Article Text
Abstract
Objectives Dysregulated chondrocyte metabolism is closely associated with the pathogenesis of osteoarthritis (OA). Suppressing chondrocyte catabolism to restore cartilage homeostasis has been extensively explored, whereas far less effort has been invested toward enhancing chondrocyte anabolism. This study aimed to repurpose clinically approved drugs as potential stimulators of chondrocyte anabolism in treating OA.
Methods Screening of a Food and Drug Administration-approved drug library; Assays for examining the chondroprotective effects of digoxin in vitro; Assays for defining the therapeutic effects of digoxin using a surgically-induced OA model; A propensity-score matched cohort study using The Health Improvement Network to examine the relationship between digoxin use and the risk of joint OA-associated replacement among patients with atrial fibrillation; identification and characterisation of the binding of digoxin to low-density lipoprotein receptor-related protein 4 (LRP4); various assays, including use of CRISPR-Cas9 genome editing to delete LRP4 in human chondrocytes, for examining the dependence on LRP4 of digoxin regulation of chondrocytes.
Results Serial screenings led to the identification of ouabain and digoxin as stimulators of chondrocyte differentiation and anabolism. Ouabain and digoxin protected against OA and relieved OA-associated pain. The cohort study of 56 794 patients revealed that digoxin use was associated with reduced risk of OA-associated joint replacement. LRP4 was isolated as a novel target of digoxin, and deletion of LRP4 abolished digoxin’s regulations of chondrocytes.
Conclusions These findings not only provide new insights into the understanding of digoxin’s chondroprotective action and underlying mechanisms, but also present new evidence for repurposing digoxin for OA.
- osteoarthritis
- chondrocytes
- therapeutics
Data availability statement
All data relevant to the study are included in the article. Not applicable.
Statistics from Altmetric.com
Key messages
What is already known about this subject?
Cardenolides, including digoxin, have been used in treating heart disease for centuries, and have been reported to enhance chondrocyte function in vitro, but how they regulate chondrocytes and whether they are therapeutic against osteoarthritis (OA) remains unknown.
What does this study add?
This study identifies digoxin as a new chondroprotective factor that protects against OA and reduces OA-associated pain in a surgically-induced mouse OA model.
Digoxin use is associated with reduced risk of knee or hip OA-associated joint replacement among patients with atrial fibrillation.
This study isolates lipoprotein receptor-related protein 4 (LRP4) as a new target of digoxin, thus advancing our understanding of digoxin’s action and underlying mechanisms and providing a solid foundation for future discoveries relating to the digoxin/LRP4 interaction in various conditions.
How might this impact on clinical practice or future developments?
The chondroprotective effects of digoxin on OA support the concept that cardenolides, particularly digoxin, may be a new option to treat patients with OA in clinics.
Introduction
Osteoarthritis (OA) is the most common joint disease and the leading cause of disability in older adults.1 2 OA incurs significant financial burden with the medical cost of the disease estimated to account for between 1% and 2.5% of the gross domestic product of various high-income countries.3 Although the complex pathogenesis of OA is not fully understood, the progressive degeneration of articular cartilage is considered the major hallmark.4 5Articular cartilage is commonly known as a physiologically non-self-renewing avascular tissue comprised predominately of extracellular matrix (ECM) maintained through feedback from restricted population of chondrocytes, which is composed mainly of type II collagen and aggrecan.6 7 It is increasingly understood that OA is an active dynamic alteration arising from an imbalance between the repair and destruction of joint tissues, and not a passive degeneration or so-called wear and tear disease as commonly described. Therefore, in addition to the well-known phenomena of increased cartilage-degrading metalloproteinases and cartilage erosion, attention should be also paid to synthesis of matrix molecules and ECM remodelling in OA.8 9Beyond that, articular cartilage has been shown to contain a population of stem cells or progenitor cells, similar to those found in many other adult tissues, that are thought to be involved in the maintenance of tissue homeostasis and in ECM remodelling in OA.10 11
To date, there is no safe treatment available that can halt OA progression. The main goals of the current disease management are pain control and functional improvement with avoidance of therapeutic toxicity.12 Identifying disease-modifying treatment for OA remain an urgent unmet clinical need. Considering the huge costs in terms of time and money associated with drug development, identification of new uses for old drugs is desirable.13–15Taking that view, we screened a drug library composed of Food and Drug Administration (FDA)-approved drugs by use of mesenchymal stem cells and chondrocytes. After preliminary validation, we found that cardenolides, represented by ouabain and digoxin, may have potential therapeutic effects against OA.
