Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
Add more filters











Database
Language
Publication year range
1.
Connect Tissue Res ; 62(6): 615-628, 2021 Nov.
Article in English | MEDLINE | ID: mdl-33043724

ABSTRACT

PURPOSE: Transposable elements are known to remodel gene structure and provide a known source of genetic variation. Retrotransposon gag-like-3 (RTL3) is a mammalian retrotransposon-derived transcript (MART) whose function in the skeletal tissue is unknown. This study aimed to elucidate the biological significance of RTL3 in chondrogenesis and type-II collagen (COL2A1) gene expression in chondrocytes. MATERIALS AND METHODS: Expression of RTL3, SOX-9 and COL2A1 mRNAs was determined by TaqMan assays and the protein expression by immunoblotting. RTL3 and Sox-9 depletion in human chondrocytes was achieved using validated siRNAs. An RTL3 mutant (∆RTL3) lacking the zinc finger domain was created using in vitro mutagenesis. Forced expression of RTL3, ∆RTL3, and SOX-9 was achieved using CMV promoter containing expression plasmids. CRISPR-Cas9 was utilized to delete Rtl3 and create a stable ATDC5Rlt3-/- cell line. Matrix deposition and Col2a1 quantification during chondrogenesis were determined by Alcian blue staining and Sircol™ Soluble Collagen Assay, respectively. RESULTS: RTL3 is not ubiquitously expressed but showed strong expression in cartilage, chondrocytes and synoviocytes but not in muscle, brain, or other tissues analyzed. Loss-of-function and gain-of-function studies demonstrated a critical role of RTL3 in the regulation of SOX-9 and COL2A1 expression and matrix synthesis during chondrogenesis. Both RTL3 and SOX-9 displayed co-regulated expression in chondrocytes. Gene regulatory activity of RTL3 requires the c-terminal CCHC zinc-finger binding domain. CONCLUSIONS: Our results identify a novel regulatory mechanism of COL2A1 expression in chondrocytes that may help to further understand the skeletal development and the pathogenesis of diseases with altered COL2A1 expression.


Subject(s)
Chondrocytes , Retroelements , Animals , Cell Differentiation , Chondrocytes/metabolism , Chondrogenesis/genetics , Collagen/metabolism , Collagen Type II/genetics , Collagen Type II/metabolism , Humans , Mammals/genetics , Mammals/metabolism , SOX9 Transcription Factor/genetics , SOX9 Transcription Factor/metabolism
2.
J Cell Sci ; 133(22)2020 11 30.
Article in English | MEDLINE | ID: mdl-33097606

ABSTRACT

Mitochondrial function is impaired in osteoarthritis (OA) but its impact on cartilage catabolism is not fully understood. Here, we investigated the molecular mechanism of mitochondrial dysfunction-induced activation of the catabolic response in chondrocytes. Using cartilage slices from normal and OA cartilage, we showed that mitochondrial membrane potential was lower in OA cartilage, and that this was associated with increased production of mitochondrial superoxide and catabolic genes [interleukin 6 (IL-6), COX-2 (also known as PTGS2), MMP-3, -9, -13 and ADAMTS5]. Pharmacological induction of mitochondrial dysfunction in chondrocytes and cartilage explants using carbonyl cyanide 3-chlorophenylhydrazone increased mitochondrial superoxide production and the expression of IL-6, COX-2, MMP-3, -9, -13 and ADAMTS5, and cartilage matrix degradation. Mitochondrial dysfunction-induced expression of catabolic genes was dependent on the JNK (herein referring to the JNK family)/activator protein 1 (AP1) pathway but not the NFκB pathway. Scavenging of mitochondrial superoxide with MitoTEMPO, or pharmacological inhibition of JNK or cFos and cJun, blocked the mitochondrial dysfunction-induced expression of the catabolic genes in chondrocytes. We demonstrate here that mitochondrial dysfunction contributes to OA pathogenesis via JNK/AP1-mediated expression of catabolic genes. Our data shows that AP1 could be used as a therapeutic target for OA management.This article has an associated First Person interview with the first author of the paper.


