Open Access

NR4A1 promotes TNF‑α‑induced chondrocyte death and migration injury via activating the AMPK/Drp1/mitochondrial fission pathway

Retraction in: /10.3892/ijmm.2023.5288

  • Authors:
    • Zhibo Zheng
    • Shuai Xiang
    • Yingjie Wang
    • Yulei Dong
    • Zeng Li
    • Yongbo Xiang
    • Yanyan Bian
    • Bin Feng
    • Bo Yang
    • Xisheng Weng
  • View Affiliations

  • Published online on: November 8, 2019     https://doi.org/10.3892/ijmm.2019.4398
  • Pages: 151-161
  • Copyright: © Zheng et al. This is an open access article distributed under the terms of Creative Commons Attribution License.

Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Nuclear receptor subfamily 4 group A member 1 (NR4A1)‑induced chondrocyte death plays a critical role in the development of osteoarthritis through poorly defined mechanisms. The present study aimed to investigate the role of NR4A1 in regulating chondrocyte death in response to tumor necrosis factor‑α (TNF‑α) and cycloheximide (CHX) treatment, with a focus on mitochondrial fission and the AMP‑activated protein kinase (AMPK) signaling pathway. The results demonstrated that NR4A1 was significantly upregulated in TNF‑α and CHX exposed chondrocytes. Increased NR4A1 triggered mitochondrial fission via the AMPK/dynamin‑related protein 1 (Drp1) pathway, resulting in mitochondrial dysfunction, and mitochondrial permeability transition pore (mPTP) opening‑related cell death. Furthermore, excessive mitochondrial fission impaired chondrocyte migration through imbalance of F‑actin homeostasis. Inhibiting NR4A1 attenuated TNF‑α and CHX‑induced mitochondrial fission and, thus, reduced mitochondrial dysfunction in chondrocytes, mPTP opening‑related cell death and migration injury. Altogether, the present data confirmed that mitochondrial fission was involved in NR4A1‑mediated chondrocyte injury via regulation of mitochondrial dysfunction, mPTP opening‑induced cell death and F‑actin‑related migratory inhibition.

Introduction

One of the hallmarks of osteoarthritis (OA) is the death of chondrocytes (1,2). Chondrocytes, the only cells in the articular cartilage that synthesize chondrocyte matrix, are essential to maintain the normal structure and function of the articular cartilage. A large number of studies have demonstrated an association between chondrocyte death and OA (1,3). On the one hand, loss of chondrocytes induced by inflammatory cytokines reduces the production of chondrocyte matrix and increases cartilage damage (4). Furthermore, intracellular components released by dead chondrocytes increase the generation of reactive oxygen species (ROS) and inflammatory factors, and aggravate the disturbance of the microenvironment in the cartilage (5,6). Previous studies have revealed that tumor necrosis factor-α (TNF-α) is upregulated in OA and contributes to chondrocyte death (7,8). Thus, exploring pertinent molecular mechanisms of chondrocyte death is critical for advancing understanding of OA and in developing novel therapeutic agents against the disease.

As the main organelle of eukaryotic cells, the mitochondria occupy approximately one fifth of cell volume and are widely involved in the regulation of cellular energy metabolism, proliferation and differentiation, aging and death (9). Mitochondrial dysfunction is implicated in the pathological progression of OA via a number of pathways, including oxidative stress, cartilage matrix synthesis and degradation disorder, cytokine mediated inflammatory response activation and chondrocyte death (10,11). Recent studies have highlighted an important role of dynamin-related protein 1 (Drp1)-mediated mitochondrial fission in mitochondrial homeostasis (12,13). Mechanistically, excessive mitochondrial fission contributes to mitochondrial dysfunction as indicated by loss of mitochondrial DNA integrity, reduction in ATP generation, mitochondrial ROS outburst, and loss of mitochondrial membrane potential (13). Furthermore, excessive fission triggers mitochondrial permeability transition pore (mPTP) opening via regulating voltage-dependent anion channel 1 (VDAC1) and hexokinase 2 (HK2), which plays a critical role in cell death (14). However, it remains unclear whether mitochondrial fission contributes to chondrocyte death in OA.

In addition to cellular death, chondrocyte motility deficiency is also an important pathogenic factor in OA (15). Several studies have shown that the balance of F/G-actin plays an important role in regulating cellular migration (16-18), suggesting that mitochondrial fission may regulate cell migration through its function on F/G-actin homeostasis. However, to the best of our knowledge, it is not known whether mitochondrial fission is involved in chondrocyte migration in OA.

The orphan nuclear receptor subfamily 4 group A member 1 (NR4A1), also termed NUR77, TR3, NGFI-B or NAK-1, is involved in the regulation of glucose and lipid metabolism, inflammatory responses and vascular homeostasis (19,20). Previous findings indicated that NR4A1 contributes to chondrocyte death by promoting mitochondrial dysfunction in OA (7,21). However, the underlying mechanism is unclear. Furthermore, NR4A1 has been reported to contribute to mitochondrial dysfunction and cellular death underlying the pathogenesis of non-alcoholic fatty liver disease via regulation of the p53/mitochondrial fission pathway (22). Accordingly, the present study hypothesized that NR4A1 may serve a role in chondrocyte mitochondrial fission and death in OA.

The present results demonstrated that NR4A1 promoted chondrocyte mitochondrial fission via the AMP-activated protein kinase (AMPK)/Drp1 pathway in OA. Furthermore, activation of mitochondrial fission triggered chondrocyte death via mitochondrial function damage and mPTP opening. Excessive mitochondrial fission also reduced chondrocyte migration by the imbalance of F/G-actin homeostasis.

