Stem cell therapy in premature ovarian insufficiency

Authors

  • Shalala Mammadova Ondokuz Mayıs University Faculty of Medicine, Department of Obstetrics and Gynecology, Samsun.

Keywords:

premature ovarian insufficiency, stem cell, infertility, amenorrhea, oligomenorrhea, genetics, apoptosis

Abstract

Premature ovarian insufficiency (POI) is defined as decreased ovarian function before the expected age of menopause. It is well known that POI increases the risk of cardiovascular diseases, osteoporosis, cognitive and mood disorders. Appropriate hormone replacement reduces these adverse risks and improves quality of life, but does not affect overall survival. Potential etiologies of POI include chromosomal abnormalities, genetic mutations, autoimmune factors, iatrogenic causes such as surgery, chemotherapy, and radiation therapy. It has been suggested that there is a significant relationship between reproduction and DNA damage. DNA damage in ovarian granulosa cells is involved in the pathogenesis of POI. Depletion of oocytes with damaged DNA occurs through different mechanisms of cell death, such as apoptosis, autophagy, and necroptosis, mediated by phosphatase and tensin homolog (PTEN)/phosphoinositide 3-kinase (PI3K)/protein kinase B (AKT)/forkhead transcription. Mesenchymal stem cells are characterized by their ability to self-renew and play an important role in the regeneration of injured tissues. Recent advances in stem cell therapy may be promising new therapeutic options in POI patients. In this article, we reviewed the topic of stem cell therapy in premature ovarian failure.

References

Coulam CB. Premature gonadal failure. Fertil Steril 1982; 38(6): 645-55.

Rocca WA, Grossardt BR, de Andrade M, Malkasian GD, Melton LJ, 3rd. Survival patterns after oophorectomy in premenopausal women: a population-based cohort study. Lancet Oncol (2006) 7:821–8.

ESHRE POI Guideline Development Group . Management of women with premature ovarian insufficiency. Guideline Eur Soc Hum Reprod Embryol (2015) p.30.

Coulam CB, Adamson SC, Annegers JF. Incidence of premature ovarian failure. Obstet Gynecol (1986).

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) 18:199–206.

Lagergeren K, Hammar M, Nedstrand E, Bladh M, Sydsjö G. The prevalence of primary ovarian insufficiency in Sweden; a national register study. BMC Womens Health (2018) 18:175.

Golezar S, Ramezani TF, Khazaei S, Ebadi A, Keshavarz Z. The global prevalence of primary ovarian insufficiency and early menopause: a meta-analysis. Climacteric (2019) 22:403–11.

Panay N, Kalu E. Management of Premature Ovarian Failure. Best Practice & Research Clinical Obstetrics & Gy- naecology. 2009;23(1):129-40.

Knauff EAH, Eijkemans MJC, Lambalk CB, ten Kate-Booij MJ, HoPek A, Beerendonk CCM, et al. Anti-Mullerian Hormone, İnhibin B, And Antral Follicle Count in Young Women With Ovarian Failure. J Clin Endocrinol Metab. 2009;94(3):786-92.

He C, Kraft P, Chen C, Buring JE, -Paré G, Hankinson SE, et al. Genome-Wide Associa- tion Studies İdentify Loci Associated with Age at Menarche and Age at Natural Menopause. Nature Genetics. 2009;41(6): 724-8.

Perry JR, Stolk L, Franceschini N, Lunetta KL, Zhai G, McArdle PF, et al. Meta-analysis of Genome- wide Association Data İdentifies Two Loci İnflu- encing Age at Menarche. Nature Genetics. 2009;41(6):648-50.

Fassnacht W, Mempel A, Strowitzki T, Vogt P. Premature Ovarian Failure (POF) Syndrome: Towards the Molecular Clinical Analysis of İts Genetic Complexity. Current Medicinal Chem-serve And Menopausal Age. Reprod Sci. 2007;14(8):780-5.

Crowley WF. Pathogenesis And Causes Of Spontaneous Primary Ovarian İnsufficiency (Premature Ovarian Failure). 2020;1-22

Anasti JN. Premature Ovarian Failure: An Update. Fertility and Sterility. 1998; 70(1): 1-15.

