Skip to main content
Erschienen in: BMC Musculoskeletal Disorders 1/2018

Open Access 01.12.2018 | Research article

The hsa-miR-181a-5p reduces oxidation resistance by controlling SECISBP2 in osteoarthritis

verfasst von: Jianli Xue, Zixin Min, Zhuqing Xia, Bin Cheng, Binshang Lan, Fujun Zhang, Yan Han, Kunzheng Wang, Jian Sun

Erschienen in: BMC Musculoskeletal Disorders | Ausgabe 1/2018

Abstract

Background

The phenotypes of osteoarthritis (OA) consist of cartilage extracellular matrix (ECM) metabolism disorder and the breakdown of cartilage homeostasis, which are induced by pro-inflammatory factors and oxidative stress. Selenoproteins regulated by selenocysteine insertion sequence binding protein 2 (SBP2) are highly effective antioxidants, but their regulatory mechanisms, particularly the involvement of miRNAs, are not fully understood.

Methods

To explore whether miR-181a-5p and SBP2 are involved in OA pathogenesis, we established an IL-1β model using the chondrocyte SW1353 cell line. Next, we up- or down-regulated SBP2 and miRNA-181a-5p expression in the cells. Finally, we measured the expression of miRNA-181a-5p, SBP2 and three selenoproteins in OA cartilage and peripheral blood.

Results

The results showed that IL-1β increased hsa-miR-181a-5p and decreased SBP2 in a time- and dose-dependent manner. GPX1 and GPX4, which encode crucial glutathione peroxidase antioxidant enzymes, were up-regulated along with SBP2 and miR-181a-5p. Furthermore, SBP2 showed a significant negative correlation with miR-181a-5p during induced ATDC5 cell differentiation. There was lower GPX1 and GPX4 mRNA expression and SBP2 protein expression in damaged cartilage than in smooth cartilage from the same OA sample, and hsa-miR-181a-5p expression on the contrary. Similar results were observed in peripheral blood. In conclusion, we have reported a novel pathway in which pro-inflammatory factors, miRNA, SBP2 and selenoproteins are associated with oxidation resistance in cartilage.

Conclusion

Overall, this study provides the first comprehensive evidence that pro-inflammatory factors cause changes in the cartilage antioxidant network and describes the discovery of novel mediators of cartilage oxidative stress and OA pathophysiology. Our data suggest that miR-181a-5p may be used to develop novel early-stage diagnostic and therapeutic strategies for OA.
Hinweise

Electronic supplementary material

The online version of this article (https://​doi.​org/​10.​1186/​s12891-018-2273-6) contains supplementary material, which is available to authorized users.
Abkürzungen
ACAN
Aggrecan
COL2
type-II collagen
ECM metabolic
Extracellular matrix
HRP
Horseradish peroxidase
IL-1β
Including interleukin-1
IL-6
Interleukin-6
inhibitor-181a-5p
miR-181a-5p inhibitor
mimic-181a-5p
miR-181a-5p mimics
OA
Osteoarthritis
PBS
Phosphate buffered saline
ROS
Reactive oxygen species
Se
Selenium
Sec
Selenocysteine
SECIS
Sec insertion sequence
SECISBP2 or SBP2
Sec insertion sequence binding protein 2
Sel
Selenoprotein
TKR
Total knee replacement
TNF-α
Tumour necrosis factor-α

Background

Osteoarthritis (OA) may be a response to superfluous mechanical stress or inflammation, and pro-inflammatory factors, including interleukin-1 (IL-1β), interleukin-6 (IL-6), and tumour necrosis factor-α (TNF-α), are involved in OA pathogenesis [1, 2]. The phenotypes of cartilage injury processes induced by pro-inflammatory factors are cartilage extracellular matrix (ECM) metabolic disorder, the disruption of cartilage homeostasis, and enhanced expression of matrix degradation enzymes such as MMP13 [3]. MMP13, a major enzyme hydrolysing type-II collagen (COL2), is a dominant protein component of the cartilage ECM [4, 5] and a biomarker for arthritis progression and therapeutic effects [68].
Reactive oxygen species (ROS) are products of aerobic metabolism that injure DNA, proteins, and cellular membranes [911]. Oxidative stress plays important roles in the pathogenesis of OA and cartilage degradation, which is induced by ROS, and traumatic loading increases cartilage oxidation and causes cell death [12]. In addition, oxidative stress-mediated regulation of the expression of redox-sensitive proteins is regarded as a key mechanism underlying age-related cellular dysfunction and disease progression [13].
Selenoproteins (Sel) are important members of a network of antioxidant enzymatic systems and minimize damage induced by ROS. They contain selenocysteine (Sec), the 21st proteinogenic amino acid, which is named after the essential biological trace element selenium (Se) and acts as an active-site residue essential for the catalytic activity of selenoproteins [911]. The genetic code ‘UGA’, commonly a termination codon in cells, encodes Sec into selenoproteins [14]. Several special cis-trans elements and trans-acting factors, typically the Sec insertion sequence (SECIS) and Sec insertion sequence binding protein 2 (SECISBP2 or SBP2), regulate selenoprotein biosynthesis [15, 16]. SECIS, which is located in the selenoprotein mRNA 3′-untranslated region (3′-UTR), binds with SBP2. The function of SBP2 is to carry Sec-tRNASec into the ribosome ‘A site’ to recognize ‘UGA’ as the Sec codon during selenoprotein synthesis [15, 16].
Intriguingly, osteo-chondroprogenitor-specific deletion of the selenocysteinyl tRNASec gene results in dyschondroplasia phenotypes, particularly those showing abnormal skeletal development in mice [17]. ‘UGA’ is recognized as a termination codon, and inactive truncated selenoproteins are produced in the presence of insufficient Sec-tRNASec [18]. Similarly, the TrxR1 short inactive fragment, a two-amino-acid truncated C-terminal motif, leads to the death of human lung carcinoma A549 cells [19]. However, little is known about how selenoprotein biosynthesis regulates OA cartilage. In particular, the pathway from pro-inflammatory factors to selenoprotein biosynthesis mediated by SBP2 in cartilage is poorly understood.
Moreover, more than 20 miRNAs, such as the cartilage-specific miR-140-5p, participate in chondrogenesis, cartilage homeostasis and degradation, and chondrocyte metabolism, which are closely associated with OA development [2022]. Further, miR-9, miR-34a and miR-146a are related with oxidative stress in OA chondrocytes [23, 24]. In a previous study, we identified a repertoire of miRNAs during the development of rat femoral articular cartilage [25] and demonstrated that miR-337 regulates chondrogenesis through a direct target, TGFBR2 [26]. Specifically, miR-181a-5p, a member of the miR-181 family, which is organized into three clusters (miR-181a/b-1, miR-181a/b-2, and miR-181c/d), is positively correlated with chondrogenesis [25]. Meanwhile, non-hypertrophic articular and hypertrophic MSC-derived chondrocytes showed differential expression of miR-181a-5p, suggesting that its expression is altered during successive differentiation stages [27]. Moreover, miR-181a-5p is predicted to be a target of hSBP2 by TargetScanHuman7.1, and it may inhibit the expression of the important ECM protein aggrecan (ACAN) in chondrocytes, simultaneously acquiring a negative feedback function for cartilage homeostasis [28]. However, further investigation is required to understand the oxidation resistance-associated roles of miR-181a-5p in OA.
In the present study, the glutathione peroxidase-encoding genes GPX1 and GPX4 and the selenoprotein S-encoding gene SELS were examined due to their regulation by SBP2. Hence, we investigated the detailed regulatory relationships among pro-inflammatory factors, miRNA, SBP2 and selenoproteins in the context of oxidation resistance in cartilage. Overall, this study provides the first comprehensive evidence for changes in pro-inflammatory factors in the cartilage antioxidant network during OA and describes the discovery of novel mediators of cartilage oxidative stress and OA pathophysiology. Therefore, our data suggest that miR-181a-5p may be useful for the development of novel early-stage diagnostic and therapeutic strategies for OA.

