Skip to main content
Top
Published in: Reproductive Biology and Endocrinology 1/2020

Open Access 01-12-2020 | Research

Decellularization and recellularization of the ovary for bioengineering applications; studies in the mouse

Authors: Ahmed Baker Alshaikh, Arvind Manikantan Padma, Matilda Dehlin, Randa Akouri, Min Jong Song, Mats Brännström, Mats Hellström

Published in: Reproductive Biology and Endocrinology | Issue 1/2020

Login to get access

Abstract

Background

Fertility preservation is particularly challenging in young women diagnosed with hematopoietic cancers, as transplantation of cryopreserved ovarian cortex in these women carries the risk for re-introducing cancer cells. Therefore, the construction of a bioengineered ovary that can accommodate isolated small follicles was proposed as an alternative to minimize the risk of malignancy transmission. Various options for viable bioengineered scaffolds have been reported in the literature. Previously, we reported three protocols for producing mouse ovarian scaffolds with the decellularization technique. The present study examined these scaffolds further, specifically with regards to their extracellular composition, biocompatibility and ability to support recellularization with mesenchymal stem cells.

Material and methods

Three decellularization protocols based on 0.5% sodium dodecyl sulfate (Protocol 1; P1), or 2% sodium deoxycholate (P2), or a combination of the two detergents (P3) were applied to produce three types of scaffolds. The levels of collagen, elastin and sulfated glycosaminoglycans (sGAGs) were quantified in the remaining extracellular matrix. Detailed immunofluorescence and scanning electron microscopy imaging were conducted to assess the morphology and recellularization efficiency of the constructs after 14 days in vitro utilizing red fluorescent protein-labelled mesenchymal stem cells.

Results

All protocols efficiently removed the DNA while the elastin content was not significantly reduced during the procedures. The SDS-protocol (P1) reduced the sGAG and the collagen content more than the SDC-protocol (P2). All scaffolds were biocompatible and recellularization was successful, particularly in several P2-derived scaffolds. The cells were extensively distributed throughout the constructs, with a denser distribution observed towards the ovarian cortex. The cell density was not significantly different (400 to 550 cells/mm2) between scaffold types. However, there was a tendency towards a higher cell density in the SDC-derived constructs. Scanning electron microscope images showed fibrous scaffolds with a dense repopulated surface structure.

