Skip to main content

Advertisement

Log in

Fufang Zhenshu Tiaozhi capsule enhances bone formation and safeguards against glucocorticoid-induced osteoporosis through innovative Mekk2-mediated β-catenin deubiquitination

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

Abstract

Introduction

Bone homeostasis depends on the regulation of β-catenin in osteoblasts. Glucocorticoids (GCs) are known to diminish β-catenin activity via Wnt pathway signaling, leading to osteoporosis. Conversely, activating β-catenin in osteoblasts through mitogen-activated protein kinase kinase kinase 2 (Mekk2) offers an innovative approach to combat GC-induced osteoporosis (GIOP). Fufang Zhenshu Tiaozhi (FTZ) capsules have shown effectiveness in treating GIOP, but the mechanisms behind this are still unclear.

Materials and methods

In this study, Mekk2 knockout mice (Mekk2−/−) was generated by CRISPR/Cas9. These mice were then subjected to Alcian Blue-Alizarin Red staining and immunofluorescence to assess their bone and cartilage development. To establish models of GIOP, both Mekk2−/− and wild-type (WT) mice were treated with dexamethasone (DXMS) and subsequently given FTZ capsules. We analyzed the resulting phenotypic changes in these mice using Micro-CT scans and histomorphological studies. Primary osteoblasts, isolated from both Mekk2−/− and WT mice, underwent qRT-PCR to measure key osteogenesis markers, including Runx2, Sp7, Bgalp, Col1a1 and Alp. Cells were then exposed to treatments with either FTZ or Wnt3a and the phosphorylation levels of β-catenin and Mekk2, along with the protein expression of Runx2, were evaluated using Western blotting and immunoprecipitation. Additionally, C3H10T1/2 cells transfected with TOPflash-luciferase and Renilla luciferase reporters were treated with FTZ and Wnt3a to measure β-catenin activity.

Results

In our study, administering FTZ in vivo effectively prevented bone loss typically induced by GCs. However, it's important to note that this protective effect was substantially reduced in mice lacking Mekk2. Additionally, FTZ showed a significant ability to enhance osteogenic differentiation in primary osteoblasts, doing so by altering the expression of Mekk2. Intriguingly, the impact of FTZ on Mekk2 appears to function through a pathway separate from the traditional Wnt signaling route. Furthermore, our findings indicate that FTZ also promotes the deubiquitination of β-catenin, contributing further to its positive effects on bone health.

Conclusions

This study suggests that FTZ plays a significant role in protecting bone mass in cases of GIOP. The mechanism through which FTZ confers this benefit involves the activation of Mekk2/β-catenin signaling pathways, which represents a promising alternative strategy to counteract the deleterious effects of GIOP by augmenting osteoblastogenesis.

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
Fig. 7

Similar content being viewed by others

Data availability

The datasets used and/or analysed during the current study are available from the corresponding author on reasonable request.

Abbreviations

GC:

Glucocorticoid

FTZ:

Fufang Zhenshu Tiaozhi

GIOP:

Glucocorticoid-induced osteoporosis

Mekk2:

Mitogen-activated protein kinase kinase kinase 2

DXMS:

Dexamethasone

WT:

Wild type

ALP:

Alkaline phosphatase

References

  1. Buckley L, Humphrey MB (2018) Glucocorticoid-induced osteoporosis. N Engl J Med 379:2547–2556. https://doi.org/10.1056/NEJMcp1800214

    Article  PubMed  Google Scholar 

  2. Chotiyarnwong P, McCloskey EV (2020) Pathogenesis of glucocorticoid-induced osteoporosis and options for treatment. Nat Rev Endocrinol 16:437–447. https://doi.org/10.1038/s41574-020-0341-0

    Article  PubMed  Google Scholar 

  3. Gregson CL, Armstrong DJ, Bowden J et al (2022) UK clinical guideline for the prevention and treatment of osteoporosis. Arch Osteoporos. https://doi.org/10.1007/s11657-022-01061-5