Cox L, Liu JH. Primary Ovarian İnsufficiency: an Update. International Journal of Women's Health. 2013;6:235-43.

Familiari U, Larocca L, Tamburrini E, Antinori A, Ortona L, Capelll A. Premenopausal Cytomegalovirus Oophoritis in a Patient with AIDS. Aids. 1991;5(4):458.

Rebar RW, Connolly HV. Clinical Features of Young Women with Hypergonadotropic Amenorrhea. Obstetrical & Gynecological Survey. 1990;45(11):763-4.

Ghahremani-Nasab M, Ghanbari E, Jahanbani Y, Mehdizadeh A, Yousefi M. Premature ovarian failure and tissue engineering. J Cell Physiol 2020;235:4217-26.

Kawamura K, Kawamura N, Hsueh AJ. Activation of dormant follicles: A new treatment for premature ovarian failure? Curr Opin Obstet Gynecol 2016;28:217-22.

Mohamed SA, Shalaby SM, Abdelaziz M, Brakta S, Hill WD, Ismail N,

et al. Human mesenchymal stem cells partially reverse infertility in chemotherapy-induced ovarian failure. Reprod Sci 2018;25:51-63.

Liu J, Zhang H, Zhang Y, Li N, Wen Y, Cao F, et al. Homing and restorative effects of bone marrow-derived mesenchymal stem cells on cisplatin injured ovaries in rats. Mol Cells 2014;37:865-72.

He Y, Chen D, Yang L, Hou Q, Ma H, Xu X. The therapeutic potential of bone marrow mesenchymal stem cells in premature ovarian failure. Stem Cell Res Ther 2018;9:263.

Lu L, Chen G, Yang J, et al. Bone marrow mesenchymal stem cells suppress growth and promote the apoptosis of glioma U251 cells through downregulation of the PI3K/AKT signaling pathway. Biomed Pharmacother. 2019;112:108625.

Jiang S, Mo C, Guo S, et al. Human bone marrow mesenchymal stem cells-derived microRNA-205-containing exosomes impede the progression of prostate cancer through suppression of RHPN2. J Exp Clin Cancer Res. 2019;38(1):495.

Chen B, Yu J, Wang Q, et al. Human bone marrow mesenchymal stem cells promote gastric cancer growth via regulating c-Myc. Stem Cells Int. 2018;2018:9501747.

Neshati V, Mollazadeh S, Fazly Bazzaz BS, et al. MicroRNA-499a-5p promotes differentiation of human bone marrow-derived mesenchymal stem cells to cardiomyocytes. Appl Biochem Biotechnol. 2018;186(1):245–255.

Fillmore N, Huqi A, Jagdip S, et al. Effect of fatty acids on human bone marrow mesenchymal stem cell energy metabolism and survival. PLoS One. 2015;10(3):e0120257.

Castro-Manrreza ME, Montesinos JJ. Immunoregulation by mesenchymal stem cells: biological aspects and clinical applications. J Immunol Res. 2015;2015:394917.

Dabrowska S, Andrzejewska A, Strzemecki D, et al. Human bone marrow mesenchymal stem cell-derived extracellular vesicles attenuate neuroinflammation evoked by focal brain injury in rats. J Neuroinflammation. 2019;16(1):216.

Liu J, Zhang H, Zhang Y, et al. Homing and restorative effects of bone marrow-derived mesenchymal stem cells on cisplatin injured ovaries in rats. Mol Cells. 2014;37(12):865–872.

Besikcioglu HE, Sarıbas GS, Ozogul C, et al. Determination of the effects of bone marrow derived mesenchymal stem cells and ovarian stromal stem cells on follicular maturation in cyclophosphamide induced ovarian failure in rats. Taiwan J Obstet Gynecol. 2019;58(1):53–59.

Badawy A, Sobh MA, Ahdy M, et al. Bone marrow mesenchymal stem cell repair of cyclophosphamide-induced ovarian insufficiency in a mouse model. Int J Womens Health. 2017;9:441–447.