Methods

Cell culture

The human chondrosarcoma chondrocyte SW1353 cell line was obtained from the Chinese Academy of Sciences (Shanghai, China) and cultured in RPMI-1640 medium (HyClone, USA) with 10% foetal bovine serum (ExCell, China). The murine chondroblast ATDC5 cell line was obtained from the European Collection of Cell Cultures (ECACC) and maintained in Dulbecco’s Modified Eagle’s medium/Ham’s F12 medium (DMEM/F12, HyClone, USA) supplemented with 5% FBS (Gibco, USA). Both cell lines were maintained in a humidified incubator with 5% CO2 at 37 °C, cultured in monolayers and grown to confluence. The medium contained 1% penicillin/streptomycin (Sigma, USA). The cells were seeded in 12-multiwell plates at 7 × 104 cells/well.
For the cartilage matrix degradation model, SW1353 cells were placed in FBS-free medium for more than 10 h, and then the cells were incubated with 0 (as control), 1, 5, 10 and 20 ng/ml IL-1β (Sino Biological Inc., China) for 12 h, or 10 ng/ml IL-1β for 0 (as control), 6, 12, 24 and 48 h. For the chondrocyte differentiation model, ATDC5 cells were induced with 1× ITS supplement (1 mg/ml insulin, 0.55 mg/ml transferrin and 0.5 μg/ml selenium) added to the medium. The chondrogenic culture medium was changed every day.

Transient transfection of hsa-miR-181a-5p mimics or inhibitor sequences

SW1353 cells were seeded for 24 h, and 50 nM hsa-miR-181a-5p mimics (mimic-181a-5p) or negative control (mimic-NC) (Genepharma, China) and 200 nM hsa-miR-181a-5p inhibitor (inhibitor-181a-5p) or negative control (inhibitor-NC) (Genepharma, China) were transiently transfected into SW1353 cells by 1.5 μl/well Lipofectamine™ 2000 (Invitrogen, USA) according to the manufacturer’s instructions. Information about miR-181a-5p is provided in Tables 1 and 2.
Table 1
Information of Mature miR-181a-5p
ID
Accession
Mature sequence (5′-3′)
hsa-miR-181a-5p
MIMAT0000256
AACAUUCAACGCUGUCGGUGAGU
mmu-miR-181a-5p
MIMAT0000210
Table 2
Information of Stem-loop hsa-miR-181a
ID
Accession
Location
Stem-loop sequence (5′-3′)
hsa-mir-181a-1 (hsa-mir-213)
MI0000289
1q32.1
UGAGUUUUGAGGUUGCUUCAGUGAACAUUCAACGCUGUCGGUGAGUUUGGAAUUAAAAUCAAAACCAUCGACCGUUGAUUGUACCCUAUGGCUAACCAUCAUCUACUCCA
hsa-mir-181a-2
MI0000269
9q33.3
AGAAGGGCUAUCAGGCCAGCCUUCAGAGGACUCCAAGGAACAUUCAACGCUGUCGGUGAGUUUGGGAUUUGAAAAAACCACUGACCGUUGACUGUACCUUGGGGUCCUUA

Transient transfection of siRNAs and plasmids

The full-length human SBP2 CDS was cloned from SW1353 chondrocyte cDNA and inserted into a pEFGP-N1 vector (Invitrogen, USA). The primer sequences for the hSBP2-CDS clone are listed in Table 3. SW1353 cells were seeded for 24 h, and 1, 1.5, 2 and 4 μg of the pEFGP-mSBP2-N1 vector or empty vector were transiently transfected into cells by 1.5 μl/well Lipofectamine™ 2000 (Invitrogen, USA). The expression of exogenous and endogenous SBP2 was determined by western blotting with an anti-SBP2 antibody after transfection for 24 h.
Table 3
Information of human primers for hSBP2-CDS
Gene
Sequence (5′-3′)
hSBP2-CDS-Forward
CAGGTCGGATCCAGACCCGGGgccaccATGGCGTCGGAGGGG
hSBP2-CDS-Reverse
TCTGTAGAATTCGGTCCCGGGTAAATTCAAATTCATCAT
Additionally, hSBP2 siRNA (si-SBP2) and control siRNA (si-NC) sequences were purchased from Genepharma Biotechnology Inc. (Genepharma, China). Cell transfection was performed according to the manufacturer’s instructions. For gene knockdown, SW1353 cells were seeded for 24 h, and 50 nM si-SBP2 (5′-GAGCCACACUACAUUGAAATT-3′) or si-NC was transiently transfected into the cells by 1.5 μl/well Lipofectamine™ 2000 (Invitrogen, USA) according to the manufacturer’s instructions. Knockdown efficiency was determined by western blotting after transfection for 48 h.

