Skip to main content

Advertisement

Log in

hUMSCs Transplantation Regulates AMPK/NR4A1 Signaling Axis to Inhibit Ovarian Fibrosis in POI Rats

  • Published:
Stem Cell Reviews and Reports Aims and scope Submit manuscript

Abstract

Background

The mechanism of human Umbilical Cord Mesenchymal Stem Cells (hUMSCs) transplantation to improve ovarian function in the rats with Premature Ovarian Insufficiency (POI) is still unclear. The aim of this study is to investigate the signal axis mechanism that is involved in the ovarian function recovery of POI rats following hUMSCs transplantation.

Methods

The rat model with POI was established by intraperitoneal injection of cisplatin. The hUMSCs were transplanted by caudal vein injection into POI rats. Hematoxylin–eosin (H&E) staining was performed to examine the morphology of rat ovarian tissue. Masson staining, Sirus red staining and immunofluorescence were used to observe the fibrosis extent of ovarian tissue. The levels of serum sex hormones and the expression of fibrosis related markers in ovarian tissues were measured by enzyme-linked immunosorbent assay (ELISA). The expression of NR4A1, Phospho-NR4A1 and AMP-activated protein kinase (AMPK) signaling in rat ovarian tissues was measured by immunohistochemistry and immunofluorescence. The role of AMPK/NR4A1 signaling axis in the regulation of ovarian function recovery in POI rats following hUMSCs transplantation was further investigated by adenovirus and siRNA intervention in isolated stromal cells.

Results

The results showed that the hUMSCs transplantation significantly inhibited ovarian tissue fibrosis and restored the ovarian function in POI rats. The level of NR4A1 and AMPK expression in ovarian tissue of POI rats after hUMSCs transplantation was significantly increased compared with the control group. In the cultured ovarian stromal cells, the similar results were obtained on the expression of NR4A1 and its regulation on fibrosis related molecular markers in Cisplatin (CDDP) damaged stromal cells following hUMSCs supernatant treatment. Both hUMSCs supernatant treatment and the addition of AMPK inhibitors increased NR4A1 expression in stromal cells. And after NR4A1 molecular intervention, fibrosis-related indicators in stromal cells changed. The data suggests that the AMPK/NR4A1 signaling axis is involved in the ovarian function changes in POI rats following hUMSCs transplantation.

Conclusion

The data from this study indicate that the inhibition of tissue fibrosis and recovery of ovarian function is regulated by AMPK/NR4A1 signaling axis in POI rats following hUMSCs transplantation.

Graphical Abstract

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

All data generated and/or analyzed during this study are included in this published article.

Abbreviations

hUMSCs :

Human umbilical cord mesenchymal stem cells

POI :

Premature ovarian insufficiency

H&E :

Hematoxylin-eosin

ELISA :

Enzyme-linked immunosorbent assay

AMPK :

AMP-activated protein kinase

CDDP :

Cisplatin

E2 :

Estradiol

HRT :

Hormone replacement therapy

Collagen I :

Collagen Type I

Collagen III :

Collagen Type III

NR4A1 :

Nuclear receptor 4A1

FSH :

Follicle stimulating hormone

LH :

Luteinizing hormone

P :

Progesterone

DAB :

Diaminobenzidine

IRS :

Immune response score

SD :

Standard deviation

ANOVA :

Analysis of variance

CTGF :

Connective tissue growth factor

MSCs :

Mesenchymal stem cells

FN :

Fibronectin

References

  1. Nozaki, Y., Furubo, E., Matsuno, T., et al. (2009). Collaborative work on evaluation of ovarian toxicity. 6) Two- or four-week repeated-dose studies and fertility study of cisplatin in female rats. The Journal of Toxicological Sciences, 34(Suppl 1), SP73–SP81.

