Aberrant gene expression by Sertoli cells in infertile men with Sertoli cell-only syndrome

Darius A Paduch, Stephanie Hilz, Andrew Grimson, Peter N Schlegel, Anne E Jedlicka, William W Wright, Darius A Paduch, Stephanie Hilz, Andrew Grimson, Peter N Schlegel, Anne E Jedlicka, William W Wright

Abstract

Sertoli cell-only (SCO) syndrome is a severe form of human male infertility seemingly characterized by the lack all spermatogenic cells. However, tubules of some SCO testes contain small patches of active spermatogenesis and thus spermatogonial stem cells. We hypothesized that these stem cells cannot replicate and seed spermatogenesis in barren areas of tubule because as-of-yet unrecognized deficits in Sertoli cell gene expression disable most stem cell niches. Performing the first thorough comparison of the transcriptomes of human testes exhibiting complete spermatogenesis with the transcriptomes of testes with SCO syndrome, we defined transcripts that are both predominantly expressed by Sertoli cells and expressed at aberrant levels in SCO testes. Some of these transcripts encode proteins required for the proper assembly of adherent and gap junctions at sites of contact with other cells, including spermatogonial stem cells (SSCs). Other transcripts encode GDNF, FGF8 and BMP4, known regulators of mouse SSCs. Thus, most SCO Sertoli cells can neither organize junctions at normal sites of cell-cell contact nor stimulate SSCs with adequate levels of growth factors. We propose that the critical deficits in Sertoli cell gene expression we have identified contribute to the inability of spermatogonial stem cells within small patches of spermatogenesis in some SCO testes to seed spermatogenesis to adjacent areas of tubule that are barren of spermatogenesis. Furthermore, we predict that one or more of these deficits in gene expression are primary causes of human SCO syndrome.

Conflict of interest statement

DAP is affiliated with 'Consulting Research Services, Inc.' This service was founded and funded by DAP. Funds came from his lecture fees. DAP does not receive any financial benefit from this service and the service has no employees. Thus, this service does not present a conflict of interest with the research presented in this manuscript. The authors declare that there are no other conflicts of interest. This does not alter our adherence to PLOS ONE policies on sharing data and materials.

Figures

Fig 1. Comparison of the transcriptomes of…
Fig 1. Comparison of the transcriptomes of testes with complete spermatogenesis with the transcriptomes of testes with SCO syndrome.
(A) K means cluster analysis comparing the transcriptomes of testes with complete spermatogenesis with testes with SCO syndrome. Genes were clustered under 5 Gene Ontology terms; one or two representative terms per cluster are listed here. (B) The abundance in of a few well-characterized transcripts considered markers of three testicular somatic cells: Sertoli cells (VIM), peritubular myoid cells (ACTA2), and Leydig cells (INHA). Also shown are transcripts encoding receptors for three hormones required for normal testis function: FSHR, AR, and LHCGR. (C) Abundance of transcripts expressed by human SSCs (GFRA1, RET, ZBTB16, UCHL1, ENO2), by spermatocytes (SYCP2, SYCP3) and by round spermatids (PRM1, PRM2). Data in panels B and C are presented as CPM values in the total testis transcriptome divided by the CPM value for beta actin in the same sample. Asterisks over a pair of bars identify significant differences between testes with complete spermatogenesis and SCO testes (p

Fig 2. The expression of Sertoli cell…

Fig 2. The expression of Sertoli cell signature transcripts in testes with complete spermatogenesis and…

Fig 2. The expression of Sertoli cell signature transcripts in testes with complete spermatogenesis and in testes with SCO syndrome.
(A) PCA analysis of the expression of Sertoli cell signature transcripts in individual testes. Data were obtained from 4 testes with complete spermatogenesis (C1-4, green symbols) and seven SCO testes (SCO5, SCO7-12, red symbols) (B). A comparison of the mean level of expression of each Sertoli cell signature transcript in testes with complete spermatogenesis and in testes with SCO syndrome. Transcripts that were expressed in SCO testes at significantly higher levels (FDR<0.05), the same level or significantly lower levels are identified by blue, white and black dots, respectively.

Fig 3. Expression of Sertoli cell signature…

Fig 3. Expression of Sertoli cell signature transcripts encoding proteins that form adhesive or gap…

Fig 3. Expression of Sertoli cell signature transcripts encoding proteins that form adhesive or gap junctions or that organize and polarize the Sertoli cell plasma membrane.
(A) CLDN11 and JAM3 are tight junction adhesive proteins. Nectin2 and CDH2 are adhesive proteins of ectoplasmic specializations. GJB2 encodes the gap junction protein, Connexin 26. (B) Transcripts that encode proteins that organize or polarize the plasma membrane. CRB2 is a cell polarity component protein that defines the site of separation of the apical and basal-lateral plasma membrane domains. TJP3 is an adaptor protein that links the adhesive proteins of a tight junction to the actin cytoskeleton. SPTB, SPTPN4 and SPTPN5 are scaffolding proteins that concentrate beneath the plasma membrane, seed formation of membrane microdomains, and link those domains to the actin cytoskeleton. All data in this figure are expressed as mean + SEM of CPMs. The presence of an asterisk over a pair of bars indicates a significant difference between testes with complete spermatogenesis and testes with SCO syndrome (FDR≤0.05).