Patients and articular cartilage collection

OA patients were diagnosed according to the modified Outerbridge classification by The Second Affiliated Hospital, Xi’an Jiaotong University Health Science Center. Articular cartilage samples were obtained at the time of total knee replacement (TKR) from 10 human patients with knee OA (6 women and 4 men; mean ± SEM age: 60 ± 8.3 y) from Shaanxi province, China. All patients were diagnosed with Kellgren and Lawrence grade IV OA. After washing with sterile phosphate buffered saline (PBS), portions of cartilage with a damaged articular surface and portions with a smooth articular surface were used for RNA extraction and immunohistochemistry. Smooth cartilage samples were carefully assessed for any gross signs of degeneration or injury, and only normal-appearing smooth cartilage was used as an internal control (a self control). All cartilage samples were collected without fibrillation. Peripheral blood samples were obtained from 20 OA patients (14 women and 6 men; mean ± SD age: 66.6 ± 5.7 y) and 20 normal control patients (14 women and 6 men; mean ± SD age: 65.9 ± 3.1 y).

Total RNA extraction and quantitative PCR analysis

For RNA extraction, cartilage tissues were harvested from smooth articular surfaces and damaged articular surfaces of the same patient and chopped into pieces that were smaller than 2 × 2 mm. Then, the pieces were immediately frozen in liquid nitrogen. Total RNA was isolated from cells, tissue pieces or plasma samples using TRIzol® (Invitrogen, USA). cDNA was synthesized from 2 μg of total RNA according to the manufacturer’s instructions (RevertAid™; Fermentas, Canada) in a final volume of 20 ml and stored at − 20 °C until use. Furthermore, miRNA-cDNA was obtained using the One Step PrimeScript® miRNA cDNA Synthesis Kit (Takara, Japan).
Both mRNA and miRNA expression was tested by 10 μl real-time quantitative PCR (RT-qPCR), which was performed on an iQ5 real-time PCR detection system (Bio-Rad, Hercules, CA, USA) with SYBR® Premix Ex Taq™ II (TaKaRa, Japan). Relative gene expression was normalized against GAPDH expression in SW1353 cells or β-Actin expression in ATDC5 cells. Additionally, let-7a was used as the internal reference for miR-181a-5p. The procedure for miRNA-cDNA qPCR consisted of two-step amplification: pre-denaturation at 95 °C for 10 s, followed by PCR amplification with 40 cycles of 95 °C for 5 s and 60 °C for 20 s. Information about the primers and PCR amplification is provided in Tables 45 and 6.
Table 4
Information of miRNA-181a-5p for Real-time PCR
MicroRNAs
Accession NO.
Forward primer (5′-3′)
hsa-miRNA-181a-5p
MIMAT0000858
CGCAACATTCAACGCTGTC
hsa-let-7a
MIMAT0000774
CGCTGAGGTAGTAGGTTGT
Reverse primer: GTGCAGGGTCCGAGGT
Table 5
Information of mouse primers for Real-time PCR
Gene
Sequence (5′-3′)
Product size (bp)
Annealing temperature (°Χ)
Sbp2
Forward:CTGCTCCAAAGGCCAAAG
195
60
Reverse:GTGATTGCCCTCTGTGTCTTC
β-Actin
Forward:AACAGTCCGCCTAGAAGCAC
281
60
Reverse:CGTTGACATCCGTAAAGACC
Table 6
Information of human primers for Real-time PCR
Gene
Sequence (5′-3′)
Product size (bp)
Annealing temperature (°C)
SBP2
Forward: CCGCAGATTCAGGGATTACT
92
60
Reverse: CTTGGAAACGGACCAGTTCT
ACAN
Forward: GGCATTTCAGCGGTTCCTTCTC
135
60
Reverse: AGCAGTTGTCTCCTCTTCTACGG
MMP13
Forward: AATATCTGAACTGGGTCTTCCAAAA
102
60
Reverse: CAGACCTGGTTTCCTGAGAACAG
COL2A1
Forward: TGGACGATCAGGCGAAACC
244
62
Reverse: GCTGCGGATGCTCTCAATCT
GPx1
Forward: AAGCTCATCACCTGGTCTCC
124
60
Reverse: CGATGTCAATGGTCTGGAAG
GPx4
Forward: GCTGTGGAAGTGGATGAAGA
105
60
Reverse: TGAGGAACTGTGGAGAGACG
SELS
Forward: CACCTATGGCTGGTACATCG
130
60
Reverse: AACATCAGGTTCCACAGCAG
GAPDH
Forward: CACCCACTCCTCCACCTTTG
110
64
Reverse: CCACCACCCTGTTGCTGTAG

Protein sample preparation and western blotting

Total protein samples from SW1353 cells or ATDC5 cells (10–20 μg) were separated by 10% SDS-PAGE and transferred to PVDF membranes (EMD Millipore, Darmstadt, Germany). After blocking with 3% non-fat milk in TBST buffer, the membranes were incubated with primary antibodies followed by secondary antibodies conjugated to horseradish peroxidase (HRP) and visualized using an ECL detection system (EMD Millipore, Darmstadt, Germany) on a chemiluminescence imaging system. The primary antibodies included anti-SBP2 (1:500, CA, USA), anti-GPX1 (1:2000, CA, USA), anti-MMP13 (1:1000, Abcam, USA) and anti-β-ACTIN (1:2000, Proteintech, China). The following secondary antibodies were purchased from Beyotime Biotech (Jiangsu, China): horseradish peroxidase-coupled anti-rabbit (1:5000) and anti-mouse (1:5000).

Immunohistochemistry staining

After measuring intrinsic peroxidase activity, articular cartilage sections were blocked with 3% hydrogen peroxide (H2O2) and then incubated with 1.5% BSA for 1 h. The sections were covered with anti-SBP2 antibodies (1:250, CA, USA) and incubated at 4 °C in a wet box. After 14 h, all sections were rinsed with PBS and then sequentially incubated with biotinylated secondary antibody for 1 h and DAB reagent (Boster, Wuhan, China) for 5 min at room temperature. Chromogenic reactions were terminated once claybank regions were observed under a microscope. Rabbit IgG was used as a negative control.

Statistical analysis

Data are presented as the mean ± SEM. The statistical significance of pathological data was calculated by using the Mann-Whitney U test. Means of two groups were compared using Student’s t test, and statistical significance was achieved at P < 0.05 in all tests (*: P < 0.05, **: P < 0.01 and **: P < 0.001). All analyses were performed using GraphPad Prism 6.0 (GraphPad Software, San Diego, CA, USA).

Results

Both hsa-miR-181a-5p and SBP2 are regulated by IL-1β in chondrocytes

IL-1β was selected to stimulate SW1353 cells, and hsa-miR-181a-5p expression levels were determined by stem loop RT-qPCR. The expression of hsa-miR-181a-5p and MMP13 continuously and robustly increased after treatment with 10 ng/ml IL-1β for 0 (as a control), 6, 12, 24 and 48 h in SW1353 cells, while SBP2 and GPX1 expression was continuously and sharply reduced at the mRNA level (Fig. 1a). Meanwhile, SBP2, GPX1 and MMP13 expression at the protein level showed the same patterns observed at the mRNA level (Fig. 1b). The expression of hsa-miR-181a-5p increased, and the expression of SBP2 at the mRNA level reduced over time after treatment with 0 (as a control), 1, 5, 10 and 20 ng/ml IL-1β for 12 h (Fig. 1c).

