Derleme
BibTex RIS Kaynak Göster
Yıl 2023, Cilt: 40 Sayı: 1, 122 - 126, 18.03.2023

Öz

Kaynakça

  • 1. Zegers-Hochschild F, Adamson GD, Dyer S, Racowsky C, De Mouzon J, Sokol R, et al. The international glossary on infertility and fertility care. Hum. Reprod. 2017; 32, 1786–1801.
  • 2. Volarevic V, Bojic S, Nurkovic J, Volarevic A, Ljujic B, Arsenijevic N, et al. Stem cells as new agents for the treatment of infertility: Current and future perspectives and challenges. BioMed Res. Int. 2014; 507234.
  • 3. Abd-Allah SH, Shalaby SM, Pasha HF, Amal S, Raafat N, Shabrawy SM, et al. Mechanistic action of mesenchymal stem cell injection in the treatment of chemically induced ovarian failure in rabbits. Cytotherapy. 2013; 15, 64–75.
  • 4. 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.
  • 5. Santamaria X, Cabanillas S, Cervello I, Arbona C, Raga F, Ferro J, et al. Autologous cell therapy with CD133+ bone marrow-derived stem cells for refractory Asherman’s syndrome and endometrial atrophy: A pilot cohort study. Hum. Reprod. 2016; 31, 1087–1096.
  • 6. Manshadi MD, Navid S, Hoshino Y, Daneshi E, Noory P, Abbasi M. The effects of human menstrual blood stem cellsderived granulosa cells on ovarian follicle formation in a rat model of premature ovarian failure. Microsc. Res. Tech. 2019; 82,635–642.
  • 7. Domnina A, Novikova P, Obidina J, Fridlyanskaya I, Alekseenko L, Kozhukharova I, et al. Human mesenchymal stem cells in spheroids improve fertility in model animals with damaged endometrium. Stem Cell Res Ther. 2018; 9, 50.
  • 8. Wang S, Yu L, Sun M, Mu S, Wang C, Wang D, et al. The therapeutic potential of umbilical cord mesenchymal stem cells in mice premature ovarian failure. Biomed Res. Int. 2013; 690491.
  • 9. Fan D, Wu S, Ye S, Wang W, Guo X, Liu Z. Umbilical cord mesenchyme stem cell local intramuscular injection for treatment of uterine niche: Protocol for a prospective, randomized, double-blinded, placebo-controlled clinical trial. Medicine. 2017; 96,e8480.
  • 10. Yang X, Zhang M, Zhang Y, Li W, Yang B. Mesenchymal stem cells derived from Wharton jelly of the human umbilical cord ameliorate damage to human endometrial stromal cells. Fertil. Steril. 2011; 96, 1029–1036.E4.
  • 11. Zhu SF, Hu HB, Xu HY, Fu XF, Peng DX, Su WY, et al. Human umbilical cord mesenchymal stem cell transplantation restores damaged ovaries. J. Cell. Mol. Med. 2015; 19, 2108–2117.
  • 12. Webber L, Davies M, Anderson R, Bartlett J, Braat D, Cartwright B, et al. ESHRE Guideline: Management of women with premature ovarian insufficiency. Hum. Reprod. 2016; 31, 926–937.
  • 13. Zangmo R, Singh N, Sharma J. Diminished ovarian reserve and premature ovarian failure: A review. IVF Lite. 2016; 3, 46.
  • 14. Hoek A, Schoemaker J, Drexhage HA. Premature Ovarian Failure and Ovarian Autoimmunity. Endocr. Rev. 1997; 18, 107–134.
  • 15. Rocha AL, Oliveira FR, Azevedo RC, Silva VA, Peres TM, Candido AL, et al. Recent advances in the understanding and management of polycystic ovary syndrome. F1000Research. 2019; 8. F1000 Faculty Rev-565.
  • 16. Ndefo UA, Eaton A, Green MR. Polycystic ovary syndrome: A review of treatment options with a focus on pharmacological approaches. Pharm. Ther. 2013; 38, 336–355.
  • 17. Sasson IE, Taylor HS. Stem cells and the pathogenesis of endometriosis. Ann. N. Y. Acad. Sci. 2008; 1127, 106–115.
  • 18. Dreisler E, Kjer JJ. Asherman’s syndrome: Current perspectives on diagnosis and management. Int. J. Women’s Health. 2019; 11, 191–198.
  • 19. Kehler J, Hübner K, Garrett S, Schöler HR. Generating oocytes and sperm from embryonic stem cells. Semin. Reprod. Med. 2005; 23, 222–233.
  • 20. Wang J, Liu C, Fujino M, Tong G, Zhang Q, Li XK, et al. Stem cells as a resource for treatment of infertility-related diseases. Curr. Mol. Med. 2019; 19, 539–546.
  • 21. Takahashi K, Yamanaka S. Induction of pluripotent stem cells from mouse embryonic and adult fibroblast cultures by defined factors. Cell. 2006; 126, 663–676.
  • 22. Hou J, Yang S, Yang H, Liu Y, Liu Y, Hai Y, et al. Generation of male differentiated germ cells from various types of stem cells. Reproduction. 2014; 147, R179–R188.