Cardenolides, one of the two subgroups of cardiac glycosides, are a class of natural biologically active steroids derived from plants and have been used for the treatment of heart disease for centuries.16 Ouabain and digoxin, two FDA-approved cardenolides, are used to increase the contractile force of the heart and decrease its rate of contraction by inhibiting the cellular Na+/K+-ATPase. Besides the well-known effect of ouabain and digoxin on the cardiovascular system, compelling evidence has indicated that they also participate in the regulation of inflammation. For instance, each has been found to inhibit the expressions of pro-inflammatory cytokines such as interleukin 6 (IL-6) and IL-17 under different pathological conditions.17–20 Additionally, previous research reported that digoxin and ouabain enhanced the functional properties of tissue-engineered cartilage in vitro.21 However, no studies have investigated the effects of cardenolides on OA. In the present study, we performed comprehensive assays with cell and animal models as well as a large population-based cohort study to demonstrate the potential clinical use of digoxin in treating OA. Additionally, we also identified low-density lipoprotein receptor-related protein 4 (LRP4) as a new binding target of digoxin and demonstrated that LRP4 was required for digoxin regulation of chondrocytes.
Results
Isolation of digoxin as a potential chondroprotective drug
To isolate the small molecule drugs that may induce messenger RNA (mRNA) expression of type II collagen (Col2a1), the major component of cartilage ECM, a drug library containing 1046 FDA-approved drugs was first screened. Briefly, C3H10T1/2 mesenchymal stem cells were treated with drugs for 24 hours individually, followed by thousands of quantitative Real time PCR (qRT-PCR) assays for examining the expression of type II collagen in response to individual drug treatment. Twenty drugs that increased the expression of Col2a1 potently were identified after the first round screening (online supplemental figure 1A). These 20 drug candidates were subjected to the second round screening by treating the C28I2 chondrocytes for 24 hours separately. There were only three candidates, particularly ouabain, that could induce the expression of COL2A1 dramatically and dose-dependently (online supplemental figure 1B,C). To further evaluate their potential anabolic effects, we treated C28I2 cells with these three drugs then examined the expressions of various genes known to associated with chondrocyte differentiation and metabolism. Among three drugs analysed, only ouabain could robustly induce the expressions of anabolic marker genes, including COL2A1, aggrecan (ACAN) and cartilage oligomeric matrix protein (COMP). Intriguingly, it strongly inhibited the expression of RUNX Family Transcription Factor 2 (RUNX2), a marker gene of chondrocyte hypertrophy and OA (online supplemental figure 1D–F). As ouabain belongs to the family of cardenolides, we also treated cells with three other cardenolides: lanatoside C, cymarin and digoxin. All three cardenolides could induce the expression of COL2A1, but only digoxin could significantly increase the expressions of ACAN and COMP, two additional markers of anabolism (online supplemental figure 1G). Therefore, we ended up using ouabain and digoxin as representatives of cardenolides to conduct further experiments.
Supplemental material
Digoxin induces chondrogenesis and regulates chondrocyte metabolism
Given the stimulatory effect of ouabain and digoxin on the expression of Col2a1 in C3H10T1/2 mesenchymal stem cells, we first sought to determine whether these two drugs could induce chondrogenesis. We treated micromass cultured C3H10T1/2 mesenchymal stem cells with either drug for 7 or 14 days. As shown in figure 1A and B, alcian blue staining validated the enhanced chondrocyte differentiation in both ouabain and digoxin treated groups compared with the phosphate-buffered saline (PBS) group. Moreover, we also examined the transcriptional levels of chondrogenic marker genes. After treatment with either drug, the mRNA expressions of Col2a1, Comp, Acan, SRY-Box transcription factor 5 (Sox5), SRY-Box transcription factor 6 (Sox6) and SRY-Box transcription factor 9 (Sox9) were all significantly upregulated (figure 1C).
