte physiology. RESULTS: Two major cell clusters with distinct expression patterns were identified at the initial phase and were with heterogeneous variation that coincides with inflammation progress. They transformed into two terminal cell clusters one of which exhibited OA-phenotype and proinflammatory characteristics through two paths, "response-to-inflammation" and "atypical response-to-inflammation", respectively. The involved cell clusters exhibited intrinsic relationship with cell types within native cartilage from OA patients. Genes controlling cell transformation to OA-phenotype were relating to the tumor necrosis factor (TNF) signaling pathway via NFKB, up-regulated KRAS signaling and the IL2/STAT5 signaling pathway and pathways relating to apoptosis and reactive oxygen species. CONCLUSION: The in vitro expanded chondrocytes under IL-1ß-induced inflammatory progression behave heterogeneously. One of the initial cell clusters could transform into a proinflammatory subpopulation through a termed response-to-inflammation path, which may serve as the core target to alleviate OA progression.


Subject(s)
Chondrocytes/pathology , Gene Expression Regulation/immunology , Osteoarthritis/immunology , Signal Transduction/genetics , Cartilage, Articular/cytology , Cells, Cultured , Child , Chondrocytes/immunology , Computational Biology , Culture Media/metabolism , Humans , Interleukin-1beta/metabolism , Osteoarthritis/genetics , Osteoarthritis/pathology , Primary Cell Culture , RNA-Seq , Signal Transduction/immunology , Single-Cell Analysis
14.
Proc Natl Acad Sci U S A ; 118(13)2021 03 30.
Article in English | MEDLINE | ID: mdl-33758095

ABSTRACT

Osteoarthritis (OA) is a painful and debilitating condition of synovial joints without any disease-modifying therapies [A. M. Valdes, T. D. Spector, Nat. Rev. Rheumatol. 7, 23-32 (2011)]. We previously identified mechanosensitive PIEZO channels, PIEZO1 and PIEZO2, both expressed in articular cartilage, to function in chondrocyte mechanotransduction in response to injury [W. Lee et al., Proc. Natl. Acad. Sci. U.S.A. 111, E5114-E5122 (2014); W. Lee, F. Guilak, W. Liedtke, Curr. Top. Membr. 79, 263-273 (2017)]. We therefore asked whether interleukin-1-mediated inflammatory signaling, as occurs in OA, influences Piezo gene expression and channel function, thus indicative of maladaptive reprogramming that can be rationally targeted. Primary porcine chondrocyte culture and human osteoarthritic cartilage tissue were studied. We found that interleukin-1α (IL-1α) up-regulated Piezo1 in porcine chondrocytes. Piezo1 expression was significantly increased in human osteoarthritic cartilage. Increased Piezo1 expression in chondrocytes resulted in a feed-forward pathomechanism whereby increased function of Piezo1 induced excess intracellular Ca2+ at baseline and in response to mechanical deformation. Elevated resting state Ca2+ in turn rarefied the F-actin cytoskeleton and amplified mechanically induced deformation microtrauma. As intracellular substrates of this OA-related inflammatory pathomechanism, in porcine articular chondrocytes exposed to IL-1α, we discovered that enhanced Piezo1 expression depended on p38 MAP-kinase and transcription factors HNF4 and ATF2/CREBP1. CREBP1 directly bound to the proximal PIEZO1 gene promoter. Taken together, these signaling and genetic reprogramming events represent a detrimental Ca2+-driven feed-forward mechanism that can be rationally targeted to stem the progression of OA.


