Skip to main content
Log in

Hyperandrogenism Decreases GRP78 Protein Level and Glucose Uptake in Human Endometrial Stromal Cells

  • Original Article
  • Published:
Reproductive Sciences Aims and scope Submit manuscript

Abstract

Background

Women with polycystic ovary syndrome (PCOS) exhibit a low fertility by chronic hyperandrogenemia. Different evidence have shown that androgens could regulate the endoplasmic reticulum (ER) homeostasis and glucose metabolism. However, it is unclear whether androgens can exert these effects on human endometrial stromal cells. Our goal was to study the protein content of GRP78 (an ER homeostasis marker) in endometria from women with PCOS and healthy women and to assess the GRP78 protein levels and its relationship with glucose uptake on a human endometrial stromal cell line stimulated with testosterone.

Methods

Immunohistochemistry assays for GRP78 were performed on endometrial samples obtained from women with PCOS (n = 8) and control women subjected to hysterectomy (n = 8). Western blot analysis for GRP78 and glucose uptake was assessed in a telomerase-immortalized human endometrial stromal cell line (T-HESC) exposed to testosterone for 24 or 48 hours and challenged to an insulin short-term stimulation. Tukey test was performed for human samples comparison. Student t test or ANOVA–Bonferroni test was carried out according to the in vitro experiment. P < .05 was considered as significant.

Results

GRP78 stromal immunostaining was reduced in PCOS endometria compared to controls (P < .05). The T-HESC shows a testosterone-dependent downregulation of GRP78 protein content (P < .05), concomitant with half-reduction in glucose uptake compared to controls (P < .05). Moreover, enhanced small interfering RNA against GRP78 messenger RNA leads to a decrease in glucose uptake (P < .05). Such effects were reverted by hydroxyflutamide, an inhibitor of androgen receptor.

Conclusion

These results suggest that hyperandrogenemic PCOS environment could compromise the endometrial homeostasis confirmed by the decrease in glucose uptake induced by testosterone and exhibited by stromal cells.

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.

Similar content being viewed by others

References

  1. Giudice LC. Endometrium in PCOS: implantation and predisposition to endocrine CA. Best Pract Res Clin Endocrinol Metab. 2006;20(2):235–244.

    Article  CAS  PubMed  Google Scholar 

  2. Frolova AI, Moley KH. Glucose transporters in the uterus: an analysis of tissue distribution and proposed physiological roles. Reproduction. 2011;142(2):211–220.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  3. Frolova AI, Moley KH. Quantitative analysis of glucose transporter mRNAs in endometrial stromal cells reveals critical role of GLUT1 in uterine receptivity. Endocrinology. 2011;152(5): 2123–2128.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  4. Carvajal R, Rosas C, Kohan K, et al. Metformin augments the levels of molecules that regulate the expression of the insulin-dependent glucose transporter GLUT4 in the endometria of hyperinsulinemic PCOS patients. Hum Reprod. 2013;28(8): 2235–2244.

    Article  CAS  PubMed  Google Scholar 

  5. Kohan K, Carvajal R, Gabler F, Vantman D, Romero C, Vega M. Role of the transcriptional factors FOXO1 and PPARG on gene expression of SLC2A4 in endometrial tissue from women with polycystic ovary syndrome. Reproduction. 2010;140(1):123–131.

    Article  CAS  PubMed  Google Scholar 

  6. Diamanti-Kandarakis E, Dunaif A. Insulin resistance and the polycystic ovary syndrome revisited: an update on mechanisms and implications. Endocr Rev. 2012;33(6):981–1030.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. Rivero R, Garin CA, Ormazabal P, et al. Protein expression of PKCZ (Protein Kinase C Zeta), Munc18c, and Syntaxin-4 in the insulin pathway in endometria of patients with polycystic ovary syndrome (PCOS). Reprod Biol Endocrinol. 2012;10:17.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. Rosas C, Gabler F, Vantman D, Romero C, Vega M. Levels of Rabs and WAVE family proteins associated with translocation of GLUT4 to the cell surface in endometria from hyperinsulinemic PCOS women. Hum Reprod. 2010;25(11):2870–2877.