Next we tested their ability to promote anabolism in both human and mouse chondrocytes. We found that both ouabain and digoxin could induce the expressions of COL2A1 and ACAN in human C28I2 chondrocytes. In addition, the expression of COMP, which encodes a protein that is present in small amounts but plays a key role in matrix composition,22 23 was also enhanced by the drugs (figure 1D and E). We next explored whether ouabain and digoxin could upregulate anabolic marker gene expressions in mouse and human primary chondrocytes. As shown in figure 1F and G, both ouabain and digoxin could stimulate chondrocyte anabolism in a dose dependent manner. In addition, human OA chondrocytes appeared to respond better than normal chondrocytes to drug treatment (figure 1G).
Given that these two drugs were previously reported to inhibit the expressions of pro-inflammatory cytokines under different pathological conditions,17–20 we next determined whether ouabain and digoxin could also inhibit pro-inflammatory cytokine activated catabolism in chondrocytes.24 C28I2 chondrocytes were incubated with pro-inflammatory cytokines (tumour necrosis factor α (TNFα) or IL-1β, known to play crucial roles in the pathogenesis of OA) for 24 hours, and both ouabain and digoxin significantly reduced the mRNA expressions of catabolic markers, a disintegrin and metalloproteinases with thrombospondin type 1 motif 4 (ADAMTS4) and matrix metallopeptidase 13 (MMP13)25 (online supplemental figures 2A and 3A). Consistently, the protein levels of ADAMTS4, MMP13 and inducible nitric oxide synthase (iNOS) were also downregulated in ouabain or digoxin treated groups (online supplemental files 2D and 3D). Mouse primary chondrocytes were also treated as described above, as shown in online supplemental figures 2B,C and 3B,C, qRT-PCR revealed significant downregulation of the expressions of catabolic marker genes, mitochondrially encoded cytochrome c oxidase II, nitric oxide synthase 2 (Nos2), Mmp13, Mmp3 and a disintegrin and metalloproteinases with thrombospondin type 1 motif 5 (Adamts5), in the drug treatment groups. The decreased protein levels of iNOS, MMP13, ADAMST5 validated the occurrence of drugs-inhibited chondrocytes catabolism (online supplemental figures 2E and 3E).
Digoxin protects against OA in a surgically induced model in vivo
To examine the effect of ouabain and digoxin on OA progression, we performed destabilisation of the medial meniscus (DMM) surgery26 in 12-week-old male C57BL/6J mice with or without ouabain or digoxin administration commencing 3 days after surgery (figure 2A). We carried out histological analyses to assess knee joint damage 12 weeks after DMM surgery. As expected, cartilage degeneration was severe at 12 weeks after DMM surgery, evidenced by markedly increased Osteoarthritis Research Society International scores. The administration of both ouabain and digoxin (50 nM and 100 nM, respectively, for intra-articular injection) caused partial but significant reduction in cartilage degradation (figure 2B and C). By immunohistochemical staining, a significant decrease in type II collagen in cartilage was observed 12 weeks after DMM surgery, which was greatly inhibited by ouabain or digoxin use (figure 2D). However, the levels of the markers of chondrocytes catabolism (cleaved ACAN, MMP13 and ADAMTS5) was increased after DMM surgery, and ouabain or digoxin administration inhibited these changes (figure 2E). As illustrated in previous studies, during the evolution of OA, the subchondral bone undergoes marked changes in its composition and structural organisation.27 Micro-computerised tomography (µCT) analysis performed in this study also showed significant difference in knee joint structure of mice in different treatment groups. Increased osteophyte formation was observed in mice 12 weeks after DMM surgery, and this effect was significantly inhibited by treatment with ouabain or digoxin (figure 2F–I). The volume of calcified meniscus and synovial tissue was also quantified and exhibited similar trends to those observed in osteophytes (figure 2F, J and K). We further analysed changes in subchondral bone mass and found that subchondral bone mass was significantly increased 12 weeks after DMM surgery and ouabain or digoxin had no significant effect on DMM-induced subchondral bone mass increase (online supplemental figures 4A-E). Collectively, these results suggest that ouabain and digoxin can limit OA development in the injury-induced OA mouse model.