Fig 4. Expression of Sertoli cell signature…

Fig 4. Expression of Sertoli cell signature transcripts encoding regulators of vesicular trafficking.

(A) Expression…

Fig 4. Expression of Sertoli cell signature transcripts encoding regulators of vesicular trafficking.
(A) Expression of RRAS2 and HRAS and the catalytic subunit of phosphoinositide 3-kinase (PI3K), PIK3CA. Binding of RRAS2 and HRAS to PI3KCA stimulates its catalytic activity. (B) Expression of regulatory subunits of PI3K. (C) Small GTPase family members that regulate vesicular trafficking. RAB20 regulates endocytic trafficking; RAB40B and RAB3D regulate exocytic trafficking. Activities of these small GTPases are stimulated by PI3K. Data are expressed as mean + SEM of CPMs for each transcript. The presence of an asterisk over a pair of bars indicates a significant difference between testes with complete spermatogenesis and testes with SCO syndrome (FDR0.05).

Fig 5. Expression of Sertoli cell signature…

Fig 5. Expression of Sertoli cell signature transcripts that encode growth factors that are known…

Fig 5. Expression of Sertoli cell signature transcripts that encode growth factors that are known regulators of SSCs and/or progenitor spermatogonia.
Data are expressed as mean + SEM of CPMs. An asterisk over a pair of bars indicates a significant difference (FDR≤0.05).

Fig 6. Confirmation that FGF8 is a…

Fig 6. Confirmation that FGF8 is a product of human Sertoli cells and verification that…

Fig 6. Confirmation that FGF8 is a product of human Sertoli cells and verification that expressions of both FGF8 protein and mRNA are reduced in SCO testes.
Testes with complete spermatogenesis (A) and with SCO syndrome (B) were dispersed into single cells and then incubated with antibodies for the Sertoli cell marker SOX9 and for FGF8. Negative control cells (C) were not incubated with antibodies. Cells were then analyzed FACs. These data are representative of three independent analyses. (D) Quantification by real-time PCR of levels of FGF8 mRNA in biopsies of human testes with complete spermatogenesis and in testes with SCO syndrome. Data (mean + SEM; n = 3) were normalized for expression of beta actin in the same sample (p

Fig 7. Summary: Deficits in Sertoli cell…

Fig 7. Summary: Deficits in Sertoli cell gene expression that are associated with specific aspects…