SBP2 regulated the biosynthesis of three selenoproteins and oxidation resistance in chondrocytes

To assess the role of SBP2 in chondrocytes, we constructed recombinant hSBP2-CDS clones and si-SBP2 (Fig. 2a) and transfected these constructs into SW1353 cells. Exogenous SBP2 (122 kDa, Fig. 2b) showed remarkable concentration-dependent up-regulation with pEGFP-N1-mSBP2. Overall, taking into consideration both endogenous SBP2 (95 kDa, Fig. 2b) and exogenous SBP2, 2 μg of pEGFP-N1-mSBP2 was the most suitable treatment to achieve SBP2 over-expression. SBP2 over-expression (P = 0.0003) in SW1353 cells elevated both GPX1 (P = 0.0064) and GPX4 (P = 0.0215) mRNA levels, whereas SELS (P = 0.4532) induced no evident changes (Fig. 2c). On the other hand, when SBP2 levels were specifically reduced by si-SBP2 (P = 0.0087), both GPX1 (P = 0.0097) and GPX4 (P = 0.0431) mRNA levels, but not SELS levels (P = 0.2093), were also down-regulated significantly (Fig. 2d). Meanwhile, total GPXs activity was increased (P = 0.0097) by SBP2 over-expression, and total GPXs activity was reduced (P = 0.0023) under SBP2 knockdown conditions (Fig. 2e).

Transfection of miR-181a-5p affects chondrocyte phenotype and oxidation resistance through SBP2

To confirm the roles of miR-181a-5p in chondrocytes, a miR-181a-5p mimic (P = 0.0022) or a miR-181a-5p inhibitor (P = 0.0108) was applied to alter miR-181a-5p levels (Additional file 1: Figure S1). The expression of cartilage-specific genes such as COL2A1, ACAN and MMP13 and total GPXs activity were detected in SW1353 cells after transfection for 24 h. First, mimic-miR-181a-5p down-regulated ACAN (P = 0.0052) and up-regulated MMP13 (P = 0.0095) (Fig. 3a), while inhibitor-miR-181a-5p also significantly up-regulated MMP13 (P = 0.0319) (Fig. 3b). Furthermore, both SBP2 (P = 0.0209) and SBP2 were significantly down-regulated in SW1353 cells when miR-181a-5p was up-regulated by mimic-181a-5p (Fig. 3c and d). In contrast, neither SBP2 nor SBP2 expression changed when miR-181a-5p was down-regulated by inhibitor-181a-5p (Fig. 3c and d). Meanwhile, total GPXs activity was reduced (P = 0.0145) by miR-181a-5p over-expression, and total GPXs activity was increased (P = 0.0143) under miR-181a-5p knockdown conditions (Fig. 3e). In addition, ITS treatment was applied to cultured cells for 14 days as described previously to induce ATDC5 cells to differentiate in vitro [29], and then the expression of mmu-miR-181a-5p, Sbp2 and SBP2 was detected. With chondrocyte differentiation, the expression of mmu-miR-181a-5p showed remarkable up-regulation at D3 (P = 0.0258), D7 (P = 0.0178) and D14 (P = 0.0103), while SBP2 protein expression was significantly reduced, although the expression of Sbp2 was almost constant (Fig. 3f).

The expression of hsa-miRNA-181a-5p, SBP2 and selenoproteins in OA cartilage

Cartilage tissues were obtained from 8 OA patients to detect the expression of miRNA-181a-5p, SBP2 and three pivotal selenoproteins. OA smooth cartilage and damaged cartilage from the same patients undergoing TKR were separated (Fig. 4a). Total RNA was extracted, and RT-qPCR was performed. According to a paired Student’s t test, miRNA-181a-5p expression levels were significantly higher (P = 0.0114) in damaged cartilage than in smooth cartilage of OA patients (Fig. 4b). Meanwhile, although SBP2 mRNA expression was unattenuated in damaged cartilage (Fig. 4c), SBP2 protein expression was reduced in damaged cartilage (Fig. 4d). Furthermore, GPX1 (P = 0.0183) and GPX4 (P = 0.0149) were down-regulated in damaged OA cartilage (Fig. 4e), while SELS showed no significant changes (Fig. 4e).

The expression of hsa-miRNA-181a-5p, SBP2 and selenoproteins in peripheral blood

Peripheral blood was collected from 20 healthy controls and 20 OA patients. To detect the expression of miRNA-181a-5p, SBP2, GPX1, GPX4 and SELS, total RNA from peripheral blood was extracted, and RT-qPCR was performed. The expression of hsa-miRNA-181a-5p (P = 0.0329) in OA peripheral blood was significantly higher than that in normal controls (Fig. 5a), while SBP2 (P = 0.0061) and GPX1 (P = 0.0111) were both lower in OA peripheral blood than in normal controls (Fig. 5b and c). In addition, SELS (P = 0.8160) showed no statistically significant differences (Fig. 5d), and GPX4 was not detected (data not shown). These results suggested that hsa-miRNA-181a-5p is a potential diagnostic biomarker for OA.