    Article  CAS  PubMed  Google Scholar 

  9. Fornes R, Ormazabal P, Rosas C, et al. Changes in the expression of insulin signaling pathway molecules in endometria from polycystic ovary syndrome women with or without hyperinsulinemia. Mol Med. 2010;16(3–4):129–136.

    Article  CAS  PubMed  Google Scholar 

  10. Corbould A. Chronic testosterone treatment induces selective insulin resistance in subcutaneous adipocytes of women. J Endocrinol. 2007;192(3):585–594.

    Article  CAS  PubMed  Google Scholar 

  11. Hojlund K, Glintborg D, Andersen NR, et al. Impaired insulin-stimulated phosphorylation of Akt and AS160 in skeletal muscle of women with polycystic ovary syndrome is reversed by pioglitazone treatment. Diabetes. 2008;57(2):357–366.

    Article  PubMed  Google Scholar 

  12. Nakatsuka A, Wada J, Iseda I, et al. Vaspin is an adipokine ameliorating ER stress in obesity as a ligand for cell-surface GRP78/ MTJ-1 complex. Diabetes. 2012;61(11):2823–2832.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. Gregor MF, Yang L, Fabbrini E, et al. Endoplasmic reticulum stress is reduced in tissues of obese subjects after weight loss. Diabetes. 2009;58(3):693–700.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  14. Ozcan U, Cao Q, Yilmaz E, et al. Endoplasmic reticulum stress links obesity, insulin action, and type 2 diabetes. Science. 2004; 306(5695):457–461.

    Article  PubMed  Google Scholar 

  15. Segawa T, Nau ME, Xu LL, et al. Androgen-induced expression of endoplasmic reticulum (ER) stress response genes in prostate cancer cells. Oncogene. 2002;21(57):8749–8758.

    Article  CAS  PubMed  Google Scholar 

  16. Ma C, Yoshioka M, Boivin A, et al. Atlas of dihydrotestosterone actions on the transcriptome of prostate in vivo. Prostate. 2009; 69(3):293–316.

    Article  CAS  PubMed  Google Scholar 

  17. Zhu G, Lee AS. Role of the unfolded protein response, GRP78 and GRP94 in organ homeostasis. J Cell Physiol. 2015;230(7): 1413–1420.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. Noyes RW, Hertig AT, Rock J. Dating the endometrial biopsy. Am J Obstet Gynecol. 1975;122(2):262–263.

    Article  CAS  PubMed  Google Scholar 

  19. Azziz R, Carmina E, Dewailly D, et al. Positions statement: criteria for defining polycystic ovary syndrome as a predominantly hyperandrogenic syndrome: an Androgen Excess Society guideline. J Clin Endocrinol Metab. 2006;91(11):4237–4245.

    Article  CAS  PubMed  Google Scholar 

  20. Garcia V, Orostica L, Poblete C, et al. Endometria from obese PCOS women with hyperinsulinemia exhibit altered adiponectin signaling. Horm Metab Res. 2015;47(12):901–909.

    Article  CAS  PubMed  Google Scholar 

  21. Krikun G, Mor G, Alvero A, et al. A novel immortalized human endometrial stromal cell line with normal progestational response. Endocrinology. 2004;145(5):2291–2296.

    Article  CAS  PubMed  Google Scholar 

  22. Plaza-Parrochia F, Bacallao K, Poblete C, et al. The role of androst-5-ene-3beta,17beta-diol (androstenediol) in cell proliferation in endometrium of women with polycystic ovary syndrome. Steroids. 2014;89:11–19.

    Article  CAS  PubMed  Google Scholar 

  23. Ormazabal P, Romero C, Quest AF, Vega M. Testosterone modulates the expression of molecules linked to insulin action and glucose uptake in endometrial cells. Horm Metab Res. 2013; 45(9):640–645.

    Article  CAS  PubMed  Google Scholar 

  24. Vera JC, Reyes AM, Velasquez FV, et al. Direct inhibition of the hexose transporter GLUT1 by tyrosine kinase inhibitors. Biochemistry. 2001;40(3):777–790.

    Article  CAS  PubMed  Google Scholar 

  25. Perez A, Ojeda P, Ojeda L, et al. Hexose transporter GLUT1 harbors several distinct regulatory binding sites for flavones and tyrphostins. Biochemistry. 2011;50(41):8834–8845.

