ABSTRACT
OBJECTIVES: This study was directed towards exploring the impacts of lncRNA HOXA11-AS-mediated microRNA (miR)-506-3p on chondrocytes proliferation and apoptosis in osteoarthritis (OA). METHODS: The articular cartilages were provided by OA patients who received total knee arthroplasty, and Human Chondrocyte (HC)-OA (HCOA) was also attained. The miR-506-3p and HOXA11-AS expressions in articular cartilages from OA patients and HCOA cells were analyzed via qPCR. After gain- and loss-of-function assays in HCOA cells, MTT assay and flow cytometry (FC) were used for assessing cell viability and apoptosis, accordingly. The levels of PIK3CA, AKT, and mTOR as well as AKT and mTOR phosphorylation levels assessed using western blotting (WB). The targeting correlation of HOXA11-AS and miR-506-3p as well as miR-506-3p and PIK3CA was assessed through Dual-Luciferase Reporter gene Assay (DLRA). RESULT: The articular cartilages from OA patients and Human Chondrocyte (HC)-OA (HCOA) cells showed increased HOXA11-AS and decreased miR-506-3p. Mechanistically, HOXA11-AS was capable of binding to miR-506-3p to increase PIK3CA, the target gene of miR-506-3p. miR-506-3p suppression facilitated HCOA cell proliferation and reduced their apoptosis, which was nullified by further silencing HOXA11-AS or silencing PIK3CA. The down-regulation of HOXA11-AS disrupted the PI3K/AKT/mTOR pathway, which was counteracted by further miR-506-3p inhibition. CONCLUSION: The silencing of HOXA11-AS might block the PI3K/AKT/mTOR pathway through miR-506-3p up-regulation, thereby restricting HCOA cell proliferation and provoking apoptosis.
Subject(s)
Apoptosis , Cell Proliferation , Chondrocytes , Down-Regulation , MicroRNAs , RNA, Long Noncoding , Humans , MicroRNAs/genetics , MicroRNAs/metabolism , Chondrocytes/metabolism , Apoptosis/genetics , Cell Proliferation/genetics , RNA, Long Noncoding/genetics , RNA, Long Noncoding/metabolism , Osteoarthritis/genetics , Osteoarthritis/metabolism , Osteoarthritis/pathology , TOR Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Cartilage, Articular/metabolism , Middle Aged , Male , Female , Cells, CulturedABSTRACT
Osteoarthritis is a multifactorial joint disease characterized by degeneration, and aging stands as a significant risk factor. Autophagy, a crucial cellular homeostasis mechanism, is influenced by aging and closely linked to cartilage health. This correlation between autophagy, cell death, and OA underscores its relevance in disease progression. MicroRNAs have emerged as autophagy regulators, with miRNA-based interventions showing promise in preclinical models. Remarkably, the ethanolic extract of propolis exhibits positive effects on autophagy-related proteins and healthy cartilage markers in an in vitro osteoarthritis model. The aim of this brief report was to evaluate through in silico analysis and postulate five microRNAs that could regulate autophagy proteins (AKT1, ATG5, and LC3) and assess whether the ethanolic extract of propolis could regulate the expression of these microRNAs. Among the examined miRNAs (miR-19a, miR-125b, miR-181a, miR-185, and miR-335), the ethanolic extract of propolis induced significant changes in four of them. Specifically, miR-125b responded to EEP by counteracting IL-1ß-induced effects, while miR-181a, miR-185, and miR-335 exhibited distinct patterns of expression under EEP treatment. These findings unveil a potential link between miRNAs, EEP, and autophagy modulation in OA, offering promising therapeutic insights. Nevertheless, further validation and clinical translation are warranted to substantiate these promising observations.
Subject(s)
MicroRNAs , Osteoarthritis , Propolis , Humans , MicroRNAs/metabolism , Propolis/pharmacology , Propolis/metabolism , Chondrocytes/metabolism , Osteoarthritis/drug therapy , Osteoarthritis/genetics , Osteoarthritis/metabolism , Ethanol/pharmacology , AutophagyABSTRACT
OBJECTIVE: Osteoarthritis is a condition characterized by articular cartilage degradation. The increased expression of ß1,4-Galactosyltransferase-I (ß1,4-GalT-I) in the articular cartilage of osteoarthritis patients was related to an inflammatory response. The aim of this study was to elucidate the role of ß1,4-GalT-I in osteoarthritis. This study aimed to determine the function of 1,4-GalT-I in osteoarthritis. METHODS: The osteoarthritis mouse model with the destabilization of the medial meniscus was established by microsurgical technique. Pathological changes in articular cartilage were observed by hematoxylin and eosin staining and safranin O-fast green staining. Quantitative real-time polymerase chain reaction, western blot, and enzyme-linked immunosorbent assays were used to observe mRNA and protein expression, respectively. RNA interactions were verified by a luciferase reporter assay. SA-ß-Gal staining was used to assess chondrocyte senescence. Immunofluorescence staining was conducted to observe the localization of Nuclear Factor-kappaB (NF-κB). RESULTS: ß1,4-GalT-I and microRNA-15a (miR-15a) show high and low expression in the articular cartilage of osteoarthritis, respectively. MiR-15a inhibits the mRNA translation of ß1,4-GalT-I. ß1,4-GalT-I promotes extracellular matrix degradation, senescence, and NF-κB activation in IL-1ß-stimulated chondrocytes, which can be reversed by overexpression of miR-15a. Intra-articular injection of microRNA-15a ameliorates cartilage degeneration by inhibiting ß1,4-GalT-I and phosphorylation of NF-κB in vivo. CONCLUSION: The authors clarified that the miR-15a/ß1,4-GalT-I axis inhibits the phosphorylation of NF-κB thereby inhibiting extracellular matrix degradation and senescence in chondrocytes to alleviate cartilage degeneration in osteoarthritis. MiR-15a and ß1,4-GalT-I may serve as potentially effective targets for the future treatment of osteoarthritis.
