Skip to main content
Top
Published in: Reproductive Health 1/2021

Open Access 01-12-2021 | Infertility | Research

Successful transplantation of spermatogonial stem cells into the seminiferous tubules of busulfan-treated mice

Authors: Hossein Azizi, Amirreza Niazi Tabar, Thomas Skutella

Published in: Reproductive Health | Issue 1/2021

Login to get access

Abstract

Background

Spermatogonial stem cells (SSCs) in the testis are crucial for transferring genetic information to the next generation. Successful transplantation of SSCs to infertile men is an advanced therapeutic application in reproductive biology research.

Methods

In this experimental research, both in vitro and in vivo characterization of undifferentiated and differentiated SSCs were performed by morphology—immunocytochemistry (ICC), immunohistochemistry (IMH), Fluidigm Real-Time polymerase chain reaction (RT-PCR) and flow cytometry analysis. The isolated SSCs were finally microinjected into the rete testis of busulfan-treated mice.
The compact undifferentiated and more loosely connected round differentiated SSCs were isolated during testicular cell expansion from their specific feeder layer.

Results

ICC analysis indicated high and low expression levels of Zbtb16 in undifferentiated and differentiated germ cells. Also, IMH analysis showed different expression levels of Zbtb16 in the two different germ stem cell populations of the testicular tissue. While Fluidigm RT-PCR analysis indicated overexpression of the TAF4B germ cell gene, the expression of DAZL, VASA, and Zbtb16 were down-regulated during the differentiation of SSCs (P < 0.05). Also, flow cytometry analysis confirmed the significant downregulation of Itgb1 and Itga4 during differentiation. By transplantation of SSCs into busulfan-treated NOD/SCID mice, GFP-labeled sperm cells developed.

