Skip to main content

Thank you for visiting nature.com. You are using a browser version with limited support for CSS. To obtain the best experience, we recommend you use a more up to date browser (or turn off compatibility mode in Internet Explorer). In the meantime, to ensure continued support, we are displaying the site without styles and JavaScript.

  • Article
  • Published:

Exosomal transfer of osteoclast-derived miRNAs to chondrocytes contributes to osteoarthritis progression

Abstract

Osteoarthritis (OA) is a prevalent aging-related joint disease lacking disease-modifying therapies. Here, we identified an upregulation of circulating exosomal osteoclast (OC)-derived microRNAs (OC-miRNAs) during the progression of surgery-induced OA in mice. We found that reducing OC-miRNAs by Cre-mediated excision of the key miRNA-processing enzyme Dicer or blocking the secretion of OC-originated exosomes by short interfering RNA-mediated silencing of Rab27a substantially delayed the progression of surgery-induced OA in mice. Mechanistically, the exosomal transfer of OC-miRNAs to chondrocytes reduced the resistance of cartilage to matrix degeneration, osteochondral angiogenesis and sensory innervation during OA progression by suppressing tissue inhibitor of metalloproteinase-2 (TIMP-2) and TIMP-3. Furthermore, systemic administration of a new OC-targeted exosome inhibitor (OCExoInhib) blunted the progression of surgery-induced OA in mice. We suggest that targeting the exosomal transfer of OC-miRNAs to chondrocytes represents a potential therapeutic avenue to tackle OA progression.

This is a preview of subscription content, access via your institution

Access options

Buy this article

Prices may be subject to local taxes which are calculated during checkout

Fig. 1: Elevated OC-miRNAs in osteoclasts and circulating exosomes in early-stage OA.
Fig. 2: Genetic knockdown of OC-miRNAs retards the progression of ACLT-induced OA in mice.
Fig. 3: Blockage of OC-exosomes dampens the progression of ACLT-induced OA in mice.
Fig. 4: OC-exosomes promote chondrocyte catabolism, endothelial cell angiogenesis and axon innervation in vitro.
Fig. 5: OC-derived exosomal miRNAs inhibit TIMP-2 and TIMP-3 to reduce the resistance of cartilage to matrix degeneration, angiogenesis and sensory innervation in vitro.
Fig. 6: OC-derived exosomal miRNAs reduce the resistance of cartilage to matrix degeneration, angiogenesis and sensory innervation in OA.
Fig. 7: OCExoInhib treatment blunts OA progression in mice after ACLT.

Similar content being viewed by others

Data availability

All miRNA qPCR array data (raw Ct values) can be freely downloaded and reproduced from https://github.com/TMBJ-HKBU/NATAGING_A00041_qPCRArray/. All data supporting the findings of this study are available within the article and its Extended Data files or are available from the corresponding author upon reasonable request.

Code availability

All code for the analytic software is available from the corresponding author upon reasonable request.

References

  1. Mandl, L. A. Osteoarthritis year in review 2018: clinical. Osteoarthritis Cartilage 27, 359–364 (2019).

    Article  CAS  PubMed  Google Scholar 

  2. Glyn-Jones, S. et al. Osteoarthritis. Lancet 386, 376–387 (2015).

    Article  CAS  PubMed  Google Scholar 

  3. Bannuru, R. R. et al. OARSI guidelines for the non-surgical management of knee, hip and polyarticular osteoarthritis. Osteoarthritis Cartilage https://doi.org/10.1016/j.joca.2019.06.011 (2019).

  4. Lories, R. J. & Luyten, F. P. The bone–cartilage unit in osteoarthritis. Nat. Rev. Rheumatol. 7, 43–49 (2011).

    Article  CAS  PubMed  Google Scholar 

  5. Karsdal, M. A. et al. The coupling of bone and cartilage turnover in osteoarthritis: opportunities for bone antiresorptives and anabolics as potential treatments? Ann. Rheum. Dis. 73, 336–348 (2014).

    Article  CAS  PubMed  Google Scholar 

  6. Burr, D. B. & Gallant, M. A. Bone remodelling in osteoarthritis. Nat. Rev. Rheumatol. 8, 665–673 (2012).

    Article  CAS  PubMed  Google Scholar 

  7. Shibakawa, A. et al. The role of subchondral bone resorption pits in osteoarthritis: MMP production by cells derived from bone marrow. Osteoarthritis Cartilage 13, 679–687 (2005).