    CAS  PubMed  Google Scholar 

  2. Kalich-Philosoph, L., Roness, H., Carmely, A., et al. (2013). Cyclophosphamide triggers follicle activation and “burnout”; AS101 prevents follicle loss and preserves fertility. Science Translational Medicine, 5(185), 185ra62.

    PubMed  Google Scholar 

  3. Shirota, M., Soda, S., Katoh, C., et al. (2003). Effects of reduction of the number of primordial follicles on follicular development to achieve puberty in female rats. Reproduction, 125(1), 85–94.

    CAS  PubMed  Google Scholar 

  4. European Society for Human Reproduction and Embryology (ESHRE) Guideline Group on POI, Webber, L., Davies, M., et al. (2016). ESHRE Guideline: management of women with premature ovarian insufficiency. Human Reproduction, 31(5), 926–937.

  5. Jankowska, K. (2017). Premature ovarian failure. Prz Menopauzalny., 16(2), 51–56.

    PubMed  PubMed Central  Google Scholar 

  6. Iorio, R., Castellucci, A., Ventriglia, G., et al. (2014). Ovarian toxicity: From environmental exposure to chemotherapy. Current Pharmaceutical Design, 20(34), 5388–5397.

    CAS  PubMed  Google Scholar 

  7. Ebrahimi, M., & Akbari, A. F. (2011). Pathogenesis and causes of premature ovarian failure: An update. International Journal of Fertility and Sterility, 5(2), 54–65.

    PubMed  PubMed Central  Google Scholar 

  8. Beral, V., Banks, E., & Reeves, G. (2002). Evidence from randomised trials on the long-term effects of hormone replacement therapy. Lancet, 360(9337), 942–944.

    PubMed  Google Scholar 

  9. Miller, J., Chan, B. K., & Nelson, H. D. (2002). Postmenopausal estrogen replacement and risk for venous thromboembolism: A systematic review and meta-analysis for the U.S. Preventive Services Task Force [published correction appears in Ann Intern Med. 2003 Feb 18;138(4):360.]. Annals of Internal Medicine, 136(9), 680–690.

    CAS  PubMed  Google Scholar 

  10. Ling, L., Feng, X., Wei, T., et al. (2019). Human amnion-derived mesenchymal stem cell (hAD-MSC) transplantation improves ovarian function in rats with premature ovarian insufficiency (POI) at least partly through a paracrine mechanism. Stem Cell Research & Therapy, 10(1), 46.

    CAS  Google Scholar 

  11. Bahrehbar, K., Rezazadeh Valojerdi, M., Esfandiari, F., et al. (2020). Human embryonic stem cell-derived mesenchymal stem cells improved premature ovarian failure. World Journal of Stem Cells, 12(8), 857–878.

    PubMed  PubMed Central  Google Scholar 

  12. Yoon, S. Y., Yoon, J. A., Park, M., et al. (2020). Recovery of ovarian function by human embryonic stem cell-derived mesenchymal stem cells in cisplatin-induced premature ovarian failure in mice. Stem Cell Research & Therapy, 11(1), 255.

    CAS  Google Scholar 

  13. Cui, L., Bao, H., Liu, Z., et al. (2020). hUMSCs regulate the differentiation of ovarian stromal cells via TGF-β1/Smad3 signaling pathway to inhibit ovarian fibrosis to repair ovarian function in POI rats. Stem Cell Research & Therapy, 11(1), 386.

    CAS  Google Scholar 

  14. Safe, S., Jin, U. H., Hedrick, E., et al. (2014). Minireview: Role of orphan nuclear receptors in cancer and potential as drug targets. Molecular Endocrinology, 28(2), 157–172.

    PubMed  Google Scholar 

  15. Pearen, M. A., & Muscat, G. E. (2010). Minireview: Nuclear hormone receptor 4A signaling: Implications for metabolic disease. Molecular Endocrinology, 24(10), 1891–1903.