Fig 7. Summary: Deficits in Sertoli cell gene expression that are associated with specific aspects of human SCO syndrome.
(A) Human testis with complete spermatogenesis. Polarity complex proteins (red) and tight junctions (gray) polarize the plasma membrane thereby generating apical and basal-lateral domains (identified by up and down arrows). Additional tight junctions, ectoplasmic specialization (yellow) and gap junctions (blue and green) are concentrated at sites of cell-cell contact. Vesicular trafficking (VT; tan circles) of adhesive and gap junction proteins facilitates the assembly and disassembly of junctions, controlled by a pathway that includes Ras GTPases, PI3K and RAB small GTPases (purple shading). Sertoli cells express normal amounts of GDNF, FGF8 and BMP4, growth factors that regulate replication and differentiation of SSCs. (B) The infertile SCO testis. Inadequate expression of polarity and adaptor proteins renders Sertoli cells incapable of properly polarizing their plasma membranes. There is reduced, inefficient vesicular trafficking of proteins that form tight junctions, ectoplasmic specializations and gap junctions, stranding these proteins in vesicles within the cytoplasm. Finally, there is reduced expression of GDNF, FGF8 and BMP4. (C) The effects of GDNF, FGF8 and BMP4 on SSCs and progenitor spermatogonia. GDNF and FGF8 promote self-renewing replication of SSCs, while BMP4 stimulates differentiation of SSCs into progenitor spermatogonia and progenitors into fully differentiated B spermatogonia.
All figures (7)
Similar articles
Cited by
References
    1. Irvine DS. Epidemiology and aetiology of male infertility. Hum Reprod. 1998;13 Suppl 1:33–44. Epub 1998/07/15. . - PubMed
    1. Schlegel PN, Palermo GD, Goldstein M, Menendez S, Zaninovic N, Veeck LL, et al. Testicular sperm extraction with intracytoplasmic sperm injection for nonobstructive azoospermia. Urology. 1997;49(3):435–40. Epub 1997/03/01. 10.1016/S0090-4295(97)00032-0 . - DOI - PubMed
    1. Nagano M, Ryu BY, Brinster CJ, Avarbock MR, Brinster RL. Maintenance of mouse male germ line stem cells in vitro. Biol Reprod. 2003;68(6):2207–14. Epub 2003/02/28. 10.1095/biolreprod.102.014050 . - DOI - PubMed
    1. Ogawa T, Ohmura M, Yumura Y, Sawada H, Kubota Y. Expansion of murine spermatogonial stem cells through serial transplantation. Biol Reprod. 2003;68(1):316–22. Epub 2002/12/21. . - PubMed
    1. Oatley JM, Brinster RL. The germline stem cell niche unit in mammalian testes. Physiol Rev. 2012;92(2):577–95. Epub 2012/04/27. 10.1152/physrev.00025.2011 - DOI - PMC - PubMed
Show all 80 references
Publication types
MeSH terms
Related information
[x]
Cite
Copy Download .nbib
Format: AMA APA MLA NLM
Fig 2. The expression of Sertoli cell…
Fig 2. The expression of Sertoli cell signature transcripts in testes with complete spermatogenesis and in testes with SCO syndrome.
(A) PCA analysis of the expression of Sertoli cell signature transcripts in individual testes. Data were obtained from 4 testes with complete spermatogenesis (C1-4, green symbols) and seven SCO testes (SCO5, SCO7-12, red symbols) (B). A comparison of the mean level of expression of each Sertoli cell signature transcript in testes with complete spermatogenesis and in testes with SCO syndrome. Transcripts that were expressed in SCO testes at significantly higher levels (FDR<0.05), the same level or significantly lower levels are identified by blue, white and black dots, respectively.
Fig 3. Expression of Sertoli cell signature…
Fig 3. Expression of Sertoli cell signature transcripts encoding proteins that form adhesive or gap junctions or that organize and polarize the Sertoli cell plasma membrane.
(A) CLDN11 and JAM3 are tight junction adhesive proteins. Nectin2 and CDH2 are adhesive proteins of ectoplasmic specializations. GJB2 encodes the gap junction protein, Connexin 26. (B) Transcripts that encode proteins that organize or polarize the plasma membrane. CRB2 is a cell polarity component protein that defines the site of separation of the apical and basal-lateral plasma membrane domains. TJP3 is an adaptor protein that links the adhesive proteins of a tight junction to the actin cytoskeleton. SPTB, SPTPN4 and SPTPN5 are scaffolding proteins that concentrate beneath the plasma membrane, seed formation of membrane microdomains, and link those domains to the actin cytoskeleton. All data in this figure are expressed as mean + SEM of CPMs. The presence of an asterisk over a pair of bars indicates a significant difference between testes with complete spermatogenesis and testes with SCO syndrome (FDR≤0.05).
Fig 4. Expression of Sertoli cell signature…
Fig 4. Expression of Sertoli cell signature transcripts encoding regulators of vesicular trafficking.
(A) Expression of RRAS2 and HRAS and the catalytic subunit of phosphoinositide 3-kinase (PI3K), PIK3CA. Binding of RRAS2 and HRAS to PI3KCA stimulates its catalytic activity. (B) Expression of regulatory subunits of PI3K. (C) Small GTPase family members that regulate vesicular trafficking. RAB20 regulates endocytic trafficking; RAB40B and RAB3D regulate exocytic trafficking. Activities of these small GTPases are stimulated by PI3K. Data are expressed as mean + SEM of CPMs for each transcript. The presence of an asterisk over a pair of bars indicates a significant difference between testes with complete spermatogenesis and testes with SCO syndrome (FDR0.05).
Fig 5. Expression of Sertoli cell signature…
Fig 5. Expression of Sertoli cell signature transcripts that encode growth factors that are known regulators of SSCs and/or progenitor spermatogonia.
Data are expressed as mean + SEM of CPMs. An asterisk over a pair of bars indicates a significant difference (FDR≤0.05).
Fig 6. Confirmation that FGF8 is a…
Fig 6. Confirmation that FGF8 is a product of human Sertoli cells and verification that expressions of both FGF8 protein and mRNA are reduced in SCO testes.
Testes with complete spermatogenesis (A) and with SCO syndrome (B) were dispersed into single cells and then incubated with antibodies for the Sertoli cell marker SOX9 and for FGF8. Negative control cells (C) were not incubated with antibodies. Cells were then analyzed FACs. These data are representative of three independent analyses. (D) Quantification by real-time PCR of levels of FGF8 mRNA in biopsies of human testes with complete spermatogenesis and in testes with SCO syndrome. Data (mean + SEM; n = 3) were normalized for expression of beta actin in the same sample (p

Fig 7. Summary: Deficits in Sertoli cell…

Fig 7. Summary: Deficits in Sertoli cell gene expression that are associated with specific aspects…

