Derleme
BibTex RIS Kaynak Göster

MEZENKİMAL KÖK HÜCRE VE KOŞULLANDIRILMIŞ BESİYERİNİN OVARYUM HASARI ÜZERİNDEKİ TEDAVİ EDİCİ ETKİLERİ

Yıl 2021, Cilt: 28 Sayı: 1, 179 - 185, 11.03.2021
https://doi.org/10.17343/sdutfd.654926

Öz

Yapılan pek çok yeni çalışmada, mezenkimal kök hücrelerin (MSC) farklı hastalık modellerinde terapötik etkileri gösterilmiştir. İnfertilite, yaygınlığı yüksek küresel bir hastalık olup erkek faktörü, azalmış over rezervi, ovulatuar faktör, tubal faktör, uterin faktör, pelvik faktör ve açıklanamayan infertilite nedeniyle ortaya çıkabilir. MSC'lerin, farklılaşma kapasiteleri ve parakrin etki özellikleri nedeniyle, ovaryan yetmezlik tedavisindeki etkilerini belirlemek için birçok çalışma yapılmıştır. Araştırmalarda farklı kaynaklardan elde edilen MSC'lerin germ hücrelerine farklılaşma kapasiteleri in vitro’da belirlenirken over nişi üzerindeki etkileri MSC'lerin transplantasyonu ile belirlenmiştir. Bu derlemede, ovaryum hasarına bağlı gelişen ovaryan yetmezlik sebeplerini ele alarak, MSC'lerin ve koşullandırılmış besiyerinin ovaryum hasarı üzerindeki terapötik etkilerini belirlemek için mevcut çalışmaları özetledik.