Subject(s)
Cartilage, Articular , MicroRNAs , Osteoarthritis , Animals , Mice , Cartilage, Articular/pathology , Chondrocytes/pathology , Interleukin-1beta , MicroRNAs/genetics , NF-kappa B/metabolism , Osteoarthritis/genetics , Signal TransductionABSTRACT
The purpose of this study is to evaluate the effects of photobiomodulation (PBM) therapy in chondrocyte response by in vitro experiments and cartilage repair using an experimental model of osteoarthritis (OA) in the knee of rats. The in vitro experiment was performed with chondrocyte cells, and they were divided into two groups: non-irradiated and irradiated with PBM (808 nm; 0.8 J or 1.4 J). Then, cell proliferation was evaluated after 1, 3, and 5 days. The experimental model of osteoarthritis (OA) was performed in the knee of 64 Wistar rats, and they were assorted into control group (CG), PBM (808 nm; 1.4 J). The results of in vitro showed that PBM 1.4 J increased cell proliferation, on days 1 and 5. However, after 3 days was demonstrated a significant increase in cell proliferation in PBM 0.8 J. The in vivo experiment results demonstrated, on histological analysis, that PBM presented less intense signs of tissue degradation with an initial surface discontinuity at the superficial zone and disorganization of the chondrocytes in the cartilage region when compared to CG, after 4 and 8 weeks. These findings were confirmed by immunohistochemistry and qRT-PCR analysis which showed that PBM increased IL-4, IL-10, COL-2, Aggrecan, and TGF-ß which are anabolic factors and acts on extracellular matrix. Also, PBM reduces the IL1-ß, an inflammatory marker that operates as a catabolic factor on articular cartilage. In conclusion, these results suggest that PBM may have led to a return to tissue homeostasis, promoting chondroprotective effects and stimulating the components of the articular tissue.
Subject(s)
Cartilage, Articular , Low-Level Light Therapy , Osteoarthritis, Knee , Osteoarthritis , Animals , Cartilage, Articular/pathology , Chondrocytes/pathology , Disease Models, Animal , Osteoarthritis/genetics , Osteoarthritis/metabolism , Osteoarthritis/radiotherapy , Osteoarthritis, Knee/genetics , Osteoarthritis, Knee/radiotherapy , Rats , Rats, WistarABSTRACT
Articular chondral lesions, caused either by trauma or chronic cartilage diseases such as osteoarthritis, present very low ability to self-regenerate. Thus, their current management is basically symptomatic, progressing very often to invasive procedures or even arthroplasties. The use of amniotic fluid stem cells (AFSCs), due to their multipotentiality and plasticity, associated with scaffolds, is a promising alternative for the reconstruction of articular cartilage. Therefore, this study aimed to investigate the chondrogenic potential of AFSCs in a micromass system (high-density cell culture) under insulin-like growth factor 1 (IGF-1) stimuli, as well as to look at their potential to differentiate directly when cultured in a porous chitosan-xanthan (CX) scaffold. The experiments were performed with a CD117 positive cell population, with expression of markers (CD117, SSEA-4, Oct-4 and NANOG), selected from AFSCs, after immunomagnetic separation. The cells were cultured in both a micromass system and directly in the scaffold, in the presence of IGF-1. Differentiation to chondrocytes was confirmed by histology and by using immunohistochemistry. The construct cell-scaffold was also analyzed by scanning electron microscopy (SEM). The results demonstrated the chondrogenic potential of AFSCs cultivated directly in CX scaffolds and also in the micromass system. Such findings support and stimulate future studies using these constructs in osteoarthritic animal models.
Subject(s)
Adult Stem Cells/cytology , Cartilage, Articular/drug effects , Chondrogenesis/genetics , Osteoarthritis/genetics , Tissue Scaffolds/chemistry , Adult Stem Cells/transplantation , Amniotic Fluid/cytology , Cartilage, Articular/growth & development , Cartilage, Articular/ultrastructure , Cell Culture Techniques , Cell Differentiation/drug effects , Chitosan/pharmacology , Gene Expression Regulation, Developmental/drug effects , Humans , Insulin-Like Growth Factor I/pharmacology , Microscopy, Electron, Scanning , Osteoarthritis/pathology , Osteoarthritis/therapy , Polysaccharides, Bacterial/pharmacology , Proto-Oncogene Proteins c-kit/genetics , Tissue Engineering/methodsABSTRACT
Autophagy is an intracellular mechanism that maintains cellular homeostasis in different tissues. This process declines in cartilage due to aging, which is correlated with osteoarthritis (OA), a multifactorial and degenerative joint disease. Several studies show that microRNAs regulate different steps of autophagy but only a few of them participate in OA. Therefore, epigenetic modifications could represent a therapeutic opportunity during the development of OA. Besides, polyphenols are bioactive components with great potential to counteract diseases, which could reverse altered epigenetic regulation and modify autophagy in cartilage. This review aims to analyze epigenetic mechanisms that are currently associated with autophagy in OA, and to evaluate whether polyphenols are used to reverse the epigenetic alterations generated by aging in the autophagy pathway.