    Article  CAS  PubMed  Google Scholar 

  26. Bacallao K, Plaza-Parrochia F, Cerda A, et al. Levels of regulatory proteins associated with cell proliferation in endometria from untreated patients having polycystic ovarian syndrome with and without endometrial hyperplasia. Reprod Sci. 2016;23(2): 211–218.

    Article  CAS  PubMed  Google Scholar 

  27. DeFronzo RA, Matsuda M. Reduced time points to calculate the composite index. Diabetes Care. 2010;33(7):e93.

    Article  PubMed  Google Scholar 

  28. McAuley KA, Mann JI, Chase JG, Lotz TF, Shaw GM. Point: HOMA—satisfactory for the time being: HOMA: the best bet for the simple determination of insulin sensitivity, until something better comes along. Diabetes Care. 2007;30(9):2411–2413.

    Article  PubMed  Google Scholar 

  29. Mozzanega B, Mioni R, Granzotto M, et al. Obesity reduces the expression of GLUT4 in the endometrium of normoinsulinemic women affected by the polycystic ovary syndrome. Ann N Y Acad Sci. 2004;1034:364–374.

    Article  CAS  PubMed  Google Scholar 

  30. Biswas N, Friese RS, Gayen JR, Bandyopadhyay G, Mahata SK, O’Connor DT. Discovery of a novel target for the dysglycemic chromogranin A fragment pancreastatin: interaction with the chaperone GRP78 to influence metabolism. PLoS One. 2014;9(1): e84132.

    Article  PubMed  PubMed Central  Google Scholar 

  31. Yamagishi N, Ueda T, Mori A, Saito Y, Hatayama T. Decreased expression of endoplasmic reticulum chaperone GRP78 in liver of diabetic mice. Biochem Biophys Res Commun. 2012;417(1): 364–370.

    Article  CAS  PubMed  Google Scholar 

  32. Perrini S, Natalicchio A, Laviola L, et al. Dehydroepiandrosterone stimulates glucose uptake in human and murine adipocytes by inducing GLUT1 and GLUT4 translocation to the plasma membrane. Diabetes. 2004;53(1):41–52.

    Article  CAS  PubMed  Google Scholar 

  33. Sato K, Iemitsu M, Aizawa K, Ajisaka R. Testosterone and DHEA activate the glucose metabolism-related signaling pathway in skeletal muscle. Am J Physiol Endocrinol Metab. 2008;294(5): E961–E968.

    Article  CAS  PubMed  Google Scholar 

  34. Sato K, Iemitsu M, Aizawa K, Ajisaka R. DHEA improves impaired activation of Akt and PKC zeta/lambda-GLUT4 pathway in skeletal muscle and improves hyperglycaemia in streptozotocin-induced diabetes rats. Acta Physiol (Oxf). 2009; 197(3):217–225.

    Article  CAS  Google Scholar 

  35. Zhang L, Liao Q. Effects of testosterone and metformin on glucose metabolism in endometrium. Fertil Steril. 2010;93(7): 2295–2298.

    Article  CAS  PubMed  Google Scholar 

  36. Allemand MC, Irving BA, Asmann YW, et al. Effect of testosterone on insulin stimulated IRS1 Ser phosphorylation in primary rat myotubes—a potential model for PCOS-related insulin resistance. PLoS One. 2009;4(1):e4274.

    Article  PubMed  PubMed Central  Google Scholar 

  37. Chen X, Li X, Huang HY, Li X, Lin JF. Effects of testosterone on insulin receptor substrate-1 and glucose transporter 4 expression in cells sensitive to insulin [in Chinese]. Zhonghua Yi Xue Za Zhi. 2006;86(21):1474–1477.

    CAS  PubMed  Google Scholar 

Download references

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Margarita Vega PhD.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Rosas, C., Oróstica, L., Poblete, C. et al. Hyperandrogenism Decreases GRP78 Protein Level and Glucose Uptake in Human Endometrial Stromal Cells. Reprod. Sci. 23, 761–770 (2016). https://doi.org/10.1177/1933719115618283

Download citation

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1177/1933719115618283

Keywords

Navigation