Subject(s)
Cartilage, Articular , Transcription Factor AP-1 , Cartilage, Articular/metabolism , Cells, Cultured , Chondrocytes/metabolism , Humans , Interleukin-1beta/metabolism , MAP Kinase Signaling System , Mitochondria , Reactive Oxygen Species/metabolism , Transcription Factor AP-1/genetics , Transcription Factor AP-1/metabolism
3.
Biomed Pharmacother ; 129: 110452, 2020 Sep.
Article in English | MEDLINE | ID: mdl-32768946

ABSTRACT

Osteoarthritis (OA) is the most prevalent joint degenerative disease leading to irreversible structural and functional changes in the joint and is a major cause of disability and reduced life expectancy in ageing population. Despite the high prevalence of OA, there is no disease modifying drug available for the management of OA. Oxidative stress, a result of an imbalance between the production of reactive oxygen species (ROS) and their clearance by antioxidant defense system, is high in OA cartilage and is a major cause of chronic inflammation. Inflammatory mediators, such as interleukin-1ß (IL-1ß), tumor necrosis factor-α (TNF-α) and interleukin-6 (IL-6) are highly upregulated in OA joints and induce ROS production and expression of matrix degrading proteases leading to cartilage extracellular matrix degradation and joint dysfunction. ROS and inflammation are interdependent, each being the target of other and represent ideal target/s for the treatment of OA. Plant polyphenols possess potent antioxidant and anti-inflammatory properties and can inhibit ROS production and inflammation in chondrocytes, cartilage explants and in animal models of OA. The aim of this review is to discuss the chondroprotective effects of polyphenols and modulation of different molecular pathways associated with OA pathogenesis and limitations and future prospects of polyphenols in OA treatment.


Subject(s)
Anti-Infective Agents/pharmacology , Antioxidants/pharmacology , Antirheumatic Agents/therapeutic use , Inflammation Mediators/antagonists & inhibitors , Joints/drug effects , Osteoarthritis/drug therapy , Oxidative Stress/drug effects , Polyphenols/pharmacology , Animals , Humans , Inflammation Mediators/metabolism , Joints/metabolism , Joints/pathology , Osteoarthritis/metabolism , Osteoarthritis/pathology , Reactive Oxygen Species/metabolism , Signal Transduction
4.
Int Immunopharmacol ; 85: 106612, 2020 Aug.
Article in English | MEDLINE | ID: mdl-32450530

ABSTRACT

Joint inflammation is a key player in the pathogenesis of osteoarthritis (OA). Imperatorin, a plant-derived small molecule has been reported to have anti-inflammatory properties; however, its effect on chondrocytes is not known. Here, we investigated the effects of Imperatorin on interleukin-1ß (IL-1ß) induced expression of inducible nitric oxide synthase (iNOS) and nitric oxide production in primary human OA chondrocytes and cartilage explants culture under pathological conditions and explored the associated signaling pathways. We pretreated chondrocytes or explants with Imperatorin (50 µM) followed by IL-1ß (1 ng/ml), and the culture supernatant was used to determine the levels of nitrite production by Griess assay and chondrocytes were harvested to prepare cell lysate or RNA for gene expression analysis of iNOS by Western blot or qPCR and in explants by immunohistochemistry (IHC). Pretreatment of primary chondrocytes and cartilage explants with Imperatorin suppressed IL-1ß induced expression of iNOS and NO production. Imperatorin blocked the IL-1ß-induced phosphorylation of ERK-MAPK/AP1 signaling pathway to suppress iNOS expression. The role of ERK in the regulation of iNOS expression was verified by using ERK inhibitor. Interestingly, we also found that Imperatorin binds to iNOS protein and inhibits its activity in vitro. Our data demonstrated that Imperatorin possess strong anti-inflammatory activity and may be developed as a therapeutic agent for the management of OA.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Extracellular Signal-Regulated MAP Kinases/metabolism , Furocoumarins/pharmacology , Nitric Oxide Synthase Type II/metabolism , Osteoarthritis/prevention & control , Transcription Factor AP-1/metabolism , Anti-Inflammatory Agents/therapeutic use , Cartilage/drug effects , Cell Survival/drug effects , Chondrocytes/cytology , Chondrocytes/drug effects , Chondrocytes/ultrastructure , Furocoumarins/therapeutic use , Humans , Interleukin-1beta/antagonists & inhibitors , Interleukin-1beta/toxicity , Molecular Docking Simulation , Nitric Oxide/metabolism , Nitric Oxide Synthase Type II/chemistry , Nitric Oxide Synthase Type II/genetics , Nitrites/analysis , Primary Cell Culture , Proteome/drug effects , Signal Transduction/drug effects , Up-Regulation
5.
J Cell Physiol ; 235(10): 6366-6376, 2020 10.
Article in English | MEDLINE | ID: mdl-32017079