Subject(s)
Autophagy/genetics , Epigenesis, Genetic , Osteoarthritis/drug therapy , Osteoarthritis/genetics , Polyphenols/therapeutic use , Animals , Autophagy/drug effects , Epigenesis, Genetic/drug effects , Humans , Models, Biological , Polyphenols/pharmacologyABSTRACT
Pararamosis is a disease that occurs due to contact with the hairs of the larval stage of the Brazilian moth Premolis semirufa. Envenomation induces osteoarticular alterations with cartilage impairment that resembles joint synovitis. Thus, the toxic venom present in the caterpillar hairs interferes with the phenotype of the cells present in the joints, resulting in inflammation and promoting tissue injury. Therefore, to address the inflammatory mechanisms triggered by envenomation, we studied the effects of P. semirufa hair extract on human chondrocytes. We have selected for the investigation, cytokines, chemokines, matrix metalloproteinases (MMPs), complement components, eicosanoids, and extracellular matrix (ECM) components related to OA and RA. In addition, for measuring protein-coding mRNAs of some molecules associated with osteoarthritis (OA) and rheumatoid arthritis (RA), reverse transcription (RT) was performed followed by quantitative real-time PCR (RT-qPCR) and we performed the RNA-sequencing (RNA-seq) analysis of the chondrocytes transcriptome. In the supernatant of cell cultures treated with the extract, we observed increased IL-6, IL-8, MCP-1, prostaglandin E2, metalloproteinases (MMP-1, MMP-2, MMP-3 and MMP-13), and complement system components (C3, C4, and C5). We noticed a significant decrease in both aggrecan and type II collagen and an increase in HMGB1 protein in chondrocytes after extract treatment. RNA-seq analysis of the chondrocyte transcriptome allowed us to identify important pathways related to the inflammatory process of the disease, such as the inflammatory response, chemotaxis of immune cells and extracellular matrix (ECM) remodeling. Thus, these results suggest that components of Premolis semirufa hair have strong inflammatory potential and are able to induce cartilage degradation and ECM remodeling, promoting a disease with an osteoarthritis signature. Modulation of the signaling pathways that were identified as being involved in this pathology may be a promising approach to develop new therapeutic strategies for the control of pararamosis and other inflammatory joint diseases.
Subject(s)
Cartilage/pathology , Chondrocytes/physiology , Inflammation/immunology , Joint Diseases/immunology , Osteoarthritis/genetics , Animals , Arthropod Venoms/metabolism , Cells, Cultured , Cytokines/metabolism , Extracellular Matrix/metabolism , Humans , Inflammation Mediators/metabolism , Joint Diseases/chemically induced , Moths/metabolism , Rainforest , Signal TransductionABSTRACT
Articular cartilage is an avascular tissue with a structure that allows it to support and cushion the overload of the surfaces in contact. It maintains its metabolic functions due to the contribution of different signaling pathways. However, several factors play a role in its deterioration, allowing to the development of osteoarthritis (OA), and one of the major factors is genetic. Our goal was to identify gene-gene interactions (epistasis) between five signaling pathways involved in the articular cartilage metabolism as possible indicators of OA risk. We applied the Multifactor-Dimensionality Reduction (MDR) method to identify and characterize the epistasis between 115 SNPs located in 73 genes related to HIF-1α, Wnt/ß-catenin, cartilage extracellular matrix metabolism, oxidative stress, and uric acid transporters. Ninety three patients diagnosed with primary knee OA and 150 healthy controls were included in the study. Genotyping was performed with the OpenArray system, the statistical analysis was carried out with the STATA software v14, and epistasis was analyzed with the MDR software v3.0.2. The MDR analysis revealed that the best interaction model was between polymorphisms rs17786744 of the STC1 gene and rs2615977 of the COL11A1 gene, with an entropy value of 4.44%, CVC 8/10, OR 5.60, 95% CI 3.27-9.59, p < 0.0001. Under this interaction model, we identified high and low risk genotypes involved in OA development. Our results suggest complex interactions between STC1 and COL11A1 genes that might have an impact on genetic susceptibility to develop OA. Further studies are required to confirm it.