Fig 7. Summary: Deficits in Sertoli cell gene expression that are associated with specific aspects of human SCO syndrome.
(A) Human testis with complete spermatogenesis. Polarity complex proteins (red) and tight junctions (gray) polarize the plasma membrane thereby generating apical and basal-lateral domains (identified by up and down arrows). Additional tight junctions, ectoplasmic specialization (yellow) and gap junctions (blue and green) are concentrated at sites of cell-cell contact. Vesicular trafficking (VT; tan circles) of adhesive and gap junction proteins facilitates the assembly and disassembly of junctions, controlled by a pathway that includes Ras GTPases, PI3K and RAB small GTPases (purple shading). Sertoli cells express normal amounts of GDNF, FGF8 and BMP4, growth factors that regulate replication and differentiation of SSCs. (B) The infertile SCO testis. Inadequate expression of polarity and adaptor proteins renders Sertoli cells incapable of properly polarizing their plasma membranes. There is reduced, inefficient vesicular trafficking of proteins that form tight junctions, ectoplasmic specializations and gap junctions, stranding these proteins in vesicles within the cytoplasm. Finally, there is reduced expression of GDNF, FGF8 and BMP4. (C) The effects of GDNF, FGF8 and BMP4 on SSCs and progenitor spermatogonia. GDNF and FGF8 promote self-renewing replication of SSCs, while BMP4 stimulates differentiation of SSCs into progenitor spermatogonia and progenitors into fully differentiated B spermatogonia.
All figures (7)
Fig 7. Summary: Deficits in Sertoli cell…
Fig 7. Summary: Deficits in Sertoli cell gene expression that are associated with specific aspects of human SCO syndrome.
(A) Human testis with complete spermatogenesis. Polarity complex proteins (red) and tight junctions (gray) polarize the plasma membrane thereby generating apical and basal-lateral domains (identified by up and down arrows). Additional tight junctions, ectoplasmic specialization (yellow) and gap junctions (blue and green) are concentrated at sites of cell-cell contact. Vesicular trafficking (VT; tan circles) of adhesive and gap junction proteins facilitates the assembly and disassembly of junctions, controlled by a pathway that includes Ras GTPases, PI3K and RAB small GTPases (purple shading). Sertoli cells express normal amounts of GDNF, FGF8 and BMP4, growth factors that regulate replication and differentiation of SSCs. (B) The infertile SCO testis. Inadequate expression of polarity and adaptor proteins renders Sertoli cells incapable of properly polarizing their plasma membranes. There is reduced, inefficient vesicular trafficking of proteins that form tight junctions, ectoplasmic specializations and gap junctions, stranding these proteins in vesicles within the cytoplasm. Finally, there is reduced expression of GDNF, FGF8 and BMP4. (C) The effects of GDNF, FGF8 and BMP4 on SSCs and progenitor spermatogonia. GDNF and FGF8 promote self-renewing replication of SSCs, while BMP4 stimulates differentiation of SSCs into progenitor spermatogonia and progenitors into fully differentiated B spermatogonia.

