130 research outputs found

    Small RNA signatures of the anterior cruciate ligament from patients with knee joint osteoarthritis

    Get PDF
    ABSTRACT The anterior cruciate ligaments are susceptible to degeneration, resulting in pain, reduced mobility and development of the degenerative joint disease osteoarthritis. There is currently a paucity of knowledge on how anterior cruciate ligament degeneration and disease can lead to osteoarthritis. Small non-coding RNAs (sncRNAs), such as microRNAs, and small nucleolar RNA, are important regulators of gene expression. We aimed to identify sncRNA profiles of human anterior cruciate ligaments to provide novel insights into their roles in osteoarthritis. RNA was extracted from the anterior cruciate ligaments of non-osteoarthritic knee joints (control) and end-stage osteoarthritis knee joints, used for small RNA sequencing and significantly differentially expressed sncRNAs defined. Bioinformatic analysis was undertaken on the differentially expressed miRNAs and their putative target mRNAs to investigate pathways and biological processes affected. Our analysis identified 184 sncRNA that were differentially expressed between control ACLs derived from osteoarthritic joints with a false discovery adjusted p value<0.05; 68 small nucleolar RNAs, 26 small nuclear RNAs and 90 microRNAs. We identified both novel and previously identified (miR-206, –101, –365 and –29b and –29c) osteoarthritis-related microRNAs and other sncRNAs (including SNORD74, SNORD114, SNORD72) differentially expressed in ligaments derived from osteoarthritic joints. Significant cellular functions deduced by the differentially small nuclear RNAs and 90 microRNAs. We identified expressed miRNAs included differentiation of muscle (P<0.001), inflammation (P<1.42E-10), proliferation of chondrocytes (P<0.03), fibrosis (P<0.001) and cell viability (P<0.03). Putative mRNAs were associated with the canonical pathways ‘Hepatic Fibrosis Signalling’ (P<3.7E-32), and ‘Osteoarthritis’ (P<2.2E-23). Biological processes included apoptosis (P<1.7E-85), fibrosis (P<1.2E-79), inflammation (P<3.4E-88), necrosis (P<7.2E-88) and angiogenesis (P<5.7E-101). SncRNAs are important regulators of anterior cruciate disease during osteoarthritis and may be used as therapeutic targets to prevent and manage anterior cruciate ligament disease and the resultant osteoarthritis

    MicroRNA Profiling in Cartilage Ageing

    Get PDF
    Osteoarthritis (OA) is the most common age-related joint disorder in man. MicroRNAs (miRNA), a class of small noncoding RNAs, are potential therapeutic targets for regulating molecular mechanisms in both disease and ageing. Whilst there is an increasing amount of research on the roles of miRNAs in ageing, there has been scant research on age-related changes in miRNA in a cartilage. We undertook a microarray study on young and old human cartilages. Findings were validated in an independent cohort. Contrasts between these samples identified twenty differentially expressed miRNAs in a cartilage from old donors, derived from an OA environment which clustered based on OA severity. We identified a number of recognised and novel miRNAs changing in cartilage ageing and OA including miR-126: a potential new candidate with a role in OA pathogenesis. These analyses represent important candidates that have the potential as cartilage ageing and OA biomarkers and therapeutic targets

    Activation of NF-κB/p65 Facilitates Early Chondrogenic Differentiation during Endochondral Ossification

    Get PDF
    BACKGROUND: NF-κB/p65 has been reported to be involved in regulation of chondrogenic differentiation. However, its function in relation to key chondrogenic factor Sox9 and onset of chondrogenesis during endochondral ossification is poorly understood. We hypothesized that the early onset of chondrogenic differentiation is initiated by transient NF-κB/p65 signaling. METHODOLOGY/PRINCIPAL FINDINGS: The role of NF-κB/p65 in early chondrogenesis was investigated in different in vitro, ex vivo and in vivo endochondral models: ATDC5 cells, hBMSCs, chicken periosteal explants and growth plates of 6 weeks old mice. NF-κB/p65 activation was manipulated using pharmacological inhibitors, RNAi and activating agents. Gene expression and protein expression analysis, and (immuno)histochemical stainings were employed to determine the role of NF-κB/p65 in the chondrogenic phase of endochondral development. Our data show that chondrogenic differentiation is facilitated by early transient activation of NF-κB/p65. NF-κB/p65-mediated signaling determines early expression of Sox9 and facilitates the subsequent chondrogenic differentiation programming by signaling through key chondrogenic pathways. CONCLUSIONS/SIGNIFICANCE: The presented data demonstrate that NF-κB/p65 signaling, as well as its intensity and timing, represents one of the transcriptional regulatory mechanisms of the chondrogenic developmental program of chondroprogenitor cells during endochondral ossification. Importantly, these results provide novel possibilities to improve the success of cartilage and bone regenerative techniques