Subject(s)
Collagen Type XI/genetics , Glycoproteins/genetics , Osteoarthritis, Knee/genetics , Adult , Alleles , Case-Control Studies , Epistasis, Genetic/genetics , Female , Gene Frequency , Genetic Predisposition to Disease/genetics , Genotype , Humans , Male , Middle Aged , Multifactor Dimensionality Reduction/methods , Osteoarthritis/genetics , Polymorphism, Single Nucleotide/genetics , Risk Factors , SoftwareABSTRACT
BACKGROUND: Osteoarthritis (OA) is one of the most common rheumatic diseases of which clinical symptoms includes swelling, synovitis and inflammatory pain, affect patients' daily life. It was reported that non-coding RNAs play vital roles in OA. However, the regulation mechanism of ncRNA in OA pathogenesis has not been fully elucidated. METHODS: The expression of SNHG7, miR-34a-5p and SYVN1 was detected using qRT-PCR in tissues, serum and cells. The protein expression of SYVN1, PCNA, cleavage-caspase 3, beclin1 and LC3 were measured using western blot. The RNA immunoprecipitation (RIP), RNA pulldown, and luciferase reporter assays were used to verify the relationship between SNHG7, miR-34a-5p and SYVN1. The MTT and flow cytometry assay was performed to detected cell proliferation and cell apoptosis respectively. RESULTS: In this study, SNHG7 and SYVN1 expression were down-regulated, but miR-34a-5p was up-regulated in OA tissues and IL-1ß treated cells compared with normal tissues and chondrocyte. Functional investigation revealed that up-regulated SNHG7 or down-regulated miR-34a-5p could promote cell proliferation and inhibit cell apoptosis and autophagy in OA cells. More than that, RIP, pulldown and luciferase reporter assay was applied to determine that miR-34a-5p was a target miRNA of SNHG7 and SYVN1 was a target mRNA of miR-34-5p. Rescue experiments showed that overexpression of miR-34a reversed high expression of SNHG7-mediated suppression of apoptosis and autophagy as well as promotion of proliferation, while its knockdown inhibited cell apoptosis and autophagy and promoted cell proliferation which could be impaired by silencing SYVN1. In addition, SNHG7 regulated SYVN1 through sponging miR-34a-5p. CONCLUSION: SNHG7 sponged miR-34a-5p to affect cell proliferation, apoptosis and autophagy through targeting SYVN1 which provides a novel sight into the pathogenesis of OA.
Subject(s)
Apoptosis/physiology , Autophagy/physiology , MicroRNAs/metabolism , Osteoarthritis/metabolism , RNA, Long Noncoding/metabolism , Ubiquitin-Protein Ligases/metabolism , Apoptosis/genetics , Autophagy/genetics , Blotting, Western , Cell Proliferation , Down-Regulation , Enzyme-Linked Immunosorbent Assay , Humans , MicroRNAs/genetics , Osteoarthritis/genetics , RNA, Long Noncoding/genetics , Real-Time Polymerase Chain Reaction , Ubiquitin-Protein Ligases/genetics , Up-RegulationABSTRACT
INTRODUCTION: The study of our genome has played an important role in the field of personalized medicine and clinical practice becoming a useful tool to assist the medical community in the early diagnosis and treatment of countless diseases; osteoarthritis (OA) is a complex chronic degenerative joint disease, despite the high prevalence of this disease and its great impact on public health, little is currently known about its etiology and risk of progression. The purpose of this review is to show the advances in genetics in the study of osteoartrosis. METHODS: The present is a review of the literature of the different aspects in which genetics has developed in the study of osteoartrosis, its scopes and its possible impact on prevention and treatment. CONCLUSION: The identification of a high number of candidate genes confirms the complex nature of the disease, it seems clear that the degree of expression of different genes is altered between an arthrosic patient and a healthy one. A deeper understanding of the link between the entire genome sequence and the association with well-characterized OA phenotypes will enable the development of biomarkers, report the risk of disease progression and allow better guidance of treatments.
INTRODUCCIÓN: El estudio de nuestro genoma ha jugado un papel importante en el campo de la medicina personalizada y la práctica clínica, lo que la convierte en una herramienta útil para ayudar a la comunidad médica en el diagnóstico y tratamiento temprano de innumerables enfermedades. La osteoartrosis (OA) es una enfermedad articular degenerativa crónica compleja; a pesar de su alta prevalencia y gran impacto en la salud pública, actualmente se sabe poco sobre su etiología y riesgo de progresión. El objeto de la presente revisión es mostrar los avances de la genética en el estudio de la osteoartrosis. MÉTODOS: Revisión de la literatura sobre los diferentes aspectos en donde la genética se ha desarrollado en el estudio de la osteoartrosis, sus alcances y sus posibles repercusiones en la prevención y tratamiento. CONCLUSIÓN: La identificación de un elevado número de genes candidatos nos confirma la compleja naturaleza de la enfermedad, parece claro que el grado de expresión de diferentes genes está alterado entre un paciente artrósico y uno sano. Una comprensión más profunda del vínculo entre la secuencia de todo el genoma y la asociación con fenotipos bien caracterizados de la OA, permitirá el desarrollo de biomarcadores, informar el riesgo de progresión de la enfermedad y permitir una mejor orientación de los tratamientos.