    Article  PubMed  PubMed Central  Google Scholar 

  4. Adami G, Saag KG (2019) Glucocorticoid-induced osteoporosis update. Curr Opin Rheumatol 31:388–393. https://doi.org/10.1097/BOR.0000000000000608

    Article  CAS  PubMed  Google Scholar 

  5. Watts NB, GLOW investigators, (2014) Insights from the global longitudinal study of osteoporosis in women (GLOW). Nat Rev Endocrinol 10:412–422. https://doi.org/10.1038/nrendo.2014.55

    Article  PubMed  Google Scholar 

  6. Silverman S, Curtis J, Saag K et al (2015) International management of bone health in glucocorticoid-exposed individuals in the observational GLOW study. Osteoporos Int 26:419–420. https://doi.org/10.1007/s00198-014-2883-2

    Article  CAS  PubMed  Google Scholar 

  7. Meszaros K, Patocs A (2020) Glucocorticoids Influencing Wnt/β-Catenin Pathway; Multiple Sites. Heterogeneous Eff Mol 25:1489. https://doi.org/10.3390/molecules25071489

    Article  CAS  Google Scholar 

  8. Komori T (2016) Glucocorticoid signaling and bone biology. Horm Metab Res 48:755–763. https://doi.org/10.1055/s-0042-110571

    Article  CAS  PubMed  Google Scholar 

  9. Greenblatt MB, Shin DY, Oh H et al (2016) MEKK2 mediates an alternative β-catenin pathway that promotes bone formation. Proc Natl Acad Sci U S A 113:E1226-1235. https://doi.org/10.1073/pnas.1600813113

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Yamashita M, Ying S-X, Zhang G-M et al (2005) Ubiquitin ligase Smurf1 controls osteoblast activity and bone homeostasis by targeting MEKK2 for degradation. Cell 121:101–113. https://doi.org/10.1016/j.cell.2005.01.035

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. Lawson LY, Brodt MD, Migotsky N et al (2022) Osteoblast-specific Wnt secretion is required for skeletal homeostasis and loading-induced bone formation in adult mice. J Bone Miner Res 37:108–120. https://doi.org/10.1002/jbmr.4445

    Article  CAS  PubMed  Google Scholar 

  12. Karner CM, Long F (2017) Wnt signaling and cellular metabolism in osteoblasts. Cell Mol Life Sci 74:1649–1657. https://doi.org/10.1007/s00018-016-2425-5

    Article  CAS  PubMed  Google Scholar 

  13. Chen X, Wang J, Tang L et al (2022) The therapeutic effect of Fufang Zhenshu Tiaozhi (FTZ) on osteoclastogenesis and ovariectomized-induced bone loss: evidence from network pharmacology, molecular docking and experimental validation. Aging (Albany NY) 14:5727–5748. https://doi.org/10.18632/aging.204172

    Article  CAS  PubMed  Google Scholar 

  14. Luo D, Li J, Chen K et al (2018) Untargeted metabolomics reveals the protective effect of Fufang Zhenshu Tiaozhi (FTZ) on aging-induced osteoporosis in mice. Front Pharmacol 9:1483. https://doi.org/10.3389/fphar.2018.01483

    Article  CAS  PubMed  Google Scholar 

  15. Huang X, Zhan H, Yang J et al (2021) Long-term effect of Zhenzhu Tiaozhi Capsule (FTZ) on hyperlipidemia: 2-year results from a retrospective study using electronic medical records. Evid Based Complement Alternat Med 2021:6264414. https://doi.org/10.1155/2021/6264414

    Article  PubMed  PubMed Central  Google Scholar 

  16. Luo D, Chen K, Li J et al (2020) Gut microbiota combined with metabolomics reveals the metabolic profile of the normal aging process and the anti-aging effect of FuFang Zhenshu TiaoZhi(FTZ) in mice. Biomed Pharmacother 121:109550. https://doi.org/10.1016/j.biopha.2019.109550