Subject(s)
Chondrocytes/metabolism , Interleukin-1alpha/metabolism , Ion Channels/genetics , Mechanotransduction, Cellular/immunology , Osteoarthritis/immunology , Activating Transcription Factor 2/metabolism , Animals , Calcium/metabolism , Cartilage, Articular/cytology , Cartilage, Articular/immunology , Cartilage, Articular/pathology , Cells, Cultured , Chondrocytes/immunology , Female , Gene Knockdown Techniques , Humans , Ion Channels/metabolism , Mechanotransduction, Cellular/genetics , Osteoarthritis/genetics , Osteoarthritis/pathology , Primary Cell Culture , Promoter Regions, Genetic/genetics , Sus scrofa , Up-Regulation/immunology
15.
Int Immunopharmacol ; 94: 107489, 2021 May.
Article in English | MEDLINE | ID: mdl-33774357

ABSTRACT

The objective of this study is to investigate the role of IL-38 in osteoarthritis (OA). IL-38 levels in serum and synovial fluid (SF) of patients with OA were examined to identify the correlation between IL-38 expression and OA activity and to determine its anti-inflammatory effects in IL-1ß-induced chondrocytes. A total of 75 patients with OA who underwent joint replacement surgery and 25 age- and sex-matched healthy volunteers were recruited. The levels of IL-38 in serum and SF are shown to be significant elevated in OA patients compared with that of healthy controls. Serum and SF IL-38 levels of OA patients are positively correlated with Kellgren-Lawrence (K-L) grades 2 to 3, as well as with pro-inflammatory cytokines IL-6, IL-23, and TNF-α, but are negatively correlated with the anti-inflammatory cytokine IL-10 in K-L grades 3 to 4. Furthermore, overexpression of IL-38 in vitro is shown to attenuate the expression of pro-inflammatory cytokines such as COX-2, IL-6, IL-8, IL-36Ra, IL-36α/ß/γ, iNOS, and TNF-α, as well as matrix degrading enzymes such as MMP3, MMP13, and ADAMTS5, and apoptosis-related indicators Bax/Bcl-2, cleaved caspase 3/pro-caspase 3, and cleaved caspase 9/pro-caspase 9. IL-38 overexpression also reduces expression of the signaling proteins p-p38, p-p65, p-JNK, and RhoA significantly. Taken together, our results show that expression of IL-38 is increased in OA tissues and OA rat chondrocytes, and is positively correlated with early disease activity. This increased IL-38 expression lead to the inactivation of MAPK, NF-κB, JNK, and RhoA signaling pathways, which might have impletion on OA chondrocytes apoptosis, degradation and inflammatory effect. Thus, IL-38 probably serves as a novel therapeutic target for the treatment of OA.


Subject(s)
Chondrocytes/immunology , Cytokines/immunology , Osteoarthritis/immunology , Aged , Animals , Cartilage, Articular/cytology , Cytokines/blood , Cytokines/genetics , Female , Hip Joint , Humans , Knee Joint , Male , Middle Aged , Mitogen-Activated Protein Kinases/immunology , NF-kappa B/immunology , Osteoarthritis/blood , Rats, Sprague-Dawley , Signal Transduction , rhoA GTP-Binding Protein/immunology
16.
Int Immunopharmacol ; 95: 107495, 2021 Jun.
Article in English | MEDLINE | ID: mdl-33684877

ABSTRACT

BACKGROUND: Osteoarthritis (OA) is characterized by chondrocyte injury and dysfunction, such as excessive apoptosis, inflammatory response and extracellular matrix (ECM) degradation. Circular RNA (circRNA) deregulation is reported to be involved in OA. Our study aimed to explore the role of circ_0134111 in OA. METHODS: Human chondrocytes were treated with interleukin-1ß (IL-1ß) to mimic OA cell model. The expression of circ_0134111, miR-515-5p and suppressor of cytokine signaling 1 (SOCS1) mRNA was measured by real-time quantitative polymerase chain reaction (RT-qPCR), and the protein levels of SOCS1 and apoptosis-/inflammation-/ECM-related markers were determined by western blot. Cell proliferation and cell apoptosis were assessed using cell counting kit-8 (CCK-8) and flow cytometry assay, respectively. For mechanism analysis, the predicted interaction between miR-515-5p and circ_0134111 or SOCS1 was verified by dual-luciferase reporter assay, pull-down assay and RNA immunoprecipitation (RIP) assay. Rescue experiments were performed to explore the interplay between miR-515-5p and circ_0134111 or SOCS1. RESULTS: Circ_0134111 was overexpressed in OA cartilage tissues and IL-1ß-induced chondrocytes. IL-1ß-induced chondrocyte apoptosis, inflammatory responses and ECM degradation were alleviated by circ_0134111 knockdown or miR-515-5p restoration. Circ_0134111 acted as miR-515-5p sponge to regulate miR-515-5p expression, and miR-515-5p deficiency reversed the effects of circ_0134111 knockdown in IL-1ß-induced chondrocytes. MiR-515-5p directly bound to SOCS1, and circ_0134111 decoyed miR-515-5p to increase SOCS1 level. MiR-515-5p restoration alleviated IL-1ß-induced chondrocyte apoptosis, inflammatory responses and ECM degradation, While SOCS1 overexpression partly abolished these effects. CONCLUSION: Circ_0134111 knockdown alleviated apoptosis, inflammatory responses and ECM degradation in OA cell model by mediating the miR-515-5p-SOCS1 network, hinting that circ_0134111 was involved in OA progression.