Subject(s)
Osteoarthritis , Biomarkers , Disease Progression , Humans , Osteoarthritis/genetics , PhenotypeABSTRACT
BACKGROUND: Osteoarthritis (OA) is one of the most common rheumatic diseases of which clinical symptoms includes swelling, synovitis and inflammatory pain, affect patients' daily life. It was reported that non-coding RNAs play vital roles in OA. However, the regulation mechanism of ncRNA in OA pathogenesis has not been fully elucidated. METHODS: The expression of SNHG7, miR-34a-5p and SYVN1 was detected using qRT-PCR in tissues, serum and cells. The protein expression of SYVN1, PCNA, cleavage-caspase 3, beclinl and LC3 were measured using western blot. The RNA immunoprecipitation (RIP), RNA pulldown, and luciferase reporter assays were used to verify the relationship between SNHG7, miR-34a-5p and SYVN1. The MTT and flow cytometry assay was performed to detected cell proliferation and cell apoptosis respectively. RESULTS: In this study, SNHG7 and SYVN1 expression were down-regulated, but miR-34a-5p was up-regulated in OA tissues and IL-1P treated cells compared with normal tissues and chondrocyte. Functional investigation revealed that up-regulated SNHG7 or down-regulated miR-34a-5p could promote cell proliferation and inhibit cell apoptosis and autophagy in OA cells. More than that, RIP, pulldown and luciferase reporter assay was applied to determine that miR-34a-5p was a target miRNA of SNHG7 and SYVN1 was a target mRNA of miR-34-5p. Rescue experiments showed that overexpression of miR-34a reversed high expression of SNHG7-mediated suppression of apoptosis and autophagy as well as promotion of proliferation, while its knockdown inhibited cell apoptosis and autophagy and promoted cell proliferation which could be impaired by silencing SYVN1. In addition, SNHG7 regulated SYVN1 through sponging miR-34a-5p. CONCLUSION: SNHG7 sponged miR-34a-5p to affect cell proliferation, apoptosis and autophagy through targeting SYVN1 which provides a novel sight into the pathogenesis of OA.
Subject(s)
Humans , Osteoarthritis/metabolism , Autophagy/physiology , Apoptosis/physiology , MicroRNAs/metabolism , Ubiquitin-Protein Ligases/metabolism , RNA, Long Noncoding/metabolism , Osteoarthritis/genetics , Autophagy/genetics , Enzyme-Linked Immunosorbent Assay , Down-Regulation , Up-Regulation , Blotting, Western , Apoptosis/genetics , MicroRNAs/genetics , Ubiquitin-Protein Ligases/genetics , Cell Proliferation , Real-Time Polymerase Chain Reaction , RNA, Long Noncoding/geneticsABSTRACT
BACKGROUND: Osteoarthritis (OA) is a major musculoskeletal disease with high prevalence in the elderly. The study of genetic polymorphisms of inflammatory mediators involved in OA may contribute to the elucidation of the complex pathophysiology of this disease and identification of susceptibility individuals. AIM: This study aimed to evaluate the association between polymorphism at tumor necrosis factor alpha gene (SNP - 308 G/A TNFA) with presence, severity and functional status of osteoarthritis in elderly. METHODS: This study was characterized as case-control and encompassed 257 physically independent elderly (Mean Age: 68.55 ± 5.2; Minimum age: 60 and Maximum age: 82) were recruited. After this selection, the groups were divided in: 92 elderly individuals with osteoarthritis (case group) and 165 without the disease (control group). METHODS: The individuals were genotyped by the TaqMan real-time PCR system. The subjects were classified based on the degree of radiological impairment according to the criteria of Kellgren-Laurence and regarding functional impairment using the WOMAC and LEQUESNE questionnaires. RESULTS: TNFA gene polymorphic individuals (subjects harboring allele A) are more affected by OA (χ2 = 8.7, p = 0.003), once they have major radiological lesion both in hip (Fisher-Freeman-Halton Test = 3.9, p = 0.04) and knee (Fisher-Freeman-Halton Test = 4.0, p = 0.04) as well as worse functional status assessed by the Lequesne questionnaire (Mann-Whitney, p = 0.04). At the multivariate analysis, after adjustment for age, gender, body mass index, the presence of rare allele for TNFA (allele A) increases the susceptibility to OA development [OR: 1.87 (95% CI: 1.1-3.2)]. CONCLUSION: We conclude that the SNP - 308 G/A of TNFA gene may affect osteoarthritis susceptibility, severity and functional status of individuals with osteoarthritis.
Subject(s)
Osteoarthritis, Hip/genetics , Osteoarthritis, Knee/genetics , Osteoarthritis/genetics , Physical Functional Performance , Polymorphism, Single Nucleotide , Severity of Illness Index , Tumor Necrosis Factor-alpha/genetics , Aged , Aged, 80 and over , Alleles , Case-Control Studies , Female , Genetic Predisposition to Disease , Genotype , Humans , Male , Middle Aged , Osteoarthritis/diagnostic imaging , Osteoarthritis/physiopathology , Osteoarthritis, Hip/diagnostic imaging , Osteoarthritis, Hip/physiopathology , Osteoarthritis, Knee/diagnostic imaging , Osteoarthritis, Knee/physiopathology , Real-Time Polymerase Chain ReactionABSTRACT
BACKGROUND: Currently, two pathogenic pathways describe the role of obesity in osteoarthritis (OA); one through biomechanical stress, and the other by the contribution of systemic inflammation. The aim of this study was to evaluate the effect of free fatty acids (FFA) in human chondrocytes (HC) expression of proinflammatory factors and reactive oxygen species (ROS). METHODS: HC were exposed to two different concentrations of FFA in order to evaluate the secretion of adipokines through cytokines immunoassays panel, quantify the protein secretion of FFA-treated chondrocytes, and fluorescent cytometry assays were performed to evaluate the reactive oxygen species (ROS) production. RESULTS: HC injury was observed at 48 h of treatment with FFA. In the FFA-treated HC the production of reactive oxygen species such as superoxide radical, hydrogen peroxide, and the reactive nitrogen species increased significantly in a at the two-dose tested (250 and 500 µM). In addition, we found an increase in the cytokine secretion of IL-6 and chemokine IL-8 in FFA-treated HC in comparison to the untreated HC. CONCLUSION: In our in vitro model of HC, a hyperlipidemia microenvironment induces an oxidative stress state that enhances the inflammatory process mediated by adipokines secretion in HC.