Conclusions

While there were differences in the key structural macromolecules between protocols, all scaffolds were biocompatible and showed effective recellularization. The results indicate that our SDC-protocol might be better than our SDS-protocol. However, additional studies are necessary to determine their suitability for attachment of small follicles and folliculogenesis.
Literature
1.
go back to reference Pereira N, Schattman GL. Fertility preservation and sexual health after Cancer therapy. J Oncol Pract. 2017;13:643–51.PubMed Pereira N, Schattman GL. Fertility preservation and sexual health after Cancer therapy. J Oncol Pract. 2017;13:643–51.PubMed
2.
go back to reference Wallace WH, Thomson AB, Kelsey TW. The radiosensitivity of the human oocyte. Hum Reprod. 2003;18:117–21.PubMed Wallace WH, Thomson AB, Kelsey TW. The radiosensitivity of the human oocyte. Hum Reprod. 2003;18:117–21.PubMed
3.
go back to reference Diaz-Garcia C, Domingo J, Garcia-Velasco JA, Herraiz S, Mirabet V, Iniesta I, Cobo A, Remohi J, Pellicer A. Oocyte vitrification versus ovarian cortex transplantation in fertility preservation for adult women undergoing gonadotoxic treatments: a prospective cohort study. Fertil Steril. 2018. Diaz-Garcia C, Domingo J, Garcia-Velasco JA, Herraiz S, Mirabet V, Iniesta I, Cobo A, Remohi J, Pellicer A. Oocyte vitrification versus ovarian cortex transplantation in fertility preservation for adult women undergoing gonadotoxic treatments: a prospective cohort study. Fertil Steril. 2018.
4.
go back to reference Moravek MB, Confino R, Smith KN, Kazer RR, Klock SC, Lawson AK, Gradishar WJ, Pavone ME. Long-term outcomes in cancer patients who did or did not pursue fertility preservation. Fertil Steril. 2018;109:349–55.PubMedPubMedCentral Moravek MB, Confino R, Smith KN, Kazer RR, Klock SC, Lawson AK, Gradishar WJ, Pavone ME. Long-term outcomes in cancer patients who did or did not pursue fertility preservation. Fertil Steril. 2018;109:349–55.PubMedPubMedCentral
5.
go back to reference Donnez J, Dolmans MM, Pellicer A, Diaz-Garcia C, Sanchez Serrano M, Schmidt KT, Ernst E, Luyckx V, Andersen CY. Restoration of ovarian activity and pregnancy after transplantation of cryopreserved ovarian tissue: a review of 60 cases of reimplantation. Fertil Steril. 2013;99:1503–13.PubMed Donnez J, Dolmans MM, Pellicer A, Diaz-Garcia C, Sanchez Serrano M, Schmidt KT, Ernst E, Luyckx V, Andersen CY. Restoration of ovarian activity and pregnancy after transplantation of cryopreserved ovarian tissue: a review of 60 cases of reimplantation. Fertil Steril. 2013;99:1503–13.PubMed
6.
go back to reference Ribeiro R, Rebolho JC, Tsumanuma FK, Brandalize GG, Trippia CH, Saab KA. Uterine transposition: technique and a case report. Fertil Steril. 2017;108:320–4 e321.PubMed Ribeiro R, Rebolho JC, Tsumanuma FK, Brandalize GG, Trippia CH, Saab KA. Uterine transposition: technique and a case report. Fertil Steril. 2017;108:320–4 e321.PubMed
7.
go back to reference Donnez J, Dolmans MM. Fertility preservation in women. N Engl J Med. 2017;377:1657–65.PubMed Donnez J, Dolmans MM. Fertility preservation in women. N Engl J Med. 2017;377:1657–65.PubMed
8.
go back to reference Dolmans MM, Marinescu C, Saussoy P, Van Langendonckt A, Amorim C, Donnez J. Reimplantation of cryopreserved ovarian tissue from patients with acute lymphoblastic leukemia is potentially unsafe. Blood. 2010;116:2908–14.PubMed Dolmans MM, Marinescu C, Saussoy P, Van Langendonckt A, Amorim C, Donnez J. Reimplantation of cryopreserved ovarian tissue from patients with acute lymphoblastic leukemia is potentially unsafe. Blood. 2010;116:2908–14.PubMed