Subject(s)
Interleukin-1beta/immunology , MicroRNAs/immunology , Osteoarthritis/immunology , RNA, Circular , Suppressor of Cytokine Signaling 1 Protein/immunology , Apoptosis/genetics , Apoptosis/immunology , Cartilage/immunology , Chondrocytes/immunology , Extracellular Matrix/immunology , Female , Humans , Male , Middle Aged
17.
J Cell Physiol ; 236(6): 4244-4257, 2021 06.
Article in English | MEDLINE | ID: mdl-33605451

ABSTRACT

The purpose of this study was to investigate the feasibility of adipose-derived stem cells (ADSCs) as the seed cells of cartilage tissue engineering. ADSCs were isolated from adipose tissue that was harvested under sterile conditions from the inguen fold of porcines and cultured in vitro. Acellular cartilage extracellular matrix (ACECM) scaffolds of pigs were then constructed. Moreover, inflammatory cells, as well as cellular and humoral immune responses, were detected using hematoxylin and eosin staining staining, immunohistochemical staining, and western blot analysis. The results showed that the cartilage complex constructed by ADSCs and ACECM through tissue engineering successfully repaired the cartilage defect of the pig knee joint. The in vivo repair experiment showed no significant difference between chondrocytes, ADSCs, and induced ADSCs, indicating that ADSCs do not require in vitro induction and have the potential for chondrogenic differentiation in the environment around the knee joint. In addition, pig-derived acellular cartilage scaffolds possess no obvious immune inflammatory response when used in xenotransplantation. ADSCs may serve as viable seed cells for cartilage tissue engineering.


Subject(s)
Cartilage Diseases/surgery , Cartilage, Articular/surgery , Chondrocytes/transplantation , Chondrogenesis , Mesenchymal Stem Cell Transplantation , Mesenchymal Stem Cells , Regeneration , Tissue Scaffolds , Adipose Tissue/cytology , Animals , Cartilage Diseases/immunology , Cartilage Diseases/metabolism , Cartilage Diseases/pathology , Cartilage, Articular/immunology , Cartilage, Articular/metabolism , Cartilage, Articular/pathology , Cells, Cultured , Chondrocytes/immunology , Chondrocytes/metabolism , Disease Models, Animal , Immunity, Humoral , Mesenchymal Stem Cells/immunology , Mesenchymal Stem Cells/metabolism , Rabbits , Swine , Swine, Miniature , Tissue Engineering
18.
FEBS Open Bio ; 11(2): 435-445, 2021 02.
Article in English | MEDLINE | ID: mdl-33326693