    Article  CAS  PubMed  Google Scholar 

  8. Zhu, S. et al. Subchondral bone osteoclasts induce sensory innervation and osteoarthritis pain. J. Clin. Invest. 129, 1076–1093 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  9. Zhen, G. et al. Inhibition of TGF-β signaling in mesenchymal stem cells of subchondral bone attenuates osteoarthritis. Nat. Med. 19, 704–712 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Goldring, S. R. & Goldring, M. B. Changes in the osteochondral unit during osteoarthritis: structure, function and cartilage–bone cross-talk. Nat. Rev. Rheumatol. 12, 632–644 (2016).

    Article  PubMed  Google Scholar 

  11. Kim, Y-. K., Kim, B. & Kim, V. N. Re-evaluation of the roles of DROSHA, Export in 5, and DICER in microRNA biogenesis. Proc. Natl Acad. Sci. USA 113, E1881–E1889 (2016).

    Article  CAS  PubMed  Google Scholar 

  12. Mizoguchi, F. et al. Osteoclast-specific Dicer gene deficiency suppresses osteoclastic bone resorption. J. Cell. Biochem. 109, 866–875 (2010).

    CAS  PubMed  Google Scholar 

  13. Liu, J. et al. A delivery system specifically approaching bone resorption surfaces to facilitate therapeutic modulation of microRNAs in osteoclasts. Biomaterials 52, 148–160 (2015).

    Article  CAS  PubMed  Google Scholar 

  14. Liang, C. et al. Tumor cell-targeted delivery of CRISPR–Cas9 by aptamer-functionalized lipopolymer for therapeutic genome editing of VEGFA in osteosarcoma. Biomaterials 147, 68–85 (2017).

    Article  CAS  PubMed  Google Scholar 

  15. Bryden, L. A., Nicholson, J. R., Doods, H. & Pekcec, A. Deficits in spontaneous burrowing behavior in the rat bilateral monosodium iodoacetate model of osteoarthritis: an objective measure of pain-related behavior and analgesic efficacy. Osteoarthritis Cartilage 23, 1605–1612 (2015).

    Article  CAS  PubMed  Google Scholar 

  16. Ostrowski, M. et al. Rab27a and Rab27b control different steps of the exosome secretion pathway. Nat. Cell Biol. 12, 19–30 (2010).

    Article  CAS  PubMed  Google Scholar 

  17. Sun, W. et al. Osteoclast-derived microRNA-containing exosomes selectively inhibit osteoblast activity. Cell Discov. 2, 16015 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. Li, D. et al. Osteoclast-derived exosomal miR-214-3p inhibits osteoblastic bone formation. Nat. Commun. 7, 10872 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. Liu, J. et al. Osteoclastic miR-214 targets TRAF3 to contribute to osteolytic bone metastasis of breast cancer. Sci. Rep. 7, 40487 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Zhuo, Z. et al. A loop-based and AGO-incorporated virtual screening model targeting AGO-mediated miRNA–mRNA interactions for drug discovery to rescue bone phenotype in genetically modified mice. Adv. Sci. 7, 1903451 (2020).

    Article  CAS  Google Scholar 

  21. Eisenstein, R., Sorgente, N., Soble, L. W., Miller, A. & Kuettner, K. E. The resistance of certain tissues to invasion: penetrability of explanted tissues by vascularized mesenchyme. Am. J. Pathol. 73, 765–774 (1973).

    CAS  PubMed  PubMed Central  Google Scholar 

  22. Datta, A. et al. High-throughput screening identified selective inhibitors of exosome biogenesis and secretion: a drug repurposing strategy for advanced cancer. Sci. Rep. 8, 8161 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  23. Johnson, J. L. et al. Identification of neutrophil exocytosis inhibitors (Nexinhibs), small-molecule inhibitors of neutrophil exocytosis and inflammation: druggability of the small GTPase Rab27a. J. Biol. Chem. 291, 25965–25982 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. Birch, C. E., Mensch, K. S., Desarno, M. J., Beynnon, B. D. & Tourville, T. W. Subchondral trabecular bone integrity changes following ACL injury and reconstruction: a cohort study with a nested, matched case–control analysis. Osteoarthritis Cartilage 26, 762–769 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Xie, H. et al. PDGF-BB secreted by preosteoclasts induces angiogenesis during coupling with osteogenesis. Nat. Med. 20, 1270–1278 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Saunders, W. B. et al. Coregulation of vascular tube stabilization by endothelial cell TIMP-2 and pericyte TIMP-3. J. Cell Biol. 175, 179–191 (2006).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. Seo, D. W. et al. TIMP-2-mediated inhibition of angiogenesis: an MMP-independent mechanism. Cell 114, 171–180 (2003).

