Volume 22, Issue 3 (2019)                   mjms 2019, 22(3): 149-157 | Back to browse issues page

XML Persian Abstract Print


Download citation:
BibTeX | RIS | EndNote | Medlars | ProCite | Reference Manager | RefWorks
Send citation to:

Abedy Ghehi F, Fathi R, Abtahi N, Eivazkhani F, Bahrehbar K, Abed Heidari E, et al . Germ cells Markers Expression in Mouse Premature Ovarian Failure Model. mjms 2019; 22 (3) :149-157
URL: http://mjms.modares.ac.ir/article-30-29850-en.html
1- Embryology Department, Reproductive Biomedicine Research Center, Institute for Biology & Reproductive Medicine, Royan Institute, Academic Center for Education, Culture and Research (ACECR), Tehran, Iran
2- Embryology Department, Reproductive Biomedicine Research Center, Institute for Biology & Reproductive Medicine, Royan Institute, Academic Center for Education, Culture and Research (ACECR), Tehran, Iran , rfathi79@royaninstitute.org
3- Stem Cells & Developmental Biology Department, Cell Science Research Center, Royan Institute for Stem Cell Biology & Technology, Academic Center for Education, Culture and Research (ACECR), Tehran, Iran
Abstract:   (7365 Views)

Aims: Premature ovarian failure is a syndrome causing amenorrhea, infertility, and increases gonadotropin levels before age 40. The use of chemotherapy drugs can be one of the reasons that lead to this disorder. The purpose of this study was to evaluate the presence of germ cells markers in mice model of premature ovarian failure following chemotherapy drugs.
Materials and Methods: In this study, 24 mature female mice were used to create a premature ovarian failure model, different amount of cyclophosphamide and busulfan were applied (experimental groups 1 to 5). Bodyweight change, vaginal smear, morphological alternation of ovarian tissue in both experimental and control (without treatment) groups were evaluated and for the best model, hormonal evaluation (FSH, E2), and expression of germline markers (Oct4, Dazl) were examined.
Findings: Since, in the second group estrus cycle disorder, the significant decrease in weight and ovarian reserve (p˂0.05) were observed, compared to the control group, so this group was chosen as the best model. An increase in FSH level and reduction in estradiol level in the second group, compared with the control group (p˂0.05), confirmed creation of the POF model. Also, genes expression of Oct-4 and Dazl showed an increase (p˂0.05) in the second group compared with the control one.
Conclusion: The presence of germ cells markers in a mouse model of premature ovarian failure following the use of chemotherapy drugs can be a new hope in the treatment of infertility in cancer patients after chemotherapy.
 

Full-Text [PDF 1060 kb]   (1985 Downloads)    
Article Type: Original Research | Subject: Embryology
Received: 2019/01/27 | Accepted: 2019/07/12

