Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2017 Jun 7.
Published in final edited form as: Andrology. 2016 Feb 4;4(2):189–212. doi: 10.1111/andr.12165

The Sertoli cell: one hundred fifty years of beauty and plasticity

L R França 1,2, R A Hess 3, J M Dufour 4, M C Hofmann 5, M D Griswold 6
PMCID: PMC5461925  NIHMSID: NIHMS843887  PMID: 26846984

SUMMARY

It has been one and a half centuries since Enrico Sertoli published the seminal discovery of the testicular ‘nurse cell’, not only a key cell in the testis, but indeed one of the most amazing cells in the vertebrate body. In this review, we begin by examining the three phases of morphological research that have occurred in the study of Sertoli cells, because microscopic anatomy was essentially the only scientific discipline available for about the first 75 years after the discovery. Biochemistry and molecular biology then changed all of biological sciences, including our understanding of the functions of Sertoli cells. Immunology and stem cell biology were not even topics of science in 1865, but they have now become major issues in our appreciation of Sertoli cell’s role in spermatogenesis. We end with the universal importance and plasticity of function by comparing Sertoli cells in fish, amphibians, and mammals. In these various classes of vertebrates, Sertoli cells have quite different modes of proliferation and epithelial maintenance, cystic vs. tubular formation, yet accomplish essentially the same function but in strikingly different ways.

Keywords: amniotes, anamniotes, blood–testis barrier, germ cell, immune privilege, immune tolerance, Sertoli cell, spermatogenesis, stem cell niche, testis

INTRODUCTION

The 150th year anniversary of the publication reporting the discovery of the Sertoli cell is to be celebrated (Sertoli, 1865). Although Enrico Sertoli (Fig. 1) went on to explore numerous other subjects, including physiology of blood proteins, tissue carbonic acid in the respiratory system and smooth muscle contraction, he was given the highest honor for his study of the human testis, when Von Ebner first called these cells, ‘the Sertoli cells’ (Ebner, 1888). Research on the Sertoli cell started out slowly, with only about 25 manuscripts published up to 1950. However, rapid advancement in our knowledge of this unique cell was soon realized with the invention of the electron microscope and discoveries of DNA, RNA and new methods of biochemistry, which permitted the incorporation of histochemistry and immunochemistry in molecular biology studies (see Table 1 for timeline of advances). Now there are nearly 500 papers published each year describing the intricate relationships established by Sertoli cells in the testis. Presented here are highlights that emphasize the superb scientific beauty and intrinsic plasticity of the cell, to which three books and numerous reviews have been devoted (Russell & Griswold, 1993b; Skinner & Griswold, 2005; Griswold, 2015b) (See Table 2 and Supplement Table S1 for lists of important reviews and books). In this brief review, we first present the basic structure of the Sertoli cell and then show how molecular biology has given us insight into the complicated mechanisms involved in its nurse cell function. We then discuss how this information is applied to current ongoing studies on the role of Sertoli cells in the spermatogonial stem cell niche and in the maintenance of testicular immune privilege. Finally, a comparative view across vertebrate species is summarized to show common but also divergent pathways that have developed to allow the same cell to establish its germ cell interactions in both cystic and tubular modes of organization. Our review cannot be totally inclusive but will highlight the passionate pursuits of the authors in the hope that we can stimulate even more interest in the complexities and importance of the Sertoli cell.

Figure 1.

Figure 1

Photo of Professor Enrico Sertoli, published in honor of his retirement after 36 years of teaching and research (Negrini et al., 1908) and drawings (Sertoli, 1865) from Sertoli’s original publication (Fig. 1A–D).

Table 1.