    Article  CAS  PubMed  Google Scholar 

  28. Kashiwagi, M., Tortorella, M., Nagase, H. & Brew, K. TIMP-3 is a potent inhibitor of aggrecanase 1 (ADAM-TS4) and aggrecanase 2 (ADAM-TS5). J. Biol. Chem. 276, 12501–12504 (2001).

    Article  CAS  PubMed  Google Scholar 

  29. Mi, M. et al. TIMP-2-deficient mice develop accelerated osteoarthritis via promotion of angiogenesis upon destabilization of the medial meniscus. Biochem. Biophys. Res. Commun. 423, 366–372 (2012).

    Article  CAS  PubMed  Google Scholar 

  30. Sahebjam, S., Khokha, R. & Mort, J. S. Increased collagen and aggrecan degradation with age in the joints of Timp3−/− mice. Arthritis Rheum. 56, 905–909 (2007).

    Article  CAS  PubMed  Google Scholar 

  31. Jiang, M. et al. Exosomes from MiR-30d-5p-ADSCs reverse acute ischemic stroke-induced, autophagy-mediated brain injury by promoting M2 microglial/macrophage polarization. Cell. Physiol. Biochem. 47, 864–878 (2018).

    Article  CAS  PubMed  Google Scholar 

  32. Huang, F. et al. miR-148a-3p mediates Notch signaling to promote the differentiation and M1 activation of macrophages. Front. Immunol. 8, 1327 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  33. Ruckerl, D. et al. Induction of IL-4Rα-dependent microRNAs identifies PI3K/Akt signaling as essential for IL-4-driven murine macrophage proliferation in vivo. Blood 120, 2307–2316 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. Chen, Y. et al. Macrophages in osteoarthritis: pathophysiology and therapeutics. Am. J. Transl. Res. 12, 261–268 (2020).

    CAS  PubMed  PubMed Central  Google Scholar 

  35. Simon, D. et al. The relationship between anterior cruciate ligament Injury and osteoarthritis of the knee. Adv. Orthop. 2015, 928301 (2015).

    Article  PubMed  PubMed Central  Google Scholar 

  36. Weber, J., Koch, M., Angele, P. & Zellner, J. The role of meniscal repair for prevention of early onset of osteoarthritis. J. Exp. Orthop. 5, 10 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  37. Glasson, S. S., Blanchet, T. J. & Morris, E. A. The surgical destabilization of the medial meniscus (DMM) model of osteoarthritis in the 129/SvEv mouse. Osteoarthritis Cartilage 15, 1061–1069 (2007).

    Article  CAS  PubMed  Google Scholar 

  38. Deacon, R. Assessing burrowing, nest construction and hoarding in mice. J. Vis. Exp. https://doi.org/10.3791/2607 (2012).

  39. Liang, C. et al. Aptamer-functionalized lipid nanoparticles targeting osteoblasts as a novel RNA interference-based bone anabolic strategy. Nat. Med. 21, 288–294 (2015).

    Article  PubMed  PubMed Central  Google Scholar 

  40. Wang, X. et al. miR-214 targets ATF4 to inhibit bone formation. Nat. Med. 19, 93–100 (2013).

    Article  PubMed  Google Scholar 

  41. Pritzker, K. P. et al. Osteoarthritis cartilage histopathology: grading and staging. Osteoarthritis Cartilage 14, 13–29 (2006).

    Article  CAS  PubMed  Google Scholar 

  42. Basappa et al. Antitumor activity of a novel HS-mimetic-vascular endothelial growth factor binding small molecule. PLoS ONE 7, e39444 (2012).

