Skip to main content

Advertisement

Log in

miR-92a-3p-inspired shRNA exhibits pro-chondrogenic and chondrocyte protective effects in osteoarthritis treatment through targeting SMAD6/7

  • Original Article
  • Published:
Journal of Bone and Mineral Metabolism Aims and scope Submit manuscript

Abstract

Introduction

Osteoarthritis (OA) compromises patients’ quality of life and requires further study. Although miR-92a-3p was reported to possess chondroprotective effects, the underlying mechanism requires further clarification. The objectives of this study were to elucidate the mechanism by which miR-92a-3p alleviates OA and to examine the efficacy of shRNA-92a-3p, which was designed based on mature miR-92a-3p.

Materials and methods

TargetScan and luciferase reporter assay were used to predict the target of miR-92a-3p. Adipose-derived stem cells (ADSCs) were transfected with miR-92a-3p/miR-NC mimic for the analysis of chondrogenic biomarkers and SMAD proteins. ADSCs and osteoarthritic chondrocytes were transduced with shRNA-92a-3p for the analysis of chondrogenic biomarkers and SMAD proteins. OA was surgically induced in C57BL/6JJcl mice, and ADSCs with/without shRNA-92a-3p transduction were intra-articularly injected for the assessment of cartilage damage.

Results

SMAD6 and SMAD7 were predicted as direct targets of miR-92a-3p by TargetScan and luciferase reporter assay. Transfection of the miR-92a-3p mimic resulted in a decrease in SMAD6 and SMAD7 levels and an increase in phospho-SMAD2/3, phospho-SMAD1/5/9, SOX9, collagen type II, and aggrecan levels in ADSCs. Furthermore, shRNA-92a-3p decreased SMAD6 and SMAD7 levels, and increased phospho-SMAD2/3, phospho-SMAD1/5/9, SOX9, collagen type II, and aggrecan levels in ADSCs and osteoarthritic chondrocytes. Additionally, ADSC-shRNA-92a-3p-EVs reduced the rate of decrease of SOX9, collagen type II, and aggrecan in osteoarthritic chondrocytes. In mice with surgically induced OA, shRNA-92a-3p-treated ADSCs alleviated cartilage damage more effectively than nontreated ADSCs.

Conclusions

miR-92a-3p and shRNA-92a-3p exhibit therapeutic effects in treating OA by targeting SMAD6 and SMAD7, thereby enhancing TGF-β signaling.

This is a preview of subscription content, log in via an institution to check access.

Access this article

Price excludes VAT (USA)
Tax calculation will be finalised during checkout.

Instant access to the full article PDF.

Fig. 1
Fig. 2
Fig. 3
Fig. 4
Fig. 5
Fig. 6

Similar content being viewed by others

References

  1. Richard FL, Steven RG, Carla RS, Mary BG (2012) Osteoarthritis: a disease of the joint as an organ. Arthritis Rheum 64:1697–1707

    Article  Google Scholar 

  2. Mary BG, Steven RG (2007) Osteoarthritis. J Cell Physiol 213:626–634

    Article  Google Scholar 

  3. Maroudas AI (1976) Balance between swelling pressure and collagen tension in normal and degenerate cartilage. Nature 260:808–809

    Article  ADS  CAS  PubMed  Google Scholar 

  4. Bollet AJ, Nance JL (1966) Biochemical findings in normal and osteoarthritic articular cartilage. II. Chondroitin sulfate concentration and chain length, water, and ash content. J Clin Invest 45:1170–1177

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  5. Kyung-Su P, Min-Jung P, Mi-La C, Seung-Ki K, Ji HJ, Hyeok-Jae K, Sung-Hwan P, Ho-Youn K (2009) Type II collagen oral tolerance; mechanism and role in collagen-induced arthritis and rheumatoid arthritis. Mod Rheumatol 19:581–589

    Article  Google Scholar 

  6. Fulya B, Onur A, Merih O, Funda T, Ozge B, Ozkan A (2016) Effects of native type ii collagen treatment on knee osteoarthritis: a randomized controlled trial. Eurasian J Med 48:95–101

    Article  Google Scholar 

  7. Chris K, Liwen C, Yao Jiong W, Albert JY, Burton BY (2002) Structure and function of aggrecan. Cell Res 12:19–32

    Article  Google Scholar 

  8. Doege KJ, Sasaki M, Kimura T, Yamada Y (1991) Complete coding sequence and deduced primary structure of the human cartilage large aggregating proteoglycan, aggrecan. Human-specific repeats, and additional alternatively spliced forms. J Biol Chem 266:894–902

    Article  CAS  PubMed  Google Scholar 

  9. Roughley P, Martens D, Rantakokko J, Alini M, Mwale F, Antoniou J (2006) The involvement of aggrecan polymorphism in degeneration of human intervertebral disc and articular cartilage. Eur Cell Mater 11:1–7