ABSTRACT

Accumulating evidence suggests that inflammation has a key role in the pathogenesis of osteoarthritis (OA). Nitric oxide (NO) has been established as one of the major inflammatory mediators in OA and drives many pathological changes during the development and progression of OA. Excessive production of NO in chondrocytes promotes cartilage destruction and cellular injury. The synthesis of NO in chondrocytes is catalyzed by inducible NO synthase (iNOS), which is thereby an attractive therapeutic target for the treatment of OA. A number of direct and indirect iNOS inhibitors, bioactive compounds, and plant-derived small molecules have been shown to exhibit chondroprotective effects by suppressing the expression of iNOS. Many of these iNOS inhibitors hold promise for the development of new, disease-modifying therapies for OA; however, attempts to demonstrate their success in clinical trials are not yet successful. Many plant extracts and plant-derived small molecules have also shown promise in animal models of OA, though further studies are needed in human clinical trials to confirm their therapeutic potential. In this review, we discuss the role of iNOS in OA pathology and the effects of various iNOS inhibitors in OA.


Subject(s)
Nitric Oxide Synthase Type II/metabolism , Osteoarthritis/metabolism , Osteoarthritis/pathology , Animals , Humans , Inflammation/drug therapy , Inflammation/metabolism , Inflammation/pathology , Inflammation Mediators/metabolism , Osteoarthritis/drug therapy , Plant Extracts/pharmacology
6.
Arthritis Rheumatol ; 71(4): 583-593, 2019 04.
Article in English | MEDLINE | ID: mdl-30302948

ABSTRACT

OBJECTIVE: Cytokine expression is tightly regulated posttranscriptionally, but high levels of interleukin-6 (IL-6) in patients with osteoarthritis (OA) indicate that regulatory mechanisms are disrupted in this disorder. The enzyme ZCCHC6 (zinc-finger CCHC domain-containing protein 6; TUT-7) has been implicated in posttranscriptional regulation of inflammatory cytokine expression, but its role in OA pathogenesis is unknown. The present study was undertaken to investigate whether ZCCHC6 directs the expression of IL-6 and influences OA pathogenesis in vivo. METHODS: Human and mouse chondrocytes were stimulated with recombinant IL-1ß. Expression of ZCCHC6 in human chondrocytes was knocked down using small interfering RNAs. IL-6 transcript stability was determined by actinomycin D chase, and 3'-uridylation of microRNAs was determined by deep sequencing. Zcchc6-/- mice were produced by gene targeting. OA was surgically induced in the knee joints of mice, and disease severity was scored using a semiquantitative grading system. RESULTS: ZCCHC6 was markedly up-regulated in damaged cartilage from human OA patients and from wild-type mice with surgically induced OA. Overexpression of ZCCHC6 induced the expression of IL-6, and its knockdown reduced IL-6 transcript stability and IL-1ß-induced IL-6 expression in chondrocytes. Reintroduction of Zcchc6 in Zcchc6-/- mouse chondrocytes rescued the IL-1ß-induced IL-6 expression. Knockdown of ZCCHC6 reduced the population of micro-RNA 26b (miR-26b) with 3'-uridylation by 60%. Zcchc6-/- mice with surgically induced OA produced low levels of IL-6 and exhibited reduced cartilage damage and synovitis in the joints. CONCLUSION: These findings indicate that ZCCHC6 enhances IL-6 expression in chondrocytes through transcript stabilization and by uridylating miR-26b, which abrogates repression of IL-6. Inhibition of IL-6 expression and significantly reduced OA severity in Zcchc6-/- mice identify ZCCHC6 as a novel therapeutic target to inhibit disease pathogenesis.