    Article  CAS  PubMed Central  Google Scholar 

  43. Sleigh, J. N., Weir, G. A. & Schiavo, G. A simple, step-by-step dissection protocol for the rapid isolation of mouse dorsal root ganglia. BMC Res Notes 9, 82 (2016).

    Article  PubMed  PubMed Central  Google Scholar 

  44. Newton, P. T. et al. Chondrogenic ATDC5 cells: an optimised model for rapid and physiological matrix mineralisation. Int. J. Mol. Med. 30, 1187–1193 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  45. Arnaoutova, I. & Kleinman, H. K. In vitro angiogenesis: endothelial cell tube formation on gelled basement membrane extract. Nat. Protoc. 5, 628–635 (2010).

    Article  CAS  PubMed  Google Scholar 

  46. Eisen, M. B., Spellman, P. T., Brown, P. O. & Botstein, D. Cluster analysis and display of genome-wide expression patterns. Proc. Natl Acad. Sci. USA 95, 14863–14868 (1998).

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgements

We thank technical staff (Y. S. Cheung, W. K. Chan, S. Lee and C. L. Chan) from Law Sau Fai Institute for Advancing Translational Medicine in Bone and Joint Diseases, Hong Kong Baptist University for providing technical support on micro-CT, confocal imaging and flow cytometry analysis. We thank Guangzhou Biogene Biotechnology Co., Ltd. for providing technical support on mouse genotyping and AAV packaging. This work was supported by the funds from National Key R&D Program of China (2018YFA0800804 to L.C.) and National Natural Science Foundation Council of China (81702189 to J. Liu; 81803374 to J. Lu., 81802187 to G.H., 81703049 to F.L. and 81700780 to C.L.), the Theme-based Research Scheme from the Research Grants Council of Hong Kong (T12-201/20-R to A.L.), the General Research Funds from the Research Grants Council of Hong Kong (12102914 to G.Z., 12101117 to G.Z., 12136616 and 12103519 to J. Liu, 12101018 to F.L., 12102120 to Y.Y., 12102518 and 12100719 to A.L. and 14112915 to B.-T.Z.), the Basic and Applied Basic Research Fund from Department of Science and Technology of Guangdong Province (2019B1515120089 to G.Z.), the Natural Science Fund of Guangdong Province (2018030310355 to G.H.), the Shenzhen Science and Technology innovation fund (JCYJ20180302174121208 to J. Lu.), the Interdisciplinary Research Clusters Matching Scheme of Hong Kong Baptist University (RC-IRCs/17-18/02 to G.Z.; RC-IRCs/17-18/04 and RC-IRMS/15-16/01 to A.L.), the Inter-institutional Collaborative Research Scheme from Hong Kong Baptist University (RC-ICRS/19-20/01 to A.L.)

Author information

Authors and Affiliations

Authors

Contributions

J. Liu, A.L. and G.Z. jointly supervised the project and wrote most of the manuscript. J. Liu, X.W., J. Lu, L.D., H.Z. and C.Z. performed the majority of the experiments, analyzed data and prepared the manuscript. G.H. collected and analyzed the clinical samples. Z.Z and D.L. assisted in the animal breeding and sample collection. F.L., C.L. and Y.Y. provided technical instructions. B.-T.Z. and L.C. provided their professional expertise.

Corresponding authors

Correspondence to Jin Liu, Aiping Lu or Ge Zhang.

Ethics declarations

Competing interests

The authors declare no competing interests.

Additional information

Peer review information Nature Aging thanks Mary Goldring, Michiel Pegtel and the other, anonymous, reviewer(s) for their contribution to the peer review of this work.

Publisher’s note Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Extended data

Extended Data Fig. 1 Characterization for bone marrow-derived OCs and serum OC-exosomes.