Conclusions

In the current study, we performed a transplantation technique that could be useful for the future microinjection of SSCs during infertility treatment and for studying in vivo differentiation of SSCs into sperm.
Literature
1.
go back to reference Kanamori M, et al. Mammalian germ cell migration during development, growth, and homeostasis. Reprod Med Biol. 2019;18(3):247–55.CrossRef Kanamori M, et al. Mammalian germ cell migration during development, growth, and homeostasis. Reprod Med Biol. 2019;18(3):247–55.CrossRef
2.
go back to reference Kubota H, Brinster RL. Spermatogonial stem cells. Biol Reprod. 2018;99(1):52–74.CrossRef Kubota H, Brinster RL. Spermatogonial stem cells. Biol Reprod. 2018;99(1):52–74.CrossRef
3.
go back to reference Lord T, Oatley JM. Functional assessment of spermatogonial stem cell purity in experimental cell populations. Stem cell research. 2018;29:129–33.CrossRef Lord T, Oatley JM. Functional assessment of spermatogonial stem cell purity in experimental cell populations. Stem cell research. 2018;29:129–33.CrossRef
4.
go back to reference Crisóstomo L, et al. Molecular mechanisms and signaling pathways involved in the nutritional support of spermatogenesis by Sertoli cells. In: Sertoli cells. Springer; 2018. p. 129–55.CrossRef Crisóstomo L, et al. Molecular mechanisms and signaling pathways involved in the nutritional support of spermatogenesis by Sertoli cells. In: Sertoli cells. Springer; 2018. p. 129–55.CrossRef
5.
go back to reference Hermo L, Lalli M, Clermont Y. Arrangement of connective tissue components in the walls of seminiferous tubules of man and monkey. Am J Anat. 1977;148(4):433–45.CrossRef Hermo L, Lalli M, Clermont Y. Arrangement of connective tissue components in the walls of seminiferous tubules of man and monkey. Am J Anat. 1977;148(4):433–45.CrossRef
6.
go back to reference Neto FT, et al. Spermatogenesis in humans and its affecting factors. Semin Cell Dev Biol. 2016;59:10–26.CrossRef Neto FT, et al. Spermatogenesis in humans and its affecting factors. Semin Cell Dev Biol. 2016;59:10–26.CrossRef
7.
go back to reference Schell C, et al. 15-Deoxy-delta 12–14-prostaglandin-J2 induces hypertrophy and loss of contractility in human testicular peritubular cells: implications for human male fertility. Endocrinology. 2010;151(3):1257–68.CrossRef Schell C, et al. 15-Deoxy-delta 12–14-prostaglandin-J2 induces hypertrophy and loss of contractility in human testicular peritubular cells: implications for human male fertility. Endocrinology. 2010;151(3):1257–68.CrossRef
8.
go back to reference Haider SG. Cell biology of Leydig cells in the testis. Int Rev Cytol. 2004;233:181–241.CrossRef Haider SG. Cell biology of Leydig cells in the testis. Int Rev Cytol. 2004;233:181–241.CrossRef
9.
go back to reference Payne AH, Hales DB. Overview of steroidogenic enzymes in the pathway from cholesterol to active steroid hormones. Endocr Rev. 2004;25(6):947–70.CrossRef Payne AH, Hales DB. Overview of steroidogenic enzymes in the pathway from cholesterol to active steroid hormones. Endocr Rev. 2004;25(6):947–70.CrossRef
10.
go back to reference Luca G, et al. Sertoli cells for cell transplantation: pre-clinical studies and future perspectives. Andrology. 2018;6(3):385–95.CrossRef Luca G, et al. Sertoli cells for cell transplantation: pre-clinical studies and future perspectives. Andrology. 2018;6(3):385–95.CrossRef
11.
go back to reference Su L, et al. Testin regulates the blood-testis barrier via disturbing occludin/ZO-1 association and actin organization. J Cell Physiol. 2020;235(9):6127–38.CrossRef Su L, et al. Testin regulates the blood-testis barrier via disturbing occludin/ZO-1 association and actin organization. J Cell Physiol. 2020;235(9):6127–38.CrossRef
12.
go back to reference Giudice MG, Vermeulen M, Wyns C. Blood testis barrier and somatic cells impairment in a series of 35 adult klinefelter syndrome patients. Int J Mol Sci. 2019;20:22.CrossRef Giudice MG, Vermeulen M, Wyns C. Blood testis barrier and somatic cells impairment in a series of 35 adult klinefelter syndrome patients. Int J Mol Sci. 2019;20:22.CrossRef
13.
go back to reference Qu N, et al. Immunological microenvironment in the testis. Reprod Med Biol. 2020;19(1):24–31.CrossRef Qu N, et al. Immunological microenvironment in the testis. Reprod Med Biol. 2020;19(1):24–31.CrossRef
14.
go back to reference Haghmorad D, et al. Improvement of fertility parameters with Tribulus Terrestris and Anacyclus Pyrethrum treatment in male rats. Int Braz J Urol. 2019;45(5):1043–54.CrossRef Haghmorad D, et al. Improvement of fertility parameters with Tribulus Terrestris and Anacyclus Pyrethrum treatment in male rats. Int Braz J Urol. 2019;45(5):1043–54.CrossRef
15.
go back to reference Dann CT, et al. Spermatogonial stem cell self-renewal requires OCT4, a factor downregulated during retinoic acid-induced differentiation. Stem Cells. 2008;26(11):2928–37.CrossRef Dann CT, et al. Spermatogonial stem cell self-renewal requires OCT4, a factor downregulated during retinoic acid-induced differentiation. Stem Cells. 2008;26(11):2928–37.CrossRef
16.
go back to reference Sharma M, Braun RE. Cyclical expression of GDNF is required for spermatogonial stem cell homeostasis. Development. 2018;145(5):dev151555.CrossRef Sharma M, Braun RE. Cyclical expression of GDNF is required for spermatogonial stem cell homeostasis. Development. 2018;145(5):dev151555.CrossRef
17.
go back to reference Tahara N, et al. Temporal changes of Sall4 lineage contribution in developing embryos and the contribution of Sall4-lineages to postnatal germ cells in mice. Sci Rep. 2018;8(1):16410.CrossRef Tahara N, et al. Temporal changes of Sall4 lineage contribution in developing embryos and the contribution of Sall4-lineages to postnatal germ cells in mice. Sci Rep. 2018;8(1):16410.CrossRef
18.
go back to reference Dong L-H, et al. emopenPostnatal germ cell development in cryptorchid boysemclose. Asian J Androl. 2019;22(3):258.CrossRef Dong L-H, et al. emopenPostnatal germ cell development in cryptorchid boysemclose. Asian J Androl. 2019;22(3):258.CrossRef
