MiR-33b-3p promotes chondrocyte proliferation and inhibits chondrocyte apoptosis and cartilage ECM degradation by targeting DNMT3A in osteoarthritis

2019 ◽  
Vol 519 (2) ◽  
pp. 430-437 ◽  
Author(s):  
Fangnan Ma ◽  
Guangheng Li ◽  
Yangyi Yu ◽  
Jianzhong Xu ◽  
Xuejian Wu
Author(s):  
Chongzhi Pan ◽  
Wenzhou Huang ◽  
Qi Chen ◽  
Jiu Xu ◽  
Guoyu Yao ◽  
...  

Purpose: Extracellular Vesicles (EVs) derived from hMSCs, have the potential to alleviate cartilage damage and inflammation. We aimed to explore the effects of EVs derived from lncRNA malat‐1-overexpressing human mesenchymal stem cells (hMSCs) on chondrocytes.Material and Methods: hMSCs-derived Extracellular Vesicles (hMSCs-EVs) were identified by transmission electron microscopy and western blot. We used a Sprague-Dawley (SD) rat model of CollagenaseⅡ-induced osteoarthritis (OA) as well as IL-1β-induced OA chondrocytes. Lentiviral vectors were used to overexpress lncRNA malat‐1 in hMSCs. Chondrocyte proliferation, inflammation, extracellular matrix degradation, and cell migration were measured by Edu staining, ELISA, western blot analysis, and transwell assay. Chondrocyte apoptosis was evaluated by flow cytometry, Hoechst 33342/PI Staining, and western blot. Safranine O-fast green (S-O) staining and HE staining were used to assess morphologic alterations of the rat knee joint.Results: hMSCsmalat−1-EVs decreased MMP-13, IL-6, and Caspase-3 expression in IL-1β-induced OA chondrocytes. Moreover, hMSCsmalat−1-EVs promoted chondrocyte proliferation and migration, suppressed apoptosis, and attenuated IL-1β-induced chondrocyte injury. Our animal experiments suggested that hMSCsmalat−1-EVs were sufficient to prevent cartilage degeneration.Conclusion: Our findings show that lncRNA malat-1from hMSCs‐delivered EVs can promote chondrocyte proliferation, alleviate chondrocyte inflammation and cartilage degeneration, and enhance chondrocyte repair. Overall, hMSCsmalat−1-EVs might be a new potential therapeutic option for patients with OA.


2014 ◽  
Vol 8 (4) ◽  
pp. 1065-1074 ◽  
Author(s):  
YING XU ◽  
GUO-JING DAI ◽  
QIAN LIU ◽  
ZHEN-LI LIU ◽  
ZHI-QIAN SONG ◽  
...  

2007 ◽  
Vol 282 (46) ◽  
pp. 33698-33706 ◽  
Author(s):  
Shufang Wu ◽  
Janna K. Flint ◽  
Geoffrey Rezvani ◽  
Francesco De Luca

NF-κB is a group of transcription factors involved in cell proliferation, differentiation, and apoptosis. Mice deficient in the NF-κB subunits p50 and p52 have retarded growth, suggesting that NF-κB is involved in bone growth. Yet, it is not clear whether the reduced bone growth of these mice depends on the lack of NF-κB activity in growth plate chondrocytes. Using cultured rat metatarsal bones and isolated growth plate chondrocytes, we studied the effects of two NF-κB inhibitors (pyrrolidine dithiocarbamate (PDTC) or BAY11-7082 (BAY)), p65 short interference RNA (siRNA), and of the overexpression of p65 on chondrocyte proliferation, differentiation, and apoptosis. To further define the underlying mechanisms, we studied the functional interaction between NF-κB p65 and BMP-2 in chondrocytes. PDTC and BAY suppressed metatarsal linear growth. Such growth inhibition resulted from decreased chondrocyte proliferation and differentiation and from increased chondrocyte apoptosis. In cultured chondrocytes, the inhibition of NF-κB p65 activation (by PDTC and BAY) and expression (by p65 siRNA) led to the same findings observed in cultured metatarsal bones. In contrast, overexpression of p65 in cultured chondrocytes induced chondrocyte proliferation and differentiation and prevented apoptosis. Although PDTC, BAY, and p65 siRNA reduced the expression of BMP-2 in cultured growth plate chondrocytes, the overexpression of p65 increased it. The addition of Noggin, a BMP-2 antagonist, neutralized the stimulatory effects of p65 on chondrocyte proliferation and differentiation, as well as its anti-apoptotic effect. In conclusion, our findings indicate that NF-κB p65 expressed in growth plate chondrocytes facilitates growth plate chondrogenesis and longitudinal bone growth by inducing BMP-2 expression and activity.


2021 ◽  
Author(s):  
Xue Bai ◽  
Hongyan Sun ◽  
Lina Jia ◽  
Junjie Xu ◽  
Peng Zhang ◽  
...  

Abstract Background: Cartilage destruction caused by inflammation is a clinical challenge. Many studies have investigated cartilage destruction in adults, but little research was conducted on children. Results: The gaps without chondrocytes and ECM between the proliferative and hypertrophy zones of the GP cartilage were formation after the treatment of LPS, but the gaps were not observed in the AuNPs + LPS group. This finding can be attributed to the capability of AuNPs to target to the chondrocytes and reduce the release of inflammatory cytokines and secretion of ECM degradation factors induced by LPS. And then, the LPS-induced apoptosis rate of mouse chondrocytes and ECM degradation rate were inhibited. Finally, the balance of catabolic and anabolic factors in the ECM was maintained.Conclusion: These findings indicate that AuNPs can partially protect the cartilage of children from inflammatory damage by suppressing chondrocyte apoptosis and ECM degradation.