  • 23. Eguizabal C, Montserrat N, Vassena R, Barragan M, Garreta E, Garcia-Quevedo L, et al. Complete meiosis from human induced pluripotent stem cells. Stem Cells. 2011; 29, 1186–1195.
  • 24. Ramathal C, Durruthy-Durruthy J, Sukhwani M, Arakaki JE, Turek PJ, Orwig KE, et al. Fate of iPSCs derived from azoospermic and fertile men following xenotransplantation to murine seminiferous tubules. Cell Rep. 2014; 7, 1284–1297.
  • 25. Sasaki K, Yokobayashi S, Nakamura T, Okamoto I, Yabuta Y, Kurimoto K, et al. Robust In Vitro induction of human germ cell fate from pluripotent stem cells. Cell Stem Cell. 2015; 17, 178–194.
  • 26. Easley CA IV, Phillips BT, McGuire MM, Barringer JM, Valli H, Hermann BP, et al. Direct differentiation of human pluripotent stem cells into haploid spermatogenic cells. Cell Rep. 2012; 2, 440–446.
  • 27. Yamashiro C, Sasaki K, Yabuta Y, Kojima Y, Nakamura T, Okamoto I, et al. Generation of human oogonia from induced pluripotent stem cells In Vitro. Science. 2018; 362, 356–360.
  • 28. Gell JJ, Clark AT. Restoring fertility with human induced pluripotent stem cells: Are we there yet?. Cell Stem Cell. 2018; 23, 777–779.
  • 29. Lee AS, Tang C, Rao MS, Weissman IL, Wu JC. Tumorigenicity as a clinical hurdle for pluripotent stem cell therapies. Nat. Med. 2013; 19, 998–1004.
  • 30. Wu P, Deng G, Sai X, Guo H, Huang H, Zhu P. Maturation strategies and limitations of induced pluripotent stem cell-derived cardiomyocytes. Biosci. Rep. 2020; 25;41.
  • 31. Fernandez TdS, de Souza Fernandez C, Mencalha AL. Human induced pluripotent stem cells from basic research to potential clinical applications in cancer. BioMed Res. Int. 2013; 430290.
  • 32. Dominici M, Le Blanc K, Mueller I, Slaper-Cortenbach I, Marini F, Krause D, et al. Minimal criteria for defining multipotent mesenchymal stromal cells. The International Society for Cellular Therapy position statement. Cytotherapy. 2006; 8, 315–317.
  • 33. Fehrer C, Lepperdinger G. Mesenchymal stem cell aging. Exp. Gerontol. 2005; 40, 926–930.
  • 34. Guillot PV, Gotherstrom C, Chan J, Kurata H, Fisk NM. Human first-trimester fetal MSC express pluripotency markers and grow faster and have longer telomeres than adult MSC. Stem Cells. 2007; 25, 646–654.
  • 35. Brady K, Dickinson SC, Guillot PV, Polak J, Blom AW, Kafienah W, et al. Human fetal and adult bone marrow-derived mesenchymal stem cells use different signaling pathways for the initiation of chondrogenesis. Stem Cells Dev. 2014; 23, 541–554.
  • 36. Huang B, Qian C, Ding C, Meng Q, Zou Q, Li H. Fetal liver mesenchymal stem cells restore ovarian function in premature ovarian insufficiency by targeting MT1. Stem Cell Res. Ther. 2019; 10, 362.
  • 37. Owen M, Friedenstein A. Stromal stem cells: Marrow-derived osteogenic precursors. Ciba Found. Symp. 1988; 136, 42–60.
  • 38. Jing Z, Qiong Z, Yonggang W, Yanping L. Rat bone marrow mesenchymal stem cells improve regeneration of thin endometrium in rat. Fertil. Steril. 2014; 101, 587–594.E3.
  • 39. Wang J, Ju B, Pan C, Gu Y, Zhang Y, Sun L, et al. Application of bone marrow-derived mesenchymal stem cells in the treatment of intrauterine adhesions in rats. Cell. Physiol. Biochem. 2016; 39, 1553–1560.
  • 40. Liu T, Huang Y, Zhang J, Qin W, Chi H, Chen J, et al. Transplantation of human menstrual blood stem cells to treat premature ovarian failure in mouse model. Stem Cells Dev. 2014; 23, 1548–1557.
  • 41. Zheng SX, Wang J, Wang XL, Ali A, Wu LM, Liu YS. Feasibility analysis of treating severe intrauterine adhesions by transplanting menstrual blood-derived stem cells. Int. J. Mol. Med. 2018; 41, 2201–2212.
  • 42. Tan J, Li P, Wang Q, Li Y, Li X, Zhao D, et al. Autologous menstrual blood-derived stromal cells transplantation for severe Asherman’s syndrome. Hum. Reprod. 2016; 31, 2723–2729.
  • 43. de Miguel-Gómez L, López-Martínez S, Francés-Herrero E, Rodríguez-Eguren A, Pellicer A, Cervelló I. Stem cells and the endometrium: From the discovery of adult stem cells to pre-clinical models. Cells. 2021; 10, 595.