    Article  CAS  PubMed  Google Scholar 

  10. Berenbaum F (2013) Osteoarthritis as an inflammatory disease (osteoarthritis is not osteoarthrosis!). Osteoarthritis Cartilage 21:16–21

    Article  CAS  PubMed  Google Scholar 

  11. Glyn-Jones S, Palmer AJR, Agricola R, Price AJ, Vincent TL, Weinans H, Carr AJ (2015) Osteoarthritis. The Lancet 386:376–387

    Article  CAS  Google Scholar 

  12. Yves-Marie P, Lars R, Rosanna F, Oliver P, Christophe D et al (2016) Adipose mesenchymal stromal cell-based therapy for severe osteoarthritis of the knee: a phase I dose-escalation trial. Stem Cells Transl Med 5:847–856

    Article  Google Scholar 

  13. Chris HJ, Young GL, Won HS, Hyang K, Jee WC, Eui CJ, Ji EK, Hackjoon S, Ji SS, Il SS, Jeong CR, Sohee O, Kang SY (2014) Intra-articular injection of mesenchymal stem cells for the treatment of osteoarthritis of the knee: a proof-of-concept clinical trial. Stem Cells 32:1254–1266

    Article  Google Scholar 

  14. Sun Q, Zhang L, Xu T, Ying J, Xia B, Jing H, Tong P (2018) Combined use of adipose derived stem cells and TGF-β3 microspheres promotes articular cartilage regeneration in vivo. Biotech Histochem 93:168–176

    Article  CAS  PubMed  Google Scholar 

  15. Li-Bo J, Soomin L, Yang W, Qin-Tong X, De-Hua M, Jian Z (2016) Adipose-derived stem cells induce autophagic activation and inhibit catabolic response to pro-inflammatory cytokines in rat chondrocytes. Osteoarthritis Cartilage 24:1071–1081

    Article  Google Scholar 

  16. Victoria LJ, David JH (2014) The epidemiology of osteoarthritis. Best Pract Res Clin Rheumatol 28:5–15

    Article  Google Scholar 

  17. Jai-Hong C, Chieh-Cheng H, Shan-Ling H, Wen-Yi C, Yi-No W, Chun-En AK, Tsai-Chin H, Li-Yen S, Shun-Wun J (2021) Adipose-derived mesenchymal stem cells-conditioned medium modulates the expression of inflammation induced bone morphogenetic protein-2, -5 and -6 as well as compared with shockwave therapy on rat knee osteoarthritis. Biomedicines 9:1399

    Article  Google Scholar 

  18. Shuangpeng J, Guangzhao T, Xu L, Zhen Y, Fuxin W, Zhuang T, Bo H, Fu W, Kangkang Z, Zhiqiang S, Xiang S, Shuyun L, Weimin G, Quanyi G (2021) Research progress on stem cell therapies for articular cartilage regeneration. Stem Cells Int 2021:8882505

    Google Scholar 

  19. Xiaoting L, Yue D, Yuelin Z, Hung-Fat T, Qizhou L (2014) Paracrine mechanisms of mesenchymal stem cell-based therapy: current status and perspectives. Cell Transplant 23:1045–1059

    Article  Google Scholar 

  20. Liwen C, Edward ET, Philip YGW, Yaojiong W (2008) Paracrine factors of mesenchymal stem cells recruit macrophages and endothelial lineage cells and enhance wound healing. PLoS ONE 3:e1886

    Article  Google Scholar 

  21. Abir NM, Omar SET, Ashraf AS, Laila AR, Dina S, Abeer MES (2011) Homing and reparative effect of intra-articular injection of autologus mesenchymal stem cells in osteoarthritic animal model. BMC Musculoskelet Disord 12:259

    Article  Google Scholar 

  22. Mokbel A, El-Tookhy O, Shamaa AA, Sabry D, Rashed L, Mostafa A (2011) Homing and efficacy of intra-articular injection of autologous mesenchymal stem cells in experimental chondral defects in dogs. Clin Exp Rheumatol 29:275–284

    CAS  PubMed  Google Scholar 

  23. Jonathan CB, Gordana VN (2016) Should we use cells, biomaterials, or tissue engineering for cartilage regeneration? Stem Cell Res Ther 7:56

    Article  Google Scholar 

  24. Sheng Z, Song C, Qing J, Ming P (2019) Determinants of stem cell lineage differentiation toward chondrogenesis versus adipogenesis. Cell Mol Life Sci 76:1653–1680

    Article  Google Scholar 

  25. Tao W, Puwapong N, Christopher AS, Aixin C, Timothy EH, Susan JK (2019) Enhanced chondrogenesis from human embryonic stem cells. Stem cell research 39:101497