Kaynakça

  • Rowe PJ, Comhaire FH, Hargreave TB, Mellows HJ, Organization WH. WHO manual for the standardized investigation and diagnosis of the infertile couple. 1993.
  • Gnoth C, Godehardt E, Frank-Herrmann P, Friol K, Tigges J, Freundl G. Definition and prevalence of subfertility and infertility. Hum Reprod. 2005;20(5):1144-7.
  • Wang X, Chen C, Wang L, Chen D, Guang W, French J. Conception, early pregnancy loss, and time to clinical pregnancy: a population-based prospective study. Fertil Steril. 2003;79(3):577-84.
  • Zinaman MJ, Clegg ED, Brown CC, O'Connor J, Selevan SG. Estimates of human fertility and pregnancy loss. Fertil Steril. 1996;65(3):503-9.
  • Boivin J, Bunting L, Collins JA, Nygren KG. International estimates of infertility prevalence and treatment-seeking: potential need and demand for infertility medical care. Hum Reprod. 2007;22(6):1506-12.
  • Johnson JA, Tough S, Sogc Genetics C. Delayed child-bearing. J Obstet Gynaecol Can. 2012;34(1):80-93.
  • Hendershot GE, Mosher WD, Pratt WF. Infertility and age: an unresolved issue. Fam Plann Perspect. 1982;14(5):287-9.
  • Menken J, Trussell J, Larsen U. Age and infertility. Science. 1986;233(4771):1389-94.
  • Bradshaw K, Carr B. Modern diagnostic evaluation of the infertile couple. Textbook of Reproductive Medicine East Norwalk, CT: Appleton & Lange. 1993:443-52.
  • Ray A, Shah A, Gudi A, Homburg R. Unexplained infertility: an update and review of practice. Reprod Biomed Online. 2012;24(6):591-602.
  • Hoffman BL, Schorge JO, Bradshaw KD, Halvorson LM, Schaffer JI, Corton MM. Williams Gynecology, Third Edition: McGraw-Hill Education; 2016.
  • Ross MH, Pawlina W. Histology: A Text and Atlas : with Correlated Cell and Molecular Biology: Wolters Kluwer Health; 2016.
  • Hall JE. Guyton and Hall textbook of medical physiology e-Book: Elsevier Health Sciences; 2015.
  • Kierszenbaum AL, Tres L. Histology and Cell Biology: an introduction to pathology E-Book: Elsevier Health Sciences; 2015.
  • Committee on Gynecologic P. Committee opinion no. 618: Ovarian reserve testing. Obstet Gynecol. 2015;125(1):268-73.
  • Billig H, Chun S-Y, Eisenhauer K, Hsueh AJ. Gonadal cell apoptosis: hormone-regulated cell demise. Human reproduction update. 1996;2(2):103-17.
  • Laml T, Schulz-Lobmeyr I, Obruca A, Huber J, Hartmann B. Premature ovarian failure: etiology and prospects. Gynecological endocrinology. 2000;14(4):292-302.
  • Amsterdam A, Dantes A, Selvaraj N, Aharoni D. Apoptosis in steroidogenic cells: structure-function analysis. Steroids. 1997;62(1):207-11.
  • Nelson LM. Clinical practice. Primary ovarian insufficiency. N Engl J Med. 2009;360(6):606-14.
  • Albright F, Smith PH, Fraser R. A syndrome characterized by primary ovarian insufficiency and decreased stature: report of 11 cases with a digression on hormonal control of axillary and pubic hair. 1942.
  • Cooper AR, Baker VL, Sterling EW, Ryan ME, Woodruff TK, Nelson LM. The time is now for a new approach to primary ovarian insufficiency. Fertil Steril. 2011;95(6):1890-7.
  • Rebar RW. Premature ovarian failure. Obstet Gynecol. 2009;113(6):1355-63.
  • van Kasteren YM, Schoemaker J. Premature ovarian failure: a systematic review on therapeutic interventions to restore ovarian function and achieve pregnancy. Hum Reprod Update. 1999;5(5):483-92.
  • Cobo A, Meseguer M, Remohi J, Pellicer A. Use of cryo-banked oocytes in an ovum donation programme: a prospective, randomized, controlled, clinical trial. Hum Reprod. 2010;25(9):2239-46.
  • Oktay K, Oktem O. Ovarian cryopreservation and transplantation for fertility preservation for medical indications: report of an ongoing experience. Fertil Steril. 2010;93(3):762-8.
  • Zhang C. The roles of different stem cells on premature ovarian failure. Current stem cell research & therapy. 2019.
  • Sheikhansari G, Aghebati-Maleki L, Nouri M, Jadidi-Niaragh F, Yousefi M. Current approaches for the treatment of premature ovarian failure with stem cell therapy. Biomed Pharmacother. 2018;102:254-62.
  • Majumdar MK, Thiede MA, Mosca JD, Moorman M, Gerson SL. Phenotypic and functional comparison of cultures of marrow-derived mesenchymal stem cells (MSCs) and stromal cells. J Cell Physiol. 1998;176(1):57-66.
  • Alonso L, Fuchs E. Stem cells of the skin epithelium. Proceedings of the National Academy of Sciences of the United States of America. 2003;100 Suppl 1:11830-5.
  • Harada S, Rodan GA. Control of osteoblast function and regulation of bone mass. Nature. 2003;423(6937):349-55.
  • Radtke F, Clevers H. Self-renewal and cancer of the gut: two sides of a coin. Science. 2005;307(5717):1904-9.
  • Stocum DL. Tissue restoration through regenerative biology and medicine. Advances in anatomy, embryology, and cell biology. 2004;176:III-VIII, 1-101, back cover.