Discussion

To explore whether miR-181a-5p and SBP2 are involved in OA pathogenesis, we established an IL-1β model using the chondrocyte SW1353 cell line. The results showed that IL-1β increased hsa-miR-181a-5p and decreased SBP2 in a time- and dose-dependent manner, while both hsa-miR-181a-5p and SBP2 seemed to participate in the catabolism pathway and oxidative stress in chondrocytes induced by IL-1β. This finding is in line with our expectation that pro-inflammatory cytokines induce miR-181a-5p up-regulation in chondrocytes along with SBP2 down-regulation. Coincidentally, miR-181a-5p up-regulates the expression of caspase-3, PARP, MMP-2, and MMP-9 while repressing chondrocyte proliferation and promoting chondrocyte apoptosis in OA [22, 30].
Next, we used recombinant plasmids and siRNA sequences targeting SBP2 to up- or down-regulate the expression of this gene in SW1353 cells. To investigate SBP2-mediated selenoprotein synthesis, GPX1, GPX4 and SELS were selected as representative selenoproteins expressed by chondrocytes in this study not only because these proteins exhibit differential cellular localization and fulfil different functions in physiological and pathological processes in various cells but also because the affinity of their SECIS binding with ‘UGA’ recoding has been categorized as strong, moderate and weak, respectively [31, 32]
As crucial antioxidant enzymes, GPX1 and GPX4 were regulated by SBP2 up- or down-regulation, while SELS expression levels were always stabilized; these expression patterns are attributable to the differential SECIS affinities and SBP2 binding efficiencies of these proteins. Our findings suggest that SBP2 expression did not align with selenoprotein expression regulation, which affected total GPXs activity and oxidation resistance in chondrocytes. Oxidative damage due to the concomitant overproduction of ROS is present in ageing and OA cartilage [33]. Predictably, oxidative stress destroys normal physiological signalling and contributes to OA [13]. The synergy between blocked selenoprotein expression and disordered metabolism of the articular cartilage ECM induces chondrocyte apoptosis and contributes to cartilage destruction [9, 34] In summary, selenoprotein biosynthesis leads to decreased antioxidant stress.
Additionally, we modulated miR-181a-5p expression by using mimic and inhibitor sequences in SW1353 cells. The expression of miR-181a-5p showed remarkable up-regulation, while SBP2 protein expression was significantly reduced. Unexpectedly, SBP2 expression did not change after miR-181a-5p knockdown, which implies that a very complex regulatory network and multiple modulators are involved in SBP2 expression. Furthermore, SBP2 showed a significant negative correlation with miR-181a-5p during the induced differentiation of ATDC5 cells. These results suggest that hsa-miR-181a-5p affects the chondrocyte phenotype by altering oxidation resistance.
The most effective antioxidants are members of the GPx family, but the mechanisms underlying their effects on OA chondrocytes under oxidative stress are not yet fully understood [9]. Our results established that miR-181a-5p regulated total GPXs activity by decreasing the expression of SBP2 in cartilage, leading to chondrocyte apoptosis and cellular damage induced by ROS. SBP2 is required for protection against ROS-induced cellular damage and increased cell survival [35]. For instance, gene mutations in SBP2 decreased the expression of several selenoproteins, resulting in a complex multisystem selenoprotein deficiency disorder in humans [36], and lipid peroxidation products mediated by free radicals increased in the blood [37]. Further, miR-34a, miR-146a, SOD2, CAT, GPXs and NRF2 are subjected to H2O2 stimulus in OA chondrocytes [24]. Meanwhile, miR-9 is a OA-related effects of oxidative stress in chondrocytes through targets SIRT1 [23].
Finally, we discovered that miRNA-181a-5p expression was increased, and SBP2 protein and GPX1 and GPX4 mRNA expression were reduced in damaged cartilage. These results suggest that hsa-miRNA-181a-5p, GPX1, GPX4 and SBP2 all participate in the OA cartilage damage process to a certain extent. Despite the inadequate number of samples, our peripheral blood data partly support the hypothesis that miR-181a-5p is released in plasma and may facilitate early-stage diagnosis of OA because it induces ROS to damage cartilage proteins. Currently, few blood-based tests are used for the detection of early-stage OA.

Conclusions

We have reported a novel pathway in cartilage. Pro-inflammatory factors mediate miR-181a-5p expression, and then miR-181a-5p regulates the pivotal selenoproteins GPX1 and GPX4 through its target SBP2, resulting in alterations to the overall activity of GPXs, which are the most important oxidation resistance proteins in cartilage. Oxidation resistance involves a series of antioxidants that overcome ROS-related stress to maintain ECM metabolism balance and protect the essential physiological functions of cartilage.

Funding

This work was supported by grants from the National Natural Science Foundation of China (Project No. 81371986, 81772410), the Shaanxi province science and technology project (2016SF-187).

Availability of data and materials

Not applicable.

Authors’ information

Kunzheng Wang, the corresponding author, is the Chairman of Chinese Orthopedic Association.
This study was performed with the approval of the Ethics Committee of the Xi’an Jiaotong University Health Science Center.
All of 10 donors of OA cartilage provided full written informed consent and “Consent to publish” before the operative procedure.
Not applicable.

Competing interests

The authors declare that they have no competing interests.