ABSTRACT

Osteoarthritis (OA) is an age-related, chronic degenerative disease. With the increasing median age of the population, this disease has become an important public health problem. New, disease-modifying therapies are needed. A potential novel molecular target is phospholipase Cγ1 (PLCγ1), a critical enzyme with important functions including calcium signaling regulation and cell proliferation. In rat chondrocytes treated with IL-1ß (20 ng·mL-1 for 36 h), inhibition of PLCγ1 with U73122 (2 µm for 12 h) increased levels and expression of the cartilage matrix components Collagen2 and Aggrecan. This beneficial effect of PLCγ1 inhibition was counteracted by increased chondrocyte apoptosis and necroptosis, increased cell death, and increase levels of ROS, all potentially negative for OA. Combined treatment of IL-1ß + U73122-treated chondrocytes with inhibitors of apoptosis (Z-VAD, 10 µm) and necroptosis (Nec-1, 30 µm) enhanced the increases in levels and expression of Collagen2 and Aggrecan, and prevented the increases in cell death and ROS levels. These results suggest that PLCγ1 inhibition may be a viable approach for an OA therapy, if combined with targeted inhibition of chondrocyte apoptosis and necroptosis.


Subject(s)
Interleukin-1beta/immunology , Osteoarthritis/drug therapy , Phospholipase C gamma/antagonists & inhibitors , Animals , Animals, Newborn , Apoptosis/drug effects , Apoptosis/immunology , Cartilage, Articular/cytology , Cartilage, Articular/drug effects , Cartilage, Articular/immunology , Cartilage, Articular/pathology , Chondrocytes/cytology , Chondrocytes/drug effects , Chondrocytes/immunology , Chondrocytes/pathology , Disease Models, Animal , Drug Therapy, Combination/methods , Estrenes/pharmacology , Estrenes/therapeutic use , Extracellular Matrix/drug effects , Extracellular Matrix/metabolism , Humans , Imidazoles/pharmacology , Imidazoles/therapeutic use , Indoles/pharmacology , Indoles/therapeutic use , Interleukin-1beta/metabolism , Necroptosis/drug effects , Necroptosis/immunology , Oligopeptides/pharmacology , Oligopeptides/therapeutic use , Osteoarthritis/immunology , Osteoarthritis/pathology , Phospholipase C gamma/metabolism , Primary Cell Culture , Pyrrolidinones/pharmacology , Pyrrolidinones/therapeutic use , Rats , Recombinant Proteins/immunology , Recombinant Proteins/metabolism
19.
Biol Pharm Bull ; 44(2): 232-237, 2021 Feb 01.
Article in English | MEDLINE | ID: mdl-33268698

ABSTRACT

Osteoarthritis (OA) is a chronic joint disease with high prevalence. However, effective treatment options for OA are still lacking. It was previously reported that LINC00473 was upregulated in patients with OA and upregulated LINC00473 might be associated with the progression of OA; however, the role of LINC00473 in OA remains to be investigated. CHON-001 human chondrocyte cells stimulated with 10 ng/mL interleukin (IL)-1ß were utilized to mimic OA in vitro. Protein expression, cell apoptosis and cell proliferation of CHON-001 cells were investigated by Western blot, Annexin V and propidium iodide (PI) double staining, cell counting-8 kit assay and immunofluorescence staining respectively. The result indicated IL-1ß triggered viability decrease and apoptosis in CHON-001 cells, which was alleviated by LINC00473 knockdown. Meanwhile, IL-1ß-induced upregulation of cleaved caspase 3 and Bax were ameliorated by LINC00473 knockdown. Likewise, IL-1ß-induced downregulation of X-linked inhibitor of apoptosis protein was alleviated by LINC00473 knockdown. In addition, LINC00473 knockdown protected CHON-001 cells against IL-1ß by inhibiting the methylation of LIM mineralization protein (LMP)-1 gene. Moreover, c-Jun N-terminal kinase (JNK)/nuclear factor-kappaB (NF-κB) signaling pathway was proved to be involved in the cell protective effect of LINC00473 knockdown in IL-1ß treated CHON-001 cells. Taken together, LINC00473 knockdown defended CHON-001 cells from IL-1ß induced cell injury via inhibition of the methylation of LMP-1. Thus, LINC00473 might possibly act as a novel therapeutic target for OA.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Cartilage, Articular/drug effects , Interleukin-1beta/metabolism , Osteoarthritis/drug therapy , RNA, Long Noncoding/antagonists & inhibitors , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/metabolism , Anti-Inflammatory Agents/therapeutic use , Cartilage, Articular/immunology , Cartilage, Articular/pathology , Cell Line , Chondrocytes/drug effects , Chondrocytes/immunology , Chondrocytes/pathology , Cytoskeletal Proteins/genetics , Cytoskeletal Proteins/metabolism , Gene Knockdown Techniques , Humans , LIM Domain Proteins/genetics , LIM Domain Proteins/metabolism , MAP Kinase Signaling System/drug effects , MAP Kinase Signaling System/genetics , MAP Kinase Signaling System/immunology , Methylation/drug effects , Molecular Targeted Therapy/methods , Osteoarthritis/genetics , Osteoarthritis/immunology , Osteoarthritis/pathology , RNA, Long Noncoding/genetics , RNA, Long Noncoding/metabolism , Up-Regulation
20.
J Cell Physiol ; 236(6): 4369-4386, 2021 06.
Article in English | MEDLINE | ID: mdl-33164235

