Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 4 de 4
Filter
Add more filters











Database
Language
Publication year range
1.
Osteoarthritis Cartilage ; 29(3): 372-379, 2021 03.
Article in English | MEDLINE | ID: mdl-33347923

ABSTRACT

OBJECTIVES: To investigate the role of zinc finger protein 440 (ZNF440) in the pathophysiology of cartilage degeneration during facet joint (FJ) and knee osteoarthritis (OA). METHODS: Expression of ZNF440 in FJ and knee cartilage was determined by immunohistochemistry, quantitative (q)PCR, and Western blotting (WB). Human chondrocytes isolated from FJ and knee OA cartilage were cultured and transduced with ZNF440 or control plasmid, or transfected with ZNF440 or control small interfering RNA (siRNA), with/without interleukin (IL)-1ß. Gene and protein levels of catabolic, anabolic and apoptosis markers were determined by qPCR or WB, respectively. In silico analyses were performed to determine compounds with potential to inhibit expression of ZNF440. RESULTS: ZNF440 expression was increased in both FJ and knee OA cartilage compared to control cartilage. In vitro, overexpression of ZNF440 significantly increased expression of MMP13 and PARP p85, and decreased expression of COL2A1. Knockdown of ZNF440 with siRNA partially reversed the catabolic and cell death phenotype of human knee and FJ OA chondrocytes stimulated with IL-1ß. In silico analysis followed by validation assays identified scriptaid as a compound with potential to downregulate the expression of ZNF440. Validation experiments showed that scriptaid reduced the expression of ZNF440 in OA chondrocytes and concomitantly reduced the expression of MMP13 and PARP p85 in human knee OA chondrocytes overexpressing ZNF440. CONCLUSIONS: The expression of ZNF440 is significantly increased in human FJ and knee OA cartilage and may regulate cartilage degenerative mechanisms. Furthermore, scriptaid reduces the expression of ZNF440 and inhibits its destructive effects in OA chondrocytes.


Subject(s)
Cartilage, Articular/metabolism , Chondrocytes/metabolism , DNA-Binding Proteins/physiology , Knee Joint , Osteoarthritis, Knee/genetics , Osteoarthritis, Spine/genetics , Zinc Fingers/genetics , Zygapophyseal Joint , Adult , Aged , Aged, 80 and over , Apoptosis/drug effects , Apoptosis/genetics , Chondrocytes/drug effects , Collagen Type II/genetics , Computer Simulation , DNA-Binding Proteins/genetics , Female , Gene Knockdown Techniques , Histone Deacetylase Inhibitors/pharmacology , Humans , Hydroxylamines/pharmacology , Immunohistochemistry , In Vitro Techniques , Inflammation/genetics , Male , Matrix Metalloproteinase 13/genetics , Metabolism/drug effects , Metabolism/genetics , Middle Aged , Osteoarthritis, Knee/metabolism , Osteoarthritis, Spine/metabolism , Quinolines/pharmacology , Young Adult , Zinc Fingers/drug effects
2.
Osteoarthritis Cartilage ; 28(11): 1471-1481, 2020 11.
Article in English | MEDLINE | ID: mdl-32738291

ABSTRACT

OBJECTIVE: MicroRNAs act locally and systemically to impact osteoarthritis (OA) pathophysiology, but comprehensive profiling of the circulating miRNome in early vs late stages of OA has yet to be conducted. Sequencing has emerged as the preferred method for microRNA profiling since it offers high sensitivity and specificity. Our objective was to sequence the miRNome in plasma from 91 patients with early [Kellgren-Lawrence (KL) grade 0 or 1 (n = 41)] or late [KL grade 3 or 4 (n = 50)] symptomatic radiographic knee OA to identify unique microRNA signatures in each disease state. DESIGN: MicroRNA libraries were prepared using the QIAseq miRNA Library Kit and sequenced on the Illumina NextSeq 550. Counts were produced for microRNAs captured in miRBase and for novel microRNAs. Statistical, bioinformatics, and computational biology approaches were used to refine and interpret the final list of microRNAs. RESULTS: From 215 differentially expressed microRNAs (FDR < 0.01), 97 microRNAs showed an increase or decrease in expression in ≥85% of samples in the early OA group as compared to the median expression in the late OA group. Increasing this threshold to ≥95%, seven microRNAs were identified: hsa-miR-335-3p, hsa-miR-199a-5p, hsa-miR-671-3p, hsa-miR-1260b, hsa-miR-191-3p, hsa-miR-335-5p, and hsa-miR-543. Four novel microRNAs were present in ≥50% of early OA samples and had 27 predicted gene targets in common with the prioritized set of predicted gene targets from the 97 microRNAs, suggesting common underlying mechanisms. CONCLUSION: Sequencing of well-characterized patient cohorts produced unbiased profiling of the circulating miRNome and identified a unique panel of 11 microRNAs in early radiographic knee OA.