Significant Sertoli cell milestones

1865 Enrico Sertoli’s first publication (Sertoli, 1865)
1888 The branched cells first called ‘Sertoli cells’ (Ebner, 1888)
1899 Sertoli and germ cells thought to have common origin (Regaud, 1899)
1901 Sertoli cells thought to be syncytial (Regaud, 1901)
1901 Sertoli cells phagocytize degenerative germ cells (Regaud, 1901; Clermont & Morgentaler, 1955; Russell & Clermont, 1977)
1902 Sertoli cells do not divide in adult testis (Ebner, 1888)
1942 Sertoli cell tumors or tubular adenomas described (Innes, 1942)
1948 ‘Sertoli cell only’ syndrome first described (Heller et al., 1948)
1949 Sertoli cells produce estrogen (Teilum, 1949; Armstrong et al., 1975)
1950 Cyclical changes in Sertoli cell morphology (Elftman, 1950; Leblond & Clermont, 1952; Brökelmann, 1963; Kerr & De Kretser, 1975)
1952 Description of changes in Sertoli cell nucleus by Stage (Leblond & Clermont, 1952)
1956 First Electron Microscopic description; Sertoli cells are not syncytial (Fawcett & Burgos, 1956)
1963 Silver method used to show changes in Sertoli cell cytoplasm morphology by Stage (Elftman, 1963)
1964 First primary cultures of Sertoli cells (Steinberger et al., 1964; Steinberger & Steinberger, 1970)
1965 Sertoli unique nucleolus and heterochromatin (Monesi, 1965; Jean et al., 1983)
1967 Junctional Specializations described (Flickinger & Fawcett, 1967)
1967 Blood–testis barrier defined physiologically (Setchell, 1967)
1968 Sertoli cell as a major factor in testicular secretions (Setchell et al., 1968; Setchell, 1974)
1969 Sertoli cell role in spermiation ultrastructure (Fawcett & Phillips, 1969; Russell, 1984)
1970 Sertoli cell nucleolus used as a constant for counting cells (Bustos-Obregon, 1970)
1970 Blood–testis barrier defined ultrastructurally between Sertoli cell junctions; basal and adluminal compartments described (Dym & Fawcett, 1970)
1970 Sertoli cell toxicant described (Kierszenbaum, 1970)
1970 Sertoli cell secretion of fluid (Setchell, 1970; Setchell et al., 1978; Wilson & Griswold, 1979)
1972 Transillumination allowed first biochemical studies of the cyclic activities (Parvinen & Vanha-Perttula, 1972)
1975 First detailed review of Sertoli cell ultrastructure (Fawcett, 1975)
1975 FSH regulation of Sertoli cells (Tung et al., 1975; Fritz et al., 1976; Means et al., 1976; Griswold, 1993a)
1975 Sertoli cell production of androgen-binding protein (Sanborn et al., 1975; Tindall et al., 1975; Fritz et al., 1976)
1976 Tubulobulbar complex described (Russell & Clermont, 1976)
1977 Sertoli ectoplasmic specialization junctions named (Russell, 1977)
1979 Golgi of Sertoli cell in 3-D (Rambourg et al., 1979)
1979 Sertoli cell production of inhibin (Sertoli cell factor) (Chowdhury et al., 1978; Labrie et al., 1978; Demoulin et al., 1979; Steinberger, 1979)
1980 Sertoli cell volume is enormous (Roosen-Runge, 1955; Cavicchia & Dym, 1977; Weber et al., 1983; Wong & Russell, 1983; Russell et al., 1986, 1990b; Kerr, 1988a,b; Sinha Hikim et al., 1989)
1980 Sertoli cell production of transferrin (Skinner & Griswold, 1980)
1981 Sertoli cell production of proteins is Stage specific (Lacroix et al., 1981; Parvinen, 1982; Ritzen et al., 1982; Mather et al., 1983; Wright et al., 1983)
1982 Sertoli cell proliferation peaks just before birth and ceases at puberty (Orth, 1982)
1983 Three-dimensional reconstruction of the Sertoli cell (Russell et al., 1983; Weber et al., 1983; Wong & Russell, 1983)
1983 Sertoli cell numbers may change in human and horse species (Johnson & Thompson, 1983; Johnson et al., 1984)
1983 Importance of Sertoli cell microtubules and other cytoskeletal elements in germ cell transport and attachment (Vogl et al., 1983a,b, 1993, 2008; Vogl & Soucy, 1985; Russell et al., 1989)
1984 Sertoli cell production of anti-Mullerian hormone (Picard & Josso, 1984)
1984 Sertoli cell interaction with peritubular myoid cell (Tung et al., 1984)
1984 FSH increases Sertoli cell proliferation (Orth, 1984)
1986 Sertoli cell production of Cyclic Protein-2 (CP-2) (Wright & Luzarraga, 1986)
1987 Vitamin A deficiency and stage synchronization (Morales & Griswold, 1987)
1987 Sertoli cell production of testibumin (Cheng et al., 1987)
1988 Autoantigenic germ cells located outside blood–testis barrier suggesting Sertoli cells secrete immunoregulatory factors (Yule et al., 1988)
1989 Sertoli cell production of growth factors (Skinner et al., 1989)
1990 Sertoli cell production of a2-macroglobulin (Cheng et al., 1990)
1990 Discovery of SRY (Koopman et al., 1990)
1991 Sertoli cell expresses Wilms’ tumor gene WT1 (Pelletier et al., 1991)
1993 Transplanted allogeneic Sertoli cells survive and protect islet allografts (Selawry & Cameron, 1993)
1993 Thyroid hormone contributes to terminal differentiation of Sertoli cells and testis size (Hess et al., 1993; van Haaster et al., 1993)
1994 GATA1 transcription factor in Sertoli cells (Yomogida et al., 1994)
1996 Sertoli cell production of SOX9 (Kent et al., 1996)
1998 GATA4 transcription factor in Sertoli cells (Viger et al., 1998; Ketola et al., 1999)
1998 Identification of DMRT1 (Raymond et al., 1998, 1999)
2000 Sertoli cell production of GDNF regulates spermatogonial stem cells (Meng et al., 2000)
2004 Sertoli cell based gene therapy is proposed (Dufour et al., 2004)
2006 Wt1 is required for Sertoli cell expression of Sox9 (Gao et al., 2006)
2008 Establishment of the SRY/SOX9 axis (Sekido & Lovell-Badge, 2008)
2009 Sertoli cell junctional complex internalization hypothesis (Young et al., 2009, 2012; Du et al., 2013; Vogl et al., 2013, 2014; Lyon et al., 2015)
2012 Movement of germ cell syncytium across Sertoli cell tight junction (Smith & Braun, 2012)
2013 INSR and IGF1R are required for FSH-mediated SC proliferation (Pitetti et al., 2013)
2015 Retinoic acid initiation of spermatogenesis and the cycle (Griswold, 2015a)