References

    1. Irvine DS. Epidemiology and aetiology of male infertility. Hum Reprod. 1998;13 Suppl 1:33–44. Epub 1998/07/15. .
    1. Schlegel PN, Palermo GD, Goldstein M, Menendez S, Zaninovic N, Veeck LL, et al. Testicular sperm extraction with intracytoplasmic sperm injection for nonobstructive azoospermia. Urology. 1997;49(3):435–40. Epub 1997/03/01. 10.1016/S0090-4295(97)00032-0 .
    1. Nagano M, Ryu BY, Brinster CJ, Avarbock MR, Brinster RL. Maintenance of mouse male germ line stem cells in vitro. Biol Reprod. 2003;68(6):2207–14. Epub 2003/02/28. 10.1095/biolreprod.102.014050 .
    1. Ogawa T, Ohmura M, Yumura Y, Sawada H, Kubota Y. Expansion of murine spermatogonial stem cells through serial transplantation. Biol Reprod. 2003;68(1):316–22. Epub 2002/12/21. .
    1. Oatley JM, Brinster RL. The germline stem cell niche unit in mammalian testes. Physiol Rev. 2012;92(2):577–95. Epub 2012/04/27. 10.1152/physrev.00025.2011
    1. Parker N, Falk H, Singh D, Fidaleo A, Smith B, Lopez MS, et al. Responses to glial cell line-derived neurotrophic factor change in mice as spermatogonial stem cells form progenitor spermatogonia which replicate and give rise to more differentiated progeny. Biol Reprod. 2014;91(4):92 Epub 2014/08/29. 10.1095/biolreprod.114.119099
    1. Wright WW. Stage-specific gene expression by Sertoli Cells In: Griswold M, editor. Sertoli Cell Biology. Amsterdam, Netherlands: Elsevier; 2015. p. 273–306.
    1. Stanley EL, Johnston DS, Fan J, Papadopoulos V, Chen H, Ge RS, et al. Stem Leydig cell differentiation: gene expression during development of the adult rat population of Leydig cells. Biol Reprod. 2011;85(6):1161–6. Epub 2011/08/13. 10.1095/biolreprod.111.091850
    1. Jan SZ, Vormer TL, Jongejan A, Roling MD, Silber SJ, de Rooij DG, et al. Unraveling transcriptome dynamics in human spermatogenesis. Development. 2017;144(20):3659–73. Epub 2017/09/25. 10.1242/dev.152413
    1. Lottrup G, Belling K, Leffers H, Nielsen JE, Dalgaard MD, Juul A, et al. Comparison of global gene expression profiles of microdissected human foetal Leydig cells with their normal and hyperplastic adult equivalents. Mol Hum Reprod. 2017;23(5):339–54. Epub 2017/03/24. 10.1093/molehr/gax012 .
    1. Paniagua R, Codesal J, Nistal M, Rodriguez MC, Santamaria L. Quantification of cell types throughout the cycle of the human seminiferous epithelium and their DNA content. A new approach to the spermatogonial stem cell in man. Anat Embryol (Berl). 1987;176(2):225–30. Epub 1987/01/01. .
    1. Petersen PM, Seieroe K, Pakkenberg B. The total number of Leydig and Sertoli cells in the testes of men across various age groups—a stereological study. J Anat. 2015;226(2):175–9. Epub 2014/12/30. 10.1111/joa.12261
    1. Camatini M, Franchi E, Decurtis I. Permeability to lanthanum of blood testis barrier in human germinal aplasia. Anat Rec. 1981;200(3):293–7. Epub 1981/07/01. 10.1002/ar.1092000308 .
    1. Matsuo Y, Nomata K, Eguchi J, Aoki D, Hayashi T, Hishikawa Y, et al. Immunohistochemical analysis of connexin43 expression in infertile human testes. Acta Histochem Cytochem. 2007;40(3):69–75. Epub 2007/07/27. 10.1267/ahc.07001
    1. Mruk DD, Cheng CY. The Mammalian Blood-Testis Barrier: Its Biology and Regulation. Endocr Rev. 2015;36(5):564–91. Epub 2015/09/12. 10.1210/er.2014-1101
    1. Luo J, Megee S, Dobrinski I. Asymmetric distribution of UCH-L1 in spermatogonia is associated with maintenance and differentiation of spermatogonial stem cells. J Cell Physiol. 2009;220(2):460–8. Epub 2009/04/24. 10.1002/jcp.21789
    1. Spradling A, Fuller MT, Braun RE, Yoshida S. Germline stem cells. Cold Spring Harb Perspect Biol. 2011;3(11):a002642 Epub 2011/07/28. 10.1101/cshperspect.a002642
    1. Dovere L, Fera S, Grasso M, Lamberti D, Gargioli C, Muciaccia B, et al. The niche-derived glial cell line-derived neurotrophic factor (GDNF) induces migration of mouse spermatogonial stem/progenitor cells. PLoS One. 2013;8(4):e59431 Epub 2013/04/25. 10.1371/journal.pone.0059431
    1. Singh D, Paduch DA, Schlegel PN, Orwig KE, Mielnik A, Bolyakov A, et al. The production of glial cell line-derived neurotrophic factor by human sertoli cells is substantially reduced in sertoli cell-only testes. Hum Reprod. 2017;32(5):1108–17. Epub 2017/04/04. 10.1093/humrep/dex061 .
    1. Schlegel PN. Causes of azoospermia and their management. Reprod Fertil Dev. 2004;16(5):561–72. Epub 2004/09/16. 10.10371/RD03087 .
    1. Li H, Durbin R. Fast and accurate short read alignment with Burrows-Wheeler transform. Bioinformatics. 2009;25(14):1754–60. Epub 2009/05/20. 10.1093/bioinformatics/btp324