    Article  Google Scholar 

  26. Yoo JU, Barthel TS, Nishimura K, Solchaga L, Caplan AI, Goldberg VM, Johnstone B (1998) The chondrogenic potential of human bone-marrow-derived mesenchymal progenitor cells. J Bone Joint Surg Am 80:1745–1757

    Article  CAS  PubMed  Google Scholar 

  27. Hongjun K, Shibi L, Jiang P, Qiang Y, Shuyun L, Li Z, Jingxiang H, Xiang S, Bin Z, Aiyuan W, Wenjing X, Quanyi G, Qing S (2015) In vivo construction of tissue-engineered cartilage using adipose-derived stem cells and bioreactor technology. Cell Tissue Bank 16:123–133

    Article  Google Scholar 

  28. Caldwell KL, Wang J (2015) Cell-based articular cartilage repair: the link between development and regeneration. Osteoarthritis Cartilage 23:351–362

    Article  CAS  PubMed  Google Scholar 

  29. Richard WC, Erik JS (2009) Origins and mechanisms of miRNAs and siRNAs. Cell 136:642–655

    Article  Google Scholar 

  30. Jun L, Ming-Liang J, Xue-Jun Z, Pei-Liang S, Hao W, Chen W, Hee-Jeong I (2017) MicroRNA-218-5p as a potential target for the treatment of human osteoarthritis. Mol Ther 25:2676–2688

    Article  Google Scholar 

  31. Wei Z, Biao Z, Chi Z, Congfeng L, Yulin Z (2018) miR-373 regulates inflammatory cytokine-mediated chondrocyte proliferation in osteoarthritis by targeting the P2X7 receptor. FEBS Open Bio 8:325–331

    Article  Google Scholar 

  32. Changhe H, Zibo Y, Yan K, Ziji Z, Ming F, Aishan H, Zhiqi Z, Weiming L (2015) MiR-193b regulates early chondrogenesis by inhibiting the TGF-beta2 signaling pathway. FEBS Lett 589:1040–1047

    Article  Google Scholar 

  33. Zhen Y, Jie H, Zhen-Ming H (2015) MicroRNA expression profiles in human adipose-derived stem cells during chondrogenic differentiation. Int J Mol Med 35:579–586

    Article  Google Scholar 

  34. Zhang Z, Kang Y, Zhang Z, Zhang H, Duan X, Liu J, Li X, Liao W (2012) Expression of microRNAs during chondrogenesis of human adipose-derived stem cells. Osteoarthritis Cartilage 20:1638–1646

    Article  CAS  PubMed  Google Scholar 

  35. Guping M, Ziji Z, Shu H, Zhiqi Z, Zongkun C, Zhiyu H, Weiming L, Yan K (2018) Exosomes derived from miR-92a-3p overexpressing human mesenchymal stem cells enhance chondrogenesis and suppress cartilage degradation via targeting WNT5A. Stem Cell Res Ther 9:247

    Article  Google Scholar 

  36. Mao G, Zhang Z, Huang Z, Chen W, Huang G, Meng F, Zhang Z, Kang Y (2017) MicroRNA-92a-3p regulates the expression of cartilage-specific genes by directly targeting histone deacetylase 2 in chondrogenesis and degradation. Osteoarthritis Cartilage 25:521–532

    Article  CAS  PubMed  Google Scholar 

  37. Changhe H, Ziji Z, Zhiqi Z, Peihui W, Xiaoyi Z, Ming F, Puyi S, Yan K, Weiming L (2015) Presence and function of microRNA-92a in chondrogenic ATDC5 and adipose-derived mesenchymal stem cells. Mol Med Rep 12:4877–4886

    Article  Google Scholar 

  38. Joshua M, Delaine KC, Przemek AG, Prabhu M, Jack FM, Sophia IS, Francesca S, Joshua MH, Susmita S, Roger JH, Kevin DC (2018) Exosomal microRNA-21-5p mediates mesenchymal stem cell paracrine effects on human cardiac tissue contractility. Circ Res 122:933–944

    Article  Google Scholar 

  39. Shi-Cong T, Ting Y, Yue-Lei Z, Wen-Jing Y, Shang-Chun G, Chang-Qing Z (2017) Exosomes derived from miR-140-5p-overexpressing human synovial mesenchymal stem cells enhance cartilage tissue regeneration and prevent osteoarthritis of the knee in a rat model. Theranostics 7:180–195

    Article  Google Scholar 

  40. Caroline AS, Wayne SR, Kevin WE (2012) NIH image to imageJ: 25 years of image analysis. Nat Methods 9:671–675

    Article  Google Scholar 

  41. Guangju Z, Jules D, Proton R (2015) TGF-β signal transduction pathways and osteoarthritis. Rheumatol Int 35:1283–1292