    Article  CAS  PubMed  Google Scholar 

  17. Diao H, Cheng J, Huang X et al (2022) The Chinese medicine Fufang Zhenzhu Tiaozhi capsule protects against atherosclerosis by suppressing EndMT via modulating Akt1/β-catenin signaling pathway. J Ethnopharmacol 293:115261. https://doi.org/10.1016/j.jep.2022.115261

    Article  CAS  PubMed  Google Scholar 

  18. Wang H, Tan H, Zhan W et al (2021) Molecular mechanism of Fufang Zhenzhu Tiaozhi capsule in the treatment of type 2 diabetes mellitus with nonalcoholic fatty liver disease based on network pharmacology and validation in minipigs. J Ethnopharmacol 274:114056. https://doi.org/10.1016/j.jep.2021.114056

    Article  CAS  PubMed  Google Scholar 

  19. Guo J, Bei W, Hu Y et al (2011) A new TCM formula FTZ lowers serum cholesterol by regulating HMG-CoA reductase and CYP7A1 in hyperlipidemic rats. J Ethnopharmacol 135:299–307. https://doi.org/10.1016/j.jep.2011.03.012

    Article  PubMed  Google Scholar 

  20. Xu Y, Tang J, Guo Q et al (2021) Traditional Chinese medicine formula FTZ protects against polycystic ovary syndrome through modulating adiponectin-mediated fat-ovary crosstalk in mice. J Ethnopharmacol 268:113587. https://doi.org/10.1016/j.jep.2020.113587

    Article  CAS  PubMed  Google Scholar 

  21. Komori T (2015) Animal models for osteoporosis. Eur J Pharmacol 759:287–294. https://doi.org/10.1016/j.ejphar.2015.03.028

    Article  CAS  PubMed  Google Scholar 

  22. Lane NE (2019) Glucocorticoid-Induced Osteoporosis: New Insights into the Pathophysiology and Treatments. Curr Osteoporos Rep 17:1–7. https://doi.org/10.1007/s11914-019-00498-x

    Article  PubMed  PubMed Central  Google Scholar 

  23. Canalis E, Mazziotti G, Giustina A, Bilezikian JP (2007) Glucocorticoid-induced osteoporosis: pathophysiology and therapy. Osteoporos Int 18:1319–1328. https://doi.org/10.1007/s00198-007-0394-0

    Article  CAS  PubMed  Google Scholar 

  24. Kenkre JS, Bassett J (2018) The bone remodelling cycle. Ann Clin Biochem 55:308–327. https://doi.org/10.1177/0004563218759371

    Article  CAS  PubMed  Google Scholar 

  25. An J, Yang H, Zhang Q et al (2016) Natural products for treatment of osteoporosis: the effects and mechanisms on promoting osteoblast-mediated bone formation. Life Sci 147:46–58. https://doi.org/10.1016/j.lfs.2016.01.024

    Article  CAS  PubMed  Google Scholar 

  26. Wang T, Liu Q, Tjhioe W et al (2017) Therapeutic potential and outlook of alternative medicine for osteoporosis. Curr Drug Targets 18:1051–1068. https://doi.org/10.2174/1389450118666170321105425

    Article  CAS  PubMed  Google Scholar 

  27. Jiang Y, Lu Y, Jiang X et al (2020) Glucocorticoids induce osteoporosis mediated by glucocorticoid receptor-dependent and -independent pathways. Biomed Pharmacother 125:109979. https://doi.org/10.1016/j.biopha.2020.109979

    Article  CAS  PubMed  Google Scholar 

  28. Messina OD, Vidal M, Torres JAM et al (2022) Evidence based Latin American guidelines of clinical practice on prevention, diagnosis, management and treatment of glucocorticoid induced osteoporosis. A 2022 update : This manuscript has been produced under the auspices of the committee of national societies (CNS) and the committee of scientific advisors (CSA) of the international osteoporosis foundation (IOF). Aging Clin Exp Res 34:2591–2602. https://doi.org/10.1007/s40520-022-02261-2