Materials and methods

Cell culture

Primary rat articular chondrocytes were isolated from knee joint cartilage slices of 5-week old male Sprague-Dawley rats (n=40; 130-150 g) using the enzyme dissociation method as described previously (7,23). The rats were housed in an environmentally controlled facility at 23°C, with a 12/12-h light/dark cycle, and provided food and water ad libitum. The animal protocols were approved by the Animal Care and Use Committee of Peking Union Medical College Hospital. Chondrocytes were cultured in DMeM (Gibco; Thermo Fisher Scientific, Inc.) supplemented with 10% fetal bovine serum (HyClone; Ge Healthcare life Sciences) at 37°C with 5% CO2 and 95% air. At 70-80% confluence, chondrocytes were incubated with TNF-α (50 ng/ml; Sigma-Aldrich; Merck KGaA) and cycloheximide (CHX; 10 µg/ml; Sigma-Aldrich; Merck KGaA) to induce cellular death as previously described (24). Chondrocytes were treated with the mitochondrial fission inhibitor mdivi1 (10 mM; Sigma-Aldrich; Merck KGaA) for 12 h at 37°C. Cyclosporine A (10 µM; Sigma-Aldrich; Merck KGaA), an mPTP blocker, was used to pre-treat chondrocytes prior to TNF-α and CHX treatment. Jasplakinolide (2 µM; Abcam; cat. no. ab141409) was used 2 h before TNF-α and CHX treatment to inhibit the F-action degradation at 37°C. AMPK inhibitor (Compound C; 10 µM) or AMPK activator (AICAR; 500 µM; Merck KGaA) were used to pre-treat chondrocytes prior to TNF-α and CHX treatment for 12 h at 37°C.

Western blot analysis

Chondrocytes were lysed in a lysis buffer containing 20 mM Tris (pH 7.4), 150 mM NaCl, 1 mM EDTA, 1 mM EGTA, 1% Triton, 0.1% sodium dodecyl sulfate (SDS) and a cocktail of protease inhibitors at 4°C for 10 min. The protein content of the lysate was measured using the BCA method. Proteins (50 or 100 µg per lane) were resolved by 10% SDS-PAGe and then transferred to a PVDF membrane. The membrane was blocked with 5% non-fat milk for 1 h at room temperature and then incubated with primary antibodies overnight at 4°C against the following: NR4A1 (Cell Signaling Technology, Inc.; cat. no. 5095; 1:1,000), GADPH (Cell Signaling Technology, Inc.; cat. no. 5174; 1:1,000), phosphorylated (phospho)-Drp1 (Ser616) (Cell Signaling Technology, Inc.; cat. no. 3455; 1:1,000), Drp1 (Cell Signaling Technology, Inc.; cat. no. 8570; 1:1,000), complex III subunit core (Cell Signaling Technology, Inc.; cat. no. 459220, 1:1,000), complex II (Abcam; cat. no. ab110410; 1:1,000), complex IV subunit II (Abcam; ab110268; 1:1,000), complex I subunit NDUFB8 (CI-20; Abcam; cat. no. ab110242; 1:1,000), G-actin (Abcam; cat. no. ab123034; 1:1,000), F-actin (Abcam; cat. no. ab205; 1:1,000), AMPK (Abcam; cat. no. ab131512; 1:1,000) and phoshpo-AMPK (Abcam; cat. no. ab23875; 1:1,000). Secondary antibodies including horseradish peroxidase (HRP)-conjugated anti-mouse IgG (1:1,000; cat. no. 14709) and HRP-conjugated anti-rabbit IgG (1:1,000; cat. no. 14708) were purchased form Cell Signaling Technology, Inc. The membrane was incubated with secondary antibodies for 1 h at room temperature. The blots were detected with an enhanced chemiluminescence substrate kit (Thermo Fisher Scientific, Inc.), and band intensity levels were analyzed using Quantity One 4.6 software (Bio-Rad laboratories, Inc.).

Cell transfection

NR4A1-specific siRNA (5′-CCA AGT ACA TCT GCC TGG CAA ACA A-3′) and scrambled control siRNA (5′-UUC UCC GAA CGU GUC ACA TGA UGU-3′) were synthesized by RiboBio (Guangzhou, China). A total of 20 nM siNR4A1 or control siRNA was used to transfect chondrocytes cells (2×106 cells/well) with lipofectamine® 2000 (Thermo Fisher Scientific, Inc.) for 48 h in 6-well plates, and the transfection efficiency was determined by western blotting.

Flow cytometry

Cell viability was measured by MTT assay (MTT cell proliferation and cytotoxicity detection kit; cat. no. C0009; Beyotime Institute of Biotechnology) and lactate dehydrogenase (LDH) release assay (LDH cytotoxicity assay kit; cat. no. C0016; Beyotime Institute of Biotechnology) as previously described (25). Cell viability was also measured by Annexin V/propidium iodide and calcein AM/ethidium homodimer-1 (ethD-1) flow cytometric analysis. For calcein AM/ethD-1 flow cytometry, cells were seeded in 6-well cell-culture plates at a density of 106/well and incubated at 37°C for 24 h. Following TNF-α and CHX treatments, cells were collected and washed with PBS three times. Subsequently, the samples were simultaneously stained with 5 µl EthD-1 (2 mM) and 2 µl calcein AM working solution in the dark for 15-20 min with the lIVe/DeAD Viability/Cytotoxicity kit (cat. no. l3224; Molecular Probes; Thermo Fisher Scientific, Inc.). The stained cells were subsequently analyzed using a BD FACS-Calibur cytometer (BD Biosciences). The data were analyzed by FlowJo software (version 7.6.5; FlowJo llC).

Mitochondrial membrane potential measurements, mPTP opening detection and ATP measurements

Mitochondrial membrane potential (ΔΨm) was measured using a mitochondrial membrane potential assay kit with JC-1 (cat. no. C2006; Beyotime Institute of Biotechnology), according to the manufacturer's protocol. Images were captured under a fluorescence microscope (Olympus Corporation) and analyzed with Image-Pro Plus 6.0 (Media Cybernetics, Inc.). In addition, mPTP opening was detected as a rapid dissipation of tetramethylrhodamine ethylester fluorescence as previously described (25). Cellular ATP levels were measured using an ATP assay kit (cat. no. S0026; Beyotime Institute of Biotechnology), according to the manufacturer's protocol.