Table 2.

Major reviews of the Sertoli cell

Sertoli Cell Booksa
1993 The Sertoli Cell (Russell & Griswold, 1993b)
2005 Sertoli Cell Biology I (Skinner & Griswold, 2005)
2015 Sertoli Cell Biology II (Griswold, 2015b)
Reviews of the Sertoli Cell
1865 Enrico Sertoli published the ‘cellule ramificate’ (Sertoli, 1865)
1865–1965 Sertoli cell named (Ebner, 1888)
Sertoli cell morphology (Regaud, 1899; Heller et al., 1948)
Sertoli cell development (Walker & Embleton, 1906; Montgomery, 1911)
Sertoli cell and the cycle (Elftman, 1963)
1966–1980 Sertoli cell ultrastructure (Fawcett, 1975)
Blood–testis barrier (Setchell, 1974; Setchell & Main, 1975)
Sertoli–germ cell interactions; ectoplasmic specialization (Russell, 1980; Russell et al., 1980)
Sertoli cell and FSH (Means et al., 1976; Means et al., 1980)
1981–1990 Sertoli cell morphology; cell junctions; cytoskeleton; spermiation (Tindall et al., 1983; Vogl et al., 1983a; Russell, 1984; Russell & Peterson, 1985; Clermont et al., 1987; Russell et al., 1987; Kerr, 1988b; Vogl, 1989)
Sertoli cell physiology; hormonal control (Burger & de Kretser, 1981; Ritzén et al., 1981; Fritz, 1982; Mather et al., 1983; Rich & de Kretser, 1983; Tindall et al., 1985; Sanborn et al., 1987; Skinner, 1987; Bardin et al., 1988; Griswold, 1988; Griswold et al., 1988; Sharpe, 1988; Ewing & Robaire, 1989; Robaire & Bayly, 1989; Wright et al., 1989; Jegou, 1991)
Sertoli cell through the cycle (Parvinen et al., 1986; Wrobel & Schimmel, 1989; de Kretser, 1990; Ueno & Mori, 1990)
Sertoli cell development (Steinberger & Steinberger, 1987; Magrek & Jost, 1991)
Sertoli cell in vitro (Russell & Steinberger, 1989; Jegou, 1992b)
Sertoli cell pathology and toxicology (Boekelheide et al., 1989)
1991–2004 General review (Jegou, 1992a; Clermont, 1993; Jegou, 1993; Kerr, 1995; Griswold, 1998)
Sertoli Cell Biochemistry and the Cycle (Toppari et al., 1991; Morales & Clermont, 1993; Parvinen, 1993)
Sertoli cell morphology (Pelletier & Byers, 1992; Russell, 1993a; Schulze & Holstein, 1993a; Mulholland et al., 2001)
Sertoli cell cytoskeleton and germ cell translocation (Vogl et al., 1991; Vogl et al., 1993)
Blood–testis barrier (Pelletier & Byers, 1992; Grier, 1993; Lui et al., 2003; Wong & Cheng, 2005)
Sertoli-germ cell communication (Jegou, 1991, 1993; Mullaney & Skinner, 1991; Vogl et al., 1991, 2000; Byers et al., 1993a,b; Russell, 1993b,c; McGuinness & Griswold, 1994; Griswold, 1995; Cheng & Mruk, 2002; Mruk & Cheng, 2004a,b)
Sertoli cell physiology (Jegou, 1992a; Josso, 1992; Sharpe, 1992; Dorrington & Khan, 1993; Griswold, 1993a,c; Heckert & Griswold, 1993; Hinton & Setchell, 1993; Kim & Wang, 1993; Sar et al., 1993; Sharpe, 1993; Skinner, 1993b; Sylvester, 1993; Dym, 1994; Andersson, 2001; de Kretser et al., 2001; Silva et al., 2002; Walker, 2003b)
Sertoli cell secretions (Fritz et al., 1993; Griswold, 1993b; Skinner, 1993a; Sylvester, 1993; McKinnell et al., 1995)