9.
go back to reference Chiti MC, Dolmans MM, Donnez J, Amorim CA. Fibrin in reproductive tissue engineering: a review on its application as a biomaterial for fertility preservation. Ann Biomed Eng. 2017. Chiti MC, Dolmans MM, Donnez J, Amorim CA. Fibrin in reproductive tissue engineering: a review on its application as a biomaterial for fertility preservation. Ann Biomed Eng. 2017.
10.
go back to reference Fisch B, Abir R. Female fertility preservation: past, present and future. Reproduction. 2018;156:F11–27.PubMed Fisch B, Abir R. Female fertility preservation: past, present and future. Reproduction. 2018;156:F11–27.PubMed
11.
go back to reference McLaughlin M, Albertini DF, Wallace WHB, Anderson RA, Telfer EE. Metaphase II oocytes from human unilaminar follicles grown in a multi-step culture system. Mol Hum Reprod. 2018;24:135–42.PubMed McLaughlin M, Albertini DF, Wallace WHB, Anderson RA, Telfer EE. Metaphase II oocytes from human unilaminar follicles grown in a multi-step culture system. Mol Hum Reprod. 2018;24:135–42.PubMed
12.
go back to reference Luyckx V, Dolmans MM, Vanacker J, Scalercio SR, Donnez J, Amorim CA. First step in developing a 3D biodegradable fibrin scaffold for an artificial ovary. J Ovarian Res. 2013;6:83.PubMedPubMedCentral Luyckx V, Dolmans MM, Vanacker J, Scalercio SR, Donnez J, Amorim CA. First step in developing a 3D biodegradable fibrin scaffold for an artificial ovary. J Ovarian Res. 2013;6:83.PubMedPubMedCentral
13.
go back to reference Vanacker J, Dolmans MM, Luyckx V, Donnez J, Amorim CA. First transplantation of isolated murine follicles in alginate. Regen Med. 2014;9:609–19.PubMed Vanacker J, Dolmans MM, Luyckx V, Donnez J, Amorim CA. First transplantation of isolated murine follicles in alginate. Regen Med. 2014;9:609–19.PubMed
14.
go back to reference Xu M, Kreeger PK, Shea LD, Woodruff TK. Tissue-engineered follicles produce live, fertile offspring. Tissue Eng. 2006;12:2739–46.PubMedPubMedCentral Xu M, Kreeger PK, Shea LD, Woodruff TK. Tissue-engineered follicles produce live, fertile offspring. Tissue Eng. 2006;12:2739–46.PubMedPubMedCentral
15.
go back to reference Shikanov A, Smith RM, Xu M, Woodruff TK, Shea LD. Hydrogel network design using multifunctional macromers to coordinate tissue maturation in ovarian follicle culture. Biomaterials. 2011;32:2524–31.PubMedPubMedCentral Shikanov A, Smith RM, Xu M, Woodruff TK, Shea LD. Hydrogel network design using multifunctional macromers to coordinate tissue maturation in ovarian follicle culture. Biomaterials. 2011;32:2524–31.PubMedPubMedCentral
16.
go back to reference Laronda MM, Rutz AL, Xiao S, Whelan KA, Duncan FE, Roth EW, Woodruff TK, Shah RN. A bioprosthetic ovary created using 3D printed microporous scaffolds restores ovarian function in sterilized mice. Nat Commun. 2017;8:15261.PubMedPubMedCentral Laronda MM, Rutz AL, Xiao S, Whelan KA, Duncan FE, Roth EW, Woodruff TK, Shah RN. A bioprosthetic ovary created using 3D printed microporous scaffolds restores ovarian function in sterilized mice. Nat Commun. 2017;8:15261.PubMedPubMedCentral
17.
go back to reference Shea LD, Woodruff TK, Shikanov A. Bioengineering the ovarian follicle microenvironment. Annu Rev Biomed Eng. 2014;16:29–52.PubMedPubMedCentral Shea LD, Woodruff TK, Shikanov A. Bioengineering the ovarian follicle microenvironment. Annu Rev Biomed Eng. 2014;16:29–52.PubMedPubMedCentral
18.