Immunofluorescence staining

Cells were fixed with 4% paraformaldehyde for 15 min at room temperature and then washed three times with PBS. Following blocking with 5% bovine serum albumin (Sigma-Aldrich; Merck KGaA) in PBS for 1 h at room temperature, the cells were then incubated with primary antibodies against F-actin (Abcam; cat. no. ab205; 1:500) and TOM20 (Cell Signaling Technology, Inc.; cat. no. 42406; 1:500) at 4°C overnight. After three washes with PBS, the cells were stained with secondary antibodies at 37°C for 1 h. The Alexa Fluor® secondary antibodies, anti-mouse IgG (1:500; cat. no. 4408) and anti-rabbit IgG (1:500; cat. no. 4412), were purchased from Cell Signaling Technology, Inc. DAPI (5 mg/ml; Sigma-Aldrich; Merck KGaA) was used to stain the nucleus at room temperature for 3 min. Furthermore, MitoSOX red mitochondrial superoxide indicator (Molecular Probes; Thermo Fisher Scientific, Inc.) was used to identify mitochondria-reactive oxygen species (mROS). The cells were incubated with MitoSOX (25 µM) in PBS at 37°C for 30 min. Images were captured using a laser confocal microscope (magnification, ×600; TcS SP5; leica Microsystems, Inc.).

Mitochondrial DNA (mtDNA) strand breaks, copy numbers and transcription level detection

mtDNA strand breaks were detected. Briefly, a 200-µl cell (1×106) suspension was centrifuged at 15,000 × g at 4°C for 20 min. The supernatant was discarded and the cells were suspended in a 400 µl solution (0.25 mmol/l inositol, 10 mmol/l Na3PO4 and 1 mmol/l MgCl2; pH 7.2) at 4°C for 30 min. The relative amounts of mtDNA and nuclear DNA content were used to assess the mtDNA copy numbers via quantitative polymerase chain reaction. The mtDNA and nuclear amplicons were generated from a complex IV segment and GAPDH segment, respectively. The mtDNA primer sequences were as follows: Forward, 5′-CTA TGT CGT GTC CAG AG-3′; and reverse, 5′-CAT GTT GTC CCG TGT CAT G-3′. The GAPDH primers, chosen as the internal standards, were as follows: Forward, 5′-CTC AGT CGT ATT CGA GTG GTC CT-3′; and reverse, 5′-CCT GTG GAA GTC CAC AAC ATG TC-3′. The transcript level of mtDNA was reflected by two different components: NADH dehydrogenase subunit 1 (ND1) and cytochrome c oxidase subunit I. The primers for cytochrome c oxidase subunit I were: Forward, 5′-ATC GTT CGG TGA GGT CGT G-3′; and reverse, 5′-CGC CGG TGT CAT TAT CGT ATA-3′. The primers for ND1 were: Forward, 5′-TTG CCG TAT ATT CAG TAT C-3′; and reverse, 5′-ATC CTG TTG CCC AGT CCA GT-3′.

Assays for respiratory chain complex activities

ETCx activities were analyzed via ELISA according to the manufacturer's protocols. The ELISA assay kits for ETCx I, II, and V were purchased from Beyotime Institute of Biotechnology (cat. nos. S0052, S0101 and S0052, respectively). Mitochondrial respiratory function was measured by polarography at 30°C using a Biological Oxyge Monitor system (Hansatech Instruments, Ltd.) and a Clarktype oxygen electrode (Hansatech DW1; Hansatech Instruments, ltd.). Mitochondrial respiration was initiated by adding glutamate/malate to a final concentration of 5 and 2.5 mmol/l for 5 min, respectively. State 3 respiration was initiated by adding ADP (150 nmol/l) for 5 min; state 4 was measured as the rate of oxygen consumption after ADP phosphorylation. The respiratory control ratio (state 3/state 4) and the ADP/O ratio (number of nmol ADP phosphorylated to atoms of oxygen consumed) were calculated.

Transwell assays

Chondrocyte migration was evaluated using 24-well Transwell chambers (Corning, Inc.). First, 1×105 chondrocytes were seeded in the upper chamber containing serum-free DMEM. The chemotactic agent stromal cell-derived factor 1 (100 ng/ml; Sigma-Aldrich; Merck KGaA) together with DMEM supplemented with 10% FBS was added to the lower chamber to induce chondrocyte migration. After a 24-h incubation at 37°C, non-migrating cells in the upper chamber were carefully removed using a cotton swab, cells that traversed through the membrane were fixed in methanol and stained with 0.05% crystal violet at room temperature. The cellular migration was imaged under a light microscope (magnification, ×100; leica Microsystems, Inc.).

Statistical analysis

All analyses were performed with SPSS 20.0 software (IBM Corp.). All experiments were performed at least three times independently. Data are presented as the mean ± standard deviation and statistical significance for each variable was estimated by a one-way analysis of variance followed by Tukey's test for post-hoc analysis. P<0.05 was considered to indicate a statistically significant difference.

Results

NR4A1 upregulation is associated with chondrocyte death under TNF-α and CHX treatment

Chondrocyte death was induced by treatment with TNF-α (50 ng/ml) plus CHX (10 µg/ml) as previously described (7). TNF-α and CHX treatment significantly decreased the viability of chondrocytes compared with the control group (Fig. 1A). In addition, western blot analysis were used to detect NR4A1 expression before and after TNF-α and CHX treatment. As presented in Fig. 1B and C, NR4A1 expression was significantly increased by TNF-α and CHX treatment, suggesting that NR4A1 upregulation was associated with chondrocyte death. TNF-α and CHX treatment for 24 h was used in the subsequent experiments, as the expression of NR4A1 was the highest at 24 h after treatment. To investigate the role of NR4A1 in TNF-α and CHX-induced chondrocyte death, chondrocytes were transfected with siRNA against NR4A1. Transfection efficiency was confirmed via western blot analysis (Fig. 1D and E), and cellular viability was measured following transfection (Fig. 1F). NR4A1-knockdown had no effect on chondrocyte viability. NR4A1 downregulation significantly decreased TNF-α and CHX-induced chondrocyte death as indicated by reduced LDH release (Fig. 1G), increased cell viability (Fig. 1H) and fewer Annexin V/PI cells (Fig. 1I and J). Together, these results indicate that NR4A1 upregulation contributes to chondrocyte death under TNF-α and CHX treatment.