Subject(s)
Hyperlipidemias/drug therapy , Inflammation/drug therapy , Obesity/drug therapy , Osteoarthritis/drug therapy , Adipokines/genetics , Cells, Cultured , Chondrocytes/drug effects , Chondrocytes/metabolism , Fatty Acids, Nonesterified/administration & dosage , Humans , Hydrogen Peroxide/metabolism , Hyperlipidemias/complications , Hyperlipidemias/genetics , Hyperlipidemias/metabolism , Inflammation/complications , Inflammation/genetics , Inflammation/metabolism , Obesity/complications , Obesity/genetics , Obesity/metabolism , Osteoarthritis/complications , Osteoarthritis/genetics , Osteoarthritis/metabolism , Oxidative Stress/drug effects , Reactive Oxygen Species/metabolismABSTRACT
Osteoarthritis (OA) triggers increased levels of inflammatory markers, including prostaglandin (PG) E2 and proinflammatory cytokines. The elevation of cytokine levels is closely associated with increased articular tissue degeneration. Thus, the use of combination therapies may presumably be able to enhance the effects on the modulation of inflammatory markers. The present study aimed to evaluate and compare the effects of photobiomodulation therapy (PBMT), physical exercise, and topical nonsteroidal anti-inflammatory drug (NSAID) use on the inflammatory process after they were applied either alone or in different combinations. OA was induced by intra-articular papain injection in the knee of rats. After 21 days, the animals began treatment with a topical NSAID and/or with physical exercise and/or PBMT. Treatments were performed three times a week for eight consecutive weeks, totaling 24 therapy sessions. Analysis of real-time polymerase chain reaction (RT-PCR) gene expression; interleukin (IL)-1ß, IL-6, and tumor necrosis factor alpha (TNF-α) protein expression; and PGE2 levels by enzyme-linked immunosorbent assay (ELISA) was conducted. Our results showed that PBMT alone and Exerc + PBMT significantly reduced IL-1ß gene expression (p < 0.05) while no treatment changed both IL-6 and TNF-α gene expression. Treatment with NSAID alone, PBMT alone, Exerc + PBMT, and NSAID + PBMT reduced IL-1ß protein expression (p < 0.05). All therapies significantly reduced IL-6 and TNF-α protein expression (p < 0.05) compared with the OA group. Similarly, all therapies, except Exerc, reduced the levels of PGE2 (p < 0.05) compared with the OA group. The results from the present study indicate that treatment with PBMT is more effective in modulating the inflammatory process underlying OA when compared with the other therapies tested.
Subject(s)
Inflammation/pathology , Low-Level Light Therapy , Osteoarthritis/pathology , Osteoarthritis/therapy , Physical Conditioning, Animal , Animals , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Anti-Inflammatory Agents, Non-Steroidal/therapeutic use , Combined Modality Therapy , Dinoprostone/blood , Disease Models, Animal , Enzyme-Linked Immunosorbent Assay , Gene Expression Regulation/drug effects , Humans , Interleukin-1beta/genetics , Interleukin-1beta/metabolism , Interleukin-6/genetics , Interleukin-6/metabolism , Knee Joint/metabolism , Male , Osteoarthritis/blood , Osteoarthritis/genetics , Rats, Wistar , Real-Time Polymerase Chain Reaction , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/metabolismABSTRACT
The objective of this study was to evaluate the effects of photobiomodulation therapy (PBMT) on inflammatory indicators, i.e., inflammatory mediators (TNF-α and CINC-1), and pain characterized by hyperalgesia and B1 and B2 receptor activation at 6, 24, and 48 h after papain-induced osteoarthritis (OA) in rats. Fifty-four rats were subjected to hyperalgesia evaluations and then divided randomly into three groups-a control group and two groups OA and OA PBMT group by using laser parameters at wavelength (808 nm), output power (50 mW), energy per point (4 Joules), power density (1.78 W/cm2), laser beam (0.028 cm2), and energy density (144 J/cm2)-the induction of osteoarthritis was then performed with 20-µl injections of a 4 % papain solution dissolved in 10 µl of saline solution, to which 10 µl of cysteine solution (0.03 M). The statistical analysis was performed using two-way ANOVA with Bonferroni's post hoc test for comparisons between the 6, 24, and 48 h and team points within each group, and between the control, injury, and PBMT groups, and p < 0.05 was considered to indicate a significant difference. The hyperalgesia was evaluated at 6, 24, and 48 h after the injury. PBMT at a wavelength of 808 nm and doses of 4 J, administered afterward, promotes increase at the threshold of pressure stimulus at 6, 24, and 48 h after application and promote cytokine attenuation levels (TNF and CINC-1) and bradykinin receptor (B1 and B2) along the experimental period. We conclude that photobiomodulation therapy was able to promote the reduction of proinflammatory cytokines such as TNF-α and CINC-1, to reduce the gene and protein expression of the bradykinin receptor (B1 and B2), as well as increasing the stimulus response threshold of pressure in an experimental model of acute osteoarthritis.