  • 44. Saha S, Roy P, Corbitt C, Kakar SS. Application of Stem Cell Therapy for Infertility. Cells. 2021 Jun 28;10(7):1613.
  • 45. Mohamed SA, Shalaby S, Brakta S, Elam L, Elsharoud A, Al-Hendy A. Umbilical cord blood mesenchymal stem cells as an infertility treatment for chemotherapy induced premature ovarian insufficiency. Biomedicines. 2019; 7, 7.
  • 46. 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; 2517514.
  • 47. Xie Q, Xiong X, Xiao N, He K, Chen M, Peng J, et al. Mesenchymal stem cells alleviate DHEA-Induced polycystic ovary syndrome (PCOS) by inhibiting inflammation in mice. Stem Cells Int. 2019; 9782373.
  • 48. Zhang C. The Roles of Different Stem Cells in Premature Ovarian Failure. Curr. Stem Cell Res. Ther. 2020; 15, 473–481.
  • 49. Zhang L, Li Y, Guan C-Y, Tian S, Lv X-D, Li J-H, et al. Therapeutic effect of human umbilical cord-derived mesenchymal stem cells on injured rat endometrium during its chronic phase. Stem Cell Res. Ther. 2018; 9, 36.
  • 50. Xu L, Ding L, Wang L, Cao Y, Zhu H, Lu J, et al. Umbilical cord-derived mesenchymal stem cells on scaffolds facilitate collagen degradation via upregulation of MMP-9 in rat uterine scars. Stem Cell Res. Ther. 2017; 8, 84.
  • 51. Ding L, Yan G, Wang B, Xu L, Gu Y, Ru T, et al. Transplantation of UC-MSCs on collagen scaffold activates follicles in dormant ovaries of POF patients with long history of infertility. Sci. China Life Sci. 2018; 61, 1554–1565.
  • 52. Xiao G-Y, Liu I-H, Cheng C-C, Chang C-C, Lee Y-H, Cheng WT-K, et al. Amniotic fluid stem cells prevent follicle atresia and rescue fertility of mice with premature ovarian failure induced by chemotherapy. PLoS ONE. 2014; 9, e106538.
  • 53. Liu T, Huang Y, Guo L, Cheng W, Zou G. CD44+/CD105+ human amniotic fluid mesenchymal stem cells survive and proliferate in the ovary long-term in a mouse model of chemotherapy-induced premature ovarian failure. Int. J. Med. Sci. 2012; 9, 592.
  • 54. Ling L, Feng X, Wei T, Wang Y, Wang Y, Wang Z, et al. 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 Res. Ther. 2019; 10, 46.
  • 55. Feng X, Ling L, Zhang W, Liu X, Wang Y, Luo Y, et al. Effects of human amnion–derived mesenchymal stem cell (hAD-MSC) transplantation In Situ on primary ovarian insufficiency in SD rats. Reprod. Sci. 2020; 27, 1502–1512.
  • 56. Yin N, Wang Y, Lu X, Liu R, Zhang L, Zhao W, et al. hPMSC transplantation restoring ovarian function in premature ovarian failure mice is associated with change of Th17/Tc17 and Th17/Treg cell ratios through the PI3K/Akt signal pathway. Stem Cell Res. Ther. 2018; 9, 37.
  • 57. Li H, Zhao W, Wang L, Luo Q, Yin N, Lu X, et al. Human placenta-derived mesenchymal stem cells inhibit apoptosis of granulosa cells induced by IRE1α pathway in autoimmune POF mice. Cell Biol. Int. 2019; 43, 899–909.
  • 58. Li H, Zhao W, Wang L, Luo Q, Yin N, Lu X, et al. Human placenta-derived mesenchymal stem cells inhibit apoptosis of granulosa cells induced by IRE1α pathway in autoimmune POF mice. Cell Biol. Int. 2019; 43, 899–909.
  • 59. Damous LL, Nakamuta JS, de Carvalho AES, Carvalho KC, Soares JMJr, de Jesus Simões M, et al. Does adipose tissue-derived stem cell therapy improve graft quality in freshly grafted ovaries? Reprod. Biol. Endocrinol. 2015; 13, 108.
  • 60. Sun M, Wang S, Li Y, Yu L, Gu F, Wang C, et al. Adipose-derived stem cells improved mouse ovary function after chemotherapy-induced ovary failure. Stem Cell Res. Ther. 2013; 4,80.
  • 61. Kilic S, Yuksel B, Pinarli F, Albayrak A, Boztok B, Delibasi T. Effect of stem cell application on Asherman syndrome, an experimental rat model. J. Assist. Reprod. Genet. 2014; 31, 975–982.
  • 62. White YA, Woods DC, Takai Y, Ishihara O, Seki H, Tilly JL. Oocyte formation by mitotically active germ cells purified from ovaries of reproductive-age women. Nat. Med. 2012; 18, 413.
  • 63. Qamar AY, Hussain T, Rafique MK, Bang S, Tanga BM, Seong G, et al. The Role of Stem Cells and Their Derived Extracellular Vesicles in Restoring Female and Male Fertility. Cells. 2021; 17;10:2460.