We also performed a series of tests to determine if ouabain and digoxin could lower OA pain sensitivity. The results of von Frey tests showed significantly reduced paw withdrawal response thresholds after DMM surgery in mice. The administration of ouabain and digoxin could significantly increase paw withdrawal response threshold, reflecting reduced pain sensitivity in mice (figure 3A). We also tested spontaneous activity of the mice in response to DMM surgery and cardenolide treatment by performing an open field test. We found that travel distance, maximum walking speed, active time and absolute turn angle decreased over time after DMM surgery. Treatment with ouabain or digoxin could significantly reverse reduced spontaneous activity caused by DMM surgery (figure 3B–F). Taken together, ouabain and digoxin were associated with reduced mechanical pain sensitivity and enhanced spontaneous ambulatory activity relative to untreated controls.
Digoxin use is associated with reduced risk of knee or hip OA-associated joint replacement among patients with atrial fibrillation
To determine whether use of digoxin associates with OA in human patients, we performed a sequential propensity score matched cohort study using data from The Health Improvement Network (THIN) in the UK. The flow charts depict the selection process of individuals are shown in online supplemental file 5). The baseline characteristics of each propensity score matched cohort are shown in online supplemental table 2). After propensity score matching, 56 794 patients were included in the analysis (n=28 397 for each group). The mean age was 74.1 (SD: 9.8) years among the digoxin cohort and 75.1 (SD: 9.7) years among the non-user cohort; the female proportions were 49.1% and 51.5%, respectively. Overall, the characteristics among the propensity score matched cohorts were well-balanced, with all standardised differences <0.1. Digoxin initiators had a lower risk of knee or hip OA-associated joint replacement than non-users (figure 4). As shown in table 1, 739 cases of knee or hip OA-associated joint replacement occurred among 28 397 digoxin users (5.8 per 1000 person-years) and 854 cases occurred among 28 397 non-users (6.8 per 1000 person-years). The rate difference in knee or hip OA-associated joint replacement between digoxin initiators and non-users was −0.9 (95% CI: −1.5 to −0.3) per 1000 person-years and the HR was 0.85 (95% CI: 0.77 to 0.93). Results from the sensitivity analyses (ie, excluding participants who had knee or hip replacement within 3 months after the index date, excluding participants who had extreme propensity scores and missing data imputation analysis) did not change substantially (table 1)
Digoxin regulates chondrocyte metabolism through ERK1/2, AKT and NF-κB signalling pathways
It is widely accepted that the extracellular signalling-regulated kinases (ERK) and protein kinase B (PKB/AKT) signalling pathways play an essential role in chondrocyte anabolism.28 29 To investigate whether ouabain and digoxin activated anabolism through ERK1/2 and AKT signalling pathways, we treated chondrocytes with ouabain or digoxin at different concentrations or different time points, and then performed Western blot analysis for total and phosphorylated ERK1/2 and AKT. As shown in online supplemental figure 6A,B, ouabain and digoxin could induce the phosphorylation of ERK1/2 and AKT in a dose and time dependent manner. Notably, ouabain and digoxin mediated activation of ERK1/2 and AKT signalling pathways and expressions of anabolic marker genes, including COL2A1, ACAN and COMP, were abolished by U0126 (ERK inhibitor) and wortmannin (AKT inhibitor), respectively, (online supplemental figure 6C-F). These results revealed that ouabain and digoxin activated chondrocyte anabolism through ERK1/2 and AKT signalling pathways.
Pro-inflammatory cytokines TNFα and IL-1β play key roles by activating p38 mitogen-activated protein kinase (MAPK) and c-Jun N-terminal kinase (JNK) signalling and the transcription factor nuclear factor kappa B (NF-κB), in the pathogenesis of OA.30 Ouabain and digoxin could inhibit both TNFα and IL-1β-induced chondrocytes catabolism promoted us to determine whether these two drugs could also affect these signalling pathways. Both ouabain and digoxin had no effect on the activation of p38 MAPK and JNK activated by TNFα, while dramatically inhibit NF-κB phosphorylation and transcriptional activity (online supplemental figure 7A-E). As illustrated in online supplemental figure 7F, p65 translocation from the cytoplasmic to the nuclear compartment on stimulation with TNFα was almost abolished in the presence of ouabain or digoxin.