(a) QPCR analysis of the mRNAs of Ctsk and Acp5 in osteoclasts (OSCAR+CTR+OCs), OC precursors (OSCAR+CTR-OCPs) and non-osteoclastic cells (OSCAR-CTR-cells) isolated from mouse hindlimb bone marrow cells by magnetic activated cell sorting (MACS). (b) Representative TRAP staining images and the number of TRAP+ multinucleated cells in the culture of the aforementioned OCs, OCPs and non-OCs. Scale bars: 100 μm. c, Nanoparticle tracking analysis showing the size distribution of the serum exosomes. (d) Representative transmission electronic microscope (TEM) images of serum exosomes. Scale bars: 200nm. (e) Western blot analysis of the exosome markers CD63, CD9, TSG101, ALIX, the osteoclast-specific markers OSCAR and CTR, and the cytoplasma proteins GM130 and cytochrome C in serum exosomes. g, Gating strategy for characterization of the circulating OC-exosomes by flow cytometry analysis. The circulating exosomes were harvested from the serum, purified by the CD63 magnetic beads and visualized by the FITC staining. The CD63+ fraction was further gated for sorting the OC-exosomes co-expressing OSCAR (PE) and CTR (APC). (g) Representative TEM images of serum OC-derived exosomes expressing OSCAR and CTR (indicated by red arrows). Scale bars: 200nm. (h) QPCR analysis of sera exosomal miR-223-3p and miR-34a-5p from ACLT and Sham mice at 2 weeks after surgery, respectively. (i) The exosome quantitation (per 106 cells) in cell culture supernatant of OCs differentiated from bone marrow monocyte/macrophage (BMMs) from ACLT and Sham mice at 2w after surgery, respectively. Data are shown as mean ± s.d. All experiments were performed in at least biologically independent triplicates in each group. The exact p values are shown in figures. In a & h, Two-way analysis of variance (ANOVA) with either Turkey’s (a) or Bonferroni’s (h) multiple comparisons test was used for statistical analysis. In b, One-way ANOVA with Turkey’s multiple comparisons was used for statistical analysis. In i, student’s t-test was used for statistical analysis.

Source data

Extended Data Fig. 2 Characterization of OCmiR-KD mice.

(a) Representative genotyping image of the Dicerfl/fl mice, Rosa-YFP mice and Dicerfl/fl; Rosa-YFP mice. (b) Representative confocal fluorescent micrographs of YFP expression after Cre-mediated recombination in TRAP+ OCs in the cryosection of subchondral bone from the Dicerfl/fl; Rosa-YFP mice administered with Cre-expressing plasmid encapsulated within the OC-TDS. (c) Gating strategy for the detection of Cre-mediated recombination in OCs in vivo by flow cytometry analysis. The dead cells were excluded by 7-aminoactinomycin D (7-AAD) staining, while the live cells were gated for identifying the OCs with the co-expression of OSCAR (PE) and CTR (APC). The Cre-mediated recombination in OCs was further confirmed by the YFP expression in OCs. (d) Microscopic images of multinucleated osteoclast formation with TRAP staining in BMMs with or without infection of Cre-expressing lentivirus (e) The exosome quantitation (per 106 cells) in cell culture supernatant of OCs differentiated from BMMs with or without infection of Cre-expressing lentivirus. (f-g) Western blot analysis of cellular protein expression of Rab27a and GAPDH (f) and exosomal protein expression of CD63 and CD9 (g). All scale bars: 100 μm. Data are shown as mean ± s.d. All experiments were performed in biologically independent triplicates in each group. The exact p values are shown in figures. Student’s t-test was used for statistical analysis.

Source data

Extended Data Fig. 3 Effect of OC-exosome blockage on osteoblasts in mice after ACLT.

(a) Representative fluorescent images of the Runx2 (red) immunostaining at tibial articular cartilage and subchondral bone. (b) The number of Runx2+ cells at subchondral bone (SCB) area or growth plate (GP) area of mice from the indicated groups at 4w after ACLT. Scale bar: 100 μm. Data are shown as mean ± s.d. N=8 mice in each group. The exact p values are shown in figures. Two-way ANOVA with Turkey’s multiple comparisons test was used for statistical analysis.

Source data

Extended Data Fig. 4 The transfer of OC-exosomes to OA cartilage in vivo.

The representative confocal images of the PKH26-labelled OC-exosomes in tibial cartilage, subchondral bone and metaphysis area in ACLT mice or Sham mice at 2w after surgery. The mice were intravenously injected with OC-exosomes (20 μg per mouse) 3 days before euthanasia. Left: PKH26-labelled OC-exosomes indicated by red fluorescent signal. Middle left: Endomucin+ vessels indicated by green fluorescent signal. Middle right: Merged images with DAPI-stained nucleates (blue) and bright field. Right: 20x zoom-in of the cartilage layer. Dotted lines indicate the osteochondral junction. Arrows indicate the PKH26-labelled OC-exosomes at osteochondral junction and cartilage layer. Scale bar: 100 μm. All experiments were performed in biologically independent triplicates in each group.