References
1. Welt CK. Primary ovarian insufficiency: A more accurate term for premature ovarian failure. Clin Endocrinol. 2008;68(4):499-509. [Link] [DOI:10.1111/j.1365-2265.2007.03073.x]
2. Kalu E, Panay N. Spontaneous premature ovarian failure: Management challenges. Gynecol Endocrinol. 2008;24(5):273-9. [Link] [DOI:10.1080/09513590801990764]
3. Bhartiya D, Singh J. FSH-FSHR3-Stem cells in ovary surface epithelium: Basis for adult ovarian biology, failure, aging and cancer. Reproduction. 2015;149(1):R35-48. [Link] [DOI:10.1530/REP-14-0220]
4. De Moraes-Ruehsen M, Jones GS. Premature ovarian failure. Fertil Steril. 1967;18(4):440-61. [Link] [DOI:10.1016/S0015-0282(16)36362-2]
5. Panay N, Fenton A. Premature ovarian failure: A growing concern. Climacteric. 2008;11(1):1-3. [Link] [DOI:10.1080/13697130701878635]
6. Lai D, Wang F, Yao X, Zhang Q, Wu X, Xiang C. Human endometrial mesenchymal stem cells restore ovarian function through improving the renewal of germline stem cells in a mouse model of premature ovarian failure. J Transl Med. 2015;13(1):155. [Link] [DOI:10.1186/s12967-015-0516-y]
7. Larsen EC, Müller J, Schmiegelow K, Rechnitzer C, Andersen AN. Reduced ovarian function in long-term survivors of radiation-and chemotherapy-treated childhood cancer. Obstet Gynecol Surv. 2004;59(5):354-5. [Link] [DOI:10.1097/00006254-200405000-00021]
8. Schover LR. Premature ovarian failure and its consequences: Vasomotor symptoms, sexuality, and fertility. J Clin Oncol. 2008;26(5):753-8. [Link] [DOI:10.1200/JCO.2007.14.1655]
9. Johnson J, Canning J, Kanedo T, Pru JK, Tilly JL. Germline stem cells and follicular renewal in the postnatal mammalian ovary. Nature. 2004;428(6979):145-50. [Link] [DOI:10.1038/nature02316]
10. Forabosco A, Sforza C, De Pol A, Vizzotto L, Marzona L, Ferrario VF. Morphometric study of the human neonatal ovary. Anat Rec. 1991;231(2):201-8. [Link] [DOI:10.1002/ar.1092310208]
11. Chemaitilly W, Mertens AC, Mitby P, Whitton J, Stovall M, Yasui Y, et al. Acute ovarian failure in the childhood cancer survivor study. J Clin Endocrinol Metab. 2006;91(5):1723-8. [Link] [DOI:10.1210/jc.2006-0020]
12. Salama M, Winkler K, Murach KF, Seeber B, Ziehr SC, Wildt L. Female fertility loss and preservation: Threats and opportunities. Ann Oncol. 2013;24(3):598-608. [Link] [DOI:10.1093/annonc/mds514]
13. Meirow D, Dor J, Kaufman B, Shrim A, Rabinovici J, Schiff E, et al. Cortical fibrosis and blood-vessels damage in human ovaries exposed to chemotherapy. Potential mechanisms of ovarian injury. Hum Reprod. 2007;22(6):1626-33. [Link] [DOI:10.1093/humrep/dem027]
14. Meirow D, Biederman H, Anderson RA, Wallace WH. Toxicity of chemotherapy and radiation on female reproduction. Clin Obstet Gynecol. 2010;53(4):727-39. [Link] [DOI:10.1097/GRF.0b013e3181f96b54]
15. Burnell M, Levine MN, Chapman JAW, Bramwell V, Gelmon K, Walley B, et al. Cyclophosphamide, epirubicin, and fluorouracil versus dose-dense epirubicin and cyclophosphamide followed by paclitaxel versus doxorubicin and cyclophosphamide followed by paclitaxel in node-positive or high-risk node-negative breast cancer. J Clin Oncol. 2010;28(1):77-82. [Link] [DOI:10.1200/JCO.2009.22.1077]
16. Behringer K, Mueller H, Goergen H, Thielen I, Eibl AD, Stumpf V, et al. Gonadal function and fertility in survivors after Hodgkin lymphoma treatment within the German Hodgkin study group HD13 to HD15 trials. J Clin Oncol. 2013;31(2):231-9. [Link] [DOI:10.1200/JCO.2012.44.3721]
17. Devi HP, Mazumder PB. Methanolic extract of Curcuma caesia Roxb. prevents the toxicity caused by cyclophosphamide to bone marrow cells, liver and kidney of mice. Pharmacogn Res. 2016;8(1):43-9. [Link] [DOI:10.4103/0974-8490.171106]
18. Song Y, Zhang C, Wang C, Zhao L, Wang Z, Dai Z, et al. Ferulic acid against cyclophosphamide-induced heart toxicity in mice by inhibiting NF-κB pathway. Evid Based Complement Altern Med. 2016;2016:1261270. [Link] [DOI:10.1155/2016/1261270]