LRP4 is a novel target of digoxin
Cardenolides bind to Na+/K+-ATPase and inhibit its activity. Accordingly, we sought to investigate whether ouabain and digoxin mediated chondrocyte metabolism through Na+/K+-ATPase. We thus suppressed the activity of Na+/K+-ATPase by treating C28I2 cells with Na+/K+-ATPase activity inhibitor istaroxime hydrochloride. To our surprise, the inhibition of Na+/K+-ATPase activity did not affect either drug enhancement of anabolism or inhibition of catabolism (online supplemental figure 8A,B). This finding indicates that ouabain and digoxin may have targets other than Na+/K+-ATPase in mediation of chondrocyte homeostasis.
To address this issue, we performed the drug affinity responsive target stability (DARTS) assay and separated proteins by sodium dodecyl sulfate–polyacrylamide gel electrophoresis. A band with the molecular weight of around 200 kDa was shown to be clearly protected by ouabain (figure 5A). This band was excised for the unbiased protein identification by mass spectrometry (figure 5B), which identified LRP4 as a potential candidate. LRP4 is known to antagonise LRP5/6 signalling and mediate bone homeostasis.31 To confirm whether LRP4 is the target of ouabain and digoxin, we employed DARTS assay using a series of concentrations of protease to digest cell lysate with or without ouabain or digoxin incubation, and found that both drugs could protect LRP4 against enzymatic digestion (figure 5C).
To further confirm the interaction between LRP4 and ouabain and digoxin, we performed the cellular thermal shift assay (CETSA),32 33 which shows the change in thermal denaturation temperature for a target protein in the presence of various drug dosages. Both ouabain and digoxin prevented the denaturation of LRP4 at various temperatures, especially at 49°C, compared with control group (figure 5D). The melting curve showed a robust change of melting temperature (Tm) in the presence of ouabain or digoxin with Tm for control, ouabain and digoxin conditions 42.63, 46.86°C and 49.28°C, respectively (figure 5D). We also performed CETSA at 49°C with different concentrations of drugs and demonstrated that ouabain and digoxin prevented LRP4 denaturation in a dose-dependent manner with the EC50 of 5.98E-01 and 5.74E-02, respectively (figure 5E).
To further unravel the associations of LRP4 with ouabain and digoxin, we then employed molecular docking simulations. The structures of ouabain and digoxin were docked with three monomers of homology modelled human LRP4, respectively. Glide XP docking showed that both ouabain and digoxin had relatively lower binding free energy when they were docked into LRP4 monomer aa 146–737 compared with the other two monomers, which was reflected by the more negative values of docking scores (online supplemental figure 9), suggesting that ouabain and digoxin may have better affinity to the region from residue 146 to 737 of LRP4. Induced-fit docking (IFD) was then performed for ouabain and digoxin with LRP4 monomer aa146-737 to get optimal binding simulation. From IFD simulation, both ouabain and digoxin were predicted to majorly interact with LRP4 with docking scores of −12.496 kcal/mol and −10.149 kcal/mol (figure 5F). The binding pocket for ouabain and digoxin was lined by residues 232–239, 243–252 and 340–348 of LRP4. With abundant hydroxyl groups in the structure, ouabain and digoxin could form multiple hydrogen bonding interactions with LRP4, which may contribute to the good binding affinity. All the hydroxyl groups of ouabain are predicted to be involved in hydrogen bonding interactions with LRP4, which bonded with residues Arg232, Glu235, Phe236, Met237, Cys238, Arg249, Asn340 and Ser344, respectively. Several hydroxyl groups of digoxin served as hydrogen bond donors interacting with the side chains of Phe236, Asp239, Asp342, Glu343, and Asn348 of LRP4. Additionally, digoxin was exhibited to be involved in hydrogen bonds as acceptor of LRP4 at residues Asn340 and Asn348 (figure 5G–J).
We next mutated the sites of LRP4 mentioned above and performed DARTS to determine key site(s) for the binding between LRP4 and drugs (figure 5K). Results showed that ouabain largely lost, while digoxin totally lost, its protective effect on the Glu-343 mutant of LRP4 (figure 5L and M), demonstrating that Glu-343 is the critical amino acid for the drugs targeting to LRP4.