    1. Anders S, Pyl PT, Huber W. HTSeq—a Python framework to work with high-throughput sequencing data. Bioinformatics. 2015;31(2):166–9. Epub 2014/09/28. 10.1093/bioinformatics/btu638
    1. Robinson MD, McCarthy DJ, Smyth GK. edgeR: a Bioconductor package for differential expression analysis of digital gene expression data. Bioinformatics. 2010;26(1):139–40. Epub 2009/11/17. 10.1093/bioinformatics/btp616
    1. Johnston DS, Wright WW, Dicandeloro P, Wilson E, Kopf GS, Jelinsky SA. Stage-specific gene expression is a fundamental characteristic of rat spermatogenic cells and Sertoli cells. Proc Natl Acad Sci U S A. 2008;105(24):8315–20. Epub 2008/06/12. 10.1073/pnas.0709854105
    1. Green CD, Ma Q, Manske GL, Shami AN, Zheng X, Marini S, et al. A Comprehensive Roadmap of Murine Spermatogenesis Defined by Single-Cell RNA-Seq. Dev Cell. 2018;46(5):651–67 e10. Epub 2018/08/28. 10.1016/j.devcel.2018.07.025 .
    1. Hasegawa K, Saga Y. Retinoic acid signaling in Sertoli cells regulates organization of the blood-testis barrier through cyclical changes in gene expression. Development. 2012;139(23):4347–55. Epub 2012/10/26. 10.1242/dev.080119 .
    1. Cowan G, Childs AJ, Anderson RA, Saunders PT. Establishment of long-term monolayer cultures of somatic cells from human fetal testes and expansion of peritubular myoid cells in the presence of androgen. Reproduction. 2010;139(4):749–57. Epub 2010/01/22. 10.1530/REP-09-0532 .
    1. Marchetti C, Hamdane M, Mitchell V, Mayo K, Devisme L, Rigot JM, et al. Immunolocalization of inhibin and activin alpha and betaB subunits and expression of corresponding messenger RNAs in the human adult testis. Biol Reprod. 2003;68(1):230–5. Epub 2002/12/21. .
    1. Szklarczyk D, Morris JH, Cook H, Kuhn M, Wyder S, Simonovic M, et al. The STRING database in 2017: quality-controlled protein-protein association networks, made broadly accessible. Nucleic Acids Res. 2017;45(D1):D362–D8. Epub 2016/12/08. 10.1093/nar/gkw937
    1. Guo J, Grow EJ, Yi C, Mlcochova H, Maher GJ, Lindskog C, et al. Chromatin and Single-Cell RNA-Seq Profiling Reveal Dynamic Signaling and Metabolic Transitions during Human Spermatogonial Stem Cell Development. Cell Stem Cell. 2017;21(4):533–46 e6. Epub 2017/10/07. 10.1016/j.stem.2017.09.003
    1. Wang YM, Sullivan PM, Petrusz P, Yarbrough W, Joseph DR. The androgen-binding protein gene is expressed in CD1 mouse testis. Mol Cell Endocrinol. 1989;63(1–2):85–92. Epub 1989/05/01. .
    1. Selva DM, Hammond GL. Human sex hormone-binding globulin is expressed in testicular germ cells and not in sertoli cells. Horm Metab Res. 2006;38(4):230–5. Epub 2006/05/16. 10.1055/s-2006-925336 .
    1. Wang XN, Li ZS, Ren Y, Jiang T, Wang YQ, Chen M, et al. The Wilms tumor gene, Wt1, is critical for mouse spermatogenesis via regulation of sertoli cell polarity and is associated with non-obstructive azoospermia in humans. PLoS Genet. 2013;9(8):e1003645 Epub 2013/08/13. 10.1371/journal.pgen.1003645
    1. Clark AM, Garland KK, Russell LD. Desert hedgehog (Dhh) gene is required in the mouse testis for formation of adult-type Leydig cells and normal development of peritubular cells and seminiferous tubules. Biol Reprod. 2000;63(6):1825–38. Epub 2000/11/25. .
    1. Das DS, Wadhwa N, Kunj N, Sarda K, Pradhan BS, Majumdar SS. Dickkopf homolog 3 (DKK3) plays a crucial role upstream of WNT/beta-CATENIN signaling for Sertoli cell mediated regulation of spermatogenesis. PLoS One. 2013;8(5):e63603 Epub 2013/05/15. 10.1371/journal.pone.0063603
    1. Behrens A, Genoud N, Naumann H, Rulicke T, Janett F, Heppner FL, et al. Absence of the prion protein homologue Doppel causes male sterility. Embo J. 2002;21(14):3652–8. Epub 2002/07/12. 10.1093/emboj/cdf386
    1. Kidder GM, Cyr DG. Roles of connexins in testis development and spermatogenesis. Semin Cell Dev Biol. 2016;50:22–30. Epub 2016/01/19. 10.1016/j.semcdb.2015.12.019 .
    1. Cheng CY, Mruk DD. Biochemistry of Sertoli cell/gern cell junctions, germ cell transport, and spermiation in the seminiferous epithelium In: Griswold MD, editor. Sertoli Cell Biology, Second Edition Amsterdam, Netherlands: Elsevier; 2015. p. 333–83.
    1. Fink C, Weigel R, Fink L, Wilhelm J, Kliesch S, Zeiler M, et al. Claudin-11 is over-expressed and dislocated from the blood-testis barrier in Sertoli cells associated with testicular intraepithelial neoplasia in men. Histochem Cell Biol. 2009;131(6):755–64. Epub 2009/02/26. 10.1007/s00418-009-0576-2 .