Stem cell applications in female infertility – A rewiev

Yıl 2023, Cilt: 40 Sayı: 1, 122 - 126, 18.03.2023

Öz

Infertility is a problem that affects approximately 15% of couples today. Although assisted reproduction techniques are widely used today, only 40-50% of couples who apply can have children with their own genetic structure. Especially in people with premature ovarian failure, the rate of conception does not exceed 5-10% with the treatments applied today. For this reason, many studies are carried out to obtain oocyte from stem cells with their proliferation and differentiation feature. In addition, regenerative cellular therapies that can replace assisted reproductive techniques and correct impaired fertility are also being investigated in both animal and human studies. In recent years, research has been carried out on stem cell-derived extracellular vesicles, which will eliminate immunological problems. In terms of safety and efficacy, clinical studies involving large populations are needed.

Kaynakça

  • 1. Zegers-Hochschild F, Adamson GD, Dyer S, Racowsky C, De Mouzon J, Sokol R, et al. The international glossary on infertility and fertility care. Hum. Reprod. 2017; 32, 1786–1801.
  • 2. Volarevic V, Bojic S, Nurkovic J, Volarevic A, Ljujic B, Arsenijevic N, et al. Stem cells as new agents for the treatment of infertility: Current and future perspectives and challenges. BioMed Res. Int. 2014; 507234.
  • 3. Abd-Allah SH, Shalaby SM, Pasha HF, Amal S, Raafat N, Shabrawy SM, et al. Mechanistic action of mesenchymal stem cell injection in the treatment of chemically induced ovarian failure in rabbits. Cytotherapy. 2013; 15, 64–75.
  • 4. 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.
  • 5. Santamaria X, Cabanillas S, Cervello I, Arbona C, Raga F, Ferro J, et al. Autologous cell therapy with CD133+ bone marrow-derived stem cells for refractory Asherman’s syndrome and endometrial atrophy: A pilot cohort study. Hum. Reprod. 2016; 31, 1087–1096.
  • 6. Manshadi MD, Navid S, Hoshino Y, Daneshi E, Noory P, Abbasi M. The effects of human menstrual blood stem cellsderived granulosa cells on ovarian follicle formation in a rat model of premature ovarian failure. Microsc. Res. Tech. 2019; 82,635–642.
  • 7. Domnina A, Novikova P, Obidina J, Fridlyanskaya I, Alekseenko L, Kozhukharova I, et al. Human mesenchymal stem cells in spheroids improve fertility in model animals with damaged endometrium. Stem Cell Res Ther. 2018; 9, 50.
  • 8. Wang S, Yu L, Sun M, Mu S, Wang C, Wang D, et al. The therapeutic potential of umbilical cord mesenchymal stem cells in mice premature ovarian failure. Biomed Res. Int. 2013; 690491.
  • 9. Fan D, Wu S, Ye S, Wang W, Guo X, Liu Z. Umbilical cord mesenchyme stem cell local intramuscular injection for treatment of uterine niche: Protocol for a prospective, randomized, double-blinded, placebo-controlled clinical trial. Medicine. 2017; 96,e8480.
  • 10. Yang X, Zhang M, Zhang Y, Li W, Yang B. Mesenchymal stem cells derived from Wharton jelly of the human umbilical cord ameliorate damage to human endometrial stromal cells. Fertil. Steril. 2011; 96, 1029–1036.E4.
  • 11. Zhu SF, Hu HB, Xu HY, Fu XF, Peng DX, Su WY, et al. Human umbilical cord mesenchymal stem cell transplantation restores damaged ovaries. J. Cell. Mol. Med. 2015; 19, 2108–2117.