LRP4 is downregulated in OA and its deficiency abolishes digoxin regulation of chondrocyte metabolism
To explore the relationship of LRP4 and OA, we examined the expression of LRP4 in human normal and patients with OA cartilage. LRP4 protein level was reduced in human OA cartilage compared with non-arthritic controls (figure 6A–C). Furthermore, in line with the results in human cartilage, the protein expression of LRP4 was decreased after murine DMM surgery, whereas intriguingly, the administration of ouabain and digoxin prevented OA-associated LRP4 downregulation (figure 6D and E).
To define the role of LRP4 in mediating ouabain and digoxin’s effects on chondrocyte metabolism, we efficiently deleted the LRP4 gene using CRISPR-Cas9 genome editing strategy (figure 6F and G). Notably, the activation of ERK1/2, AKT signalling pathways induced by ouabain and digoxin were abolished in LRP4 knockout chondrocytes, leaving expression of the anabolic marker genes, such as COL2A1, ACAN and COMP, unchanged after drug treatment (figure 6H, I and J). Furthermore, ouabain and digoxin mediated inhibition of NF-κB activation was lost in LRP4 knockout cells, resulting in increased expression of catabolic genes, including ADAMTS4 and MMP13 (online supplemental figure 10A-D).
We next re-expressed wild type or Glu343 mutated LRP4 in LRP4 knockout C28I2 chondrocytes. Transfection of wild type LRP4 expression plasmid reinstated ouabain and digoxin induced activation of ERK1/2, AKT signalling pathways and chondrocyte anabolism (figure 6H, K and L) and also restored the anti-NF-κB effect of ouabain and digoxin and the inhibition of chondrocyte catabolism (online supplemental figure 10A,B,E,F). However, expression of LRP4 containing a Glu343 site mutation in LRP4 knockout C28I2 cells did not restore the regulatory effects of ouabain and digoxin on chondrocyte metabolism (figure 6K and L, online supplemental figure 10E,F). Taken together, these findings indicated that ouabain and digoxin facilitation of anabolism and inhibition of catabolism in chondrocytes is LRP4-dependent and Glu343 is critical for drug-mediated regulation of chondrocyte metabolism.
Discussion
We have made several key observations in the studies presented here. (1) After performance of three rounds of qRT-PCR screening of an FDA-approved drug library, we identified ouabain and digoxin as candidates with potential to promote the anabolism of chondrocyte ECM. (2) We found that the administration of ouabain or digoxin limited OA development and relieved OA-associated pain sensitivity in mice, and our large population-based cohort study provides clinical evidence that digoxin use was associated with a reduced risk of knee or hip OA-associated joint replacement among patients with atrial fibrillation. (3) Through combined use of DARTS, proteomics, CETSA, IFD and generation of point mutations, we identified LRP4 as a previously unrecognised target of digoxin and found that digoxin’s regulation of chondrocytes depends on the presence of LRP4.
No previous studies have directly investigated the effects of cardenolides use on OA. Though indirect evidence concerning the utility of these compounds in facilitating cartilage homeostasis has been provided through in vitro study which found that ouabain and digoxin could increase the functional properties of bovine articular chondrocytes. This promotion may be related to the amount of collagen cross-linking and increased Ca2+ oscillations.21 In the current study, we have implemented a murine surgically–induced OA model to provide in vivo evidence that both ouabain and digoxin can limit OA development and relieve OA-associated pain. Consistently, the anabolism of ECM of chondrocytes is promoted, while the catabolism is inhibited, by ouabain and digoxin. To explore the mechanism of this dual protective effect, we studied multiple pathways closely related to OA by using specific inhibitors of each pathway. The results confirmed that activation of AKT and ERK pathways are involved in ouabain and digoxin-mediated promotion of ECM anabolism, while these cardenolides exert inhibitory effects on NF-κB pathway activation lending to reduced chondrocyte catabolism.