Sertoli cell in vitro (Jegou, 1992b; Djakiew & Onoda, 1993; Steinberger & Jakubowiak, 1993; Dym, 1994)
Sertoli cell development (Gondos & Berndston, 1993; Pelliniemi et al., 1993; Orth et al., 2000; Walker, 2003a; Brehm & Steger, 2005)
Sertoli cell pathology and toxicology (Boekelheide, 1993; Schulze & Holstein, 1993b; Jegou et al., 2000; Sharpe et al., 2003; Toyama et al., 2003)
Species comparison (Bartke et al., 1993; Hinsch, 1993; Pudney, 1993; Guraya, 1995; McKinnell et al., 1995)
Sertoli cell and immune system (Dufour et al., 2003a)
2005–2015 Sertoli cell morphology (Hess & França, 2005; Kerr et al., 2006; O’Donnell et al., 2011; Vogl et al., 2013; Berruti & Paiardi, 2014; Hess & Vogl, 2015; Lyon et al., 2015)
Sertoli cell biochemistry and the Cycle (Hermo et al., 2010; Griswold, 2015a; Wright, 2015)
Sertoli cell numbers (Johnson et al., 2008)
Sertoli cell intracellular trafficking (Cheng & Mruk, 2009; Xiao et al., 2014b)
Sertoli cell, extracellular matrix, and polarity(Siu & Cheng, 2009; Wong & Cheng, 2009)
Sertoli cell, cytoskeleton, and germ cell interactions (Vogl et al., 2008; Cheng & Mruk, 2010, 2015; Cheng et al., 2010; Hermo et al., 2010; Kopera et al., 2010; Lie et al., 2010; Mruk & Cheng, 2010; O’Donnell et al., 2011; Su et al., 2013; O’Donnell & O’bryan, 2014; Qian et al., 2014; Xiao et al., 2014a)
Blood–testis barrier (Mital et al., 2011; Pelletier, 2011; Cheng & Mruk, 2012; Lie et al., 2013; Jiang et al., 2014; Li et al., 2015; Mruk & Cheng, 2015)
Sertoli cell physiology (Brehm & Steger, 2005; Walker & Cheng, 2005; Cheng & Mruk, 2010; Rato et al., 2010; Lucas et al., 2011; Smith & Walker, 2014; Smith et al., 2015)
Sertoli cell and transcriptional regulation (Griswold & McLean, 2005; Lui & Cheng, 2008, 2012; Cheng et al., 2010; Fok et al., 2014; Cheng & Mruk, 2015; Chojnacka & Mruk, 2015; Heckert & Agbor, 2015; Hogarth, 2015; Wright, 2015; Yan, 2015)
Sertoli cell and microRNAs (Ramaiah & Wilkinson, 2015)
Sertoli cell pathology and toxicology (Wong & Cheng, 2011; Brunocilla et al., 2012; Johnson, 2014; Murphy & Richburg, 2014; Gao et al., 2015; Reis et al., 2015)
Sertoli cell and spermatogonial stem cells (Oatley & Brinster, 2012; Garcia & Hofmann, 2013; Hai et al., 2014; de Rooij, 2015)
Sertoli cells and immune system (Mital et al., 2010; Franca et al., 2012; Kaur et al., 2012, 2014a, 2015)
Sertoli cells and cancer (Oliveira et al., 2015)
Sertoli cell and meiosis (Griswold, 2016)
Sertoli cell and development (Gassei & Schlatt, 2007; Sekido & Lovell-Badge, 2008; Barrionuevo et al., 2011; Jakob & Lovell-Badge, 2011; Nicholls et al., 2012; Tarulli et al., 2012; Escott et al., 2014; Dong et al., 2015; Loveland & Hedger, 2015; Yang & Oatley, 2015; Yao et al., 2015; Young et al., 2015)
Species comparison (Schulz et al., 2010; França et al., 2015)
a