  • 12. Webber L, Davies M, Anderson R, Bartlett J, Braat D, Cartwright B, et al. ESHRE Guideline: Management of women with premature ovarian insufficiency. Hum. Reprod. 2016; 31, 926–937.
  • 13. Zangmo R, Singh N, Sharma J. Diminished ovarian reserve and premature ovarian failure: A review. IVF Lite. 2016; 3, 46.
  • 14. Hoek A, Schoemaker J, Drexhage HA. Premature Ovarian Failure and Ovarian Autoimmunity. Endocr. Rev. 1997; 18, 107–134.
  • 15. Rocha AL, Oliveira FR, Azevedo RC, Silva VA, Peres TM, Candido AL, et al. Recent advances in the understanding and management of polycystic ovary syndrome. F1000Research. 2019; 8. F1000 Faculty Rev-565.
  • 16. Ndefo UA, Eaton A, Green MR. Polycystic ovary syndrome: A review of treatment options with a focus on pharmacological approaches. Pharm. Ther. 2013; 38, 336–355.
  • 17. Sasson IE, Taylor HS. Stem cells and the pathogenesis of endometriosis. Ann. N. Y. Acad. Sci. 2008; 1127, 106–115.
  • 18. Dreisler E, Kjer JJ. Asherman’s syndrome: Current perspectives on diagnosis and management. Int. J. Women’s Health. 2019; 11, 191–198.
  • 19. Kehler J, Hübner K, Garrett S, Schöler HR. Generating oocytes and sperm from embryonic stem cells. Semin. Reprod. Med. 2005; 23, 222–233.
  • 20. Wang J, Liu C, Fujino M, Tong G, Zhang Q, Li XK, et al. Stem cells as a resource for treatment of infertility-related diseases. Curr. Mol. Med. 2019; 19, 539–546.
  • 21. Takahashi K, Yamanaka S. Induction of pluripotent stem cells from mouse embryonic and adult fibroblast cultures by defined factors. Cell. 2006; 126, 663–676.
  • 22. Hou J, Yang S, Yang H, Liu Y, Liu Y, Hai Y, et al. Generation of male differentiated germ cells from various types of stem cells. Reproduction. 2014; 147, R179–R188.
  • 23. Eguizabal C, Montserrat N, Vassena R, Barragan M, Garreta E, Garcia-Quevedo L, et al. Complete meiosis from human induced pluripotent stem cells. Stem Cells. 2011; 29, 1186–1195.
  • 24. Ramathal C, Durruthy-Durruthy J, Sukhwani M, Arakaki JE, Turek PJ, Orwig KE, et al. Fate of iPSCs derived from azoospermic and fertile men following xenotransplantation to murine seminiferous tubules. Cell Rep. 2014; 7, 1284–1297.
  • 25. Sasaki K, Yokobayashi S, Nakamura T, Okamoto I, Yabuta Y, Kurimoto K, et al. Robust In Vitro induction of human germ cell fate from pluripotent stem cells. Cell Stem Cell. 2015; 17, 178–194.
  • 26. Easley CA IV, Phillips BT, McGuire MM, Barringer JM, Valli H, Hermann BP, et al. Direct differentiation of human pluripotent stem cells into haploid spermatogenic cells. Cell Rep. 2012; 2, 440–446.
  • 27. Yamashiro C, Sasaki K, Yabuta Y, Kojima Y, Nakamura T, Okamoto I, et al. Generation of human oogonia from induced pluripotent stem cells In Vitro. Science. 2018; 362, 356–360.
  • 28. Gell JJ, Clark AT. Restoring fertility with human induced pluripotent stem cells: Are we there yet?. Cell Stem Cell. 2018; 23, 777–779.
  • 29. Lee AS, Tang C, Rao MS, Weissman IL, Wu JC. Tumorigenicity as a clinical hurdle for pluripotent stem cell therapies. Nat. Med. 2013; 19, 998–1004.
  • 30. Wu P, Deng G, Sai X, Guo H, Huang H, Zhu P. Maturation strategies and limitations of induced pluripotent stem cell-derived cardiomyocytes. Biosci. Rep. 2020; 25;41.