    CAS  PubMed  PubMed Central  Google Scholar 

  16. He, L., Yuan, L., Yu, W., et al. (2020). A regulation loop between YAP and NR4A1 balances cell proliferation and apoptosis. Cell Reports, 33(3), 108284.

    CAS  PubMed  Google Scholar 

  17. Mohankumar, K., Li, X., Sung, N., et al. (2020). Bis-indole-derived nuclear receptor 4A1 (NR4A1, Nur77) ligands as inhibitors of endometriosis. Endocrinology, 161(4), bqaa027.

    PubMed  PubMed Central  Google Scholar 

  18. Palumbo-Zerr, K., Zerr, P., Distler, A., et al. (2015). Orphan nuclear receptor NR4A1 regulates transforming growth factor-β signaling and fibrosis. Nature Medicine, 21(2), 150–158.

    CAS  PubMed  Google Scholar 

  19. Yin, N., Zhao, W., Luo, Q., et al. (2018). Restoring ovarian function with human placenta-derived mesenchymal stem cells in autoimmune-induced premature ovarian failure mice mediated by treg cells and associated cytokines. Reproductive Sciences, 25(7), 1073–1082.

    CAS  PubMed  Google Scholar 

  20. Li, J., Mao, Q., He, J., et al. (2017). Human umbilical cord mesenchymal stem cells improve the reserve function of perimenopausal ovary via a paracrine mechanism. Stem Cell Research & Therapy, 8(1), 55.

    Google Scholar 

  21. Chen, Z. Z., & Niu, Y. Y. (2019). Stem cell therapy for Parkinson’s disease using non-human primate models. Zoological Research, 40(5), 349–357.

    PubMed  PubMed Central  Google Scholar 

  22. Voltarelli, J. C., Couri, C. E., Oliveira, M. C., et al. (2011). Stem cell therapy for diabetes mellitus. Kidney International Supplements (2011), 1(3), 94–98.

    Google Scholar 

  23. Huynh, K. (2020). Stem cell therapy improves heart function by triggering an acute immune response. Nature Reviews Cardiology, 17(2), 69.

    PubMed  Google Scholar 

  24. Salwa, K. L. (2021). Engrafted stem cell therapy for Alzheimer’s disease: A promising treatment strategy with clinical outcome. Journal of Controlled Release, 338, 837–857.

    CAS  PubMed  Google Scholar 

  25. Faiella, W., & Atoui, R. (2016). Therapeutic use of stem cells for cardiovascular disease. Clinical and Translational Medicine, 5(1), 34.

    PubMed  PubMed Central  Google Scholar 

  26. Park, T. Y., Oh, J. M., Cho, J. S., et al. (2020). Stem cell-loaded adhesive immiscible liquid for regeneration of myocardial infarction. Journal of Controlled Release, 321, 602–615.

    CAS  PubMed  Google Scholar 

  27. Sun, L., Li, D., Song, K., et al. (2017). Exosomes derived from human umbilical cord mesenchymal stem cells protect against cisplatin-induced ovarian granulosa cell stress and apoptosis in vitro. Science and Reports, 7(1), 2552.

    Google Scholar 

  28. Wang, Z., Wang, Y., Yang, T., et al. (2017). Study of the reparative effects of menstrual-derived stem cells on premature ovarian failure in mice [published correction appears in Stem Cell Res Ther. 2017 Mar 8;8(1):49]. Stem Cell Research & Therapy., 8(1), 11.

    Google Scholar 

  29. Sharma, D., & Bhartiya, D. (2022). Aged mice ovaries harbor stem cells and germ cell nests but fail to form follicles. Journal of Ovarian Research, 15(1), 37.

    CAS  PubMed  PubMed Central  Google Scholar 

  30. Bhartiya, D., Singh, P., Sharma, D., et al. (2022). Very small embryonic-like stem cells (VSELs) regenerate whereas mesenchymal stromal cells (MSCs) rejuvenate diseased reproductive tissues. Stem Cell Reviews and Reports, 18(5), 1718–1727.