2021 ◽  
Author(s):  
Wenqiang Xu ◽  
Xiaofeng Liu ◽  
Wenqing Qu ◽  
Xin Wang ◽  
Hao Su ◽  
...  

Abstract Osteoarthritis (OA) is a common joint disease with characteristics of chronic inflammation and articular cartilage degeneration. It has been proved that LSD1 was up-regulated in OA cartilage tissues, but its role and regulatory mechanism in OA are unclear. Herein, interleukin 1 beta (IL-1β)-treated human chondrocytes was performed as a cell model of OA. Then, LSD1 expression was found that up-regulated in OA cartilage tissues and IL-1β-induced chondrocytes. Knockdown of LSD1 increased cell viability, while decreased apoptosis rate and inflammatory cytokines secretion levels in IL-1β-induced chondrocytes. In addition, knockdown of LSD1 reduced the expression of catabolic proteins (MMP-13 and ADAMTS-5) and enhanced the expression of anabolic proteins (Collagen II and Aggrecan) in chondrocytes after IL-1β stimulation. Moreover, overexpression of TRIM32 repressed chondrocyte viability, while promoted IL-1β-induced chondrocyte apoptosis, inflammation and ECM degradation. The expression of LSD1 and TRIM32 in OA cartilage was positively correlated, and knockdown of LSD1 down-regulated TRIM32 expression of chondrocytes. Our data further indicated that LSD1 regulated autophagy of chondrocytes through modulating TRIM32. Overexpression of TRIM32 reduced the effect of LSD1 knockdown on IL-1β-induced chondrocytes, while activating autophagy by Rapamycin further reversed this reduction. Therefore, our study shows that knockdown of LSD1 inhibited IL-1β-induced chondrocyte apoptosis, inflammation and ECM degradation via TRIM32-mediated autophagy.


2019 ◽  
Vol 2019 ◽  
pp. 1-17 ◽  
Author(s):  
Yao Li ◽  
Yaosen Wu ◽  
Kaixia Jiang ◽  
Wen Han ◽  
Jing Zhang ◽  
...  

Osteoarthritis (OA) is an age-related degenerative disease with complicated pathology involving chondrocyte apoptosis and extracellular matrix (ECM) degradation. Previous studies have shown that moderate autophagy has a protective effect against apoptosis in chondrocyte. Mangiferin is a natural polyphenol and exerts multiple pharmacological effects on different diseases in various preclinical studies. In this study, we investigated the effects of mangiferin on OA and delineated a potential molecular mechanism. In vitro, mangiferin treatment inhibited the expression of proapoptotic proteins induced by tert-butyl hydroperoxide (TBHP), increased the expression of antiapoptotic Bcl-2, and prevented ECM degradation by inhibiting the production of matrix-degrading enzyme. Mechanistically, mangiferin enhanced autophagy by activating the AMP-activated protein kinase (AMPK) signaling pathway. On the contrary, inhibition of autophagy partly abolished the protective effects of mangiferin on antiapoptosis and ECM synthesis in TBHP-treated chondrocyte. Correspondingly, the protective effect of mangiferin was also found in a mouse OA model. In conclusion, our results suggested that mangiferin serves as a potentially applicable candidate for treating OA.


1998 ◽  
Vol 41 (9) ◽  
pp. 1632-1638 ◽  
Author(s):  
Sanshiro Hashimoto ◽  
Robert L. Ochs ◽  
Setsuro Komiya ◽  
Martin Lotz

Endocrinology ◽  
2011 ◽  
Vol 152 (4) ◽  
pp. 1423-1433 ◽  
Author(s):  
Hongzhi Sun ◽  
Weijin Zang ◽  
Bo Zhou ◽  
Lin Xu ◽  
Shufang Wu

Abstract Dehydroepiandrosterone (DHEA) is produced by the adrenal cortex and is the most abundant steroid in humans. Although in some physiological and pathological conditions the increased secretion of DHEA and its sulfated form is associated with accelerated growth rate and skeletal maturation, it is unclear whether DHEA can affect longitudinal bone growth and skeletal maturation by acting directly at the growth plate. In our study, DHEA suppressed metatarsal growth, growth plate chondrocyte proliferation, and hypertrophy/differentiation. In addition, DHEA increased the number of apoptotic chondrocytes in the growth plate. In cultured chondrocytes, DHEA reduced chondrocyte proliferation and induced apoptosis. The DHEA-induced inhibition of metatarsal growth and growth plate chondrocyte proliferation and hypertrophy/differentiation was nullified by culturing metatarsals with DHEA in the presence of ICI 182,780, an inhibitor of estrogen receptor, but not in the presence of Casodex, an inhibitor of androgen receptor. Lastly, nuclear factor-κB DNA binding activity was inhibited by the addition of DHEA in the medium of cultured chondrocyte. Our findings indicate that DHEA suppressed bone growth by acting directly at growth plate through estrogen receptor. Such growth inhibition is mediated by decreased chondrocyte proliferation and hypertrophy/differentiation and by increased chondrocyte apoptosis.


Sign in / Sign up

Export Citation Format

Share Document