  • 31. Fernandez TdS, de Souza Fernandez C, Mencalha AL. Human induced pluripotent stem cells from basic research to potential clinical applications in cancer. BioMed Res. Int. 2013; 430290.
  • 32. Dominici M, Le Blanc K, Mueller I, Slaper-Cortenbach I, Marini F, Krause D, et al. Minimal criteria for defining multipotent mesenchymal stromal cells. The International Society for Cellular Therapy position statement. Cytotherapy. 2006; 8, 315–317.
  • 33. Fehrer C, Lepperdinger G. Mesenchymal stem cell aging. Exp. Gerontol. 2005; 40, 926–930.
  • 34. Guillot PV, Gotherstrom C, Chan J, Kurata H, Fisk NM. Human first-trimester fetal MSC express pluripotency markers and grow faster and have longer telomeres than adult MSC. Stem Cells. 2007; 25, 646–654.
  • 35. Brady K, Dickinson SC, Guillot PV, Polak J, Blom AW, Kafienah W, et al. Human fetal and adult bone marrow-derived mesenchymal stem cells use different signaling pathways for the initiation of chondrogenesis. Stem Cells Dev. 2014; 23, 541–554.
  • 36. Huang B, Qian C, Ding C, Meng Q, Zou Q, Li H. Fetal liver mesenchymal stem cells restore ovarian function in premature ovarian insufficiency by targeting MT1. Stem Cell Res. Ther. 2019; 10, 362.
  • 37. Owen M, Friedenstein A. Stromal stem cells: Marrow-derived osteogenic precursors. Ciba Found. Symp. 1988; 136, 42–60.
  • 38. Jing Z, Qiong Z, Yonggang W, Yanping L. Rat bone marrow mesenchymal stem cells improve regeneration of thin endometrium in rat. Fertil. Steril. 2014; 101, 587–594.E3.
  • 39. Wang J, Ju B, Pan C, Gu Y, Zhang Y, Sun L, et al. Application of bone marrow-derived mesenchymal stem cells in the treatment of intrauterine adhesions in rats. Cell. Physiol. Biochem. 2016; 39, 1553–1560.
  • 40. Liu T, Huang Y, Zhang J, Qin W, Chi H, Chen J, et al. Transplantation of human menstrual blood stem cells to treat premature ovarian failure in mouse model. Stem Cells Dev. 2014; 23, 1548–1557.
  • 41. Zheng SX, Wang J, Wang XL, Ali A, Wu LM, Liu YS. Feasibility analysis of treating severe intrauterine adhesions by transplanting menstrual blood-derived stem cells. Int. J. Mol. Med. 2018; 41, 2201–2212.
  • 42. Tan J, Li P, Wang Q, Li Y, Li X, Zhao D, et al. Autologous menstrual blood-derived stromal cells transplantation for severe Asherman’s syndrome. Hum. Reprod. 2016; 31, 2723–2729.
  • 43. de Miguel-Gómez L, López-Martínez S, Francés-Herrero E, Rodríguez-Eguren A, Pellicer A, Cervelló I. Stem cells and the endometrium: From the discovery of adult stem cells to pre-clinical models. Cells. 2021; 10, 595.
  • 44. Saha S, Roy P, Corbitt C, Kakar SS. Application of Stem Cell Therapy for Infertility. Cells. 2021 Jun 28;10(7):1613.
  • 45. Mohamed SA, Shalaby S, Brakta S, Elam L, Elsharoud A, Al-Hendy A. Umbilical cord blood mesenchymal stem cells as an infertility treatment for chemotherapy induced premature ovarian insufficiency. Biomedicines. 2019; 7, 7.
  • 46. 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; 2517514.
  • 47. Xie Q, Xiong X, Xiao N, He K, Chen M, Peng J, et al. Mesenchymal stem cells alleviate DHEA-Induced polycystic ovary syndrome (PCOS) by inhibiting inflammation in mice. Stem Cells Int. 2019; 9782373.
  • 48. Zhang C. The Roles of Different Stem Cells in Premature Ovarian Failure. Curr. Stem Cell Res. Ther. 2020; 15, 473–481.