Mohamed SA, Shalaby SM, Abdelaziz M, et al. Human mesenchymal stem cells partially reverse infertility in chemotherapy-induced ovarian failure. Reprod Sci. 2018;25(1):51–63.

Gabr H, Rateb MA, El Sissy MH, et al. The effect of bone marrow-derived mesenchymal stem cells on chemotherapy induced ovarian failure in albino rats. Microsc Res Tech. 2016;79(10):938–947.

Zhang Y, Xia X, Yan J, et al. Mesenchymal stem cell-derived angiogenin promotes primodial follicle survival and angiogenesis in transplanted human ovarian tissue. Reprod Biol Endocrinol. 2017;15:18.

Chen X, Wang Q, Li X, et al. Heat shock pretreatment of mesenchymal stem cells for inhibiting the apoptosis of ovarian granulosa cells enhanced the repair effect on chemotherapy-induced premature ovarian failure. Stem Cell Res Ther. 2018;9:240.

Alviano F, Fossati V, Marchionni C, Arpinati M, Bonsi L, Franchina M, Lanzoni G, Cantoni S, Cavallini C, Bianchi F, Tazzari PL, Pasquinelli G, Foroni L, et al. Term amniotic membrane is a high throughput source for multipotent mesenchymal stem cells with the ability to differentiate into endothelial cells in vitro. BMC Dev Biol. 2007;7:11.

Wolbank S, Peterbauer A, Fahrner M, Hennerbichler S, van Griensven M, Stadler G, Redl H, Gabriel C. Dose-dependent immunomodulatory effect of human stem cells from amniotic membrane: a comparison with human mesenchymal stem cells from adipose tissue. Tissue Eng. 2007;13:1173–1183.

Portmann-Lanz CB, Schoeberlein A, Portmann R, Mohr S, Rollini P, Sager R, Surbek DV. Turning placenta into brain: placental mesenchymal stem cells differentiate into neurons and oligodendrocytes. Am J Obstet Gynecol. 2010; 202(294): e1–e11.

Chen CP. Placental villous mesenchymal cells trigger trophoblast invasion. Cell Adhes Migr. 2014;8(2):94–97.

Roy R, Brodarac A, Kukucka M, Kurtz A, Becher PM, Julke K, et al. Cardioprotection by placenta-derived stromal cells in a murine myocardial infarction model. J Surg Res. 2013;185:70–83.

Prather WR, Toren A, Meiron M, Ofir R, Tschope C, Horwitz EM. The role of placental-derived adherent stromal cell (PLX-PAD) in the treatment of critical limb ischemia. Cytotherapy. 2009;11:427–434.

Zahavi-Goldstein E, Blumenfeld M, Fuchs-Telem D, Pinzur L, Rubin S, Aberman Z, et al. Placenta-derived PLX-PAD mesenchymal-like stromal cells are efficacious in rescuing blood flow in hind limb ischemia mouse model by a dose- and site-dependent mechanism of action. Cytotherapy. 2017;19:1438–1446.

Liu SH, Huang JP, Lee RK, et al. Paracrine factors from human placental multipotent mesenchymal stromal cells protect endothelium from oxidative injury via STAT3 and manganese superoxide dismutase activation. Biol Reprod. 2010;82:905–913.

Kong P, Xie X, Li F, Liu Y, Lu Y. Placenta mesenchymal stem cell accelerates wound healing by enhancing angiogenesis in diabetic Goto- Kakizaki (GK) rats. Biochem Biophys Res Commun. 2013;438(2):410–419.

Komaki M, Numata Y, Morioka C, et al. Exosomes of human placenta-derived mesenchymal stem cells stimulate angiogenesis. Stem Cell Res Ther. 2017;8(1):219.

Magaña-Guerrero FS, Domínguez-López A, Martínez-Aboytes P, et al. Human amniotic membrane mesenchymal stem cells inhibit neutrophil extracellular traps through TSG-6. Sci Rep. 2017;7:12426.