    CAS  PubMed  Google Scholar 

  31. Lu, X., Bao, H., Cui, L., et al. (2020). hUMSC transplantation restores ovarian function in POI rats by inhibiting autophagy of theca-interstitial cells via the AMPK/mTOR signaling pathway. Stem Cell Research & Therapy, 11(1), 268.

    CAS  Google Scholar 

  32. Zhang, H., Luo, Q., Lu, X., et al. (2018). Effects of hPMSCs on granulosa cell apoptosis and AMH expression and their role in the restoration of ovary function in premature ovarian failure mice. Stem Cell Research & Therapy, 9(1), 20.

    CAS  Google Scholar 

  33. Wingate, A. D., & Arthur, J. S. (2006). Post-translational control of Nur77. Biochemical Society Transactions, 34(Pt 6), 1107–1109.

    CAS  PubMed  Google Scholar 

  34. Fahrner, T. J., Carroll, S. L., & Milbrandt, J. (1990). The NGFI-B protein, an inducible member of the thyroid/steroid receptor family, is rapidly modified posttranslationally. Molecular and Cellular Biology, 10(12), 6454–6459.

    CAS  PubMed  PubMed Central  Google Scholar 

  35. Li, Q. X., Ke, N., Sundaram, R., et al. (2006). NR4A1, 2, 3–an orphan nuclear hormone receptor family involved in cell apoptosis and carcinogenesis. Histology and Histopathology, 21(5), 533–540.

    CAS  PubMed  Google Scholar 

  36. Zeng, X., Yue, Z., Gao, Y., et al. (2018). NR4A1 is involved in fibrogenesis in ovarian endometriosis. Cellular Physiology and Biochemistry, 46(3), 1078–1090.

    CAS  PubMed  Google Scholar 

  37. Lee, M., Katerelos, M., Gleich, K., et al. (2018). Phosphorylation of Acetyl-CoA Carboxylase by AMPK reduces renal fibrosis and is essential for the anti-fibrotic effect of metformin. Journal of the American Society of Nephrology, 29(9), 2326–2336.

    CAS  PubMed  PubMed Central  Google Scholar 

  38. Chen, D. Q., Wang, Y. N., Vaziri, N. D., et al. (2020). Poricoic acid A activates AMPK to attenuate fibroblast activation and abnormal extracellular matrix remodelling in renal fibrosis. Phytomedicine, 72, 153232.

    CAS  PubMed  Google Scholar 

  39. Li, Y., Liu, M., Song, X., et al. (2020). Exogenous hydrogen sulfide ameliorates diabetic myocardial fibrosis by inhibiting cell aging through SIRT6/AMPK autophagy. Frontiers in Pharmacology, 11, 1150.

    PubMed  PubMed Central  Google Scholar 

  40. Feng, Y., Zhang, Y., & Xiao, H. (2018). AMPK and cardiac remodelling. Science China Life Sciences, 61(1), 14–23.

    CAS  PubMed  Google Scholar 

  41. Han, X., Song, J., Lian, L. H., et al. (2018). Ginsenoside 25-OCH3-PPD promotes activity of LXRs to ameliorate P2X7R-mediated NLRP3 inflammasome in the development of hepatic fibrosis. Journal of Agriculture and Food Chemistry, 66(27), 7023–7035.

    CAS  Google Scholar 

  42. Wang, L., Yuan, D., Zheng, J., et al. (2019). Chikusetsu saponin IVa attenuates isoprenaline-induced myocardial fibrosis in mice through activation autophagy mediated by AMPK/mTOR/ULK1 signaling. Phytomedicine, 58, 152764.

    CAS  PubMed  Google Scholar 

  43. Xiong, Y., Ran, J., Xu, L., et al. (2020). Reactivation of NR4A1 restrains chondrocyte inflammation and ameliorates osteoarthritis in rats. Frontiers in Cell and Developmental Biology, 8, 158.