ABSTRACT

The inflammatory environment and excessive chondrocyte apoptosis have been demonstrated to play crucial roles in the onset of osteoarthritis (OA). Hydrogen sulfide (H2 S), a gaseous signalling molecule, exerts an inhibitory effect on inflammation and apoptosis in several degenerative diseases. However, the protective effect of H2 S against OA has not been fully clarified, and its underlying mechanism should be examined further. In the current study, the role of endogenous H2 S in the pathogenesis of OA and its protective effects on interleukin (IL)-1ß-induced chondrocytes were identified. Our data revealed decreased H2 S expression in both human degenerative OA cartilage tissue and IL-1ß-induced chondrocytes. Pretreatment with the H2 S donor sodium hydrosulfide (NaHS) dramatically attenuated IL-1ß-induced overproduction of inflammatory cytokines and improved the balance between anabolic and catabolic chondrocyte capacities, and these effects were dependent on PI3K/AKT pathway-mediated inhibition of nuclear factor kappa B (NF-κB). Moreover, mitochondrial dysfunction-related apoptosis was significantly reversed by NaHS in IL-1ß-stimulated chondrocytes. Mechanistically, NaHS partially suppressed IL-1ß-induced phosphorylation of the mitogen-activated protein kinase (MAPK) cascades. Furthermore, in the destabilization of the medial meniscus mouse model, OA progression was ameliorated by NaHS administration. Taken together, these results suggest that H2 S may antagonize IL-1ß-induced inflammation and mitochondrial dysfunction-related apoptosis via selective suppression of the PI3K/Akt/NF-κB and MAPK signalling pathways, respectively, in chondrocytes and may be a potential therapeutic agent for the treatment of OA.


Subject(s)
Antirheumatic Agents/pharmacology , Apoptosis/drug effects , Chondrocytes/drug effects , Hydrogen Sulfide/pharmacology , Inflammation Mediators/metabolism , Interleukin-1beta/toxicity , Joints/drug effects , Mitochondria/drug effects , Osteoarthritis/drug therapy , Sulfides/pharmacology , Aged , Animals , Antirheumatic Agents/metabolism , Case-Control Studies , Cells, Cultured , Chondrocytes/immunology , Chondrocytes/metabolism , Chondrocytes/pathology , Disease Models, Animal , Female , Humans , Hydrogen Sulfide/metabolism , Joints/immunology , Joints/metabolism , Joints/pathology , Male , Mice, Inbred C57BL , Middle Aged , Mitochondria/immunology , Mitochondria/metabolism , Mitochondria/pathology , Mitogen-Activated Protein Kinases/metabolism , NF-kappa B/metabolism , Osteoarthritis/immunology , Osteoarthritis/metabolism , Osteoarthritis/pathology , Phosphatidylinositol 3-Kinase/metabolism , Phosphorylation , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction , Sulfides/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...