go back to reference Xiao S, Zhang J, Romero MM, Smith KN, Shea LD, Woodruff TK. In vitro follicle growth supports human oocyte meiotic maturation. Sci Rep. 2015;5:17323.PubMedPubMedCentral Xiao S, Zhang J, Romero MM, Smith KN, Shea LD, Woodruff TK. In vitro follicle growth supports human oocyte meiotic maturation. Sci Rep. 2015;5:17323.PubMedPubMedCentral
19.
go back to reference Campo H, Cervello I, Simon C. Bioengineering the uterus: an overview of recent advances and future perspectives in reproductive medicine. Ann Biomed Eng. 2017;45:1710–7.PubMed Campo H, Cervello I, Simon C. Bioengineering the uterus: an overview of recent advances and future perspectives in reproductive medicine. Ann Biomed Eng. 2017;45:1710–7.PubMed
20.
go back to reference Hellström M, Bandstein S, Brännström M. Uterine tissue engineering and the future of uterus transplantation. Ann Biomed Eng. 2017;45:1718–30.PubMed Hellström M, Bandstein S, Brännström M. Uterine tissue engineering and the future of uterus transplantation. Ann Biomed Eng. 2017;45:1718–30.PubMed
21.
go back to reference Peloso A, Dhal A, Zambon JP, Li P, Orlando G, Atala A, Soker S. Current achievements and future perspectives in whole-organ bioengineering. Stem Cell Res Ther. 2015;6:107.PubMedPubMedCentral Peloso A, Dhal A, Zambon JP, Li P, Orlando G, Atala A, Soker S. Current achievements and future perspectives in whole-organ bioengineering. Stem Cell Res Ther. 2015;6:107.PubMedPubMedCentral
22.
go back to reference Reing JE, Zhang L, Myers-Irvin J, Cordero KE, Freytes DO, Heber-Katz E, Bedelbaeva K, McIntosh D, Dewilde A, Braunhut SJ, Badylak SF. Degradation products of extracellular matrix affect cell migration and proliferation. Tissue Eng Part A. 2009;15:605–14.PubMed Reing JE, Zhang L, Myers-Irvin J, Cordero KE, Freytes DO, Heber-Katz E, Bedelbaeva K, McIntosh D, Dewilde A, Braunhut SJ, Badylak SF. Degradation products of extracellular matrix affect cell migration and proliferation. Tissue Eng Part A. 2009;15:605–14.PubMed
23.
go back to reference Laronda MM, Jakus AE, Whelan KA, Wertheim JA, Shah RN, Woodruff TK. Initiation of puberty in mice following decellularized ovary transplant. Biomaterials. 2015;50:20–9.PubMedPubMedCentral Laronda MM, Jakus AE, Whelan KA, Wertheim JA, Shah RN, Woodruff TK. Initiation of puberty in mice following decellularized ovary transplant. Biomaterials. 2015;50:20–9.PubMedPubMedCentral
24.
go back to reference Liu WY, Lin SG, Zhuo RY, Xie YY, Pan W, Lin XF, Shen FX. Xenogeneic Decellularized scaffold: a novel platform for ovary regeneration. Tissue Eng Part C Methods. 2017;23:61–71.PubMedPubMedCentral Liu WY, Lin SG, Zhuo RY, Xie YY, Pan W, Lin XF, Shen FX. Xenogeneic Decellularized scaffold: a novel platform for ovary regeneration. Tissue Eng Part C Methods. 2017;23:61–71.PubMedPubMedCentral
25.
go back to reference Hassanpour A, Talaei-Khozani T, Kargar-Abarghouei E, Razban V, Vojdani Z. Decellularized human ovarian scaffold based on a sodium lauryl ester sulfate (SLES)-treated protocol, as a natural three-dimensional scaffold for construction of bioengineered ovaries. Stem Cell Res Ther. 2018;9:252.PubMedPubMedCentral Hassanpour A, Talaei-Khozani T, Kargar-Abarghouei E, Razban V, Vojdani Z. Decellularized human ovarian scaffold based on a sodium lauryl ester sulfate (SLES)-treated protocol, as a natural three-dimensional scaffold for construction of bioengineered ovaries. Stem Cell Res Ther. 2018;9:252.PubMedPubMedCentral
26.