    Article  PubMed  Google Scholar 

  29. Latifyan S, Genot MT, Klastersky J (2016) Bisphosphonate-related osteonecrosis of the jaw: a review of the potential efficacy of low-level laser therapy. Support Care Cancer 24:3687–3693. https://doi.org/10.1007/s00520-016-3139-9

    Article  CAS  PubMed  Google Scholar 

  30. Janovská Z (2012) Bisphosphonate-related osteonecrosis of the jaws. A severe side effect of bisphosphonate therapy. Acta Medica (Hradec Kralove) 55:111–115. https://doi.org/10.14712/18059694.2015.47

    Article  PubMed  Google Scholar 

  31. Jiang L, Dong J, Wei J, Liu L (2022) Comparison of denosumab and oral bisphosphonates for the treatment of glucocorticoid-induced osteoporosis: a systematic review and meta-analysis. BMC Musculoskelet Disord 23:1027. https://doi.org/10.1186/s12891-022-05997-0

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. Otto S, Pautke C, Van den Wyngaert T et al (2018) Medication-related osteonecrosis of the jaw: prevention, diagnosis and management in patients with cancer and bone metastases. Cancer Treat Rev 69:177–187. https://doi.org/10.1016/j.ctrv.2018.06.007

    Article  PubMed  Google Scholar 

  33. Butz H, Mészáros K, Likó I, Patocs A (2021) Wnt-Signaling Regulated by Glucocorticoid-Induced miRNAs. Int J Mol Sci 22:11778. https://doi.org/10.3390/ijms222111778

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. Rossini M, Gatti D, Adami S (2013) Involvement of WNT/β-catenin signaling in the treatment of osteoporosis. Calcif Tissue Int 93:121–132. https://doi.org/10.1007/s00223-013-9749-z

    Article  CAS  PubMed  Google Scholar 

  35. Saidak Z, Marie PJ (2012) Strontium signaling: molecular mechanisms and therapeutic implications in osteoporosis. Pharmacol Ther 136:216–226. https://doi.org/10.1016/j.pharmthera.2012.07.009

    Article  CAS  PubMed  Google Scholar 

  36. Wan L, Zou W, Gao D et al (2011) Cdh1 regulates osteoblast function through an APC/C-independent modulation of Smurf1. Mol Cell 44:721–733. https://doi.org/10.1016/j.molcel.2011.09.024

    Article  CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

Not applicable.

Funding

This study was supported in part by the National Natural Science Foundation of China (No. 81704098, 82305265), Science and Technology Planning Project of Yunfu (No. 2020A090402), Guangdong Science and Technology Innovation Strategy Special Funding "Great Special Project + Task List" (No. 2020A090402), and China Postdoctoral Science Foundation (grant number: 2023M730809). This sponsor hasn’t participated in the design of the study or collection, analysis, or interpretation of data or in writing manuscript.

Author information

Authors and Affiliations

Authors

Contributions

Ping Sun and Qunwei Dong conceived and supervised the study; Guoju Hong designed experiments; Guoju Hong, Jiangyan Wang and Dongdong Ge performed experiments; Lin Tang provided new tools and reagents; Tianyu Zhou and Youhong Xie analysed data; Guoju Hong wrote the manuscript; Ping Sun made manuscript revisions.

Corresponding authors

Correspondence to Qunwei Dong or Ping Sun.

Ethics declarations

Conflict of interests

The authors declare that they have no competing interests.

Ethics approval and consent to participate

The animal protocol was approved by the animal ethics broad of Guangzhou University of Chinese Medicine where the animal study was performed.

Consent for publication

Not applicable.

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 (TIF 1074 KB)

Supplementary file2 (TIF 3207 KB)

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Hong, G., Tang, L., Zhou, T. et al. Fufang Zhenshu Tiaozhi capsule enhances bone formation and safeguards against glucocorticoid-induced osteoporosis through innovative Mekk2-mediated β-catenin deubiquitination. J Bone Miner Metab (2024). https://doi.org/10.1007/s00774-024-01516-4

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1007/s00774-024-01516-4

Keywords

Navigation