Digoxin is known as the only safe inotropic drug for oral use that improves hemodynamics.34 This led us to hypothesise that records related to digoxin might be accessible in general practitioner based medical records databases. Herein, we performed a sequential propensity score matched cohort study via leveraging the data from THIN. THIN contains health information on approximately 17 million patients from 790 general practices in the UK. THIN data reflects a routine medical practice environment and have been shown to be valid for use in clinical and epidemiological research studies.35–37 Excitingly, we found that digoxin use was associated with reduced risk of joint replacement surgery due to OA in a large population with atrial fibrillation. Despite the inevitable limitations of this database-based study (confounding factors, information authenticity and lack of knee or hip images), this study has significant implications for the clinical prospects of digoxin against OA.
Most of the biological activities of cardenolides are based on their ability to inhibit the membrane-bound Na+/K+-ATPase.16 Inhibition of Na+/K+-ATPase has been shown to induce cell proliferation, autophagy and even apoptosis, not only in cardiac myocytes but also other several cell lines.38 39 Under the intervention condition in this study, both ouabain and digoxin treatment did not show any toxicity (online supplemental figure 11). Neither the promoting effect of ouabain and digoxin on cartilage ECM anabolism nor the inhibitory effect on ECM catabolism was affected by inhibiting the activity of Na+/K+-ATPase with Istaroxime hydrochloride in chondrocytes. This suggested that ouabain and digoxin may regulate cartilage ECM homeostasis through a completely different target that has not yet been discovered. Therefore, through combined use of DARTS, proteomics, CETSA, IFD and point mutations, we identified LRP4 as the target and Glu-343 as the critical amino acid involved in the interaction with digoxin. By knockout and re-expression of LRP4 in chondrocytes, we demonstrated that LRP4 is indispensable in the process of both ouabain and digoxin promoting ECM synthesis by regulating ERK and AKT pathways and inhibiting ECM degradation by regulating NF-κB pathway (online supplemental figure 12).
LRP4 is a member of the low-density lipoprotein receptor family. At neuromuscular junction, neuronal agrin binds LRP4 in a complex with the muscle-specific kinase to form neuromuscular synapse.40 In recent decades, increasing reports describing mutations of members of the LRP family and their relation to bone has reinforced the important role of LRP family members in the pathogenesis of devastating diseases such as osteoporosis, rheumatoid arthritis and OA.31 LRP4 was reported to induce gene expressions of ECM proteins, as well as production of total proteoglycans in ATDC5 chondrocyte cells, whereas LRP4 knockdown had opposite effects and reduced mRNA expression of SOX9, a master regulator for chondrogenesis.41 LRP4 was also reported to upregulate SOX9 expression in primary bovine chondrocytes, and LRP4 was indispensable for the promotion of chondrocyte differentiation and cartilage formation mediated by agrin.42 These findings are consistent with our current report of reduced LRP4 expression in OA cartilage and that the dependence of the chondroprotective effect of ouabain and digoxin on LRP4. However, the specific mechanism by which LRP4 affects chondrocyte function warranties further investigations. Despite the lack of current knowledge regarding LRP4 in chondrocytes, the roles of LRP4 and other members of the LRP family in bone has long been studied. LRP4 and family-member LRP5/6 have opposite effects on bone mass regulation31 43 In OA specifically, in addition to the lack of reports on LRP4, the role of LRP5 and LRP6 is also controversial.44–47 Therefore, although LRP4 has been found to regulate Wnt and bone morphogenetic protein (BMP) pathways in other physiological and pathological processes,48–50 further studies are needed to determine whether LRP4 can affect the development of OA by regulating these pathways and thereby mediating the interaction with other LRPs. In our study, both ouabain and digoxin regulated the expression of the proteins related to AKT, ERK and NF-κB pathways in chondrocytes. This provides a new clue for the mechanism of LRP4 in chondrocyte function and the development of OA.
This study carries significance from several perspectives: This study identifies digoxin as a stimulator of chondrocyte differentiation and anabolism, and uncovers a new strategy for enhancing chondrocyte function and cartilage integrity. Thus, digoxin may be used for treating various chondrocyte-associated diseases and conditions, particularly OA. Due to the unique mechanism of action (eg, targeting LRP4 to enhance chondrocyte function), digoxin may be effective for patients with OA who fail to respond to current anti-inflammatory and anti-catabolic treatments. This study identifies LRP4 as a new target of digoxin, thus advancing our understanding of digoxin’s action and underlying mechanisms, and also providing a solid foundation for future discoveries related to this digoxin/LRP4 interaction in various conditions.