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Open AccessThis article is distributed under the terms of the Creative Commons Attribution 4.0 International License (http://​creativecommons.​org/​licenses/​by/​4.​0/​), which permits unrestricted use, distribution, and reproduction in any medium, provided you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons license, and indicate if changes were made. The Creative Commons Public Domain Dedication waiver (http://​creativecommons.​org/​publicdomain/​zero/​1.​0/​) applies to the data made available in this article, unless otherwise stated.
Literatur
1.
Zurück zum Zitat Loeser RF, Collins JA, Diekman BO. Ageing and the pathogenesis of osteoarthritis. Nat Rev Rheumatol. 2016;12(7):412–20.CrossRef Loeser RF, Collins JA, Diekman BO. Ageing and the pathogenesis of osteoarthritis. Nat Rev Rheumatol. 2016;12(7):412–20.CrossRef
2.
Zurück zum Zitat Mobasheri A, Rayman MP, Gualillo O, Sellam J, van der Kraan P, Fearon U. The role of metabolism in the pathogenesis of osteoarthritis. Nat Rev Rheumatol. 2017;13(5):302–11.CrossRef Mobasheri A, Rayman MP, Gualillo O, Sellam J, van der Kraan P, Fearon U. The role of metabolism in the pathogenesis of osteoarthritis. Nat Rev Rheumatol. 2017;13(5):302–11.CrossRef
3.
Zurück zum Zitat Kapoor M, Martel-Pelletier J, Lajeunesse D, Pelletier JP, Fahmi H. Role of proinflammatory cytokines in the pathophysiology of osteoarthritis. Nat Rev Rheumatol. 2011;7(1):33–42.CrossRef Kapoor M, Martel-Pelletier J, Lajeunesse D, Pelletier JP, Fahmi H. Role of proinflammatory cytokines in the pathophysiology of osteoarthritis. Nat Rev Rheumatol. 2011;7(1):33–42.CrossRef
4.
Zurück zum Zitat Mitchell PG, Magna HA, Reeves LM, Lopresti-Morrow LL, Yocum SA, Rosner PJ, Geoghegan KF, Hambor JE. Cloning, expression, and type II collagenolytic activity of matrix metalloproteinase-13 from human osteoarthritic cartilage. J Clin Invest. 1996;97(3):761–8.CrossRef Mitchell PG, Magna HA, Reeves LM, Lopresti-Morrow LL, Yocum SA, Rosner PJ, Geoghegan KF, Hambor JE. Cloning, expression, and type II collagenolytic activity of matrix metalloproteinase-13 from human osteoarthritic cartilage. J Clin Invest. 1996;97(3):761–8.CrossRef
5.
Zurück zum Zitat Wang M, Sampson ER, Jin H, Li J, Ke QH, Im HJ, Chen D. MMP13 is a critical target gene during the progression of osteoarthritis. Arthritis Res Ther. 2013;15(1):R5.CrossRef Wang M, Sampson ER, Jin H, Li J, Ke QH, Im HJ, Chen D. MMP13 is a critical target gene during the progression of osteoarthritis. Arthritis Res Ther. 2013;15(1):R5.CrossRef
6.
Zurück zum Zitat Kim JH, Jeon J, Shin M, Won Y, Lee M, Kwak JS, Lee G, Rhee J, Ryu JH, Chun CH, et al. Regulation of the catabolic cascade in osteoarthritis by the zinc-ZIP8-MTF1 axis. Cell. 2014;156(4):730–43.CrossRef Kim JH, Jeon J, Shin M, Won Y, Lee M, Kwak JS, Lee G, Rhee J, Ryu JH, Chun CH, et al. Regulation of the catabolic cascade in osteoarthritis by the zinc-ZIP8-MTF1 axis. Cell. 2014;156(4):730–43.CrossRef
7.
Zurück zum Zitat Corciulo C, Lendhey M, Wilder T, Schoen H, Cornelissen AS, Chang G, Kennedy OD, Cronstein BN. Endogenous adenosine maintains cartilage homeostasis and exogenous adenosine inhibits osteoarthritis progression. Nat Commun. 2017;8:15019.CrossRef Corciulo C, Lendhey M, Wilder T, Schoen H, Cornelissen AS, Chang G, Kennedy OD, Cronstein BN. Endogenous adenosine maintains cartilage homeostasis and exogenous adenosine inhibits osteoarthritis progression. Nat Commun. 2017;8:15019.CrossRef
8.
Zurück zum Zitat Jeon OH, Kim C, Laberge RM, Demaria M, Rathod S, Vasserot AP, Chung JW, Kim DH, Poon Y, David N, et al. Local clearance of senescent cells attenuates the development of post-traumatic osteoarthritis and creates a pro-regenerative environment. Nat Med. 2017;23(6):775–81.CrossRef Jeon OH, Kim C, Laberge RM, Demaria M, Rathod S, Vasserot AP, Chung JW, Kim DH, Poon Y, David N, et al. Local clearance of senescent cells attenuates the development of post-traumatic osteoarthritis and creates a pro-regenerative environment. Nat Med. 2017;23(6):775–81.CrossRef
9.
Zurück zum Zitat Labunskyy VM, Hatfield DL, Gladyshev VN. Selenoproteins: molecular pathways and physiological roles. Physiol Rev. 2014;94(3):739–77.CrossRef Labunskyy VM, Hatfield DL, Gladyshev VN. Selenoproteins: molecular pathways and physiological roles. Physiol Rev. 2014;94(3):739–77.CrossRef
10.
Zurück zum Zitat Kryukov GV, Castellano S, Novoselov SV, Lobanov AV, Zehtab O, Guigo R, Gladyshev VN. Characterization of mammalian selenoproteomes. Science. 2003;300(5624):1439–43.CrossRef Kryukov GV, Castellano S, Novoselov SV, Lobanov AV, Zehtab O, Guigo R, Gladyshev VN. Characterization of mammalian selenoproteomes. Science. 2003;300(5624):1439–43.CrossRef
11.
12.
Zurück zum Zitat Issa R, Boeving M, Kinter M, Griffin TM. Effect of biomechanical stress on endogenous antioxidant networks in bovine articular cartilage. J Orthop Res. 2018;36(2):760–9.PubMed Issa R, Boeving M, Kinter M, Griffin TM. Effect of biomechanical stress on endogenous antioxidant networks in bovine articular cartilage. J Orthop Res. 2018;36(2):760–9.PubMed
13.
Zurück zum Zitat Collins JA, Wood ST, Nelson KJ, Rowe MA, Carlson CS, Chubinskaya S, Poole LB, Furdui CM, Loeser RF. Oxidative stress promotes Peroxiredoxin Hyperoxidation and attenuates pro-survival signaling in aging chondrocytes. J Biol Chem. 2016;291(13):6641–54.CrossRef Collins JA, Wood ST, Nelson KJ, Rowe MA, Carlson CS, Chubinskaya S, Poole LB, Furdui CM, Loeser RF. Oxidative stress promotes Peroxiredoxin Hyperoxidation and attenuates pro-survival signaling in aging chondrocytes. J Biol Chem. 2016;291(13):6641–54.CrossRef
14.
Zurück zum Zitat Driscoll DM, Copeland PR. Mechanism and regulation of selenoprotein synthesis. Annu Rev Nutr. 2003;23:17–40.CrossRef Driscoll DM, Copeland PR. Mechanism and regulation of selenoprotein synthesis. Annu Rev Nutr. 2003;23:17–40.CrossRef