NR4A1 upregulation promotes chondrocyte death via activating mitochondrial fission/mPTP opening

To determine the underlying mechanism whereby NR4A1 triggers chondrocyte death, the present study focused on changes of mPTP opening, as previous studies have reported mPTP opening as an upstream trigger of cellular death (25,26). Compared with the control group, TNF-α and CHX significantly increased mPTP opening time and dissipated the mitochondrial membrane potential, which was reversed by NR4A1-knockdown (Fig. 2A-C). Furthermore, cyclosporine A promoted chondrocyte survival compared with the TNF-α and CHX group, indicating that mPTP opening contributed to NR4A1-medicated chondrocyte death (Fig. 2D).

Mitochondrial fission has been reported as an important activator of mPTP opening and contributes to NR4A1-mediated cell death (20,27). Thus, the present study examined whether mitochondrial fission was involved in NR4A1-induced mPTP opening and cellular death under TNF-α and CHX treatment. To answer this question, mdivi1, an inhibitor of mitochondrial fission, was used in chondrocytes as the negative control group. As presented in Fig. 2F, a higher number of mitochondrial fragments were observed in the TNF-α and CHX group, but not the siNR4A1 group. mdivi1 markedly reduced the number of mitochondrial fragments after TNF-α and CHX treatment (Fig. 2F). Similar results were observed in terms of the mitochondrial length (Fig. 2e). Furthermore, inhibiting mitochondrial fission also significantly reduced TNF-α and CHX-triggered mPTP opening (Fig. 2G) and cell death as indicated by LDH release (Fig. 2H) and calcein AM/ethD-1 flowcytometric analysis (Fig. 2I and J), which is consistent with NR4A1 deficiency. Collectively, these results confirmed that NR4A1 promoted mPTP opening-related cell death via activating mitochondrial fission under TNF-α and CHX stimulation.

Mitochondrial fission activation results in mitochondrial energy metabolism disorder

Besides regulating cell death, the mitochondria are also an important organelle for regulating cell energy metabolism, which has been reported to be involved in the pathological progression of OA (28,29). Thus, the present study investigated whether NR4A1 contributes to mitochondrial dysfunction in chondrocytes after TNF-α and CHX treatment. The results demonstrated that TNF-α and CHX reduced the amount of double-stranded mtDNA breaks, mtDNA copy number and mtDNA transcript levels (Fig. 3A-C). Furthermore, TNF-α and CHX inhibited the content and activity of the electron transport chain complexes (eTCx), which were coupled to state 3/4 respiratory and ATP generation (Fig. 3e-I). However, these changes were reversed by NR4A1-knockdown and application of mdivi1. Furthermore, compared with the TNF-α and CHX group, NR4A1-knockdown and application of mdivi1 also inhibited mitochondrial ROS production under TNF-α and CHX treatment (Fig. 3J). These results indicated that NR4A1 deficiency protects mitochondrial function via fission in chondrocytes after TNF-α and CHX treatment.

Excessive mitochondrial fission inhibits cellular migration via an imbalance of F-actin homeostasis

Chondrocyte motility deficiency also contributes to OA progression (15,30). However, the effect of mitochondrial fission on chondrocyte migration is unclear. To address this question, the present study first observed changes of chondrocyte migration under TNF-α and CHX treatment. As presented in Fig. 4A, TNF-α and CHX markedly decreased chondrocyte migration compared with the control group. However, loss of NR4A1 increased chondrocyte migration, which is similar to the effect of mdivi1, suggesting that mitochondrial fission facilitates TNF-α and CHX-triggered inhibition of chondrocyte migration. Given that F-actin plays an important role in the regulation of cell migration, the present study was interested in whether F-actin homeostasis is implicated in inhibition of chondrocyte migration. Fluorescence microscopy was used to observe F-actin changes. As shown in Fig. 4B, TNF-α and CHX markedly reduced the fluorescence intensity of F-actin. Because F-actin is composed of G-actin, western blot analysis were performed to examine the amount of F- and G-actin to confirm the imbalance of F-actin homeostasis. TNF-α and CHX significantly reduced the expression of F-actin and increased the expression of G-actin compared with the control group (Fig. 4C-E). However, inhibiting F-actin degradation by jasplakinolide promoted cell migration (Fig. 4A) and maintained F-actin homeostasis (Fig. 4B-E), which are similar to the results following mdivi1 treatment and NR4A1-knockdown. This verified the hypothesis that NR4A1 induced mitochondrial fission-triggered imbalance of F-actin homeostasis and contributed to impaired chondrocyte migration under TNF-α and CHX stimulation.

NR4A1 regulates mitochondrial fission via the AMPK/Drp1 pathway

What remains unclear is how NR4A1 evokes mitochondrial fission. Several studies have argued that the AMPK pathway plays a critical role in Drp1 phosphorylation activation and mitochondrial fission (31,32). Thus, the present study investigated whether the AMPK pathway was implicated in NR4A1-induced mitochondrial fission. To answer this question, AICAR, an activator of the AMPK pathway, was used as the positive control and compound C, an inhibitor of the AMPK pathway, was used as the negative control. The results indicated that TNF-α and CHX inhibited activation of the AMPK pathway as indicated by significant down-regulated phospho-AMPK expression, which was blocked by NR4A1-knockdown (Fig. 5A and B). levels of AMP and ATP were also measured to confirm the activation of the AMPK pathway (Fig. 5F). To explore the role of AMPK in mitochondrial fission activation, Drp1 post-was assessed by western blot analysis. As presented in Fig. 5E, TNF-α and CHX increased the expression of phospho-Drp1 (Ser616), which was accompanied with increased Drp1 accumulation in the mitochondria (Fig. 5D) and reduced Dpr1 expression in the cytoplasm (Fig. 5C). However, these changes were reversed by NR4A1-knockdown, which is similar to application of AICAR. Furthermore, inhibiting AMPK by compound c abolished the protective effect of NR4A1-knockdown. Together, these results revealed that NR4A1 activated mitochondrial fission via the APMK pathway.