    Physiological tonicity improves human chondrogenic marker expression through nuclear factor of activated T-cells 5 in vitro

    Get PDF
    Abstract Introduction: Chondrocytes experience a hypertonic environment compared to plasma (280 mOsm) due to the high fixed negative charge density of cartilage. Standard isolation of chondrocytes removes their hypertonic matrix, exposing them to non-physiological conditions. During in-vitro expansion, chondrocytes quickly lose their specialized phenotype, making them inappropriate for cell-based regenerative strategies. We aimed to elucidate the effects of tonicity during isolation and in-vitro expansion on chondrocyte phenotype. Methods: Human articular chondrocytes were isolated and subsequently expanded at control tonicity (280 mOsm) or at moderately elevated, physiological, tonicity (380 mOsm). The effects of physiological tonicity on chondrocyte proliferation and chondrogenic marker expression were evaluated. The role of Tonicity-responsive Enhancer Binding Protein (TonEBP/NFAT5) in response to physiological tonicity was investigated using nuclear factor of activated T-cells 5 (NFAT5) RNA interference. Results: Moderately elevated, physiological, tonicity (380 mOsm) did not affect chondrocyte proliferation, while higher tonicities inhibited proliferation and diminished cell viability. Physiological tonicity improved expression of chondrogenic markers and NFAT5 and its target genes, while suppressing dedifferentiation marker collagen type I and improving type II/type I expression ratios >100-fold. Effects of physiological tonicity were similar in osteoarthritic and ‘normal’ (non-osteoarthritic) chondrocytes, indicating a disease-independent mechanism. NFAT5 RNA interference abolished tonicity-mediated effects and revealed that NFAT5 positively regulates collagen type II expression, while suppressing type I. Conclusions: Physiological tonicity provides a simple, yet effective, means to improve phenotypical characteristics during cytokine-free isolation and in-vitro expansion of human articular chondrocytes. Our findings will lead to the development of improved cell-based repair strategies for chondral lesions and provides important insights into mechanisms underlying osteoarthritic progression

    Equine synovial fluid small non-coding RNA signatures in early osteoarthritis

    Get PDF
    Background Osteoarthritis remains one of the greatest causes of morbidity and mortality in the equine population. The inability to detect pre-clinical changes in osteoarthritis has been a significant impediment to the development of effective therapies against this disease. Synovial fluid represents a potential source of disease-specific small non-coding RNAs (sncRNAs) that could aid in the understanding of the pathogenesis of osteoarthritis. We hypothesised that early stages of osteoarthritis would alter the expression of sncRNAs, facilitating the understanding of the underlying pathogenesis and potentially provide early biomarkers. Methods Small RNA sequencing was performed using synovial fluid from the metacarpophalangeal joints of both control and early osteoarthritic horses. A group of differentially expressed sncRNAs was selected for further validation through qRT-PCR using an independent cohort of synovial fluid samples from control and early osteoarthritic horses. Bioinformatic analysis was performed in order to identify putative targets of the differentially expressed microRNAs and to explore potential associations with specific biological processes. Results Results revealed 22 differentially expressed sncRNAs including 13 microRNAs; miR-10a, miR-223, let7a, miR-99a, miR-23b, miR-378, miR-143 (and six novel microRNAs), four small nuclear RNAs; U2, U5, U11, U12, three small nucleolar RNAs; U13, snoR38, snord96, and one small cajal body-specific RNA; scarna3. Five sncRNAs were validated; miR-223 was significantly reduced in early osteoarthritis and miR-23b, let-7a-2, snord96A and snord13 were significantly upregulated. Significant cellular actions deduced by the differentially expressed microRNAs included apoptosis (P < 0.0003), necrosis (P < 0.0009), autophagy (P < 0.0007) and inflammation (P < 0.00001). A conservatively filtered list of 57 messenger RNA targets was obtained; the top biological processes associated were regulation of cell population proliferation (P < 0.000001), cellular response to chemical stimulus (P < 0.000001) and cell surface receptor signalling pathway (P < 0.000001). Conclusions Synovial fluid sncRNAs may be used as molecular biomarkers for early disease in equine osteoarthritic joints. The biological processes they regulate may play an important role in understanding early osteoarthritis pathogenesis. Characterising these dynamic molecular changes could provide novel insights on the process and mechanism of early osteoarthritis development and is critical for the development of new therapeutic approaches
    • …
    corecore