    1. Stammler A, Luftner BU, Kliesch S, Weidner W, Bergmann M, Middendorff R, et al. Highly Conserved Testicular Localization of Claudin-11 in Normal and Impaired Spermatogenesis. PLoS One. 2016;11(8):e0160349 Epub 2016/08/04. 10.1371/journal.pone.0160349
    1. Rodriguez-Boulan E, Macara IG. Organization and execution of the epithelial polarity programme. Nat Rev Mol Cell Biol. 2014;15(4):225–42. Epub 2014/03/22. 10.1038/nrm3775
    1. Gao Y, Lui WY, Lee WM, Cheng CY. Polarity protein Crumbs homolog-3 (CRB3) regulates ectoplasmic specialization dynamics through its action on F-actin organization in Sertoli cells. Sci Rep. 2016;6:28589 Epub 2016/07/01. 10.1038/srep28589
    1. Gruber M, Mathew LK, Runge AC, Garcia JA, Simon MC. EPAS1 Is Required for Spermatogenesis in the Postnatal Mouse Testis. Biol Reprod. 2010;82(6):1227–36. Epub 2010/02/26. 10.1095/biolreprod.109.079202
    1. Bennett V, Baines AJ. Spectrin and ankyrin-based pathways: metazoan inventions for integrating cells into tissues. Physiol Rev. 2001;81(3):1353–92. Epub 2001/06/28. 10.1152/physrev.2001.81.3.1353 .
    1. Machnicka B, Czogalla A, Hryniewicz-Jankowska A, Boguslawska DM, Grochowalska R, Heger E, et al. Spectrins: a structural platform for stabilization and activation of membrane channels, receptors and transporters. Biochim Biophys Acta. 2014;1838(2):620–34. Epub 2013/05/16. 10.1016/j.bbamem.2013.05.002 .
    1. Siu MK, Wong CH, Lee WM, Cheng CY. Sertoli-germ cell anchoring junction dynamics in the testis are regulated by an interplay of lipid and protein kinases. J Biol Chem. 2005;280(26):25029–47. Epub 2005/05/05. 10.1074/jbc.M501049200 .
    1. Smith BE, Braun RE. Germ cell migration across Sertoli cell tight junctions. Science. 2012;338(6108):798–802. Epub 2012/09/22. 10.1126/science.1219969
    1. Xiao X, Mruk DD, Wong EW, Lee WM, Han D, Wong CK, et al. Differential effects of c-Src and c-Yes on the endocytic vesicle-mediated trafficking events at the Sertoli cell blood-testis barrier: an in vitro study. Am J Physiol Endocrinol Metab. 2014;307(7):E553–62. Epub 2014/08/15. 10.1152/ajpendo.00176.2014
    1. Nakhaeizadeh H, Amin E, Nakhaei-Rad S, Dvorsky R, Ahmadian MR. The RAS-Effector Interface: Isoform-Specific Differences in the Effector Binding Regions. PLoS One. 2016;11(12):e0167145 Epub 2016/12/10. 10.1371/journal.pone.0167145
    1. Bhattacharya S, McElhanon KE, Gushchina LV, Weisleder N. Role of phosphatidylinositol-4,5-bisphosphate 3-kinase signaling in vesicular trafficking. Life Sci. 2016;167:39–45. Epub 2016/10/21. 10.1016/j.lfs.2016.10.018
    1. Egami Y, Araki N. Rab20 regulates phagosome maturation in RAW264 macrophages during Fc gamma receptor-mediated phagocytosis. PLoS One. 2012;7(4):e35663 Epub 2012/05/01. 10.1371/journal.pone.0035663
    1. Jacob A, Jing J, Lee J, Schedin P, Gilbert SM, Peden AA, et al. Rab40b regulates trafficking of MMP2 and MMP9 during invadopodia formation and invasion of breast cancer cells. J Cell Sci. 2013;126(Pt 20):4647–58. Epub 2013/08/02. 10.1242/jcs.126573
    1. Millar AL, Pavios NJ, Xu J, Zheng MH. Rab3D: a regulator of exocytosis in non-neuronal cells. Histol Histopathol. 2002;17(3):929–36. Epub 2002/08/10. 10.14670/HH-17.929 .
    1. Boyer A, Hermo L, Paquet M, Robaire B, Boerboom D. Seminiferous tubule degeneration and infertility in mice with sustained activation of WNT/CTNNB1 signaling in sertoli cells. Biol Reprod. 2008;79(3):475–85. Epub 2008/05/16. 10.1095/biolreprod.108.068627 .
    1. Boyer A, Yeh JR, Zhang X, Paquet M, Gaudin A, Nagano MC, et al. CTNNB1 signaling in sertoli cells downregulates spermatogonial stem cell activity via WNT4. PLoS One. 2012;7(1):e29764 Epub 2012/01/19. 10.1371/journal.pone.0029764
    1. Hasegawa K, Saga Y. FGF8-FGFR1 signaling acts as a niche factor for maintaining undifferentiated spermatogonia in the mouse. Biol Reprod. 2014;91(6):145 Epub 2014/11/02. 10.1095/biolreprod.114.121012 .
    1. Yeh JR, Zhang X, Nagano MC. Wnt5a is a cell-extrinsic factor that supports self-renewal of mouse spermatogonial stem cells. J Cell Sci. 2011;124(Pt 14):2357–66. Epub 2011/06/23. 10.1242/jcs.080903 .
    1. Pellegrini M, Grimaldi P, Rossi P, Geremia R, Dolci S. Developmental expression of BMP4/ALK3/SMAD5 signaling pathway in the mouse testis: a potential role of BMP4 in spermatogonia differentiation. J Cell Sci. 2003;116(Pt 16):3363–72. Epub 2003/07/15. 10.1242/jcs.00650 .
    1. Yang Y, Feng Y, Feng X, Liao S, Wang X, Gan H, et al. BMP4 Cooperates with Retinoic Acid to Induce the Expression of Differentiation Markers in Cultured Mouse Spermatogonia. Stem Cells Int. 2016;2016:9536192 Epub 2016/11/01. 10.1155/2016/9536192