Subject(s)
Inflammation Mediators/metabolism , Low-Level Light Therapy , Osteoarthritis/metabolism , Osteoarthritis/radiotherapy , Receptors, Bradykinin/metabolism , Acute Disease , Animals , Chemokine CXCL1 , Disease Models, Animal , Enzyme-Linked Immunosorbent Assay , Extremities/pathology , Gene Expression Regulation , Hyperalgesia/complications , Hyperalgesia/genetics , Hyperalgesia/metabolism , Hyperalgesia/pathology , Male , Osteoarthritis/complications , Osteoarthritis/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rats, Wistar , Real-Time Polymerase Chain Reaction , Tumor Necrosis Factor-alpha/metabolismABSTRACT
Peroxisome proliferator-activated receptor is closely associated with the pathogenesis of osteoarthritis. The level of exogenous advanced glycation end-products (AGEs) in articular cartilage is highly associated with the severity of osteoarthritic lesions. However, their interactions and role in promoting osteoarthritisprogression remain unclear. Here, we investigated the effect of AGEs on transforming growth factor (TGF)-ß and matrix metalloproteinase (MMP)-9 expression, and discussed the correlation between AGEs and osteoarthritis, possible signaling pathways and mechanism in rabbit chondrocytes. TGF-ß and MMP-9 mRNA and protein expression, catalase (CAT) and superoxide dismutase (SOD) activity, and malondialdehyde (MDA) and reactive oxygen species (ROS) levels were analyzed in chondrocytes treated with different concentrations of AGEs using RT-PCR and/or western blot; we detected NF-κB nuclear translocation by immunofluorescence. AGE treatment significantly increased TGF-ß and MMP-9 mRNA and protein expression compared to controls (P < 0.01) in a dose-dependent manner (highest at 100 µg/mL). AGE-induced TGF-ß and MMP-9 expressions in chondrocytes were significantly inhibited by anti-RAGE and PDTC (0.1 mM) treatment (P < 0.01). Furthermore, AGE-treatment significantly decreased CAT and SOD activity and increased MDA levels in a concentration-dependent manner compared to controls (P < 0.05), significantly promoting NF-κB nuclear translocation. AGE significantly inhibited the increased expression of TGF-ß and MMP- 9, and induced chondrocyte damage. Its mechanism is associated with RAGE activation, increased ROS expression, and activation of the NF- κB signaling pathways.
Subject(s)
Chondrocytes/metabolism , Glycation End Products, Advanced/pharmacology , Matrix Metalloproteinase 9/biosynthesis , PPAR alpha/metabolism , Transforming Growth Factor beta/biosynthesis , Animals , Cartilage, Articular/metabolism , Cells, Cultured , Down-Regulation/drug effects , Glycation End Products, Advanced/genetics , Glycation End Products, Advanced/metabolism , Male , Matrix Metalloproteinase 13/genetics , Matrix Metalloproteinase 9/genetics , Matrix Metalloproteinase 9/metabolism , NF-kappa B/genetics , NF-kappa B/metabolism , Osteoarthritis/genetics , Osteoarthritis/metabolism , PPAR alpha/genetics , RNA, Messenger/metabolism , Rabbits , Reactive Oxygen Species/metabolism , Signal Transduction/drug effects , Transforming Growth Factor beta/genetics , Transforming Growth Factor beta/metabolismABSTRACT
In this study, we investigated the differential expression profiles of cyclooxygenase-2 (COX-2) mRNA and proteins in osteoarthritis (OA) and rheumatoid arthritis (RA) patients to elucidate the role of COX-2 expression in the pathogenesis and development of these diseases and to provide novel drug targets for treating arthritis. A total of 60 patients who received arthroscopic surgeries for treating OA (N = 30) or RA (N = 30) were examined. Fifteen normal synovial tissue samples were included as the control group. Fibroblastic synovial cells in all samples were cultured in vitro and COX-2 mRNA, protein expression levels, and COX-2 levels were detected in synovial fluids by real-time polymerase chain reaction, western blotting, and enzyme-linked immunosorbent assay methods, respectively. The mRNA level of COX-2 was significantly elevated in synovial cells from OA and RA patients compared to that in control samples (P < 0.05). COX-2 mRNA level was significantly higher in synovial cells from OA patients than in those from RA patients (P < 0.05). Consistent results were obtained for COX-2 protein expression levels from patients' synovial samples. In synovial fluids, OA (P < 0.05), but not RA (P > 0.05), patients showed significantly higher COX-2 levels compared to the control group. Elevated synovial COX-2 expression facilitates the pathogenesis of OA and RA, and thus this index reflects the condition of these 2 diseases.