Discussion

Inhibiting chondrocyte death has been considered as an important treatment option for OA (1,2). Accumulating evidence has indicated that NR4A1 plays a critical role in the regulation of chondrocytes death (7,21). However, the underlying mechanism remains unclear. The present study confirmed that NR4A1 upregulation by TNF-α and CHX contributes to chondrocyte death in OA. Mechanically, upregulated NR4A1 induced mitochondrial fission via the AMPK/Drp1 pathway. Excessive mitochondrial fission exacerbated chondrocyte death by promoting mPTP opening. Mitochondrial fission also impaired cellular migration via an imbalance of F/G-actin homeostasis. To the best of our knowledge, this is the first study to explore the role of NR4A1 in OA together with Drp1-related mitochondrial fission, mPTP opening, F/G-actin-associated migration and AMPK signaling. Nevertheless, these findings are based on in vitro studies and additional studies using NR4A1−/− mice or rats will shed further light on the role of NR4A1 in vivo.

A number of studies have focused on the mechanism of chondrocyte death in OA due to the critical role of chondrocyte survival in OA treatment (1-3). Different types of chondrocyte death, including apoptosis (33) and necroptosis (24), have been shown to be involved in TNF-α-induced chondrocyte death. Therefore, searching for common upstream pathways of apoptosis and necroptosis is essential to improve chondrocyte survival and OA treatment. mPTP opening has been regarded as an important upstream regulator of both apoptosis (34,35) and necroptosis (26,36,37). The present study confirmed that inhibition of mPTP promoted chondrocyte survival under TNF-α and CHX treatment as indicated by MTT assay, LDH release, Annexin V/propidium iodide and calcein AM/ethD-1 flow cytometric analyses. Further study is needed on the role of mPTP opening in inhibiting chondrocyte apoptosis and necroptosis.

Mitochondrial fission (14), calcium overload (38), ROS (39), mROS (25) and cyclophilin D (40) have been reported to participate in mPTP opening. excessive mitochondrial fission triggers mPTP opening by promoting the dissociation of VDAC1 and HK2 on the mitochondrial outer membrane (14). The present study confirmed that NR4A1 promoted mPTP opening by evoking mitochondrial fission, which is similar to previous studies. It remains unclear whether other mPTP opening-activators contribute to NR4A1-induced mPTP opening and chondrocyte death in OA. Further experimental evidence is needed.

During mitochondrial fission, Drp1 is an important regulator of fission and the ring structure formed by Drp1 on the mitochondrial outer membrane is a key step of mitochondrial division (41). The present study confirmed that TNF-α and CHX increased the expression of phospho-Drp1 (Ser616), accompanied with increased Drp1 accumulation in the mitochondria and reduced Dpr1 expression in the cytoplasm. Furthermore, Drp1 recruitment on the mitochondria requires its corresponding receptors located on the mitochondrial outer membrane. Accumulating evidence has indicated that four proteins are involved in fission, Fis1, Mff, MiD49 and MiD51 (42,43). Further studies are required to determine which receptor participates in mitochondrial fission in chondrocytes.

In the present study, it was confirmed that overexpression of NR4A1 is involved in mitochondrial function damage via activating excessive mitochondrial fission, which damaged the structure and function of mitochondrial DNA and was responsible for the expression of ETCx, as evidenced by lower ATP generation. Consistent with the present findings, a previous study has reported that NR4A1 functions in the dissipation of mitochondrial membrane potential, cellular energy disorder and irreversible cell death via the mPTP complex ANT1-VDAC1 pathway (44).

Furthermore, the present study confirmed that excessive mitochondrial fission decreased chondrocyte migration via disruption of F-actin homeostasis. It is understood that F-actin is an important factor affecting cellular motility. In mitochondrial fission, F-actin participates in mitochondrial fission by breaking down into G-actin and then interacting with Drp1 and its receptors on the mitochondria outer member to promote the formation of a contractile ring (45). Therefore, we hypothesize that excessive mitochondrial division disrupts the F-actin balance by consuming a large amount of F-actin, which ultimately decreases cellular motility. Inhibiting mitochondrial fission protects chondrocyte mobilization under treatment of TNF-α and CHX, which is consistent with the effect of NR4A1-knockdown and jasplakinolide.

The current study has provided the first piece of evidence indicating that NR4A1 triggers Drp1-mitochondrial fission via the AMPK pathway. The effect of AMPK on mitochondrial fission remains controversial. A previous study has reported that AMPK promotes mitochondrial fission by activating Mff (46). However, other studies have suggested that AMPK inhibits mitochondrial fission by reducing the expression of drp1 in aortic endothelial cells (47). The present study demonstrated that AMPK inhibited mitochondrial fission by reducing phospho-Drp1 (Ser616) in chondrocytes under treatment of TNF-α and CHX.

In conclusion, the present study has demonstrated that NR4A1 upregulation promotes mitochondrial fission via the AMPK pathway (Fig. 6). excessive mitochondrial fragments contribute to mitochondrial dysfunction, mPTP opening and F-actin homeostasis, which finally induces chondrocyte death and impairs migration. These findings support the conclusion that NR4A1 may be an attractive therapeutic target in OA.

Acknowledgments

Not applicable.

Funding

This study was financially supported by a grant from the National Key Research and Development Program (grant. no. 2016YFA0101003).

Availability of data and materials

The datasets generated and/or analyzed during the current study are not publicly available, but data may be available from the corresponding author upon reasonable request.

Authors' contributions

ZZ and SX conceived and designed the study. YD, ZL, YW and YX analyzed and interpreted the data. ZZ and SX drafted the manuscript. BY and XW performed the experiments and critically revised the manuscript for important intellectual content. YB and BF performed statistical analysis. XW and YB obtained the funding. YD, ZL, YW and YX collected and assembled the data. All authors approved the final manuscript.