  • 49. Zhang L, Li Y, Guan C-Y, Tian S, Lv X-D, Li J-H, et al. Therapeutic effect of human umbilical cord-derived mesenchymal stem cells on injured rat endometrium during its chronic phase. Stem Cell Res. Ther. 2018; 9, 36.
  • 50. Xu L, Ding L, Wang L, Cao Y, Zhu H, Lu J, et al. Umbilical cord-derived mesenchymal stem cells on scaffolds facilitate collagen degradation via upregulation of MMP-9 in rat uterine scars. Stem Cell Res. Ther. 2017; 8, 84.
  • 51. Ding L, Yan G, Wang B, Xu L, Gu Y, Ru T, et al. Transplantation of UC-MSCs on collagen scaffold activates follicles in dormant ovaries of POF patients with long history of infertility. Sci. China Life Sci. 2018; 61, 1554–1565.
  • 52. Xiao G-Y, Liu I-H, Cheng C-C, Chang C-C, Lee Y-H, Cheng WT-K, et al. Amniotic fluid stem cells prevent follicle atresia and rescue fertility of mice with premature ovarian failure induced by chemotherapy. PLoS ONE. 2014; 9, e106538.
  • 53. Liu T, Huang Y, Guo L, Cheng W, Zou G. CD44+/CD105+ human amniotic fluid mesenchymal stem cells survive and proliferate in the ovary long-term in a mouse model of chemotherapy-induced premature ovarian failure. Int. J. Med. Sci. 2012; 9, 592.
  • 54. Ling L, Feng X, Wei T, Wang Y, Wang Y, Wang Z, et al. 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 Res. Ther. 2019; 10, 46.
  • 55. Feng X, Ling L, Zhang W, Liu X, Wang Y, Luo Y, et al. Effects of human amnion–derived mesenchymal stem cell (hAD-MSC) transplantation In Situ on primary ovarian insufficiency in SD rats. Reprod. Sci. 2020; 27, 1502–1512.
  • 56. Yin N, Wang Y, Lu X, Liu R, Zhang L, Zhao W, et al. hPMSC transplantation restoring ovarian function in premature ovarian failure mice is associated with change of Th17/Tc17 and Th17/Treg cell ratios through the PI3K/Akt signal pathway. Stem Cell Res. Ther. 2018; 9, 37.
  • 57. Li H, Zhao W, Wang L, Luo Q, Yin N, Lu X, et al. Human placenta-derived mesenchymal stem cells inhibit apoptosis of granulosa cells induced by IRE1α pathway in autoimmune POF mice. Cell Biol. Int. 2019; 43, 899–909.
  • 58. Li H, Zhao W, Wang L, Luo Q, Yin N, Lu X, et al. Human placenta-derived mesenchymal stem cells inhibit apoptosis of granulosa cells induced by IRE1α pathway in autoimmune POF mice. Cell Biol. Int. 2019; 43, 899–909.
  • 59. Damous LL, Nakamuta JS, de Carvalho AES, Carvalho KC, Soares JMJr, de Jesus Simões M, et al. Does adipose tissue-derived stem cell therapy improve graft quality in freshly grafted ovaries? Reprod. Biol. Endocrinol. 2015; 13, 108.
  • 60. Sun M, Wang S, Li Y, Yu L, Gu F, Wang C, et al. Adipose-derived stem cells improved mouse ovary function after chemotherapy-induced ovary failure. Stem Cell Res. Ther. 2013; 4,80.
  • 61. Kilic S, Yuksel B, Pinarli F, Albayrak A, Boztok B, Delibasi T. Effect of stem cell application on Asherman syndrome, an experimental rat model. J. Assist. Reprod. Genet. 2014; 31, 975–982.
  • 62. White YA, Woods DC, Takai Y, Ishihara O, Seki H, Tilly JL. Oocyte formation by mitotically active germ cells purified from ovaries of reproductive-age women. Nat. Med. 2012; 18, 413.
  • 63. Qamar AY, Hussain T, Rafique MK, Bang S, Tanga BM, Seong G, et al. The Role of Stem Cells and Their Derived Extracellular Vesicles in Restoring Female and Male Fertility. Cells. 2021; 17;10:2460.
Toplam 63 adet kaynakça vardır.