go back to reference Jakus AE, Laronda MM, Rashedi AS, Robinson CM, Lee C, Jordan SW, Orwig KE, Woodruff TK, Shah RN. "tissue papers" from organ-specific Decellularized extracellular matrices. Adv Funct Mater. 2017;27. Jakus AE, Laronda MM, Rashedi AS, Robinson CM, Lee C, Jordan SW, Orwig KE, Woodruff TK, Shah RN. "tissue papers" from organ-specific Decellularized extracellular matrices. Adv Funct Mater. 2017;27.
27.
go back to reference Oktay K, Bedoschi G, Pacheco F, Turan V, Emirdar V. First pregnancies, live birth, and in vitro fertilization outcomes after transplantation of frozen-banked ovarian tissue with a human extracellular matrix scaffold using robot-assisted minimally invasive surgery. Am J Obstet Gynecol. 2016;214:94.e91–9. Oktay K, Bedoschi G, Pacheco F, Turan V, Emirdar V. First pregnancies, live birth, and in vitro fertilization outcomes after transplantation of frozen-banked ovarian tissue with a human extracellular matrix scaffold using robot-assisted minimally invasive surgery. Am J Obstet Gynecol. 2016;214:94.e91–9.
28.
go back to reference Pors SE, Ramlose M, Nikiforov D, Lundsgaard K, Cheng J, Andersen CY, Kristensen SG. Initial steps in reconstruction of the human ovary: survival of pre-antral stage follicles in a decellularized human ovarian scaffold. Hum Reprod. 2019;34:1523–35.PubMed Pors SE, Ramlose M, Nikiforov D, Lundsgaard K, Cheng J, Andersen CY, Kristensen SG. Initial steps in reconstruction of the human ovary: survival of pre-antral stage follicles in a decellularized human ovarian scaffold. Hum Reprod. 2019;34:1523–35.PubMed
29.
go back to reference Sadr SZ, Fatehi R, Maroufizadeh S, Amorim CA, Ebrahimi B. Utilizing fibrin-alginate and Matrigel-alginate for mouse follicle development in three-dimensional culture systems. Biopreserv Biobank. 2018. Sadr SZ, Fatehi R, Maroufizadeh S, Amorim CA, Ebrahimi B. Utilizing fibrin-alginate and Matrigel-alginate for mouse follicle development in three-dimensional culture systems. Biopreserv Biobank. 2018.
30.
go back to reference Alshaikh AB, Padma AM, Dehlin M, Akouri R, Song MJ, Brannstrom M, Hellstrom M. Decellularization of the mouse ovary: comparison of different scaffold generation protocols for future ovarian bioengineering. J Ovarian Res. 2019;12:58.PubMedPubMedCentral Alshaikh AB, Padma AM, Dehlin M, Akouri R, Song MJ, Brannstrom M, Hellstrom M. Decellularization of the mouse ovary: comparison of different scaffold generation protocols for future ovarian bioengineering. J Ovarian Res. 2019;12:58.PubMedPubMedCentral
31.
go back to reference Hellström M, El-Akouri RR, Sihlbom C, Olsson BM, Lengqvist J, Bäckdahl H, Johansson BR, Olausson M, Sumitran-Holgersson S, Brännström M. Towards the development of a bioengineered uterus: comparison of different protocols for rat uterus decellularization. Acta Biomater. 2014;10:5034–42.PubMed Hellström M, El-Akouri RR, Sihlbom C, Olsson BM, Lengqvist J, Bäckdahl H, Johansson BR, Olausson M, Sumitran-Holgersson S, Brännström M. Towards the development of a bioengineered uterus: comparison of different protocols for rat uterus decellularization. Acta Biomater. 2014;10:5034–42.PubMed
32.
go back to reference Hellström M, Moreno-Moya JM, Bandstein S, Bom E, Akouri RR, Miyazaki K, Maruyama T, Brännström M. Bioengineered uterine tissue supports pregnancy in a rat model. Fertil Steril. 2016;106:487–96 e481.PubMed Hellström M, Moreno-Moya JM, Bandstein S, Bom E, Akouri RR, Miyazaki K, Maruyama T, Brännström M. Bioengineered uterine tissue supports pregnancy in a rat model. Fertil Steril. 2016;106:487–96 e481.PubMed
33.