Extended Data Fig. 5 OC-derived exosomal miR-214-3p promote cartilage matrix degeneration, angiogenesis and sensory axon innervation in vivo.

(a) Schematic diagram showing the strategy for osteoclast-targeted delivery of control ncRNA in Ctsk-Cre mice (WT, OCExo-Intact), osteoclast-specific miR-214 knockout mice (CKO, OCExo-Intact) and osteoclast-specific miR-214 knockin mice (CKI, OCExo-Intact) and osteoclast-targeted delivery of Rab27a siRNA in the CKI mice (OCExo-BLK). (b-c) QPCR analysis of the miR-214-3p in BM-OCs and serum exosomes (b) and the pri-miR-214 and miR-214-3p in cartilage (c) from the indicated mice. (d) The burrowing performance. (e) Left: Representative Safranin O staining images of tibial articular cartilage. Right: the OARSI scores. (f-g) Left: Representative fluorescent images of the Endomucin (EDMC, green) and CD31 (red) immunostaining (f) and CGRP (green) immunostaining (g) at tibial articular cartilage and subchondral bone. Right: the number of EDMChiCD31hi cells (f) and CGRP+ fibers (g) around the osteochondral junction. All scale bar: 100 μm. Data are shown as mean ± s.d. In b & c, N=4 mice per group. The exact p values are shown in figures. Two-way ANOVA with Bonferroni’s (h) multiple comparisons test was used for statistical analysis. In d-g, N=6 mice in each group. One-way ANOVA with Turkey’s multiple comparisons test was used for statistical analysis.

Source data

Extended Data Fig. 6 OC-derived exosomal miRNAs regulate TIMP2 and TIMP3 in chondrocytes in vitro.

(a) Representative confocal fluorescent micrographs showing the co-localization of FAM-labelled miR-214-3p mimics and PKH26-labelled OC-exosomes in chondrocytes. Scale bar: 100 μm. (b) QPCR analysis of the mRNA levels of TIMP1, 2 and 3 in IL-1β-stimulated chondrocytes with or without OCs co-culture. (c) Western blot analysis of the mRNA levels of TIMP1, 2 and 3 in IL-1β-stimulated chondrocytes with or without OCs co-culture. The aforementioned OCs were differentiated from BMMs with either Rab27a siRNA or ncRNA transfection. Data are shown as mean ± s.d. All experiments were performed in biologically independent triplicates in each group. The exact p values are shown in figures. Two-way ANOVA with Turkey’s multiple comparisons test was used for statistical analysis.

Source data

Extended Data Fig. 7 The development of OCExoInhib.

(a) TRAP staining analysis of the osteoclastogenesis of Raw264.7 cells treated with various exosome inhibitors at 0.5 μM. Scale bar: 100 μm. (b) MTT assay of the cell viability of Raw264.7 cells treated with Nexinhib20 or LJ001. (c) The synthetic route for d-Asp8-Nexinhib20 derivative conjugate. Reagents and conditions: (i) Br2, DCM, 0 °C to r.t., 8h; (ii) 1,2,4-triazole, Cs2CO3, acetonitrile, 80 °C, 4 h; (iii) 3-hydroxylbenzaldehyde, piperidine, toluene, 60 °C, 4 h; (iv) K2CO3, acetonitrile, 40 °C, 6 h; (v) LiOH, THF/H2O, 20 °C, 4h; (vi) N-Hydroxysuccinimide, 1,3-dicyclohexylcarbodiimide, DCM, 20 °C, 12h; (vii) Ethyldiisopropylamine, d-Asp8, DMF, 20 °C, 24h. (d) Exosome quantitation (per 106 cells) in cell culture supernatant of RANKL-induced Raw264.7 cells with the treatments indicated. (e) QPCR analysis of the mRNAs of Ctsk and Acp5 in RANKL-induced Raw264.7 cells with the treatments indicated. (f) The number of bone resorption pits in bone slices seeded with RANKL-induced Raw264.7 cells with the treatments indicated. (g) Molecular docking showing the interaction between Rab27a and Nexinhib20 (left panel), between Rab27a and LJ001 (middle panel), and between Rab27a and OCExoInhib (right panel), respectively. Red, yellow, blue and white ribbons: Rab27a. The binding surfaces are identified in gray. The structure of inhibitor is displayed by green ball-and-stick model and the structure of d-Asp8 is displayed by purple ball-and-stick model. (h) Representative biophotonic images of the distribution of the FAM-labelled LJ001 and OCExoInhib in organ levels. (i) Representative confocal images of the colocalization of FAM-labelled OCExoInhib and TRAP+ OCs (arrow indicated) at tibial subchondral bone surface. Dotted line indicates the osteochondral junction. Scale bar: 100 μm. Data are shown as mean ± s.d. All experiments were performed in biologically independent triplicates in each group. The exact p values are shown in figures unless the asterisks are annotated as follow. In a, ***: p < 0.001 vs Veh group. Two-way ANOVA (a & e) or One-way ANOVA (d & f) with Turkey’s multiple comparisons test was used for statistical analysis.