Subject(s)
Circulating MicroRNA/blood , Osteoarthritis, Knee/genetics , Transcriptome , Adult , Aged , Aged, 80 and over , Computational Biology , Disease Progression , Female , Humans , Male , Middle Aged , Osteoarthritis, Knee/blood , Osteoarthritis, Knee/diagnostic imaging , Young Adult
3.
Osteoarthritis Cartilage ; 28(10): 1394-1400, 2020 10.
Article in English | MEDLINE | ID: mdl-32683043

ABSTRACT

OBJECT: Autophagy maintains cartilage homeostasis and is compromised during osteoarthritis (OA), contributing to cartilage degeneration. We sought to determine if D-isomer TAT-Beclin-1, a potent inducer of autophagy, could attenuate post-traumatic OA in mice. METHODS: 10-week-old mice underwent destabilization of the medial meniscus (DMM) surgery to induce post-traumatic OA, or sham surgery (control), and injected intra-articularly with D-isomer TAT-Beclin-1 (0.5-2 mg/kg) or PBS 1 week post-surgery for up to 9 weeks. Mice were sacrificed at 2 or 10 weeks post-surgery. Knee joint sections were evaluated by histopathology for cartilage degeneration and synovitis, and immunostaining for key markers of autophagy (LC3B), cell proliferation (nuclear Ki67), activated fibroblasts (αSMA), and cells of hematopoietic origin (CD45). RESULTS: All D-isomer TAT-Beclin-1-treated DMM mice had no difference in the degree of cartilage degeneration compared to PBS-injected DMM mice. Surprisingly, all D-isomer TAT-Beclin-1-treated mice exhibited substantial synovial hyperplasia, with increased cellularity and ECM deposition (fibrosis-like phenotype), as compared to PBS-injected mice. Synovial effects of D-isomer TAT-Beclin-1 were dose- and injection frequency-dependent. An increased percentage of cells positive for LC3B and nuclear Ki67 were found in the synovial intima early after injection, which persisted after frequent injections. CONCLUSIONS: D-isomer TAT-Beclin-1 did not attenuate cartilage degeneration, but rather induced synovial hyperplasia associated with increased expression of key markers of autophagy and cell proliferation and a fibrosis-like phenotype, independent of markers of fibroblast activation or persistent hematopoietic-origin cell infiltration. These data suggest that, if not tissue-targeted, caution should be taken using autophagy activators due to diverse cellular responses in the joint.


Subject(s)
Autophagy/drug effects , Beclin-1/pharmacology , Cartilage, Articular/drug effects , Cell Proliferation/drug effects , Osteoarthritis, Knee/pathology , Synovial Membrane/drug effects , Animals , Cartilage, Articular/pathology , Disease Models, Animal , Fibroblasts/drug effects , Gene Products, tat/pharmacology , Hyperplasia , Injections, Intra-Articular , Menisci, Tibial/surgery , Mice , Synovial Membrane/pathology , Synovitis/pathology , Tibial Meniscus Injuries
4.
Osteoarthritis Cartilage ; 17(3): 406-13, 2009 Mar.
Article in English | MEDLINE | ID: mdl-18760941

ABSTRACT

OBJECTIVE: Connective tissue growth factor (CTGF) has been implicated in regulation of chondrocyte differentiation at multiple steps and has been implicated in the progression of diseases such as scleroderma and osteoarthritis. However, the pathways mediating the expression of CTGF/CCN2 and related factors in cartilage are not fully understood. We have previously shown that the Rho family of proteins and the actin cytoskeleton regulate both early and late chondrocyte differentiation. RESULTS: Here we demonstrate that several CTGF/Cyr61/Nov (CCN) family members are differentially affected by either inhibition of actin polymerization (cytochalasin D treatment), promotion of actin polymerization (jasplakinolide treatment), inhibition of RhoA/rho kinase (ROCK) signaling (Y27632 treatment) and Rac1 signaling. We also show that the Smad site in the CTGF/CCN2 promoter is responsive to both Rac1 inhibition and cytochalasin D treatment, suggesting a role of TGFbeta/Smad signaling in mediating the effects of actin dynamics and Rac1. CONCLUSION: Collectively, these data show that Rac1 and actin pathways control CTGF/CCN2 expression in chondrocytes which might be relevant to both skeletal development and associated diseases such as osteoarthritis.


Subject(s)
Actins/physiology , Cell Differentiation/physiology , Chondrocytes/cytology , Connective Tissue Growth Factor/genetics , Gene Expression Regulation , rac1 GTP-Binding Protein/physiology , Animals , Blotting, Western , Cytoskeleton , Female , Mice , Polymerase Chain Reaction , Pregnancy , Transforming Growth Factor beta1/physiology , rho GTP-Binding Proteins/physiology
SELECTION OF CITATIONS
SEARCH DETAIL