Ayrıntılar

Birincil Dil İngilizce
Konular Sağlık Kurumları Yönetimi
Bölüm Review Articles
Yazarlar

Ayşe Özdemir 0000-0003-4232-4794

Ayşe Şeyma Taştan 0000-0001-7336-8648

Erken Görünüm Tarihi 18 Mart 2023
Yayımlanma Tarihi 18 Mart 2023
Gönderilme Tarihi 25 Temmuz 2022
Kabul Tarihi 1 Ekim 2022
Yayımlandığı Sayı Yıl 2023 Cilt: 40 Sayı: 1

Kaynak Göster

APA Özdemir, A., & Taştan, A. Ş. (2023). Stem cell applications in female infertility – A rewiev. Journal of Experimental and Clinical Medicine, 40(1), 122-126.
AMA Özdemir A, Taştan AŞ. Stem cell applications in female infertility – A rewiev. J. Exp. Clin. Med. Mart 2023;40(1):122-126.
Chicago Özdemir, Ayşe, ve Ayşe Şeyma Taştan. “Stem Cell Applications in Female Infertility – A Rewiev”. Journal of Experimental and Clinical Medicine 40, sy. 1 (Mart 2023): 122-26.
EndNote Özdemir A, Taştan AŞ (01 Mart 2023) Stem cell applications in female infertility – A rewiev. Journal of Experimental and Clinical Medicine 40 1 122–126.
IEEE A. Özdemir ve A. Ş. Taştan, “Stem cell applications in female infertility – A rewiev”, J. Exp. Clin. Med., c. 40, sy. 1, ss. 122–126, 2023.
ISNAD Özdemir, Ayşe - Taştan, Ayşe Şeyma. “Stem Cell Applications in Female Infertility – A Rewiev”. Journal of Experimental and Clinical Medicine 40/1 (Mart 2023), 122-126.
JAMA Özdemir A, Taştan AŞ. Stem cell applications in female infertility – A rewiev. J. Exp. Clin. Med. 2023;40:122–126.
MLA Özdemir, Ayşe ve Ayşe Şeyma Taştan. “Stem Cell Applications in Female Infertility – A Rewiev”. Journal of Experimental and Clinical Medicine, c. 40, sy. 1, 2023, ss. 122-6.
Vancouver Özdemir A, Taştan AŞ. Stem cell applications in female infertility – A rewiev. J. Exp. Clin. Med. 2023;40(1):122-6.