Subject(s)
Gene Silencing , Interleukin-6/metabolism , Nucleotidyltransferases/metabolism , Osteoarthritis/genetics , Severity of Illness Index , Animals , Chondrocytes/metabolism , Disease Models, Animal , Gene Expression Regulation , Humans , Knee Joint/metabolism , Mice , RNA Nucleotidyltransferases , RNA, Small Interfering/metabolism , Up-Regulation
7.
Cell Physiol Biochem ; 49(3): 932-946, 2018.
Article in English | MEDLINE | ID: mdl-30184535

ABSTRACT

BACKGROUND/AIMS: Butein (2',3,4,4'-Tetrahydroxychalcone), a polyphenol produced by several plants including Butea monoserpma, has been reported to exert potent anti-inflammatory activity but the mechanism remains unknown. In the present work we investigated the mechanism of Butein-mediated suppression of IL-6 expression in normal and human osteoarthritis (OA) chondrocytes under pathological conditions. METHODS: Expression level of interleukin-6 (IL-6) protein in OA cartilage was analyzed by immunohistochemistry using a validated antibody. Chondrocytes derived from normal or OA cartilage by enzymatic digestion were pretreated with Butein followed by stimulation with interleukin-1ß (IL-1ß) and the levels of IL-6 mRNA were quantified by TaqMan assay and the protein levels were measured by Western immunoblotting. Autophagy activation was determined by Western blotting and confocal microscopy. Autophagy was inhibited by siRNA mediated knockdown of ATG5. RESULTS: Expression of IL-6 protein was high in the OA cartilage compared to smooth cartilage from the same patient. OA chondrocytes and cartilage explants stimulated with IL-1ß showed high level expression of IL-6 mRNA and protein. Butein increased the phosphorylation of AMPKαThr-172, TSC2Ser-1387 and ULK1Ser-317 and inhibited the phosphorylation of mTORSer-2448 and its downstream target p70S6K and increased autophagy flux that correlated with the suppression of the IL-1ß mediated expression of IL-6 in normal and OA chondrocytes. In OA chondrocytes with siRNA-mediated knockdown of ATG5 expression, treatment with Butein failed to activate autophagy and abrogated the suppression of IL-1ß induced IL-6 expression. CONCLUSION: Our findings demonstrate for the first time that Butein activate autophagy in OA chondrocytes via AMPK/TSC2/ULK1/mTOR pathway. Additionally, activation of autophagy was essential to block the IL-1ß-induced expression of IL-6 in OA chondrocytes. These data support further studies to evaluate the use of Butein or compounds derived from it for the management of OA.


Subject(s)
Autophagy/drug effects , Chalcones/pharmacology , Interleukin-1beta/pharmacology , Interleukin-6/metabolism , Signal Transduction/drug effects , AMP-Activated Protein Kinases/metabolism , Autophagy-Related Protein 5/antagonists & inhibitors , Autophagy-Related Protein 5/genetics , Autophagy-Related Protein 5/metabolism , Autophagy-Related Protein-1 Homolog/metabolism , Cartilage, Articular/cytology , Cartilage, Articular/pathology , Cell Survival/drug effects , Cells, Cultured , Chondrocytes/cytology , Chondrocytes/drug effects , Chondrocytes/metabolism , Humans , Interleukin-6/genetics , Intracellular Signaling Peptides and Proteins/metabolism , Osteoarthritis/metabolism , Osteoarthritis/pathology , Phosphorylation/drug effects , RNA Interference , RNA, Small Interfering/metabolism , Reactive Oxygen Species/metabolism , TOR Serine-Threonine Kinases/metabolism , Tuberous Sclerosis Complex 2 Protein , Tumor Suppressor Proteins/metabolism
8.
IUBMB Life ; 67(12): 897-913, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26597014