See Table S1 for complete listing.

MORPHOLOGY OF THE SERTOLI CELL

Morphological studies of the Sertoli cell have gone through three phases of investigation (Fig. 2), beginning with routine light microscopy (LM), which lasted until about 1960. The next phase began after the invention of the electron microscope (TEM), which was used to record higher resolution images of Sertoli cell organelles and membranes, and lasted until about 2000. The final phase has been immunohistochemistry and immunofluorescence (Hess & Vogl, 2015), which began prior to 2000, but has gradually become the major tool for localizing specific proteins in the testis (Hogarth & Griswold, 2013) and three-dimensional imaging of Sertoli–germ cell interactions (Fig. 3).

Figure 2.

Figure 2

Three phases observed in morphological studies of the Sertoli cell. (A) Light microscopy (LM). The image is from mouse seminiferous epithelium, Stage IV (Periodic acid-Schiff’s stain). The Sertoli cell nucleus is euchromatic with a large nucleolus and a single satellite chromocenter. An intimate association of germ cells with the Sertoli cell is displayed with pachytene spermatocytes adjacent to the Sertoli cell cytoplasm and heads of elongated spermatids that are pulled deep into the Sertoli cell crypts and lying next to the apical region of its nucleus. Round spermatids are found near the lumen. Bar = 12 lm. (B) Transmission electron microscopy (TEM). The tissue is from human testis, showing the Sertoli cell resting on the basement membrane and surrounded by germ cells. Spermatogonia (Spg); Spermatocyte (Spc). The Sertoli cell has a highly euchromatic nucleus (SCN), a large nucleolus (Nu) with two satellite chromocenters (Cc) and an indentation of the nuclear membrane (In). Plasmalemma (Pl); Ectoplasmic specialization (Eps) at the blood–testis barrier. Bar = 5 μm. (C) Immunofluorescence microscopy. The Sertoli cell was isolated in vitro with attached elongated spermatids and labeled for somatic cell-specific tubulin (green) and filamentous actin (red). Actin is labeled at the ectoplasmic specialization and the intercellular bridges. The microtubules are continuous with the apical regions and around the basal area of the nucleus and directly adjacent to the ectoplasmic specializations, attached to the spermatid heads that are drawn deep into the Sertoli cell crypts. Bar = 10 μm. Original illustration provided by Dr. A. Wayne Vogl, Department of Cellular and Physiological Sciences, Faculty of Medicine, University of British Columbia, Vancouver, Canada (Vogl et al., 1995). Modified and reprinted with permission of the Copyright © holder Elsevier.