19.
go back to reference Garbuzov A, et al. Purification of GFRα1+ and GFRα1–spermatogonial stem cells reveals a niche-dependent mechanism for fate determination. Stem Cell Rep. 2018;10(2):553–67.CrossRef Garbuzov A, et al. Purification of GFRα1+ and GFRα1–spermatogonial stem cells reveals a niche-dependent mechanism for fate determination. Stem Cell Rep. 2018;10(2):553–67.CrossRef
20.
go back to reference David G, et al. Histone deacetylase associated with mSin3A mediates repression by the acute promyelocytic leukemia-associated PLZF protein. Oncogene. 1998;16(19):2549–56.CrossRef David G, et al. Histone deacetylase associated with mSin3A mediates repression by the acute promyelocytic leukemia-associated PLZF protein. Oncogene. 1998;16(19):2549–56.CrossRef
21.
go back to reference Clotaire DZJ, et al. Functions of promyelocytic leukaemia zinc finger (Plzf) in male germline stem cell development and differentiation. Reprod Fertil Dev. 2019;31(8):1315–20.CrossRef Clotaire DZJ, et al. Functions of promyelocytic leukaemia zinc finger (Plzf) in male germline stem cell development and differentiation. Reprod Fertil Dev. 2019;31(8):1315–20.CrossRef
22.
go back to reference Buaas FW, et al. Plzf is required in adult male germ cells for stem cell self-renewal. Nat Genet. 2004;36(6):647–52.CrossRef Buaas FW, et al. Plzf is required in adult male germ cells for stem cell self-renewal. Nat Genet. 2004;36(6):647–52.CrossRef
23.
go back to reference Li H, et al. DAZL is a master translational regulator of murine spermatogenesis. Natl Sci Rev. 2018;6(3):455–68.CrossRef Li H, et al. DAZL is a master translational regulator of murine spermatogenesis. Natl Sci Rev. 2018;6(3):455–68.CrossRef
24.
go back to reference Hwang, H., et al., Novel functions of the ubiquitin-independent proteasome system in regulating Xenopus germline development. Development, 2019. 146(8): dev172700. Hwang, H., et al., Novel functions of the ubiquitin-independent proteasome system in regulating Xenopus germline development. Development, 2019. 146(8): dev172700.
25.
go back to reference Nicholls PK, et al. Mammalian germ cells are determined after PGC colonization of the nascent gonad. Proc Natl Acad Sci. 2019;116(51):25677–87.CrossRef Nicholls PK, et al. Mammalian germ cells are determined after PGC colonization of the nascent gonad. Proc Natl Acad Sci. 2019;116(51):25677–87.CrossRef
26.
go back to reference Azizi H, et al. Investigation of VASA Gene and Protein Expression in Neonate and Adult Testicular Germ Cells in Mice In Vivo and In Vitro. Cell J. 2020;22(2):171–7.PubMed Azizi H, et al. Investigation of VASA Gene and Protein Expression in Neonate and Adult Testicular Germ Cells in Mice In Vivo and In Vitro. Cell J. 2020;22(2):171–7.PubMed
27.
go back to reference Gustafson EA, et al. ZFP628 is a TAF4b-interacting transcription factor required for mouse spermiogenesis. Mol Cell Biol. 2020;40(7):e00228-19.CrossRef Gustafson EA, et al. ZFP628 is a TAF4b-interacting transcription factor required for mouse spermiogenesis. Mol Cell Biol. 2020;40(7):e00228-19.CrossRef
28.
go back to reference Yang D, et al. A novel role of Krüppel-like factor 4 in Zhikong scallop Chlamys farreri during spermatogenesis. PLoS ONE. 2017;12:6. Yang D, et al. A novel role of Krüppel-like factor 4 in Zhikong scallop Chlamys farreri during spermatogenesis. PLoS ONE. 2017;12:6.
29.
go back to reference Takahashi K, Yamanaka S. Induction of pluripotent stem cells from mouse embryonic and adult fibroblast cultures by defined factors. cell. 2006;126(4):663–76.CrossRef Takahashi K, Yamanaka S. Induction of pluripotent stem cells from mouse embryonic and adult fibroblast cultures by defined factors. cell. 2006;126(4):663–76.CrossRef
30.
go back to reference Kim YH, et al. GDNF family receptor alpha 1 is a reliable marker of undifferentiated germ cells in bulls. Theriogenology. 2019;132:172–81.CrossRef Kim YH, et al. GDNF family receptor alpha 1 is a reliable marker of undifferentiated germ cells in bulls. Theriogenology. 2019;132:172–81.CrossRef
31.
go back to reference Valdivia M, et al. Spermatogonial stem cells identified by molecular expression of PLZF, integrin beta1 and reactivity to Dolichos biflorus agglutinin in alpaca adult testes. Andrologia. 2019;51(6):e13283.CrossRef Valdivia M, et al. Spermatogonial stem cells identified by molecular expression of PLZF, integrin beta1 and reactivity to Dolichos biflorus agglutinin in alpaca adult testes. Andrologia. 2019;51(6):e13283.CrossRef
32.
go back to reference Kadam P, et al. Co-transplantation of mesenchymal stem cells improves spermatogonial stem cell transplantation efficiency in mice. Stem Cell Res Ther. 2018;9(1):317.CrossRef Kadam P, et al. Co-transplantation of mesenchymal stem cells improves spermatogonial stem cell transplantation efficiency in mice. Stem Cell Res Ther. 2018;9(1):317.CrossRef
33.
go back to reference Takashima S, Shinohara T. Culture and transplantation of spermatogonial stem cells. Stem Cell Res. 2018;29:46–55.CrossRef Takashima S, Shinohara T. Culture and transplantation of spermatogonial stem cells. Stem Cell Res. 2018;29:46–55.CrossRef
34.
go back to reference Sinha N, Whelan EC, Brinster RL. Isolation, cryopreservation, and transplantation of spermatogonial stem cells. Methods Mol Biol. 2019;2005:205–20.CrossRef Sinha N, Whelan EC, Brinster RL. Isolation, cryopreservation, and transplantation of spermatogonial stem cells. Methods Mol Biol. 2019;2005:205–20.CrossRef
Metadata
Title
Successful transplantation of spermatogonial stem cells into the seminiferous tubules of busulfan-treated mice
Authors
Hossein Azizi
Amirreza Niazi Tabar
Thomas Skutella
Publication date
01-12-2021
Publisher
BioMed Central
Published in
Reproductive Health / Issue 1/2021
Electronic ISSN: 1742-4755
DOI
https://doi.org/10.1186/s12978-021-01242-4

Other articles of this Issue 1/2021

Reproductive Health 1/2021 Go to the issue