Ethics approval and consent to participate

This study was performed in accordance with the National Institutes of Health guidelines for the use of experimental animals. The protocols were approved by the Animal Care and Use Committee of Peking Union Medical College Hospital (Beijing, China).

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

References

1 

Liu Y, Lin L, Zou R, Wen C, Wang Z and Lin F: MSC-derived exosomes promote proliferation and inhibit apoptosis of chondrocytes via lncRNA-KlF3-AS1/miR-206/GIT1 axis in osteoarthritis. Cell Cycle. 17:2411–2422. 2018. View Article : Google Scholar : PubMed/NCBI

2 

Song E K, Jeon J, Jang DG, Kim HE, Sim HJ, Kwon KY, Medina-Ruiz S, Jang HJ, Lee AR, Rho JG, et al: ITGBL1 modulates integrin activity to promote cartilage formation and protect against arthritis. Sci Transl Med. 10:eaam74862018. View Article : Google Scholar : PubMed/NCBI

3 

Scotece M, Conde J, Abella V, López V, Francisco V, Ruiz C, Campos V, Lago F, Gomez R, Pino J and Gualillo O: Oleocanthal inhibits catabolic and inflammatory mediators in lPS-activated human primary osteoarthritis (OA) chondrocytes through MAPKs/NF-kB pathways. Cell Physiol Biochem. 49:2414–2426. 2018. View Article : Google Scholar

4 

Cheleschi S, Fioravanti A, De Palma A, Corallo C, Franci D, Volpi N, Bedogni G, Giannotti S and Giordano N: Methylsulfonylmethane and mobilee prevent negative effect of IL-1β in human chondrocyte cultures via NF-kB signaling pathway. Int Immunopharmacol. 65:129–139. 2018. View Article : Google Scholar : PubMed/NCBI

5 

Kojima H and Mori C: Minocycline treatment of genital infections caused by Chlamydia trachomatis (C. trachomatis). Kansenshogaku Zasshi. 59:824–830. 1985.In Japanese. View Article : Google Scholar : PubMed/NCBI

6 

Polzer K, Schett G and Zwerina J: The lonely death: Chondrocyte apoptosis in TNF-induced arthritis. Autoimmunity. 40:333–336. 2007. View Article : Google Scholar : PubMed/NCBI

7 

Shi X, Ye H, Yao X and Gao Y: The involvement and possible mechanism of NR4A1 in chondrocyte apoptosis during osteoarthritis. Am J Transl Res. 9:746–754. 2017.PubMed/NCBI

8 

Heidari F, Bahari A, Amarlou A and Fakheri BA: Fumaric acids as a novel antagonist of TLR-4 pathway mitigates arsenic-exposed inflammation in human monocyte-derived dendritic cells. Immunopharmacol Immunotoxicol. 41:513–520. 2019. View Article : Google Scholar : PubMed/NCBI

9 

Chowdhury A, Aich A, Jain G, Wozny K, Lüchtenborg C, Hartmann M, Bernhard O, Balleiniger M, Alfar EA, Zieseniss A, et al: Defective mitochondrial cardiolipin remodeling dampens HIF-1α expression in hypoxia. Cell Rep. 25:561–570.e6. 2018. View Article : Google Scholar

10 

Blanco FJ, Valdes AM and Rego-Perez I: Mitochondrial DNA variation and the pathogenesis of osteoarthritis phenotypes. Nat Rev Rheumatol. 14:327–340. 2018. View Article : Google Scholar : PubMed/NCBI

11 

Lopez-Armada MJ, Carames B, Martin MA, Cillero-Pastor B, Lires-Dean M, Fuentes-Boquete I, Arenas J and Blanco FJ: Mitochondrial activity is modulated by TNFalpha and IL-1beta in normal human chondrocyte cells. Osteoarthritis Cartilage. 14:1011–1022. 2006. View Article : Google Scholar : PubMed/NCBI

12 

Kurihara Y, Itoh R, Shimizu A, Walenna NF, Chou B, Ishii K, Soejima T, Fujikane A and Hiromatsu K: Chlamydia trachomatis targets mitochondrial dynamics to promote intracellular survival and proliferation. Cell Microbiol. 21:e129622018. View Article : Google Scholar : PubMed/NCBI

13 

Singh M, Denny H, Smith C, Granados J and Renden R: Presynaptic loss of dynamin related protein 1 impairs synaptic vesicle release and recycling at the mouse calyx of held. J Physiol. 596:6263–6287. 2018. View Article : Google Scholar : PubMed/NCBI

14 

Zhou H, Zhang Y, Hu S, Shi C, Zhu P, Ma Q, Jin Q, Cao F, Tian F and Chen Y: Melatonin protects cardiac microvasculature against ischemia/reperfusion injury via suppression of mitochondrial fiss ion-VDAC1-HK2-mPTP-mitophagy axis. J Pineal Res. 63:2017. View Article : Google Scholar

15 

Ye D, Jian W, Feng J and Liao X: Role of long noncoding RNA ZFAS1 in proliferation, apoptosis and migration of chondrocytes in osteoarthritis. Biomed Pharmacother. 104:825–831. 2018. View Article : Google Scholar : PubMed/NCBI

16 

Xie L, Li LY, Zheng D, Xie YM, Xu XE, Tao LH, Liao LD, Xie YH, Cheng YW, Xu LY and Li EM: F806 Suppresses the invasion and metastasis of esophageal squamous cell carcinoma via downregulating F-actin assembly-related rho family proteins. Biomed Res Int. 2018:20493132018. View Article : Google Scholar : PubMed/NCBI

17 

Zhang W, Liu K, Pei Y, Ma J, Tan J and Zhao J: Mst1 regulates non-small cell lung cancer A549 cell apoptosis by inducing mitochondrial damage via ROCK1/F-actin pathways. Int J Oncol. 53:2409–2422. 2018.PubMed/NCBI

18 

D'Agostino G, Saporito A, Cecchinato V, Silvestri Y, Borgeat A, Anselmi L and Uguccioni M: Lidocaine inhibits cyto-skeletal remodelling and human breast cancer cell migration. Br J Anaesth. 121:962–968. 2018. View Article : Google Scholar : PubMed/NCBI