go back to reference Simsa R, Padma AM, Heher P, Hellstrom M, Teuschl A, Jenndahl L, Bergh N, Fogelstrand P. Systematic in vitro comparison of decellularization protocols for blood vessels. PLoS One. 2018;13:e0209269.PubMedPubMedCentral Simsa R, Padma AM, Heher P, Hellstrom M, Teuschl A, Jenndahl L, Bergh N, Fogelstrand P. Systematic in vitro comparison of decellularization protocols for blood vessels. PLoS One. 2018;13:e0209269.PubMedPubMedCentral
34.
go back to reference Sittadjody S, Enck KM, Wells A, Yoo JJ, Atala A, Saul JM, Opara EC. Encapsulation of Mesenchymal stem cells in 3D ovarian cell constructs promotes stable and long-term hormone secretion with improved physiological outcomes in a syngeneic rat model. Ann Biomed Eng. 2019. Sittadjody S, Enck KM, Wells A, Yoo JJ, Atala A, Saul JM, Opara EC. Encapsulation of Mesenchymal stem cells in 3D ovarian cell constructs promotes stable and long-term hormone secretion with improved physiological outcomes in a syngeneic rat model. Ann Biomed Eng. 2019.
35.
go back to reference Xia X, Yin T, Yan J, Yan L, Jin C, Lu C, Wang T, Zhu X, Zhi X, Wang J, et al. Mesenchymal stem cells enhance angiogenesis and follicle survival in human cryopreserved ovarian cortex transplantation. Cell Transplant. 2015;24:1999–2010.PubMed Xia X, Yin T, Yan J, Yan L, Jin C, Lu C, Wang T, Zhu X, Zhi X, Wang J, et al. Mesenchymal stem cells enhance angiogenesis and follicle survival in human cryopreserved ovarian cortex transplantation. Cell Transplant. 2015;24:1999–2010.PubMed
36.
go back to reference Herraiz S, Romeu M, Buigues A, Martinez S, Diaz-Garcia C, Gomez-Segui I, Martinez J, Pellicer N, Pellicer A. Autologous stem cell ovarian transplantation to increase reproductive potential in patients who are poor responders. Fertil Steril. 2018;110:496–505 e491.PubMed Herraiz S, Romeu M, Buigues A, Martinez S, Diaz-Garcia C, Gomez-Segui I, Martinez J, Pellicer N, Pellicer A. Autologous stem cell ovarian transplantation to increase reproductive potential in patients who are poor responders. Fertil Steril. 2018;110:496–505 e491.PubMed
37.
go back to reference Davies S, Forge A. Preparation of the mammalian organ of Corti for scanning electron microscopy. J Microsc. 1987;147:89–101.PubMed Davies S, Forge A. Preparation of the mammalian organ of Corti for scanning electron microscopy. J Microsc. 1987;147:89–101.PubMed
38.
go back to reference Hillebrandt KH, Everwien H, Haep N, Keshi E, Pratschke J, Sauer IM. Strategies based on organ decellularization and recellularization. Transpl Int. 2019;32:571–85.PubMed Hillebrandt KH, Everwien H, Haep N, Keshi E, Pratschke J, Sauer IM. Strategies based on organ decellularization and recellularization. Transpl Int. 2019;32:571–85.PubMed
39.
go back to reference Caralt M, Uzarski JS, Iacob S, Obergfell KP, Berg N, Bijonowski BM, Kiefer KM, Ward HH, Wandinger-Ness A, Miller WM, et al. Optimization and critical evaluation of decellularization strategies to develop renal extracellular matrix scaffolds as biological templates for organ engineering and transplantation. Am J Transplant. 2015;15:64–75.PubMed Caralt M, Uzarski JS, Iacob S, Obergfell KP, Berg N, Bijonowski BM, Kiefer KM, Ward HH, Wandinger-Ness A, Miller WM, et al. Optimization and critical evaluation of decellularization strategies to develop renal extracellular matrix scaffolds as biological templates for organ engineering and transplantation. Am J Transplant. 2015;15:64–75.PubMed
40.