Subject(s)
Arthritis, Rheumatoid/metabolism , Cyclooxygenase 2/genetics , Cyclooxygenase 2/metabolism , Gene Expression Regulation , Osteoarthritis/metabolism , Adult , Aged , Arthritis, Rheumatoid/genetics , Arthritis, Rheumatoid/pathology , Female , Humans , Male , Middle Aged , Osteoarthritis/genetics , Osteoarthritis/pathology , Synovial Fluid/metabolism , Synovial Membrane/metabolismABSTRACT
A rat model with cartilage chondrocyte injury was established using interleukin-1ß (IL-1ß) to investigate the effect of Ginkgo biloba extract (EGb) on matrix metalloproteinase-3 (MMP-3) expression. Rat chondrocytes were extracted and randomly divided into six groups: control group, IL-1ß (model) group, IL-1ß + dexamethasone group, and IL-1ß + EGb group (both high and low dose groups). Reverse transcriptase polymerase chain reaction and enzyme-linked immunosorbent assay were used to detect MMP-3 expression. Compared to the MMP-3 mRNA level in the control group, MMP-3 mRNA level significantly increased in the model group (P < 0.05). The application of dexamethasone or EGb significantly decreased the MMP-3 mRNA level (P < 0.05). MMP-3 mRNA and protein levels decreased in the EGb-treated group, especially in the high-dose group, compared to those in the dexamethasone group (P < 0.05). EGb may reduce MMP-3 production during IL-1ß-induced chondrocyte damage and protect chondrocytes to some extent, with better efficacy at high doses.
Subject(s)
Chondrocytes/drug effects , Chondrocytes/metabolism , Gene Expression , Ginkgo biloba/chemistry , Matrix Metalloproteinase 3/genetics , Plant Extracts/pharmacology , Animals , Cells, Cultured , Chondrocytes/pathology , Disease Models, Animal , Enzyme-Linked Immunosorbent Assay , Matrix Metalloproteinase 3/metabolism , Osteoarthritis/genetics , Osteoarthritis/metabolism , Osteoarthritis/pathology , Plant Extracts/chemistry , RNA, Messenger/genetics , RNA, Messenger/metabolism , RatsABSTRACT
The purpose of this study was to identify genes and pathways for osteoarthritis (OA) diagnosis and therapy. We downloaded the gene expression profile of OA from Gene Expression Omnibus (GEO) database including 10 early OA, 9 late OA, and 5 normal control samples. Next, we screened differentially expressed genes (DEGs) between early- and late-stage OA samples comparing with healthy control samples. Then, the Search Tool for the Retrieval of Interacting Genes/Proteins (STRING) software was used to construct protein-protein interaction (PPI) network, which was to predict the proteins that may interact with DEGs. The Gene Ontology (GO)-enrichment method was used to analyze the function of genes in the PPI networks. Meanwhile network module analysis was performed using Cytoscape. A total of 24 and 29 DEGs were identified for the early and late OA, respectively. TAC1 showed the highest degree in the PPI network. Functional annotation of the TAC1 network module indicated that this gene is associated with the G protein-coupled signal transduction pathway. In summary, TAC1, together with G protein-coupled receptors, appear to play a role in the biogenesis and progress of OA. Further analysis of this gene and pathway could therefore provide a potential target for the diagnosis and treatment of OA.
Subject(s)
Gene Expression Profiling , Osteoarthritis/genetics , Protein Interaction Maps/genetics , Case-Control Studies , Cluster Analysis , Gene Expression Regulation , Humans , Software , Statistics as TopicABSTRACT
OBJECTIVE: To analyze a set of circulating microRNA (miRNA) in plasma from patients with primary Osteoarthritis (OA) and describe the biological significance of altered miRNA in OA based on an in silico analysis of their target genes. METHODS: miRNA expression was analyzed using TaqMan Low Density Arrays and independent assays. The search for potential messenger RNA (mRNA) targets of the differentially expressed miRNA was performed by means of the miRWalk and miRecords database; we conducted the biological relevance of the predicted miRNA targets by pathway analysis with the Reactome and DAVID databases. RESULTS: We measured the expression of 380 miRNA in OA; 12 miRNA were overexpressed under the OA condition (p value, ≤0.05; fold change, >2). These results were validated by the detection of some selected miRNA by quantitative PCR (qPCR). In silico analysis showed that target messenger RNA (mRNA) were potentially regulated by these miRNA, including genes such as SMAD1, IL-1B, COL3A, VEGFA, and FGFR1, important in chondrocyte maintenance and differentiation. Some metabolic pathways affected by the miRNA: mRNA ratio are signaling Bone morphogenetic proteins (BMP), Platelet-derived growth factor (PDGF), and Nerve growth factor (NGF), these latter two involved in the process of pain. CONCLUSIONS: We identified 12 miRNA in the plasma of patients with primary OA. Specific miRNA that are altered in the disease could be released into plasma, either due to cartilage damage or to an inherent cellular mechanism. Several miRNA could regulate genes and pathways related with development of the disease; eight of these circulating miRNA are described, to our knowledge, for first time in OA.