19. Li F, Turan V, Lierman S, Cuvelier C, De Sutter P, Oktay K. Sphingosine-1-phosphate prevents chemotherapy-induced human primordial follicle death. Hum Reprod. 2014;29(1):107-13. [Link] [DOI:10.1093/humrep/det391]
20. Blumenfeld Z. Preservation of fertility and ovarian function and minimalization of chemotherapy associated gonadotoxicity and premature ovarian failure: The role of inhibin-A and-B as markers. Mol Cell Endocrinol. 2002;187(1-2):93-105. [Link] [DOI:10.1016/S0303-7207(01)00712-2]
21. Caligioni CS. Assessing reproductive status/stages in mice. Curr Protoc Neurosci. 2009;48(1):A-4I.1-8. [Link] [DOI:10.1002/0471142301.nsa04is48]
22. Tavana S, Azarnia M, Rezazadeh Valojerdi M, Shahverdi A. Hyaluronic acid-based hydrogel scaffold without angiogenic growth factors enhances ovarian tissue function after autotransplantation in rats. Biomed Mater. 2016;11(5):055006. [Link] [DOI:10.1088/1748-6041/11/5/055006]
23. Luborsky JL, Meyer P, Sowers MF, Gold EB, Santoro N. Premature menopause in a multi‐ethnic population study of the menopause transition. Hum Reprod. 2003;18(1):199-206. [Link] [DOI:10.1093/humrep/deg005]
24. Song D, Zhong Y, Qian C, Zou Q, Ou J, Shi Y, et al. Human umbilical cord mesenchymal stem cells therapy in cyclophosphamide-induced premature ovarian failure rat model. BioMed Res Int. 2016;2016:2517514. [Link] [DOI:10.1155/2016/2517514]
25. Luo Q, Yin N, Zhang L, Yuan W, Zhao W, Luan X, et al. Role of SDF-1/CXCR4 and cytokines in the development of ovary injury in chemotherapy drug induced premature ovarian failure mice. Life Sci. 2017;179:103-9. [Link] [DOI:10.1016/j.lfs.2017.05.001]
26. Nelson LM. Primary ovarian insufficiency. New Engl J Med. 2009;360:606-14. [Link] [DOI:10.1056/NEJMcp0808697]
27. Anasti JN. Premature ovarian failure: AN update. Fertil Steril. 1998;70(1):1-15. [Link] [DOI:10.1016/S0015-0282(98)00099-5]
28. Lazaros LA, Hatzi EG, Pamporaki CE, Sakaloglou PI, Xita NV, Markoula SI, et al. The ovarian response to standard gonadotrophin stimulation depends on FSHR, SHBG and CYP19 gene synergism. J Assist Reprod Genet. 2012;29(11):1185-91. [Link] [DOI:10.1007/s10815-012-9849-0]
29. Broekmans FJ, Knauff EAH, Te Velde ER, Macklon NS, Fauser BC. Female reproductive ageing: Current knowledge and future trends. Trends Endocrinol Metab. 2007;18(2):58-65. [Link] [DOI:10.1016/j.tem.2007.01.004]
30. Yacobi K, Wojtowicz A, Tsafriri A, Gross A. Gonadotropins enhance caspase-3 and-7 activity and apoptosis in the theca-interstitial cells of rat preovulatory follicles in culture. Endocrinology. 2004;145(4):1943-51. [Link] [DOI:10.1210/en.2003-1395]
31. Jung M, Peterson H, Chavez L, Kahlem P, Lehrach H, Vilo J, et al. A data integration approach to mapping OCT4 gene regulatory networks operative in embryonic stem cells and embryonal carcinoma cells. PloS One. 2010;5(5):e10709. [Link] [DOI:10.1371/journal.pone.0010709]
32. Pan GJ, Chang ZY, Schöler HR, Pei D. Stem cell pluripotency and transcription factor Oct4. Cell Res. 2002;12(5-6):321-9. [Link] [DOI:10.1038/sj.cr.7290134]
33. Wang XQ, Ongkeko WM, Chen L, Yang ZF, Lu P, Chen KK, et al. Octamer 4 (Oct4) mediates chemotherapeutic drug resistance in liver cancer cells through a potential Oct4-AKT-ATP‐binding cassette G2 pathway. Hepatology. 2010;52(2):528-39. [Link] [DOI:10.1002/hep.23692]
34. Lu CS, Shieh GS, Wang CT, Su BH, Su YC, Chen YC, et al. Chemotherapeutics-induced Oct4 expression contributes to drug resistance and tumor recurrence in bladder cancer. Oncotarget. 2017;8(19):30844-58. [Link] [DOI:10.18632/oncotarget.9602]
35. Niikura Y, Niikura T, Tilly JL. Aged mouse ovaries possess rare premeiotic germ cells that can generate oocytes following transplantation into a young host environment. Aging Albany NY. 2009;1(12):971-8. [Link] [DOI:10.18632/aging.100105]

Send email to the article author


Rights and permissions
Creative Commons License This work is licensed under a Creative Commons Attribution-NonCommercial 4.0 International License.