ABSTRACT

Advanced glycation end products (AGEs) are a cohort of heterogeneous compounds that are formed after the nonenzymatic glycation of proteins, lipids and nucleic acids. Accumulation of AGEs in the body is implicated in various pathophysiological conditions like diabetes, cardiovascular diseases and atherosclerosis. Numerous studies have reported the connecting link between AGEs and the various complications associated with diseases. Hence, detection and measurement of AGEs becomes centrally important to understand and manage the menace created by AGEs inside the body. In recent years, an increasing number of immunotechniques as well as bioanalytical techniques have been developed to efficiently measure the levels of AGEs, but most of them are still far away from being clinically consistent, as relative disparity and ambiguity masks their standardization. This article is designed to critically review the recent advances and the emerging techniques for detection of AGEs. It is an attempt to summarize the major techniques that exist currently for the detection of AGEs both qualitatively and quantitatively. This review primarily focuses on the detection and quantification of AGEs which are formed in vivo. Immunochemical approach though costly but most effective and accurate method to measure the level of AGEs. Literature review suggests that detection of autoantibody targeting AGEs is a promising way that can be utilized for detection of AGEs. Future research efforts should be dedicated to develop this method in order to push forward the clinical applications of detection of AGEs.


Subject(s)
Biochemistry/methods , Glycation End Products, Advanced/analysis , Immunohistochemistry/methods , Animals , Autoantibodies , Chromatography, Affinity/methods , Chromatography, Liquid/methods , Electrophoresis/methods , Enzyme-Linked Immunosorbent Assay/methods , Glycation End Products, Advanced/immunology , Glycation End Products, Advanced/metabolism , Humans , Mass Spectrometry/methods , Mice , Spectrometry, Fluorescence/methods
9.
Microrna ; 4(3): 209-16, 2015.
Article in English | MEDLINE | ID: mdl-26729022

ABSTRACT

UNLABELLED: Backgroud: TCF7L2 encodes for a transcription factor that plays a significant role in the Wnt signaling pathway and has been implicated in schizophrenia and blood glucose homeostasis. During embryonic development, TCF7L2 is expressed widely in the midbrain and forebrain regions of mouse brain. However, expression data of TCF7L2 and its functional studies are much scarcer for chicken brain. Our study demonstrated interesting expression pattern of TCF7L2 in the optic tectum of developing chicken embryo. Further study was undertaken to develop and test the knock down tools for TCF7L2 in the developing chicken embryo which would help in its functional characterization. METHODS: An RNAi based strategy was chosen to knock down TCF7L2A. miRNA based construct was designed and developed to target TCF7L2. However these miRNA construct may or may not be effective in knocking down their target gene, therefore the efficiency of the miRNA construct in knocking down TCF7L2 was tested. First the efficiency was measured in vitro using a sensor assay. The efficient pre-miRNA was cloned in an avian retrovirus to achieve in vivo knock down in the developing chicken embryo. RCAS-miRNA construct targeting TCF7L2, was electroporated in the tectum of embryonic day 3.5 chicken embryo. The extent of in vivo knock down of TCF7L2 by the RCAS-miRNA-TCF7L2 was examined using RNA in situ hybridization technique. RESULTS: TCF7L2 showed strong layer specific expression from embryonic day 5 to embryonic day 13 which is also the important time window for tectal development. In addition to the layer specific expression, TCF7L2 is also strongly expressed in the pre-tegmentum area of the tectum. The TCF7L2 transcript showed marked decrease in the region that was electroporated with RCAS-miRNA-TCF7L2. CONCLUSION: Therefore this study has led to the generation and validation of RCAS-pre miRNA-TCF7L2 which effectively knocks down TCF7L2 in vivo and thus has paved the way for further functional characterization of TCF7L2 in the chicken optic tectum.

SELECTION OF CITATIONS
SEARCH DETAIL