15.
Zurück zum Zitat Takeuchi A, Schmitt D, Chapple C, Babaylova E, Karpova G, Guigo R, Krol A, Allmang C. A short motif in Drosophila SECIS binding protein 2 provides differential binding affinity to SECIS RNA hairpins. Nucleic Acids Res. 2009;37(7):2126–41.CrossRef Takeuchi A, Schmitt D, Chapple C, Babaylova E, Karpova G, Guigo R, Krol A, Allmang C. A short motif in Drosophila SECIS binding protein 2 provides differential binding affinity to SECIS RNA hairpins. Nucleic Acids Res. 2009;37(7):2126–41.CrossRef
16.
Zurück zum Zitat Donovan J, Caban K, Ranaweera R, Gonzalez-Flores JN, Copeland PR. A novel protein domain induces high affinity selenocysteine insertion sequence binding and elongation factor recruitment. J Biol Chem. 2008;283(50):35129–39.CrossRef Donovan J, Caban K, Ranaweera R, Gonzalez-Flores JN, Copeland PR. A novel protein domain induces high affinity selenocysteine insertion sequence binding and elongation factor recruitment. J Biol Chem. 2008;283(50):35129–39.CrossRef
17.
Zurück zum Zitat Downey CM, Horton CR, Carlson BA, Parsons TE, Hatfield DL, Hallgrimsson B, Jirik FR. Osteo-chondroprogenitor-specific deletion of the selenocysteine tRNA gene, Trsp, leads to chondronecrosis and abnormal skeletal development: a putative model for Kashin-Beck disease. PLoS Genet. 2009;5(8):e1000616.CrossRef Downey CM, Horton CR, Carlson BA, Parsons TE, Hatfield DL, Hallgrimsson B, Jirik FR. Osteo-chondroprogenitor-specific deletion of the selenocysteine tRNA gene, Trsp, leads to chondronecrosis and abnormal skeletal development: a putative model for Kashin-Beck disease. PLoS Genet. 2009;5(8):e1000616.CrossRef
18.
Zurück zum Zitat Papp LV, Wang J, Kennedy D, Boucher D, Zhang Y, Gladyshev VN, Singh RN, Khanna KK. Functional characterization of alternatively spliced human SECISBP2 transcript variants. Nucleic Acids Res. 2008;36(22):7192–206.CrossRef Papp LV, Wang J, Kennedy D, Boucher D, Zhang Y, Gladyshev VN, Singh RN, Khanna KK. Functional characterization of alternatively spliced human SECISBP2 transcript variants. Nucleic Acids Res. 2008;36(22):7192–206.CrossRef
19.
Zurück zum Zitat Anestal K, Arner ES. Rapid induction of cell death by selenium-compromised thioredoxin reductase 1 but not by the fully active enzyme containing selenocysteine. J Biol Chem. 2003;278(18):15966–72.CrossRef Anestal K, Arner ES. Rapid induction of cell death by selenium-compromised thioredoxin reductase 1 but not by the fully active enzyme containing selenocysteine. J Biol Chem. 2003;278(18):15966–72.CrossRef
20.
Zurück zum Zitat Vicente R, Noel D, Pers YM, Apparailly F, Jorgensen C. Deregulation and therapeutic potential of microRNAs in arthritic diseases. Nat Rev Rheumatol. 2015. Vicente R, Noel D, Pers YM, Apparailly F, Jorgensen C. Deregulation and therapeutic potential of microRNAs in arthritic diseases. Nat Rev Rheumatol. 2015.
21.
Zurück zum Zitat Nugent M. MicroRNAs: exploring new horizons in osteoarthritis: Osteoarthritis and cartilage / OARS, Osteoarthritis Research Society; 2015. Nugent M. MicroRNAs: exploring new horizons in osteoarthritis: Osteoarthritis and cartilage / OARS, Osteoarthritis Research Society; 2015.
22.
Zurück zum Zitat Nakamura A, Rampersaud YR, Sharma A, Lewis SJ, Wu B, Datta P, Sundararajan K, Endisha H, Rossomacha E, Rockel JS, et al. Identification of microRNA-181a-5p and microRNA-4454 as mediators of facet cartilage degeneration. JCI insight. 2016;1(12):e86820.CrossRef Nakamura A, Rampersaud YR, Sharma A, Lewis SJ, Wu B, Datta P, Sundararajan K, Endisha H, Rossomacha E, Rockel JS, et al. Identification of microRNA-181a-5p and microRNA-4454 as mediators of facet cartilage degeneration. JCI insight. 2016;1(12):e86820.CrossRef
23.
Zurück zum Zitat D'Adamo S, Cetrullo S, Guidotti S, Borzi RM, Flamigni F. Hydroxytyrosol modulates the levels of microRNA-9 and its target sirtuin-1 thereby counteracting oxidative stress-induced chondrocyte death. Osteoarthr Cartil. 2017;25(4):600–10.CrossRef D'Adamo S, Cetrullo S, Guidotti S, Borzi RM, Flamigni F. Hydroxytyrosol modulates the levels of microRNA-9 and its target sirtuin-1 thereby counteracting oxidative stress-induced chondrocyte death. Osteoarthr Cartil. 2017;25(4):600–10.CrossRef
24.
Zurück zum Zitat Cheleschi S, De Palma A, Pascarelli NA, Giordano N, Galeazzi M, Tenti S, Fioravanti A. Could Oxidative Stress Regulate the Expression of MicroRNA-146a and MicroRNA-34a in Human Osteoarthritic Chondrocyte Cultures? Int J Mol Sci. 2017;18(12).CrossRef Cheleschi S, De Palma A, Pascarelli NA, Giordano N, Galeazzi M, Tenti S, Fioravanti A. Could Oxidative Stress Regulate the Expression of MicroRNA-146a and MicroRNA-34a in Human Osteoarthritic Chondrocyte Cultures? Int J Mol Sci. 2017;18(12).CrossRef
25.
Zurück zum Zitat Sun J, Zhong N, Li Q, Min Z, Zhao W, Sun Q, Tian L, Yu H, Shi Q, Zhang F, et al. MicroRNAs of rat articular cartilage at different developmental stages identified by Solexa sequencing. Osteoarthr Cartil. 2011;19(10):1237–45.CrossRef Sun J, Zhong N, Li Q, Min Z, Zhao W, Sun Q, Tian L, Yu H, Shi Q, Zhang F, et al. MicroRNAs of rat articular cartilage at different developmental stages identified by Solexa sequencing. Osteoarthr Cartil. 2011;19(10):1237–45.CrossRef
26.
Zurück zum Zitat Zhong N, Sun J, Min Z, Zhao W, Zhang R, Wang W, Tian J, Tian L, Ma J, Li D, et al. MicroRNA-337 is associated with chondrogenesis through regulating TGFBR2 expression. Osteoarthr Cartil. 2012;20(6):593–602.CrossRef Zhong N, Sun J, Min Z, Zhao W, Zhang R, Wang W, Tian J, Tian L, Ma J, Li D, et al. MicroRNA-337 is associated with chondrogenesis through regulating TGFBR2 expression. Osteoarthr Cartil. 2012;20(6):593–602.CrossRef
27.