19 

Wu L, Amarachintha S, Xu J, Oley F Jr and Du W: Mesenchymal COX2-PG secretome engages NR4A-WNT signalling axis in haematopoietic progenitors to suppress anti-leukaemia immunity. Br J Haematol. 183:445–456. 2018. View Article : Google Scholar : PubMed/NCBI

20 

Koenis DS, Medzikovic L, van Loenen PB, van Weeghel M, Huveneers S, Vos M, Evers-van Gogh IJ, Van den Bossche J, Speijer D, Kim Y, et al: Nuclear receptor Nur77 limits the macrophage inflammatory response through transcriptional reprogramming of mitochondrial metabolism. Cell Rep. 24:2127–2140.e7. 2018. View Article : Google Scholar : PubMed/NCBI

21 

Ryan SM, McMorrow J, Umerska A, Patel HB, Kornerup KN, Tajber L, Murphy EP, Perretti M, Corrigan OI and Brayden DJ: An intra-articular salmon calcitonin-based nanocomplex reduces experimental inflammatory arthritis. J Control Release. 167:120–129. 2013. View Article : Google Scholar : PubMed/NCBI

22 

Zhou H, Du W, Li Y, Shi C, Hu N, Ma S, Wang W and Ren J: Effects of melatonin on fatty liver disease: The role of NR4A1/DNA-PKcs/p53 pathway, mitochondrial fission, and mitophagy. J Pineal Res. 64:2018. View Article : Google Scholar

23 

Liu Z, Cai H, Zheng X, Zhang B and Xia C: The involvement of mutual inhibition of eRK and mTOR in PlCg1-mediated MMP-13 expression in human osteoarthritis chondrocytes. Int J Mol Sci. 16:17857–17869. 2015. View Article : Google Scholar : PubMed/NCBI

24 

Lee SW, Rho JH, Lee SY, Kim JH, Cheong JH, Kim HY, Jeong NY, Chung WT and Yoo YH: Leptin protects rat articular chondrocytes from cytotoxicity induced by TNF-α in the presence of cyclohexamide. Osteoarthritis Cartilage. 23:2269–2278. 2015. View Article : Google Scholar : PubMed/NCBI

25 

Zhou H, Hu S, Jin Q, Shi C, Zhang Y, Zhu P, Ma Q, Tian F and Chen Y: Mff-dependent mitochondrial fission contributes to the pathogenesis of cardiac microvasculature ischemia/reperfusion injury via induction of mROS-mediated cardiolipin oxidation andHK2/VDAC1 disassociation-involved mPTP opening. J Am Heart Assoc. 6:e0053282017. View Article : Google Scholar

26 

Zhu P, Hu S, Jin Q, Li D, Tian F, Toan S, Li Y, Zhou H and Chen Y: Ripk3 promotes ER stress-induced necroptosis in cardiac IR injury: A mechanism involving calcium overload/XO/ROS/mPTP pathway. Redox Biol. 16:157–168. 2018. View Article : Google Scholar : PubMed/NCBI

27 

Jeanneteau F, Barrere C, Vos M, De Vries CJM, Rouillard C, Levesque D, Dromard Y, Moisan MP, Duric V, Franklin TC, et al: The stress-induced transcription factor NR4A1 adjusts mitochondrial function and synapse number in prefrontal cortex. J Neurosci. 38:1335–1350. 2018. View Article : Google Scholar : PubMed/NCBI

28 

Yao X, Zhang J, Jing X, Ye Y, Guo J, Sun K and Guo F: Fibroblast growth factor 18 exerts anti-osteoarthritic effects through PI3K-AKT signaling and mitochondrial fusion and fission. Pharmacol Res. 139:314–324. 2018. View Article : Google Scholar : PubMed/NCBI

29 

Bolduc JA, Collins JA and Loeser RF: Reactive oxygen species, aging and articular cartilage homeostasis. Free Radic Biol Med. 132:73–82. 2019. View Article : Google Scholar

30 

Tsuyuguchi Y, Nakasa T, Ishikawa M, Miyaki S, Matsushita R, Kanemitsu M and Adachi N: The benefit of minced cartilage over isolated chondrocytes in atelocollagen gel on chondrocyte proliferation and migration. Cartilage. 19476035188052052018.Epub ahead of print. PubMed/NCBI

31 

Liu J, Yan W, Zhao X, Jia Q, Wang J, Zhang H, Liu C, He K and Sun Z: Sirt3 attenuates post-infarction cardiac injury via inhibiting mitochondrial fission and normalization of AMPK-Drp1 pathways. Cell Signal. 53:1–13. 2019. View Article : Google Scholar

32 

Jeong HY, Kang JM, Jun HH, Kim DJ, Park SH, Sung MJ, Heo JH, Yang DH, Lee SH and Lee SY: Chloroquine and amodiaquine enhance AMPK phosphorylation and improve mitochondrial fragmentation in diabetic tubulopathy. Sci Rep. 8:87742018. View Article : Google Scholar : PubMed/NCBI

33 

Lepetsos P, Papavassiliou KA and Papavassiliou AG: Redox and NF-kB signaling in osteoarthritis. Free Radic Biol Med. 132:90–100. 2019. View Article : Google Scholar

34 

Jang Y, Kwon I, Song W, Cosio-Lima LM, Taylor S and Lee Y: Modulation of mitochondrial phenotypes by endurance exercise contributes to neuroprotection against a MPTP-induced animal model of PD. Life Sci. 209:455–465. 2018. View Article : Google Scholar : PubMed/NCBI

35 

Alizadeh AM, Faghihi M, Khori V, Sohanaki H, Pourkhalili K, Mohammadghasemi F and Mohsenikia M: Oxytocin protects cardiomyocytes from apoptosis induced by ischemia-reperfusion in rat heart: Role of mitochondrial ATP-dependent potassium channel and permeability transition pore. Peptides. 36:71–77. 2012. View Article : Google Scholar : PubMed/NCBI