    Article  Google Scholar 

  42. Erfan AE, Ming L, Patricia EH, Jules D, Glynn M, Andrew F, Roger G, Proton R, Guangju Z (2015) Overexpression of MMP13 in human osteoarthritic cartilage is associated with the SMAD-independent TGF-β signalling pathway. Arthritis Res Ther 17:264

    Article  Google Scholar 

  43. Jie S, Shan L, Di C (2014) TGF-β signaling and the development of osteoarthritis. Bone Res 2:14002

    Article  Google Scholar 

  44. Ude CC, Chen HC, Norhamdan MY, Azizi BM, Aminuddin BS, Ruszymah BHI (2017) The evaluation of cartilage differentiations using transforming growth factor beta3 alone and with combination of bone morphogenetic protein-6 on adult stem cells. Cell Tissue Banking 18:355–367

    Article  CAS  PubMed  Google Scholar 

  45. Zhi-Gao Y, Ruo-Fu T, Yi-Ying Q, Wei-Ping C, Yan X, Li-Dong W (2020) Restoration of cartilage defects using a super paramagnetic iron oxide-labeled adipose-derived mesenchymal stem cell and TGF-β3-loaded bilayer PLGA construct. Regen Med 15:1735–1747

    Article  Google Scholar 

  46. Serra R, Johnson M, Filvaroff EH, LaBorde J, Sheehan DM, Derynck R, Moses HL (1997) Expression of a truncated, kinase-defective TGF-beta type II receptor in mouse skeletal tissue promotes terminal chondrocyte differentiation and osteoarthritis. J Cell Biol 139:541–552

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  47. Baek D, Villén J, Shin C, Camargo FD, Gygi SP, Bartel DP (2008) The impact of microRNAs on protein output. Nature 455:64–71

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  48. Selbach M, Schwanhäusser B, Thierfelder N, Fang Z, Khanin R, Rajewsky N (2008) Widespread changes in protein synthesis induced by microRNAs. Nature 455:58–63

    Article  ADS  CAS  PubMed  Google Scholar 

  49. Derynck R, Zhang Y, Feng XH (1998) Transcriptional activators of TGF-β Responses: SMADs. Cell 95:737–740

    Article  CAS  PubMed  Google Scholar 

  50. Joan M, Joan S, David W (2005) SMAD transcription factors. Genes Dev 19:2783–2810

    Article  Google Scholar 

  51. Joan M (2012) TGFβ signalling in context. Nat Rev Mol Cell Biol 13:616–620

    Article  Google Scholar 

  52. Young-Kook K, Boseon K, Kim VN (2016) Re-evaluation of the roles of DROSHA, Export in 5, and DICER in microRNA biogenesis. Proc Natl Acad Sci USA 113:E1881–E1889

    Google Scholar 

Download references

Acknowledgements

This work is supported by Japan Society for the Promotion of Science KAKENHI [grant numbers #19K22700, #21H03136]. P0 Human ADSCs were kindly provided by a previous laboratory member Dr. Inaki (Miyagi National Hospital). P0 mouse ADSCs were kindly provided by Dr. IGARASHI (the department of Sensory and Motor System Medicine, the University of Tokyo). P0 Human OA chondrocytes were kindly provided by Dr. Saito (the department of orthopedic surgery and spinal surgery, the University of Tokyo Hospital).

Author information

Authors and Affiliations

Authors

Contributions

All authors contributed to the study conception and design. CZ prepared materials, performed the majority of the experiments described in this manuscript, and participated in the analysis of the histological data. AH and KH supervised the studies, provided technical support, and directed the interpretations of the result. Also, the first draft of the manuscript was written by CZ and all authors commented on previous versions of the manuscript. All authors read and approved the final manuscript.

Corresponding author

Correspondence to Atsuhiko Hikita.

Ethics declarations

Conflict of interest

Atsuhiko Hikita held an endowed chair supported by FUJISOFT INCORPORATED (until 31 October 2020) and an endowed chair supported by CPC corporation, Kyowa Co., Ltd., Kanto Chemical Co., Inc., and Nichirei Corporation (from 1 July, 2021, to 30 June, 2022), and is affiliated with the social cooperation program of Kohjin Bio Co., Ltd. (since 1 July, 2022). All other authors have no conflicts of interest.

Additional information

Publisher's Note

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

Supplementary Information

Below is the link to the electronic supplementary material.

Supplementary file1 (DOCX 287 KB)

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Zheng, C., Hoshi, K. & Hikita, A. miR-92a-3p-inspired shRNA exhibits pro-chondrogenic and chondrocyte protective effects in osteoarthritis treatment through targeting SMAD6/7. J Bone Miner Metab 42, 1–16 (2024). https://doi.org/10.1007/s00774-023-01474-3

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1007/s00774-023-01474-3

Keywords

Navigation