  • Odorico JS, Kaufman DS, Thomson JA. Multilineage differentiation from human embryonic stem cell lines. Stem cells. 2001;19(3):193-204.
  • Stocum DL. Stem cells in regenerative biology and medicine. Wound repair and regeneration : official publication of the Wound Healing Society [and] the European Tissue Repair Society. 2001;9(6):429-42.
  • Friedenstein AJ, Piatetzky S, II, Petrakova KV. Osteogenesis in transplants of bone marrow cells. Journal of embryology and experimental morphology. 1966;16(3):381-90.
  • Friedenstein AJ, Chailakhjan RK, Lalykina KS. The development of fibroblast colonies in monolayer cultures of guinea-pig bone marrow and spleen cells. Cell and tissue kinetics. 1970;3(4):393-403.
  • Caplan AI. Mesenchymal stem cells. Journal of orthopaedic research : official publication of the Orthopaedic Research Society. 1991;9(5):641-50.
  • Prockop DJ. Marrow stromal cells as stem cells for nonhematopoietic tissues. Science. 1997;276(5309):71-4.
  • Liu ZJ, Zhuge Y, Velazquez OC. Trafficking and differentiation of mesenchymal stem cells. J Cell Biochem. 2009;106(6):984-91.
  • 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(4):315-7.
  • Bianco P, Robey PG, Simmons PJ. Mesenchymal stem cells: revisiting history, concepts, and assays. Cell Stem Cell. 2008;2(4):313-9.
  • Zuk PA, Zhu M, Ashjian P, De Ugarte DA, Huang JI, Mizuno H, et al. Human adipose tissue is a source of multipotent stem cells. Mol Biol Cell. 2002;13(12):4279-95.
  • Pittenger MF, Mackay AM, Beck SC, Jaiswal RK, Douglas R, Mosca JD, et al. Multilineage potential of adult human mesenchymal stem cells. Science. 1999;284(5411):143-7.
  • Barry FP, Murphy JM. Mesenchymal stem cells: clinical applications and biological characterization. Int J Biochem Cell Biol. 2004;36(4):568-84.
  • Minguell JJ, Erices A, Conget P. Mesenchymal stem cells. Exp Biol Med (Maywood). 2001;226(6):507-20.
  • Baglio SR, Pegtel DM, Baldini N. Mesenchymal stem cell secreted vesicles provide novel opportunities in (stem) cell-free therapy. Front Physiol. 2012;3:359.
  • Makridakis M, Roubelakis MG, Vlahou A. Stem cells: insights into the secretome. Biochim Biophys Acta. 2013;1834(11):2380-4.
  • Yang D, Wang W, Li L, Peng Y, Chen P, Huang H, et al. The relative contribution of paracine effect versus direct differentiation on adipose-derived stem cell transplantation mediated cardiac repair. PLoS One. 2013;8(3):e59020.
  • Fu X, He Y, Xie C, Liu W. Bone marrow mesenchymal stem cell transplantation improves ovarian function and structure in rats with chemotherapy-induced ovarian damage. Cytotherapy. 2008;10(4):353-63.
  • Wang L, Ying YF, Ouyang YL, Wang JF, Xu J. VEGF and bFGF increase survival of xenografted human ovarian tissue in an experimental rabbit model. J Assist Reprod Genet. 2013;30(10):1301-11.
  • Pawitan JA. Prospect of stem cell conditioned medium in regenerative medicine. Biomed Res Int. 2014;2014:965849.
  • Bhang SH, Lee S, Shin JY, Lee TJ, Jang HK, Kim BS. Efficacious and clinically relevant conditioned medium of human adipose-derived stem cells for therapeutic angiogenesis. Mol Ther. 2014;22(4):862-72.
  • Takehara Y, Yabuuchi A, Ezoe K, Kuroda T, Yamadera R, Sano C, et al. The restorative effects of adipose-derived mesenchymal stem cells on damaged ovarian function. Lab Invest. 2013;93(2):181-93.
  • Lai D, Wang F, Chen Y, Wang L, Wang Y, Cheng W. Human amniotic fluid stem cells have a potential to recover ovarian function in mice with chemotherapy-induced sterility. BMC Dev Biol. 2013;13:34.
  • 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;2013:690491.
  • Lai D, Wang F, Dong Z, Zhang Q. Skin-derived mesenchymal stem cells help restore function to ovaries in a premature ovarian failure mouse model. PLoS One. 2014;9(5):e98749.
  • Wang Z, Wang Y, Yang T, Li J, Yang X. Study of the reparative effects of menstrual-derived stem cells on premature ovarian failure in mice. Stem Cell Res Ther. 2017;8(1):11.
  • Khanmohammadi N, Sameni HR, Mohammadi M, Pakdel A, Mirmohammadkhani M, Parsaie H, et al. Effect of Transplantation of Bone Marrow Stromal Cell- Conditioned Medium on Ovarian Function, Morphology and Cell Death in Cyclophosphamide-Treated Rats. Cell J. 2018;20(1):10-8.
  • Zhang Q, Xu M, Yao X, Li T, Wang Q, Lai D. Human amniotic epithelial cells inhibit granulosa cell apoptosis induced by chemotherapy and restore the fertility. Stem Cell Res Ther. 2015;6:152.
  • Zhang Q, Bu S, Sun J, Xu M, Yao X, He K, et al. Paracrine effects of human amniotic epithelial cells protect against chemotherapy-induced ovarian damage. Stem Cell Res Ther. 2017;8(1):270.
  • Zhang H, Luo Q, Lu X, Yin N, Zhou D, Zhang L, et al. 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. 2018;9(1):20.
  • 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 research & therapy. 2019;10(1):46.

THERAPEUTIC EFFECTS OF MESENCHYMAL STEM CELL AND CONDITIONED MEDIUM ON OVARIAN DAMAGE

Yıl 2021, Cilt: 28 Sayı: 1, 179 - 185, 11.03.2021
https://doi.org/10.17343/sdutfd.654926

Öz

Many recent studies have demonstrated the therapeutic effects of mesenchymal stem cells (MSC) in different disease models. Infertility is a global disease with high prevalence and may be caused by male factor, decreased ovarian reserve, ovulatory factor, tubal factor, uterine factor, pelvic factor and unexplained infertility. Many studies have been conducted to determine the effects of MSCs in the treatment of ovarian insufficiency due to their differentiation capacity and paracrine effects. The differentiation capacities of MSCs obtained from different sources to germ cells were determined in vitro and their effects on ovarian niche were determined by transplantation of MSCs. In this review, we discuss the causes of ovarian insufficiency due to ovarian damage and summarize the current studies to determine the therapeutic effects of MSCs and conditioned media on ovarian damage.

Kaynakça

  • Rowe PJ, Comhaire FH, Hargreave TB, Mellows HJ, Organization WH. WHO manual for the standardized investigation and diagnosis of the infertile couple. 1993.
  • Gnoth C, Godehardt E, Frank-Herrmann P, Friol K, Tigges J, Freundl G. Definition and prevalence of subfertility and infertility. Hum Reprod. 2005;20(5):1144-7.
  • Wang X, Chen C, Wang L, Chen D, Guang W, French J. Conception, early pregnancy loss, and time to clinical pregnancy: a population-based prospective study. Fertil Steril. 2003;79(3):577-84.
  • Zinaman MJ, Clegg ED, Brown CC, O'Connor J, Selevan SG. Estimates of human fertility and pregnancy loss. Fertil Steril. 1996;65(3):503-9.
  • Boivin J, Bunting L, Collins JA, Nygren KG. International estimates of infertility prevalence and treatment-seeking: potential need and demand for infertility medical care. Hum Reprod. 2007;22(6):1506-12.
  • Johnson JA, Tough S, Sogc Genetics C. Delayed child-bearing. J Obstet Gynaecol Can. 2012;34(1):80-93.
  • Hendershot GE, Mosher WD, Pratt WF. Infertility and age: an unresolved issue. Fam Plann Perspect. 1982;14(5):287-9.
  • Menken J, Trussell J, Larsen U. Age and infertility. Science. 1986;233(4771):1389-94.
  • Bradshaw K, Carr B. Modern diagnostic evaluation of the infertile couple. Textbook of Reproductive Medicine East Norwalk, CT: Appleton & Lange. 1993:443-52.
  • Ray A, Shah A, Gudi A, Homburg R. Unexplained infertility: an update and review of practice. Reprod Biomed Online. 2012;24(6):591-602.
  • Hoffman BL, Schorge JO, Bradshaw KD, Halvorson LM, Schaffer JI, Corton MM. Williams Gynecology, Third Edition: McGraw-Hill Education; 2016.
  • Ross MH, Pawlina W. Histology: A Text and Atlas : with Correlated Cell and Molecular Biology: Wolters Kluwer Health; 2016.
  • Hall JE. Guyton and Hall textbook of medical physiology e-Book: Elsevier Health Sciences; 2015.
  • Kierszenbaum AL, Tres L. Histology and Cell Biology: an introduction to pathology E-Book: Elsevier Health Sciences; 2015.
  • Committee on Gynecologic P. Committee opinion no. 618: Ovarian reserve testing. Obstet Gynecol. 2015;125(1):268-73.
  • Billig H, Chun S-Y, Eisenhauer K, Hsueh AJ. Gonadal cell apoptosis: hormone-regulated cell demise. Human reproduction update. 1996;2(2):103-17.
  • Laml T, Schulz-Lobmeyr I, Obruca A, Huber J, Hartmann B. Premature ovarian failure: etiology and prospects. Gynecological endocrinology. 2000;14(4):292-302.
  • Amsterdam A, Dantes A, Selvaraj N, Aharoni D. Apoptosis in steroidogenic cells: structure-function analysis. Steroids. 1997;62(1):207-11.
  • Nelson LM. Clinical practice. Primary ovarian insufficiency. N Engl J Med. 2009;360(6):606-14.
  • Albright F, Smith PH, Fraser R. A syndrome characterized by primary ovarian insufficiency and decreased stature: report of 11 cases with a digression on hormonal control of axillary and pubic hair. 1942.
  • Cooper AR, Baker VL, Sterling EW, Ryan ME, Woodruff TK, Nelson LM. The time is now for a new approach to primary ovarian insufficiency. Fertil Steril. 2011;95(6):1890-7.
  • Rebar RW. Premature ovarian failure. Obstet Gynecol. 2009;113(6):1355-63.
  • van Kasteren YM, Schoemaker J. Premature ovarian failure: a systematic review on therapeutic interventions to restore ovarian function and achieve pregnancy. Hum Reprod Update. 1999;5(5):483-92.
  • Cobo A, Meseguer M, Remohi J, Pellicer A. Use of cryo-banked oocytes in an ovum donation programme: a prospective, randomized, controlled, clinical trial. Hum Reprod. 2010;25(9):2239-46.
  • Oktay K, Oktem O. Ovarian cryopreservation and transplantation for fertility preservation for medical indications: report of an ongoing experience. Fertil Steril. 2010;93(3):762-8.
  • Zhang C. The roles of different stem cells on premature ovarian failure. Current stem cell research & therapy. 2019.
  • Sheikhansari G, Aghebati-Maleki L, Nouri M, Jadidi-Niaragh F, Yousefi M. Current approaches for the treatment of premature ovarian failure with stem cell therapy. Biomed Pharmacother. 2018;102:254-62.
  • Majumdar MK, Thiede MA, Mosca JD, Moorman M, Gerson SL. Phenotypic and functional comparison of cultures of marrow-derived mesenchymal stem cells (MSCs) and stromal cells. J Cell Physiol. 1998;176(1):57-66.
  • Alonso L, Fuchs E. Stem cells of the skin epithelium. Proceedings of the National Academy of Sciences of the United States of America. 2003;100 Suppl 1:11830-5.
  • Harada S, Rodan GA. Control of osteoblast function and regulation of bone mass. Nature. 2003;423(6937):349-55.
  • Radtke F, Clevers H. Self-renewal and cancer of the gut: two sides of a coin. Science. 2005;307(5717):1904-9.
  • Stocum DL. Tissue restoration through regenerative biology and medicine. Advances in anatomy, embryology, and cell biology. 2004;176:III-VIII, 1-101, back cover.
  • Odorico JS, Kaufman DS, Thomson JA. Multilineage differentiation from human embryonic stem cell lines. Stem cells. 2001;19(3):193-204.
  • Stocum DL. Stem cells in regenerative biology and medicine. Wound repair and regeneration : official publication of the Wound Healing Society [and] the European Tissue Repair Society. 2001;9(6):429-42.
  • Friedenstein AJ, Piatetzky S, II, Petrakova KV. Osteogenesis in transplants of bone marrow cells. Journal of embryology and experimental morphology. 1966;16(3):381-90.
  • Friedenstein AJ, Chailakhjan RK, Lalykina KS. The development of fibroblast colonies in monolayer cultures of guinea-pig bone marrow and spleen cells. Cell and tissue kinetics. 1970;3(4):393-403.
  • Caplan AI. Mesenchymal stem cells. Journal of orthopaedic research : official publication of the Orthopaedic Research Society. 1991;9(5):641-50.
  • Prockop DJ. Marrow stromal cells as stem cells for nonhematopoietic tissues. Science. 1997;276(5309):71-4.
  • Liu ZJ, Zhuge Y, Velazquez OC. Trafficking and differentiation of mesenchymal stem cells. J Cell Biochem. 2009;106(6):984-91.
  • 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(4):315-7.
  • Bianco P, Robey PG, Simmons PJ. Mesenchymal stem cells: revisiting history, concepts, and assays. Cell Stem Cell. 2008;2(4):313-9.
  • Zuk PA, Zhu M, Ashjian P, De Ugarte DA, Huang JI, Mizuno H, et al. Human adipose tissue is a source of multipotent stem cells. Mol Biol Cell. 2002;13(12):4279-95.
  • Pittenger MF, Mackay AM, Beck SC, Jaiswal RK, Douglas R, Mosca JD, et al. Multilineage potential of adult human mesenchymal stem cells. Science. 1999;284(5411):143-7.
  • Barry FP, Murphy JM. Mesenchymal stem cells: clinical applications and biological characterization. Int J Biochem Cell Biol. 2004;36(4):568-84.
  • Minguell JJ, Erices A, Conget P. Mesenchymal stem cells. Exp Biol Med (Maywood). 2001;226(6):507-20.
  • Baglio SR, Pegtel DM, Baldini N. Mesenchymal stem cell secreted vesicles provide novel opportunities in (stem) cell-free therapy. Front Physiol. 2012;3:359.
  • Makridakis M, Roubelakis MG, Vlahou A. Stem cells: insights into the secretome. Biochim Biophys Acta. 2013;1834(11):2380-4.
  • Yang D, Wang W, Li L, Peng Y, Chen P, Huang H, et al. The relative contribution of paracine effect versus direct differentiation on adipose-derived stem cell transplantation mediated cardiac repair. PLoS One. 2013;8(3):e59020.
  • Fu X, He Y, Xie C, Liu W. Bone marrow mesenchymal stem cell transplantation improves ovarian function and structure in rats with chemotherapy-induced ovarian damage. Cytotherapy. 2008;10(4):353-63.
  • Wang L, Ying YF, Ouyang YL, Wang JF, Xu J. VEGF and bFGF increase survival of xenografted human ovarian tissue in an experimental rabbit model. J Assist Reprod Genet. 2013;30(10):1301-11.
  • Pawitan JA. Prospect of stem cell conditioned medium in regenerative medicine. Biomed Res Int. 2014;2014:965849.
  • Bhang SH, Lee S, Shin JY, Lee TJ, Jang HK, Kim BS. Efficacious and clinically relevant conditioned medium of human adipose-derived stem cells for therapeutic angiogenesis. Mol Ther. 2014;22(4):862-72.
  • Takehara Y, Yabuuchi A, Ezoe K, Kuroda T, Yamadera R, Sano C, et al. The restorative effects of adipose-derived mesenchymal stem cells on damaged ovarian function. Lab Invest. 2013;93(2):181-93.
  • Lai D, Wang F, Chen Y, Wang L, Wang Y, Cheng W. Human amniotic fluid stem cells have a potential to recover ovarian function in mice with chemotherapy-induced sterility. BMC Dev Biol. 2013;13:34.
  • 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;2013:690491.
  • Lai D, Wang F, Dong Z, Zhang Q. Skin-derived mesenchymal stem cells help restore function to ovaries in a premature ovarian failure mouse model. PLoS One. 2014;9(5):e98749.
  • Wang Z, Wang Y, Yang T, Li J, Yang X. Study of the reparative effects of menstrual-derived stem cells on premature ovarian failure in mice. Stem Cell Res Ther. 2017;8(1):11.
  • Khanmohammadi N, Sameni HR, Mohammadi M, Pakdel A, Mirmohammadkhani M, Parsaie H, et al. Effect of Transplantation of Bone Marrow Stromal Cell- Conditioned Medium on Ovarian Function, Morphology and Cell Death in Cyclophosphamide-Treated Rats. Cell J. 2018;20(1):10-8.
  • Zhang Q, Xu M, Yao X, Li T, Wang Q, Lai D. Human amniotic epithelial cells inhibit granulosa cell apoptosis induced by chemotherapy and restore the fertility. Stem Cell Res Ther. 2015;6:152.
  • Zhang Q, Bu S, Sun J, Xu M, Yao X, He K, et al. Paracrine effects of human amniotic epithelial cells protect against chemotherapy-induced ovarian damage. Stem Cell Res Ther. 2017;8(1):270.
  • Zhang H, Luo Q, Lu X, Yin N, Zhou D, Zhang L, et al. 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. 2018;9(1):20.
  • 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 research & therapy. 2019;10(1):46.
Toplam 62 adet kaynakça vardır.

Ayrıntılar

Birincil Dil Türkçe
Konular Klinik Tıp Bilimleri
Bölüm Derlemeler
Yazarlar

Burak Ün 0000-0002-8885-7062

Meryem Akpolat Ferah 0000-0002-3419-1728

Büşra Çetinkaya Ün 0000-0001-9466-4278

Yayımlanma Tarihi 11 Mart 2021
Gönderilme Tarihi 4 Aralık 2019
Kabul Tarihi 21 Mayıs 2020
Yayımlandığı Sayı Yıl 2021 Cilt: 28 Sayı: 1

Kaynak Göster

Vancouver Ün B, Ferah MA, Çetinkaya Ün B. MEZENKİMAL KÖK HÜCRE VE KOŞULLANDIRILMIŞ BESİYERİNİN OVARYUM HASARI ÜZERİNDEKİ TEDAVİ EDİCİ ETKİLERİ. SDÜ Tıp Fak Derg. 2021;28(1):179-85.

                                                                                         14791


Süleyman Demirel Üniversitesi Tıp Fakültesi Dergisi/Medical Journal of Süleyman Demirel University is licensed under Creative Commons Attribution-NonCommercial-NoDerivs 4.0 International.