Source data

Extended Data Fig. 8 OCExoInhib treatment blunt the OA progression in mice after DMM.

(a) Schematic diagram showing the strategy for OCExoinhib treatment in C57BL/6 mice with DMM. (b) The burrowing performance. (c) Left: Representative Safranin O staining images of tibial articular cartilage. Right: the OARSI scores. (d-f) Left: Representative fluorescent images of the TIMP2 (green) immunostaining (d), the Endomucin (EDMC, green) and CD31 (red) immunostaining (e) or the CGRP (green) immunostaining (f) at tibial articular cartilage and SCB. Right: the number of TIMP2+ chondrocytes at cartilage layer (d), the number of EDMChiCD31hi cells (e) and CGRP+ fibers (f) around the osteochondral junction (white dotted line). All scale bars: 100 μm. Data are shown as mean ± s.d. N=8 mice per group. The exact p values are shown in figures unless the asterisks are annotated as follow. In b, ***: p < 0.001 DMM+Veh / DMM+OCExoInhib group vs Sham+Veh group, φφφ: p < 0.001 DMM+Veh group vs DMM+OCExoInhib group. Two-way ANOVA with Turkey’s multiple comparisons test was used for statistical analysis. In c-f, One-way ANOVA with Turkey’s multiple comparisons test was used for statistical analysis.

Extended Data Fig. 9 Schematic diagram of the osteoclast-chondrocyte crosstalk mediated by osteoclast-derived exosomal miRNA in OA.

Left side of the dotted line: an illustration of the health articular joint. Right side of the dotted line: an illustration of the articular joint with OA. The box illustrates the postulated exosomal miRNA-mediated osteoclast-chondrocyte crosstalk in OA.

Supplementary information

Source data

Source Data Fig. 1

Statistical source data.

Source Data Fig. 2

Statistical source data.

Source Data Fig. 3

Statistical source data.

Source Data Fig. 4

Statistical source data.

Source Data Fig. 4

Unprocessed western blots.

Source Data Fig. 5

Statistical source data.

Source Data Fig. 5

Unprocessed western blots.

Source Data Fig. 6

Statistical source data.

Source Data Fig. 7

Statistical source data.

Source Data Extended Data Fig. 1

Statistical source data.

Source Data Extended Data Fig. 1

Unprocessed western blots.

Source Data Extended Data Fig. 2

Statistical source data.

Source Data Extended Data Fig. 2

Unprocessed western blots.

Source Data Extended Data Fig. 3

Statistical source data.

Source Data Extended Data Fig. 5

Statistical source data.

Source Data Extended Data Fig. 6

Statistical source data.

Source Data Extended Data Fig. 6

Unprocessed western blots.

Source Data Extended Data Fig. 7

Statistical source data.

Source Data Extended Data Fig. 8

Statistical source data.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Liu, J., Wu, X., Lu, J. et al. Exosomal transfer of osteoclast-derived miRNAs to chondrocytes contributes to osteoarthritis progression. Nat Aging 1, 368–384 (2021). https://doi.org/10.1038/s43587-021-00050-6

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s43587-021-00050-6

This article is cited by

Search

Quick links

Nature Briefing: Translational Research

Sign up for the Nature Briefing: Translational Research newsletter — top stories in biotechnology, drug discovery and pharma.

Get what matters in translational research, free to your inbox weekly. Sign up for Nature Briefing: Translational Research