go back to reference Eivazkhani F, Abtahi NS, Tavana S, Mirzaeian L, Abedi F, Ebrahimi B, Montazeri L, Valojerdi MR, Fathi R. Evaluating two ovarian decellularization methods in three species. Mater Sci Eng C Mater Biol Appl. 2019;102:670–82.PubMed Eivazkhani F, Abtahi NS, Tavana S, Mirzaeian L, Abedi F, Ebrahimi B, Montazeri L, Valojerdi MR, Fathi R. Evaluating two ovarian decellularization methods in three species. Mater Sci Eng C Mater Biol Appl. 2019;102:670–82.PubMed
41.
go back to reference Paulo Zambon J, Atala A, Yoo JJ. Methods to generate tissue-derived constructs for regenerative medicine applications. Methods. 2019;171:3.PubMed Paulo Zambon J, Atala A, Yoo JJ. Methods to generate tissue-derived constructs for regenerative medicine applications. Methods. 2019;171:3.PubMed
42.
go back to reference Gilpin A, Yang Y. Decellularization strategies for regenerative medicine: from processing techniques to applications. Biomed Res Int. 2017;2017:9831534.PubMedPubMedCentral Gilpin A, Yang Y. Decellularization strategies for regenerative medicine: from processing techniques to applications. Biomed Res Int. 2017;2017:9831534.PubMedPubMedCentral
43.
go back to reference Rana D, Zreiqat H, Benkirane-Jessel N, Ramakrishna S, Ramalingam M. Development of decellularized scaffolds for stem cell-driven tissue engineering. J Tissue Eng Regen Med. 2017;11:942–65.PubMed Rana D, Zreiqat H, Benkirane-Jessel N, Ramakrishna S, Ramalingam M. Development of decellularized scaffolds for stem cell-driven tissue engineering. J Tissue Eng Regen Med. 2017;11:942–65.PubMed
44.
go back to reference Tiemann TT, Padma AM, Sehic E, Backdahl H, Oltean M, Song MJ, Brannstrom M, Hellstrom M. Towards uterus tissue engineering: a comparative study of sheep uterus decellularisation. Mol Hum Reprod. 2020. Tiemann TT, Padma AM, Sehic E, Backdahl H, Oltean M, Song MJ, Brannstrom M, Hellstrom M. Towards uterus tissue engineering: a comparative study of sheep uterus decellularisation. Mol Hum Reprod. 2020.
45.
go back to reference Mirzaeian L, Eivazkhani F, Hezavehei M, Moini A, Esfandiari F, Valojerdi MR, Fathi R. Optimizing the cell seeding protocol to human Decellularized ovarian scaffold: application of dynamic system for bio-engineering. Cell J. 2020;22:227–35.PubMed Mirzaeian L, Eivazkhani F, Hezavehei M, Moini A, Esfandiari F, Valojerdi MR, Fathi R. Optimizing the cell seeding protocol to human Decellularized ovarian scaffold: application of dynamic system for bio-engineering. Cell J. 2020;22:227–35.PubMed
46.
go back to reference 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;13:155.PubMedPubMedCentral 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;13:155.PubMedPubMedCentral
47.
go back to reference Dorin RP, Pohl HG, De Filippo RE, Yoo JJ, Atala A. Tubularized urethral replacement with unseeded matrices: what is the maximum distance for normal tissue regeneration? World J Urol. 2008;26:323–6.PubMed Dorin RP, Pohl HG, De Filippo RE, Yoo JJ, Atala A. Tubularized urethral replacement with unseeded matrices: what is the maximum distance for normal tissue regeneration? World J Urol. 2008;26:323–6.PubMed
Metadata
Title
Decellularization and recellularization of the ovary for bioengineering applications; studies in the mouse
Authors
Ahmed Baker Alshaikh
Arvind Manikantan Padma
Matilda Dehlin
Randa Akouri
Min Jong Song
Mats Brännström
Mats Hellström
Publication date
01-12-2020
Publisher
BioMed Central
Published in
Reproductive Biology and Endocrinology / Issue 1/2020
Electronic ISSN: 1477-7827
DOI
https://doi.org/10.1186/s12958-020-00630-y

Other articles of this Issue 1/2020

Reproductive Biology and Endocrinology 1/2020 Go to the issue