Zurück zum Zitat Gabler J, Ruetze M, Kynast KL, Grossner T, Diederichs S, Richter W. Stage-specific miRs in chondrocyte maturation: differentiation-dependent and hypertrophy-related miR clusters and the miR-181 family. Tissue Eng A. 2015;21(23–24):2840–51.CrossRef Gabler J, Ruetze M, Kynast KL, Grossner T, Diederichs S, Richter W. Stage-specific miRs in chondrocyte maturation: differentiation-dependent and hypertrophy-related miR clusters and the miR-181 family. Tissue Eng A. 2015;21(23–24):2840–51.CrossRef
28.
Zurück zum Zitat Sumiyoshi K, Kubota S, Ohgawara T, Kawata K, Abd El Kader T, Nishida T, Ikeda N, Shimo T, Yamashiro T, Takigawa M. Novel role of miR-181a in cartilage metabolism. J Cell Biochem. 2013;114(9):2094–100.CrossRef Sumiyoshi K, Kubota S, Ohgawara T, Kawata K, Abd El Kader T, Nishida T, Ikeda N, Shimo T, Yamashiro T, Takigawa M. Novel role of miR-181a in cartilage metabolism. J Cell Biochem. 2013;114(9):2094–100.CrossRef
29.
Zurück zum Zitat Xu J, Jiang C, Zhu W, Wang B, Yan J, Min Z, Geng M, Han Y, Ning Q, Zhang F, et al. NOD2 pathway via RIPK2 and TBK1 is involved in the aberrant catabolism induced by T-2 toxin in chondrocytes. Osteoarthr Cartil. 2015;23(9):1575–85.CrossRef Xu J, Jiang C, Zhu W, Wang B, Yan J, Min Z, Geng M, Han Y, Ning Q, Zhang F, et al. NOD2 pathway via RIPK2 and TBK1 is involved in the aberrant catabolism induced by T-2 toxin in chondrocytes. Osteoarthr Cartil. 2015;23(9):1575–85.CrossRef
30.
Zurück zum Zitat Wu XF, Zhou ZH, Zou J. MicroRNA-181 inhibits proliferation and promotes apoptosis of chondrocytes in osteoarthritis by targeting PTEN. Biochem Cell Biol. 2017;95(3):437–44.CrossRef Wu XF, Zhou ZH, Zou J. MicroRNA-181 inhibits proliferation and promotes apoptosis of chondrocytes in osteoarthritis by targeting PTEN. Biochem Cell Biol. 2017;95(3):437–44.CrossRef
31.
Zurück zum Zitat Latreche L, Jean-Jean O, Driscoll DM, Chavatte L. Novel structural determinants in human SECIS elements modulate the translational recoding of UGA as selenocysteine. Nucleic Acids Res. 2009;37(17):5868–80.CrossRef Latreche L, Jean-Jean O, Driscoll DM, Chavatte L. Novel structural determinants in human SECIS elements modulate the translational recoding of UGA as selenocysteine. Nucleic Acids Res. 2009;37(17):5868–80.CrossRef
32.
Zurück zum Zitat Touat-Hamici Z, Legrain Y, Bulteau AL, Chavatte L. Selective up-regulation of human selenoproteins in response to oxidative stress. J Biol Chem. 2014;289(21):14750–61.CrossRef Touat-Hamici Z, Legrain Y, Bulteau AL, Chavatte L. Selective up-regulation of human selenoproteins in response to oxidative stress. J Biol Chem. 2014;289(21):14750–61.CrossRef
33.
Zurück zum Zitat Loeser RF, Carlson CS, Del Carlo M, Cole A. Detection of nitrotyrosine in aging and osteoarthritic cartilage: correlation of oxidative damage with the presence of interleukin-1beta and with chondrocyte resistance to insulin-like growth factor 1. Arthritis Rheum. 2002;46(9):2349–57.CrossRef Loeser RF, Carlson CS, Del Carlo M, Cole A. Detection of nitrotyrosine in aging and osteoarthritic cartilage: correlation of oxidative damage with the presence of interleukin-1beta and with chondrocyte resistance to insulin-like growth factor 1. Arthritis Rheum. 2002;46(9):2349–57.CrossRef
34.
Zurück zum Zitat Min Z, Zhao W, Zhong N, Guo Y, Sun M, Wang Q, Zhang R, Yan J, Tian L, Zhang F, et al. Abnormality of epiphyseal plate induced by selenium deficiency diet in two generation DA rats. APMIS. 2015;123(8):697–705.CrossRef Min Z, Zhao W, Zhong N, Guo Y, Sun M, Wang Q, Zhang R, Yan J, Tian L, Zhang F, et al. Abnormality of epiphyseal plate induced by selenium deficiency diet in two generation DA rats. APMIS. 2015;123(8):697–705.CrossRef
35.
Zurück zum Zitat Papp LV, Lu J, Bolderson E, Boucher D, Singh R, Holmgren A, Khanna KK. SECIS-binding protein 2 promotes cell survival by protecting against oxidative stress. Antioxid Redox Signal. 2010;12(7):797–808.CrossRef Papp LV, Lu J, Bolderson E, Boucher D, Singh R, Holmgren A, Khanna KK. SECIS-binding protein 2 promotes cell survival by protecting against oxidative stress. Antioxid Redox Signal. 2010;12(7):797–808.CrossRef
36.
Zurück zum Zitat Schoenmakers E, Agostini M, Mitchell C, Schoenmakers N, Papp L, Rajanayagam O, Padidela R, Ceron-Gutierrez L, Doffinger R, Prevosto C, et al. Mutations in the selenocysteine insertion sequence-binding protein 2 gene lead to a multisystem selenoprotein deficiency disorder in humans. J Clin Invest. 2010;120(12):4220–35.CrossRef Schoenmakers E, Agostini M, Mitchell C, Schoenmakers N, Papp L, Rajanayagam O, Padidela R, Ceron-Gutierrez L, Doffinger R, Prevosto C, et al. Mutations in the selenocysteine insertion sequence-binding protein 2 gene lead to a multisystem selenoprotein deficiency disorder in humans. J Clin Invest. 2010;120(12):4220–35.CrossRef
37.
Zurück zum Zitat Saito Y, Shichiri M, Hamajima T, Ishida N, Mita Y, Nakao S, Hagihara Y, Yoshida Y, Takahashi K, Niki E, et al. Enhancement of lipid peroxidation and its amelioration by vitamin E in a subject with mutations in the SBP2 gene. J Lipid Res. 2015;56(11):2172–82.CrossRef Saito Y, Shichiri M, Hamajima T, Ishida N, Mita Y, Nakao S, Hagihara Y, Yoshida Y, Takahashi K, Niki E, et al. Enhancement of lipid peroxidation and its amelioration by vitamin E in a subject with mutations in the SBP2 gene. J Lipid Res. 2015;56(11):2172–82.CrossRef
Metadaten
Titel
The hsa-miR-181a-5p reduces oxidation resistance by controlling SECISBP2 in osteoarthritis
verfasst von
Jianli Xue
Zixin Min
Zhuqing Xia
Bin Cheng
Binshang Lan
Fujun Zhang
Yan Han
Kunzheng Wang
Jian Sun
Publikationsdatum
01.12.2018
Verlag
BioMed Central
Erschienen in
BMC Musculoskeletal Disorders / Ausgabe 1/2018
Elektronische ISSN: 1471-2474
DOI
https://doi.org/10.1186/s12891-018-2273-6

Weitere Artikel der Ausgabe 1/2018

BMC Musculoskeletal Disorders 1/2018 Zur Ausgabe

Arthropedia

Grundlagenwissen der Arthroskopie und Gelenkchirurgie. Erweitert durch Fallbeispiele, Videos und Abbildungen. 
» Jetzt entdecken

Update Orthopädie und Unfallchirurgie

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert.