Skip to main content
Top
Published in: Reproductive Biology and Endocrinology 1/2018

Open Access 01-12-2018 | Research

Expression of transcriptional factor EB (TFEB) in differentiating spermatogonia potentially promotes cell migration in mouse seminiferous epithelium

Authors: Yue Liu, Yanqin Hu, Li Wang, Chen Xu

Published in: Reproductive Biology and Endocrinology | Issue 1/2018

Login to get access

Abstract

Background

Spermatogenesis is a complex process involving the self-renewal and differentiation of spermatogonia into mature spermatids in the seminiferous tubules. During spermatogenesis, germ cells migrate from the basement membrane to cross the blood-testis barrier (BTB) and finally reach the luminal side of the seminiferous epithelium. However, the mechanism for regulating the migration of germ cells remains unclear. In this study, we focused on the expression and function of transcriptional factor EB (TFEB), a master regulator of lysosomal biogenesis, autophagy and endocytosis, in spermatogenesis.

Methods

The expression pattern of the TFEB in mouse testes were investigated by Western blotting and immunohistochemistry analyses. Either undifferentiated spermatogonia or differentiating spermatogonia were isolated from testes using magnetic-activated cell sorting based on specific cell surface markers. Differentiation of spermatogonia was induced with 100 nM retinoic acid (RA). shRNA was used to knock down TFEB in cells. TFEB expression was detected by immunofluorescence, qRT-PCR, and Western blotting. Cell migration was determined by both transwell migration assay and wound healing assay applied to a cell line of immortalized spermatogonia, GC-1 cells.

Results

During testicular development, TFEB expression was rapidly increased in the testes at the period of 7 days post-partum (dpp) to 14 dpp, whereas in adult testis, it was predominantly localized in the nucleus of spermatogonia at stages VI to VIII of the seminiferous epithelial cycle. Accordingly, TFEB was observed to be mainly expressed in differentiating spermatogonia and was activated for nuclear translocation by RA treatment. Moreover, knockdown of TFEB expression by RNAi did not affect spermatogonial differentiation, but significantly reduced cell migration in GC-1 cells.

Conclusion

These findings imply that regionally distinct expression and activation of TFEB was strongly associated with RA signaling, and therefore may promote cell migration across the BTB and transport along the seminiferous epithelium.
Appendix
Available only for authorised users
Literature
1.
2.
go back to reference Mecklenburg JM, Hermann BP. Mechanisms Regulating Spermatogonial Differentiation. In: Piprek R. (eds) Molecular Mechanisms of Cell Differentiation in Gonad Development. Results and Problems in Cell Differentiation. Springer, Cham; 2016. p253-287. Mecklenburg JM, Hermann BP. Mechanisms Regulating Spermatogonial Differentiation. In: Piprek R. (eds) Molecular Mechanisms of Cell Differentiation in Gonad Development. Results and Problems in Cell Differentiation. Springer, Cham; 2016. p253-287.
3.
go back to reference Song HW, Wilkinson MF. Transcriptional control of spermatogonial maintenance and differentiation. Semin Cell Dev Biol. 2014;30:14–26.CrossRef Song HW, Wilkinson MF. Transcriptional control of spermatogonial maintenance and differentiation. Semin Cell Dev Biol. 2014;30:14–26.CrossRef
4.
go back to reference Li L, Gao Y, Chen H, Jesus T, Tang E, Li N, Lian Q, Ge RS, Cheng CY. Cell polarity, cell adhesion, and spermatogenesis: role of cytoskeletons. F1000Res. 2017;6:1565.CrossRef Li L, Gao Y, Chen H, Jesus T, Tang E, Li N, Lian Q, Ge RS, Cheng CY. Cell polarity, cell adhesion, and spermatogenesis: role of cytoskeletons. F1000Res. 2017;6:1565.CrossRef
5.
go back to reference Xiao X, Mruk DD, Wong CKC, Cheng CY. Germ cell transport across the seminiferous epithelium during spermatogenesis. Physiology. 2014;29:286–98.CrossRef Xiao X, Mruk DD, Wong CKC, Cheng CY. Germ cell transport across the seminiferous epithelium during spermatogenesis. Physiology. 2014;29:286–98.CrossRef
6.
go back to reference Busada JT, Geyer CB. The role of retinoic acid (RA) in Spermatogonial differentiation. Biol Reprod. 2016;94:10.CrossRef Busada JT, Geyer CB. The role of retinoic acid (RA) in Spermatogonial differentiation. Biol Reprod. 2016;94:10.CrossRef
7.
go back to reference Griswold MD. Spermatogenesis: the commitment to meiosis. Physiol Rev. 2016;96:1–17.CrossRef Griswold MD. Spermatogenesis: the commitment to meiosis. Physiol Rev. 2016;96:1–17.CrossRef
8.
go back to reference Mruk DD, Cheng CY. Sertoli-Sertoli and Sertoli-germ cell interactions and their significance in germ cell movement in the seminiferous epithelium during spermatogenesis. Endocr Rev. 2004;25:747–806.CrossRef Mruk DD, Cheng CY. Sertoli-Sertoli and Sertoli-germ cell interactions and their significance in germ cell movement in the seminiferous epithelium during spermatogenesis. Endocr Rev. 2004;25:747–806.CrossRef
9.
go back to reference Wen Q, Tang EI, Xiao X, Gao Y, Chu DS, Mruk DD, Silvestrini B, Cheng CY. Transport of germ cells across the seminiferous epithelium during spermatogenesis-the involvement of both actin- and microtubule-based cytoskeletons. Tissue Barriers. 2016;4:e1265042.CrossRef Wen Q, Tang EI, Xiao X, Gao Y, Chu DS, Mruk DD, Silvestrini B, Cheng CY. Transport of germ cells across the seminiferous epithelium during spermatogenesis-the involvement of both actin- and microtubule-based cytoskeletons. Tissue Barriers. 2016;4:e1265042.CrossRef
10.
go back to reference Coly PM, Gandolfo P, Castel H, Morin F. The autophagy machinery: a new player in chemotactic cell migration. Front Neurosci. 2017;11:78.CrossRef Coly PM, Gandolfo P, Castel H, Morin F. The autophagy machinery: a new player in chemotactic cell migration. Front Neurosci. 2017;11:78.CrossRef
11.
go back to reference Kawano S, Torisu T, Esaki M, Torisu K, Matsuno Y, Kitazono T. Autophagy promotes degradation of internalized collagen and regulates distribution of focal adhesions to suppress cell adhesion. Biol Open. 2017;6:1644–53.CrossRef Kawano S, Torisu T, Esaki M, Torisu K, Matsuno Y, Kitazono T. Autophagy promotes degradation of internalized collagen and regulates distribution of focal adhesions to suppress cell adhesion. Biol Open. 2017;6:1644–53.CrossRef
12.
go back to reference Kenific CM, Wittmann T, Debnath J. Autophagy in adhesion and migration. J Cell Sci. 2016;129:3685–93.CrossRef Kenific CM, Wittmann T, Debnath J. Autophagy in adhesion and migration. J Cell Sci. 2016;129:3685–93.CrossRef
13.
go back to reference Maritzen T, Schachtner H, Legler DF. On the move: endocytic trafficking in cell migration. Cell Mol Life Sci. 2015;72:2119–34.CrossRef Maritzen T, Schachtner H, Legler DF. On the move: endocytic trafficking in cell migration. Cell Mol Life Sci. 2015;72:2119–34.CrossRef
14.
go back to reference Paul NR, Jacquemet G, Caswell PT. Endocytic trafficking of Integrins in cell migration. Curr Biol. 2015;25:1092–105.CrossRef Paul NR, Jacquemet G, Caswell PT. Endocytic trafficking of Integrins in cell migration. Curr Biol. 2015;25:1092–105.CrossRef
15.
go back to reference Schmid SL. Reciprocal regulation of signaling and endocytosis: implications for the evolving cancer cell. J Cell Biol. 2017;216:2623–32.PubMedPubMedCentral Schmid SL. Reciprocal regulation of signaling and endocytosis: implications for the evolving cancer cell. J Cell Biol. 2017;216:2623–32.PubMedPubMedCentral
16.
go back to reference Pastore N, Brady OA, Diab HI, Martina JA, Sun L, Huynh T, Lim JA, Zare H, Raben N, Ballabio A, Puertollano R. TFEB and TFE3 cooperate in the regulation of the innate immune response in activated macrophages. Autophagy. 2016;12:1240–58.CrossRef Pastore N, Brady OA, Diab HI, Martina JA, Sun L, Huynh T, Lim JA, Zare H, Raben N, Ballabio A, Puertollano R. TFEB and TFE3 cooperate in the regulation of the innate immune response in activated macrophages. Autophagy. 2016;12:1240–58.CrossRef
17.
go back to reference Sardiello M, Palmieri M, di Ronza A, Medina DL, Valenza M, Gennarino VA, Di Malta C, Donaudy F, Embrione V, Polishchuk RS, Banfi S, Parenti G, Cattaneo E, Ballabio A. A gene network regulating lysosomal biogenesis and function. Science. 2009;325:473–7.CrossRef Sardiello M, Palmieri M, di Ronza A, Medina DL, Valenza M, Gennarino VA, Di Malta C, Donaudy F, Embrione V, Polishchuk RS, Banfi S, Parenti G, Cattaneo E, Ballabio A. A gene network regulating lysosomal biogenesis and function. Science. 2009;325:473–7.CrossRef
18.
go back to reference Settembre C, Di Malta C, Polito VA, Garcia Arencibia M, Vetrini F, Erdin S, Erdin SU, Huynh T, Medina D, Colella P, et al. TFEB links autophagy to lysosomal biogenesis. Science. 2011;332:1429–33.CrossRef Settembre C, Di Malta C, Polito VA, Garcia Arencibia M, Vetrini F, Erdin S, Erdin SU, Huynh T, Medina D, Colella P, et al. TFEB links autophagy to lysosomal biogenesis. Science. 2011;332:1429–33.CrossRef
19.
go back to reference Napolitano G, Ballabio A. TFEB at a glance. J Cell Sci. 2016;129:2475–81.CrossRef Napolitano G, Ballabio A. TFEB at a glance. J Cell Sci. 2016;129:2475–81.CrossRef
20.
go back to reference Peña-Llopis S, Brugarolas J. TFEB, a novel mTORC1 effector implicated in lysosome biogenesis, endocytosis and autophagy. Cell Cycle. 2011;10:3987–8.CrossRef Peña-Llopis S, Brugarolas J. TFEB, a novel mTORC1 effector implicated in lysosome biogenesis, endocytosis and autophagy. Cell Cycle. 2011;10:3987–8.CrossRef
21.
go back to reference Song W, Wang F, Savini M, Ake A, di Ronza A, Sardiello M, Segatori L. TFEB regulates lysosomal proteostasis. Hum Mol Genet. 2013;22:1994–2009.CrossRef Song W, Wang F, Savini M, Ake A, di Ronza A, Sardiello M, Segatori L. TFEB regulates lysosomal proteostasis. Hum Mol Genet. 2013;22:1994–2009.CrossRef
22.
go back to reference Martina JA, Chen Y, Gucek M, Puertollano R. MTORC1 functions as a transcriptional regulator of autophagy by preventing nuclear transport of TFEB. Autophagy. 2012;8:903–14.CrossRef Martina JA, Chen Y, Gucek M, Puertollano R. MTORC1 functions as a transcriptional regulator of autophagy by preventing nuclear transport of TFEB. Autophagy. 2012;8:903–14.CrossRef
23.
go back to reference Medina DL, Di Paola S, Peluso I, Armani A, De Stefani D, Venditti R, Montefusco S, Scotto-Rosato A, Prezioso C, Forrester A, et al. Lysosomal calcium signalling regulates autophagy through calcineurin and TFEB. Nat Cell Biol. 2015;17:288–99.CrossRef Medina DL, Di Paola S, Peluso I, Armani A, De Stefani D, Venditti R, Montefusco S, Scotto-Rosato A, Prezioso C, Forrester A, et al. Lysosomal calcium signalling regulates autophagy through calcineurin and ​TFEB. Nat Cell Biol. 2015;17:288–99.CrossRef
24.
go back to reference Puertollano R, Ferguson SM, Brugarolas J, Ballabio A. The complex relationship between TFEB transcription factor phosphorylation and subcellular localization. EMBO J. 2018;37:e98804.CrossRef Puertollano R, Ferguson SM, Brugarolas J, Ballabio A. The complex relationship between TFEB transcription factor phosphorylation and subcellular localization. EMBO J. 2018;37:e98804.CrossRef
25.
go back to reference Settembre C, De Cegli R, Mansueto G, Saha PK, Vetrini F, Visvikis O, Huynh T, Carissimo A, Palmer D, Klisch TJ, et al. TFEB controls cellular lipid metabolism through a starvation-induced autoregulatory loop. Nat Cell Biol. 2013;15:647–58.CrossRef Settembre C, De Cegli R, Mansueto G, Saha PK, Vetrini F, Visvikis O, Huynh T, Carissimo A, Palmer D, Klisch TJ, et al. TFEB controls cellular lipid metabolism through a starvation-induced autoregulatory loop. Nat Cell Biol. 2013;15:647–58.CrossRef
26.
go back to reference Settembre C, Ballabio A. Lysosome: regulator of lipid degradation pathways. Trends Cell Biol. 2014;24:743–50.CrossRef Settembre C, Ballabio A. Lysosome: regulator of lipid degradation pathways. Trends Cell Biol. 2014;24:743–50.CrossRef
27.
go back to reference Tong Y, Song F. Intracellular calcium signaling regulates autophagy via calcineurin-mediated TFEB dephosphorylation. Autophagy. 2015;11:1192–5.CrossRef Tong Y, Song F. Intracellular calcium signaling regulates autophagy via calcineurin-mediated TFEB dephosphorylation. Autophagy. 2015;11:1192–5.CrossRef
28.
go back to reference Giatromanolaki A, Kalamida D, Sivridis E, Karagounis IV, Gatter KC, Harris AL, Koukourakis MI. Increased expression of transcription factor EB (TFEB) is associated with autophagy, migratory phenotype and poor prognosis in non-small cell lung cancer. Lung Cancer. 2015;90:98–105.CrossRef Giatromanolaki A, Kalamida D, Sivridis E, Karagounis IV, Gatter KC, Harris AL, Koukourakis MI. Increased expression of transcription factor EB (TFEB) is associated with autophagy, migratory phenotype and poor prognosis in non-small cell lung cancer. Lung Cancer. 2015;90:98–105.CrossRef
29.
go back to reference Sakamoto H, Yamashita K, Okamoto K, Kadowaki T, Sakai E, Umeda M, Tsukuba T. Transcription factor EB influences invasion and migration in oral squamous cell carcinomas. Oral Dis. 2018;24:741–8.CrossRef Sakamoto H, Yamashita K, Okamoto K, Kadowaki T, Sakai E, Umeda M, Tsukuba T. Transcription factor EB influences invasion and migration in oral squamous cell carcinomas. Oral Dis. 2018;24:741–8.CrossRef
30.
go back to reference He Z, Kokkinaki M, Jiang J, Dobrinski I, Dym M. Isolation, characterization, and culture of human spermatogonia. Biol Reprod. 2010;82:363–72.CrossRef He Z, Kokkinaki M, Jiang J, Dobrinski I, Dym M. Isolation, characterization, and culture of human spermatogonia. Biol Reprod. 2010;82:363–72.CrossRef
31.
go back to reference Liu Y, Chen G, Lu L, Sun H, Guo Q, Xue K, Fan Y, Ding Z. RNASET2 in human spermatozoa and seminal plasma: a novel relevant indicator for asthenozoospermia. Andrology. 2013;1:75–84.CrossRef Liu Y, Chen G, Lu L, Sun H, Guo Q, Xue K, Fan Y, Ding Z. RNASET2 in human spermatozoa and seminal plasma: a novel relevant indicator for asthenozoospermia. Andrology. 2013;1:75–84.CrossRef
32.
go back to reference Holembowski L, Kramer D, Riedel D, Sordella R, Nemajerova A, Dobbelstein M, Moll UM. TAp73 is essential for germ cell adhesion and maturation in testis. J Cell Biol. 2014;204:1173–90.CrossRef Holembowski L, Kramer D, Riedel D, Sordella R, Nemajerova A, Dobbelstein M, Moll UM. TAp73 is essential for germ cell adhesion and maturation in testis. J Cell Biol. 2014;204:1173–90.CrossRef
33.
go back to reference Sato Y, Yoshida K, Nozawa S, Yoshiike M, Arai M, Otoi T, Iwamoto T. Establishment of adult mouse Sertoli cell lines by using the starvation method. Reproduction. 2013;145:505–16.CrossRef Sato Y, Yoshida K, Nozawa S, Yoshiike M, Arai M, Otoi T, Iwamoto T. Establishment of adult mouse Sertoli cell lines by using the starvation method. Reproduction. 2013;145:505–16.CrossRef
34.
go back to reference Huszar JM, Payne CJ. MicroRNA 146 (Mir146) modulates spermatogonial differentiation by retinoic acid in mice. Biol Reprod. 2013;88:15.CrossRef Huszar JM, Payne CJ. MicroRNA 146 (Mir146) modulates spermatogonial differentiation by retinoic acid in mice. Biol Reprod. 2013;88:15.CrossRef
35.
go back to reference Choi NY, Park YS, Ryu JS, Lee HJ, Araúzo-Bravo MJ, Ko K, Han DW, Schöler HR, Ko K. A novel feeder-free culture system for expansion of mouse spermatogonial stem cells. Mol Cells. 2014;37:473–9.CrossRef Choi NY, Park YS, Ryu JS, Lee HJ, Araúzo-Bravo MJ, Ko K, Han DW, Schöler HR, Ko K. A novel feeder-free culture system for expansion of mouse spermatogonial stem cells. Mol Cells. 2014;37:473–9.CrossRef
36.
go back to reference Chen H, Fok KL, Yu S, Jiang J, Chen Z, Gui Y, Cai Z, Chan HC. CD147 is required for matrix metalloproteinases-2 production and germ cell migration during spermatogenesis. Mol Hum Reprod. 2011;17:405–14.CrossRef Chen H, Fok KL, Yu S, Jiang J, Chen Z, Gui Y, Cai Z, Chan HC. CD147 is required for matrix metalloproteinases-2 production and germ cell migration during spermatogenesis. Mol Hum Reprod. 2011;17:405–14.CrossRef
37.
go back to reference Boya P, Codogno P, Rodriguez-Muela N. Autophagy in stem cells: repair, remodelling and metabolic reprogramming. Development. 2018;145:146506.CrossRef Boya P, Codogno P, Rodriguez-Muela N. Autophagy in stem cells: repair, remodelling and metabolic reprogramming. Development. 2018;145:146506.CrossRef
38.
go back to reference Brunt L, Scholpp S. The function of endocytosis in Wnt signaling. Cell Mol Life Sci. 2018;75:785–95.CrossRef Brunt L, Scholpp S. The function of endocytosis in Wnt signaling. Cell Mol Life Sci. 2018;75:785–95.CrossRef
39.
go back to reference Chen X, He Y, Lu F. Autophagy in stem cell biology: a perspective on stem cell self-renewal and differentiation. Stem Cells Int. 2018;2018:9131397.PubMedPubMedCentral Chen X, He Y, Lu F. Autophagy in stem cell biology: a perspective on stem cell self-renewal and differentiation. Stem Cells Int. 2018;2018:9131397.PubMedPubMedCentral
40.
go back to reference Busada JT, Niedenberger BA, Velte EK, Keiper BD, Geyer CB. Mammalian target of rapamycin complex 1 (mTORC1) is required for mouse spermatogonial differentiation in vivo. Dev Biol. 2015;407:90–102.CrossRef Busada JT, Niedenberger BA, Velte EK, Keiper BD, Geyer CB. Mammalian target of rapamycin complex 1 (mTORC1) is required for mouse spermatogonial differentiation in vivo. Dev Biol. 2015;407:90–102.CrossRef
41.
go back to reference Hobbs RM, Seandel M, Falciatori I, Rafii S, Pandolfi PP. Plzf regulates germline progenitor self-renewal by opposing mTORC1. Cell. 2010;142:468–79.CrossRef Hobbs RM, Seandel M, Falciatori I, Rafii S, Pandolfi PP. Plzf regulates germline progenitor self-renewal by opposing mTORC1. Cell. 2010;142:468–79.CrossRef
42.
go back to reference Endo T, Freinkman E, de Rooij DG, Page DC. Periodic production of retinoic acid by meiotic and somatic cells coordinates four transitions in mouse spermatogenesis. Proc Natl Acad Sci U S A. 2017;114:10132–10141.CrossRef Endo T, Freinkman E, de Rooij DG, Page DC. Periodic production of retinoic acid by meiotic and somatic cells coordinates four transitions in mouse spermatogenesis. Proc Natl Acad Sci U S A. 2017;114:10132–10141.CrossRef
43.
go back to reference Chen SR, Liu YX. Regulation of spermatogonial stem cell self-renewal and spermatocyte meiosis by Sertoli cell signaling. Reproduction. 2015;149:159–67.CrossRef Chen SR, Liu YX. Regulation of spermatogonial stem cell self-renewal and spermatocyte meiosis by Sertoli cell signaling. Reproduction. 2015;149:159–67.CrossRef
44.
go back to reference Manku G, Culty M. Mammalian gonocyte and spermatogonia differentiation: recent advances and remaining challenges. Reproduction. 2015;149:139–57.CrossRef Manku G, Culty M. Mammalian gonocyte and spermatogonia differentiation: recent advances and remaining challenges. Reproduction. 2015;149:139–57.CrossRef
45.
go back to reference Li C, Chen S, Li H, Chen L, Zhao Y, Jiang Y, Liu Z, Liu Y, Gao S, Wang F, et al. MicroRNA-16 modulates melatonin-induced cell growth in the mouse-derived Spermatogonia cell line GC-1 spg cells by targeting Ccnd1. Biol Reprod. 2016;95:57.CrossRef Li C, Chen S, Li H, Chen L, Zhao Y, Jiang Y, Liu Z, Liu Y, Gao S, Wang F, et al. MicroRNA-16 modulates melatonin-induced cell growth in the mouse-derived Spermatogonia cell line GC-1 spg cells by targeting Ccnd1. Biol Reprod. 2016;95:57.CrossRef
46.
go back to reference Santillo A, Falvo S, Chieffi P, Di Fiore MM, Senese R, Chieffi Baccari G. D-aspartate induces proliferative pathways in Spermatogonial GC-1 cells. J Cell Physiol. 2016;231:490–5.CrossRef Santillo A, Falvo S, Chieffi P, Di Fiore MM, Senese R, Chieffi Baccari G. D-aspartate induces proliferative pathways in Spermatogonial GC-1 cells. J Cell Physiol. 2016;231:490–5.CrossRef
47.
go back to reference Zhao L, Zhu Z, Yao C, Huang Y, Zhi E, Chen H, Tian R, Li P, Yuan Q, Xue Y, et al. VEGFC/VEGFR3 signaling regulates mouse Spermatogonial cell proliferation via the activation of AKT/MAPK and cyclin D1 pathway and mediates the apoptosis by affecting caspase 3/9 and Bcl-2. Cell Cycle. 2018;17:225–39.CrossRef Zhao L, Zhu Z, Yao C, Huang Y, Zhi E, Chen H, Tian R, Li P, Yuan Q, Xue Y, et al. VEGFC/VEGFR3 signaling regulates mouse Spermatogonial cell proliferation via the activation of AKT/MAPK and cyclin D1 pathway and mediates the apoptosis by affecting caspase 3/9 and Bcl-2. Cell Cycle. 2018;17:225–39.CrossRef
48.
go back to reference Zhang J, Wong CH, Xia W, Mruk DD, Lee NP, Lee WM, Cheng CY. Regulation of Sertoli-germ cell adherens junction dynamics via changes in protein-protein interactions of the N-cadherin-beta-catenin protein complex which are possibly mediated by c-Src and myotubularin-related protein 2: an in vivo study using an androgen suppression model. Endocrinology. 2005;146:1268–84.CrossRef Zhang J, Wong CH, Xia W, Mruk DD, Lee NP, Lee WM, Cheng CY. Regulation of Sertoli-germ cell adherens junction dynamics via changes in protein-protein interactions of the N-cadherin-beta-catenin protein complex which are possibly mediated by c-Src and myotubularin-related protein 2: an in vivo study using an androgen suppression model. Endocrinology. 2005;146:1268–84.CrossRef
49.
go back to reference Hannigan MM, Zagore LL, Licatalosi DD. Ptbp2 Controls an Alternative Splicing Network Required for Cell Communication during Spermatogenesis. Cell Rep. 2017;19:2598–2612.CrossRef Hannigan MM, Zagore LL, Licatalosi DD. Ptbp2 Controls an Alternative Splicing Network Required for Cell Communication during Spermatogenesis. Cell Rep. 2017;19:2598–2612.CrossRef
50.
go back to reference Holembowski L, Kramer D, Riedel D, Sordella R, Nemajerova A, Dobbelstein M, Moll UM. TAp73 is essential for germ cell adhesion and maturation in testis. J Cell Biol. 2014;204:1173–1190.CrossRef Holembowski L, Kramer D, Riedel D, Sordella R, Nemajerova A, Dobbelstein M, Moll UM. TAp73 is essential for germ cell adhesion and maturation in testis. J Cell Biol. 2014;204:1173–1190.CrossRef
51.
go back to reference Takashima S, Kanatsu-Shinohara M, Tanaka T, Takehashi M, Morimoto H, Shinohara T. Rac mediates mouse spermatogonial stem cell homing to germline niches by regulating transmigration through the blood-testis barrier. Cell Stem Cell. 2011;9:463–75.CrossRef Takashima S, Kanatsu-Shinohara M, Tanaka T, Takehashi M, Morimoto H, Shinohara T. Rac mediates mouse spermatogonial stem cell homing to germline niches by regulating transmigration through the blood-testis barrier. Cell Stem Cell. 2011;9:463–75.CrossRef
52.
go back to reference Song HW, Bettegowda A, Lake BB, Zhao AH, Skarbrevik D, Babajanian E, Sukhwani M, Shum EY, Phan MH, Plank TM, et al. The Homeobox transcription factor RHOX10 drives mouse Spermatogonial stem cell establishment. Cell Rep. 2016;17:149–64.CrossRef Song HW, Bettegowda A, Lake BB, Zhao AH, Skarbrevik D, Babajanian E, Sukhwani M, Shum EY, Phan MH, Plank TM, et al. The Homeobox transcription factor RHOX10 drives mouse Spermatogonial stem cell establishment. Cell Rep. 2016;17:149–64.CrossRef
Metadata
Title
Expression of transcriptional factor EB (TFEB) in differentiating spermatogonia potentially promotes cell migration in mouse seminiferous epithelium
Authors
Yue Liu
Yanqin Hu
Li Wang
Chen Xu
Publication date
01-12-2018
Publisher
BioMed Central
Published in
Reproductive Biology and Endocrinology / Issue 1/2018
Electronic ISSN: 1477-7827
DOI
https://doi.org/10.1186/s12958-018-0427-x

Other articles of this Issue 1/2018

Reproductive Biology and Endocrinology 1/2018 Go to the issue