    PubMed  PubMed Central  Google Scholar 

  44. Mei, Q., Mou, H., Liu, X., et al. (2021). Therapeutic Potential of HUMSCs in female reproductive aging. Frontiers in Cell and Developmental Biology, 10(9), 650003.

    Google Scholar 

Download references

Acknowledgements

All authors are acknowledged for their contribution to the study.

Funding

This research was supported by Scientific Research Start-up funding Project of Binzhou Medical University (NO. BY2021KYQD12), the Natural Science Foundation of Shandong Province (NO. ZR2021QH149), the University and Locality Collaborative Development Program of Yantai (NO. 2021XDHZ082) and Yantai Double Hundred Program to Q.F.

Author information

Authors and Affiliations

Authors

Contributions

CLL, BHC and JZL were in charge of experimental design and literature research. CLL, ZWQ and TY were in charge of experimental studies. BHC and LQQ were in charge of data analysis and interpretation. SYR assisted with the experiments. FQ and JZL were in charge of the manuscript editing. All authors read and approved the ending version of the final manuscript.

Corresponding authors

Correspondence to Qiang Fu or Zhonglin Jiang.

Ethics declarations

Ethics Approval and Consent to Participate

Animals were treated in accordance with the Basel Declaration in the context of phase experimental animals. The use of animals was approved by the Ethics Committee of Binzhou Medical University. We have obtained the participants’ informed consent to donate the tissue for research use. The use of human tissue was approved by the Ethics committee of the Yantai Yuhuangding Hospital.

Consent for Publication

Not applicable.

Competing Interests

The authors declare that they have no competing interests.

Conflict of Interest

The authors declare no conflict 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.

12015_2022_10469_MOESM1_ESM.png

Supplementary file1 Supplementary Figure 1. hUMSCs identification. [A-a]: Microscopic morphology of hUMSCs (Scale bar=560 µm). [A-b]: hUMSCs stained with Alizarin red S to demonstrate their differentiation into osteoblasts (Scale bar=230 µm). [A-c]: hUMSCs stained with Oil Red O to demonstrate their differentiation into adipocytes (Scale bar=50 µm). [B]: The following surface markers of hUMSCs were identified: CD44, CD90, CD73, CD105, CD34, CD45 and HLA–DR. (PNG 6496 kb)

12015_2022_10469_MOESM2_ESM.png

Supplementary file2 Supplementary Figure 2. Stromal cell identification. Stromal cells were characterized by expression of cell surface markers. [A]: The isolated cells were characterized with immunofluorescence staining. The mesenchymal cells were stained by Vimentin as green, the endothelial cells were stained with Factor VIII as red and the epithelial cells were stained with cytokeratin as red. NR4A1 is stained green. The cell nucleus was stained by DAPI as blue (Scale bar=60 µm). [B]: The morphology of stromal cells under microscope(Scale bar=560 µm). [C]: Cell viability of stromal cells were measured after different concentrations of NR4A1 agonist, DIM-C-pPhOCH3 treatment. Data are expressed as the mean ± SD. *: P<0.05, **: P<0.01 and ***: P<0.001. (PNG 4197 kb)

Rights and permissions

Springer Nature or its licensor (e.g. a society or other partner) holds exclusive rights to this article under a publishing agreement with the author(s) or other rightsholder(s); author self-archiving of the accepted manuscript version of this article is solely governed by the terms of such publishing agreement and applicable law.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Cui, L., Bao, H., Zhu, W. et al. hUMSCs Transplantation Regulates AMPK/NR4A1 Signaling Axis to Inhibit Ovarian Fibrosis in POI Rats. Stem Cell Rev and Rep 19, 1449–1465 (2023). https://doi.org/10.1007/s12015-022-10469-y

Download citation

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1007/s12015-022-10469-y

Keywords

Navigation