Wu Q, Fang T, Lang H, Chen M, Shi P, Pang X, et al. Comparison of the proliferation, migration and angiogenic properties of human amniotic epithelial and mesenchymal stem cells and their effects on endothelial cells. Int J Mol Med. 2017;39:918–926.

Jeong IS, Park Y, Ryu HA, et al. Dual chemotactic factors-secreting human amniotic mesenchymal stem cells via TALEN mediated gene editing enhanced angiogenesis. Int J Cardiol. 2018;260:156162.

Li J, et al. Human amnion-derived stem cells have immunosuppressive properties on NK cells and monocytes. Cell Transplant. 2015;24:2065– 2076.

Pianta S, et al. Amniotic membrane mesenchymal cells-derived factors skew T cell polarization toward Treg and downregulate Th1 and Th17 cells subsets. Stem Cell Rev. 2015;11:394–407.

Lai D, Guo Y, Zhang Q, et al. Differentiation of human menstrual blood-derived endometrial mesenchymal stem cells into oocyte-like cells. Acta Biochim Biophys Sin Shanghai. 2016;48(11):998–1005.

Khoury M, Alcayaga-Miranda F, Illanes SE, et al. The promising potential of menstrual stem cells for antenatal diagnosis and cell therapy. Front Immunol. 2014;5:205.

Alcayaga-Miranda F, Cuenca J, Luz-Crawford P, et al. Characterization of menstrual stem cells: angiogenic effect, migration and hematopoietic stem cell support in comparison with bone marrow mesenchymal stem cells. Stem Cell Res Ther. 2015;6:32.

Cui CH, Uyama T, Miyado K, et al. Menstrual blood-derived cells confer human dystrophin expression in the murine model of Duchenne muscular dystrophy via cell fusion and myogenic transdifferentiation. Mol Biol Cell. 2007;18:1586–1594.

Uzbas E, May ID, Parisi AM, et al. Molecular physiognomies and applications of adipose-derived stem cells. Stem Cell Rev Rep. 2015;2:298– 308.

Neri S, Bourin P, Peyrafitte JA, et al. Human adipose stromal cells (ASC) for the regeneration of injured cartilage display genetic stability after in vitro culture expansion. PLoS One. 2013;8:e77895.

Hayashi K, Ogushi S, Kurimoto K, Shimamoto S, Ohta H, Saitou M. Offspring from oocytes derived from in vitro primordial germ cell-like cells in mice. Science 2012;338:971-5.

Gupta S, Lodha P, Karthick MS, Tandulwadkar SR. Role of autologous bone marrow-derived stem cell therapy for follicular recruitment in premature ovarian insufficiency: Review of literature and a case report of world’s first baby with ovarian autologous stem cell therapy in a perimenopausal woman of age 45 year. J Hum Reprod Sci 2018;11:125-30.

Kimbrel EA, Lanza R. Current status of pluripotent stem cells: Moving the first therapies to the clinic. Nat Rev DrugDiscov2015;14:681-92.

Kwong J, Chan FL, Wong KK, Birrer MJ, Archibald KM, Balkwill FR, et al. Inflammatory cytokine tumor necrosis factor alpha confers precancerous phenotype in an organoid model of normal human ovarian surface epithelialcells. Neoplasia2009;11:529-41.

Esfandyari S, Chugh RM, Park HS, Hobeika E, Ulin M, Al-Hendy A. Mesenchymal stem cells as a bio organ for treatment of female infertility. Cells 2020;9:2253.

T. Tiryaki, S. Canikyan, P. Koçak, S. Cohen, A. Sterodimas, K. U. Schlaudraff, M. Scheflan, P. Hollands. Adipose-derived Stromal Vascular Matrix (SVM): a new paradigm in regenerative medicine. CellR4 2021; 9: e3060.

Published

2023-07-21

How to Cite

Mammadova, S. (2023). Stem cell therapy in premature ovarian insufficiency. Journal of Infertility, 1(1), 22–27. Retrieved from https://journalofinfertility.org/index.php/infertility/article/view/5