36 

Zhou H, Li D, Zhu P, Ma Q, Toan S, Wang J, Hu S, Chen Y and Zhang Y: Inhibitory effect of melatonin on necroptosis via repressing the Ripk3-PGAM5-CypD-mPTP pathway attenuates cardiac microvascular ischemia-reperfusion injury. J Pineal Res. 65:e125032018. View Article : Google Scholar : PubMed/NCBI

37 

Zhang T, Zhang Y, Cui M, Jin L, Wang Y, Lv F, Liu Y, Zheng W, Shang H, Zhang J, et al: CaMKII is a RIP3 substrate mediating ischemia- and oxidative stress-induced myocardial necroptosis. Nat Med. 22:175–182. 2016. View Article : Google Scholar : PubMed/NCBI

38 

Wu KC, Liou HH, Lee CY and Lin CJ: Down-regulation of natural resistance-associated macrophage protein-1 (Nramp1) is associated with 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP)/1-methyl-4-phenylpyridinium (MPP+)-induced α-synuclein accumulation and neurotoxicity. Neuropathol Appl Neurobiol. 45:157–173. 2019. View Article : Google Scholar

39 

Kar D and Bandyopadhyay A: Targeting peroxisome proliferator activated receptor α (PPAR α) for the prevention of mitochondrial impairment and hypertrophy in cardiomyocytes. Cell Physiol Biochem. 49:245–259. 2018. View Article : Google Scholar

40 

Zhang R, Li G, Zhang Q, Tang Q, Huang J, Hu C, Liu Y, Wang Q, Liu W, Gao N and Zhou S: Hirsutine induces mPTP-dependent apoptosis through ROCK1/PTeN/PI3K/GSK3β pathway in human lung cancer cells. Cell Death Dis. 9:5982018. View Article : Google Scholar

41 

Wang Q, Zhang M, Torres G, Wu S, Ouyang C, Xie Z and Zou MH: Metformin suppresses diabetes-accelerated atherosclerosis via the inhibition of Drp1-mediated mitochondrial fission. Diabetes. 66:193–205. 2017. View Article : Google Scholar :

42 

Jin Q, Li R, Hu N, Xin T, Zhu P, Hu S, Ma S, Zhu H, Ren J and Zhou H: DUSP1 alleviates cardiac ischemia/reperfusion injury by suppressing the Mff-required mitochondrial fission and Bnip3-related mitophagy via the JNK pathways. Redox Biol. 14:576–587. 2018. View Article : Google Scholar

43 

Zhang Z, Liu L, Wu S and Xing D: Drp1, Mff, Fis1, and MiD51 are coordinated to mediate mitochondrial fission during UV irradiation-induced apoptosis. FASEB J. 30:466–476. 2016. View Article : Google Scholar

44 

Wang WJ, Wang Y, Chen HZ, Xing YZ, Li FW, Zhang Q, Zhou B, Zhang HK, Zhang J, Bian Xl, et al: Orphan nuclear receptor TR3 acts in autophagic cell death via mitochondrial signaling pathway. Nat Chem Biol. 10:133–140. 2014. View Article : Google Scholar

45 

Rascalou A, Lamartine J, Poydenot P, Demarne F and Bechetoille N: Mitochondrial damage and cytoskeleton reorganization in human dermal fibroblasts exposed to artificial visible light similar to screen-emitted light. J Dermatol Sci. S0923-1811(18)30213-30215. 2018.Epub ahead of print. View Article : Google Scholar : PubMed/NCBI

46 

Toyama EQ, Herzig S, Courchet J, Lewis TL Jr, Losón OC, Hellberg K, Young NP, Chen H, Polleux F, Chan DC and Shaw RJ: Metabolism. AMP-activated protein kinase mediates mitochondrial fission in response to energy stress. Science. 351:275–281. 2016. View Article : Google Scholar : PubMed/NCBI

47 

Kang JW, Hong JM and Lee SM: Melatonin enhances mitophagy and mitochondrial biogenesis in rats with carbon tetrachloride-induced liver fibrosis. J Pineal Res. 60:383–393. 2016. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

January-2020
Volume 45 Issue 1

Print ISSN: 1107-3756
Online ISSN:1791-244X

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Zheng Z, Xiang S, Wang Y, Dong Y, Li Z, Xiang Y, Bian Y, Feng B, Yang B, Weng X, Weng X, et al: NR4A1 promotes TNF‑α‑induced chondrocyte death and migration injury via activating the AMPK/Drp1/mitochondrial fission pathway Retraction in /10.3892/ijmm.2023.5288. Int J Mol Med 45: 151-161, 2020
APA
Zheng, Z., Xiang, S., Wang, Y., Dong, Y., Li, Z., Xiang, Y. ... Weng, X. (2020). NR4A1 promotes TNF‑α‑induced chondrocyte death and migration injury via activating the AMPK/Drp1/mitochondrial fission pathway Retraction in /10.3892/ijmm.2023.5288. International Journal of Molecular Medicine, 45, 151-161. https://doi.org/10.3892/ijmm.2019.4398
MLA
Zheng, Z., Xiang, S., Wang, Y., Dong, Y., Li, Z., Xiang, Y., Bian, Y., Feng, B., Yang, B., Weng, X."NR4A1 promotes TNF‑α‑induced chondrocyte death and migration injury via activating the AMPK/Drp1/mitochondrial fission pathway Retraction in /10.3892/ijmm.2023.5288". International Journal of Molecular Medicine 45.1 (2020): 151-161.
Chicago
Zheng, Z., Xiang, S., Wang, Y., Dong, Y., Li, Z., Xiang, Y., Bian, Y., Feng, B., Yang, B., Weng, X."NR4A1 promotes TNF‑α‑induced chondrocyte death and migration injury via activating the AMPK/Drp1/mitochondrial fission pathway Retraction in /10.3892/ijmm.2023.5288". International Journal of Molecular Medicine 45, no. 1 (2020): 151-161. https://doi.org/10.3892/ijmm.2019.4398