    1. Ohta H, Yomogida K, Dohmae K, Nishimune Y. Regulation of proliferation and differentiation in spermatogonial stem cells: the role of c-kit and its ligand SCF. Development. 2000;127(10):2125–31. Epub 2000/04/19. .
    1. Oatley JM, Oatley MJ, Avarbock MR, Tobias JW, Brinster RL. Colony stimulating factor 1 is an extrinsic stimulator of mouse spermatogonial stem cell self-renewal. Development. 2009;136(7):1191–9. Epub 2009/03/10. 10.1242/dev.032243
    1. Yang QE, Kim D, Kaucher A, Oatley MJ, Oatley JM. CXCL12-CXCR4 signaling is required for the maintenance of mouse spermatogonial stem cells. J Cell Sci. 2013;126(Pt 4):1009–20. Epub 2012/12/15. 10.1242/jcs.119826
    1. Chui K, Trivedi A, Cheng CY, Cherbavaz DB, Dazin PF, Huynh AL, et al. Characterization and Functionality of Proliferative Human Sertoli Cells. Cell Transplant. 2010. Epub 2010/11/09. 10.3727/096368910X536563 .
    1. Johnston DS, Olivas E, DiCandeloro P, Wright WW. Stage-specific changes in GDNF expression by rat Sertoli cells: a possible regulator of the replication and differentiation of stem spermatogonia. Biol Reprod. 2011;85(4):763–9. Epub 2011/06/10. 10.1095/biolreprod.110.087676
    1. Chiba K, Kondo Y, Yamaguchi K, Miyake H, Fujisawa M. Inhibition of claudin-11 and occludin expression in rat Sertoli cells by mono-(2-ethylhexyl) phthalate through p44/42 mitogen-activated protein kinase pathway. J Androl. 2012;33(3):368–74. Epub 2011/08/27. 10.2164/jandrol.111.013664 .
    1. Hartsock A, Nelson WJ. Adherens and tight junctions: structure, function and connections to the actin cytoskeleton. Biochim Biophys Acta. 2008;1778(3):660–9. Epub 2007/09/15. 10.1016/j.bbamem.2007.07.012
    1. Nusrat A, Parkos CA, Verkade P, Foley CS, Liang TW, Innis-Whitehouse W, et al. Tight junctions are membrane microdomains. J Cell Sci. 2000;113 (Pt 10):1771–81. Epub 2000/04/19. .
    1. Pradhan D, Lombardo CR, Roe S, Rimm DL, Morrow JS. alpha -Catenin binds directly to spectrin and facilitates spectrin-membrane assembly in vivo. J Biol Chem. 2001;276(6):4175–81. Epub 2000/11/09. 10.1074/jbc.M009259200 .
    1. Cheng CY, Mruk DD. Regulation of blood-testis barrier dynamics by focal adhesion kinase (FAK): an unexpected turn of events. Cell Cycle. 2009;8(21):3493–9. Epub 2009/10/14. 10.4161/cc.8.21.9833
    1. Cheng CY, Mruk DD. An intracellular trafficking pathway in the seminiferous epithelium regulating spermatogenesis: a biochemical and molecular perspective. Crit Rev Biochem Mol Biol. 2009;44(5):245–63. Epub 2009/07/23. 10.1080/10409230903061207
    1. Chiba K, Yamaguchi K, Ando M, Miyake H, Fujisawa M. Expression pattern of testicular claudin-11 in infertile men. Urology. 2012;80(5):1161 e13–7. Epub 2012/09/07. 10.1016/j.urology.2012.06.036 .
    1. Risley MS, Tan IP, Farrell J. Gap junctions with varied permeability properties establish cell-type specific communication pathways in the rat seminiferous epithelium. Biol Reprod. 2002;67(3):945–52. Epub 2002/08/24. .
    1. Decrouy X, Gasc JM, Pointis G, Segretain D. Functional characterization of Cx43 based gap junctions during spermatogenesis. J Cell Physiol. 2004;200(1):146–54. Epub 2004/05/12. 10.1002/jcp.10473 .
    1. Vitillo L, Kimber SJ. Integrin and FAK Regulation of Human Pluripotent Stem Cells. Curr Stem Cell Rep. 2017;3(4):358–65. Epub 2017/11/28. 10.1007/s40778-017-0100-x
    1. Fuchs E, Tumbar T, Guasch G. Socializing with the neighbors: stem cells and their niche. Cell. 2004;116(6):769–78. Epub 2004/03/24. .
    1. Morrison SJ, Kimble J. Asymmetric and symmetric stem-cell divisions in development and cancer. Nature. 2006;441(7097):1068–74. Epub 2006/07/01. 10.1038/nature04956 .
    1. Meng X, Lindahl M, Hyvonen ME, Parvinen M, de Rooij DG, Hess MW, et al. Regulation of cell fate decision of undifferentiated spermatogonia by GDNF. Science. 2000;287(5457):1489–93. .
    1. Huleihel M, Fadlon E, Abuelhija A, Piltcher Haber E, Lunenfeld E. Glial cell line-derived neurotrophic factor (GDNF) induced migration of spermatogonial cells in vitro via MEK and NF-kB pathways. Differentiation. 2013;86(1–2):38–47. Epub 2013/08/14. 10.1016/j.diff.2013.06.005 .
    1. Tadokoro Y, Yomogida K, Ohta H, Tohda A, Nishimune Y. Homeostatic regulation of germinal stem cell proliferation by the GDNF/FSH pathway. Mech Dev. 2002;113(1):29–39. Epub 2002/03/20. .
    1. Tindall DJ, Vitale R, Means AR. Androgen binding protein as a biochemical marker of formation of the blood-testis barrier. Endocrinology. 1975;97(3):636–48. Epub 1975/09/01. 10.1210/endo-97-3-636 .

Source: PubMed

3
Prenumerera