Limitations of this study should be also acknowledged. Although previous studies have used joint replacement as a proxy for end-stage OA and joint replacement has been generally accepted as a clinically relevant ‘hard’ outcome in cohort studies of OA,51–53 knee images and functional data were not available in THIN; thus, we were unable to examine the association between ddigoxin and the risk of progression of structural lesions and functional deterioration of OA. Although we conducted a 1:1 propensity score matched cohort study to control for many potential confounders, this may limit generalisability of our findings to patients with OA whose characteristics differ from patients with OA included in the current study. Although we have demonstrated that digoxin and ouabain can relieve OA-related pain in vivo, we failed to delve deeply into the mechanisms. The weak correlation between pain and cartilage loss, the complex classification of pain and the conduction involved,54 55 give us reason to believe that further research, richer tools and extended time points are needed to uncover a full story. Further, additional in vivo characterisation and pharmaceutic kinetic assays of the drug in OA, including dose dependence, frequency and route of delivery, as well as treatment duration, warrant further investigations with preclinical animal models and human clinical trials.
In summary, functional studies, including various cell-based assays, in vivo animal model, and analysis of a large cohort of human patients, support the concept of using digoxin as novel treatment for patients with OA in clinic. Additionally, combined use of various approaches isolates LRP4 as the new target of digoxin responsible for its chondroprotective action. Thus, this study not only provides new insights into the understanding of digoxin’s action and underlying mechanisms, but may also broaden the clinical application of digoxin.
Data availability statement
All data relevant to the study are included in the article. Not applicable.
Ethics statements
Patient consent for publication
Ethics approval
All animal procedures were carried out in accordance with institutional guidelines and approved by the Institutional Animal Care and Use Committee of New York University. Human subjects research was performed according to the Institutional Review Boards at New York University Medical Center (IRB Study Number i11-01488). This study was approved by the THIN Scientific Review Committee (20SRC003). THIN is a registered trademark of Cegedim SA in the UK and other countries. Reference made to the THIN database is intended to be descriptive of the data asset licensed by IQVIA. This work uses de-identified data provided by patients as part of their routine primary care.
Acknowledgments
The authors would like to acknowledge all laboratory members for insightful discussions. We thank Dr Mary Goldring for providing us with human C28I2 chondrocytes. We are also grateful to Dr Tanaji T Talele (St. John’s University, New York, New York, USA) for providing the computing resources for docking analysis. We also thank NYU Proteomics Laboratory and microCT core for technique support.
References
Supplementary materials
Supplementary Data
This web only file has been produced by the BMJ Publishing Group from an electronic file supplied by the author(s) and has not been edited for content.
Footnotes
Handling editor Josef S Smolen
K-dW and XD contributed equally.
Contributors K-dW and XD designed and performed the experiments, collected and analysed the data and drafted the manuscript. NJ and X-yC participated in chondrogenesis-related experiments and identification of the drug target. CZ and JW performed the cohort study. AH assisted with human sample collection and language modification. AK participated in drug screening. Z-nL and Z-sC performed the induced-fit docking simulations. G-hL had full access to the database in the study and take responsibility for the integrity of the data and the accuracy of the data analysis. C-jL had the concept, supervised all of the experiments and revised the manuscript. C-jL as the guarantor and responseible for the overall content. All authors have read, provided critical feedback on intellectual content and approved the final manuscript.
Funding This work is supported partly by NIH research grants R01AR062207, R01AR061484, R01AR076900, R01NS103931 and a DOD research grant W81XWH-16-1-0482.
Competing interests None declared.
Provenance and peer review Not commissioned; externally peer reviewed.
Supplemental material This content has been supplied by the author(s). It has not been vetted by BMJ Publishing Group Limited (BMJ) and may not have been peer-reviewed. Any opinions or recommendations discussed are solely those of the author(s) and are not endorsed by BMJ. BMJ disclaims all liability and responsibility arising from any reliance placed on the content. Where the content includes any translated material, BMJ does not warrant the accuracy and reliability of the translations (including but not limited to local regulations, clinical guidelines, terminology, drug names and drug dosages), and is not responsible for any error and/or omissions arising from translation and adaptation or otherwise.