Figure 3.

Figure 3

Schematic illustration of the Sertoli cell’s interaction with germ cells at different stages during spermatogenesis and other key functions, including: (1) transport of micronutrients across the junctional complex; (2) management of waste and recycled leftover cytoplasm during germ cell development; (3) maintenance of the blood–testis barrier (BTB); (4) establishment of germ cell adhesions and communication; (5) inhibition of immune reactions and maintenance of immune privilege; (6) initiation and response to endocrine signaling pathways; (7) initiation and regulation of the cycle of the seminiferous epithelium; and (8) maintenance of stem cell homeostasis. Most autoimmunogenic germ cells are sequestered within the adluminal compartment of the seminiferous epithelium behind the BTB, where Sertoli cells surround them. Sertoli cells secrete immunoregulatory factors (5) that modify the immune response and induce regulatory immune cells such as macrophages (M2) and T cells (Tregs). (A) Actin filaments (green) are seen along the basal Sertoli/Sertoli tight junctions but also lining the heads of elongated spermatids; (B) Claudin-11 (red) stains only the basal junctional complex; (C) Actin (green), Rab5 (red) and DAPI (blue for nucleus) show the intricate relationship of these proteins to the tubulobulbar complex; (D) Androgen receptor (brown) stains only the Sertoli cell nucleus in the hamster seminiferous epithelium. Original illustration provided by Dr. Wayne Vogl (Hess & Vogl, 2015). Modified with approval of the Copyright © holder for Sertoli Cell Biology, 2nd edition, Elsevier Academic Press.

Sertoli cell morphology represents one of the most complex, three-dimensional structures in cell biology and yet Enrico Sertoli made his historical observations without the benefit of a good fixative, thin sections of embedded testicular tissues and histological stains that are now common laboratory tools. Nevertheless, he was able to describe unique branches of the cell’s cytoplasm that supported germ cell development and the nucleus with a large nucleolus (an important morphological feature that is used for cellular recognition today). Sertoli suggested that these cells were individual, but others claimed that they were a syncytium (Ebner, 1888), which became a standing controversy that was not settled until 1955, when electron microscopy was able to reveal cellular membranes and junctional complexes (Burgos & Fawcett, 1955; Zebrun & Mollenhauer, 1960; Fawcett, 1975). Numerous reviews of Sertoli cell morphology (Table 2) have provided unique insights into the cell’s interactions within the seminiferous epithelium (Figure S1), particularly focusing on the following structures: shape of the nucleus, the thin cytoplasmic arms, the intricate physical association with germ cells, the spermatid disengagement complex and the changes observed in these features over the course of the cycle of the seminiferous epithelium (Heller et al., 1948; Elftman, 1963; Flickinger & Fawcett, 1967; Fawcett, 1975; Russell, 1980, 1984, 1993a,b; Wright et al., 1983; Kerr, 1988b; Russell et al., 1990a; Ueno & Mori, 1990; Morales & Clermont, 1993; Mruk & Cheng, 2004b, 2010; Hess & França, 2005; Wong & Cheng, 2005; Vogl et al., 2008; O’Donnell et al., 2011; Xiao et al., 2014a; Hess & Vogl, 2015). These morphological features have contributed significantly to the overall beauty of this ‘cellule madri’ or ‘mother cell.’

The Sertoli cell nucleus is one of its most distinguishable organelles (Hess & França, 2005; Hess & Vogl, 2015). It is large, euchromatic (Fig. 2), and capable of changing shape throughout the cycle of the seminiferous epithelium, often exhibiting deep invaginations (Figure S2) of the nuclear membrane that is surrounded by vimentin intermediate filaments. The nucleolus is very large and stains intensely (Schulze et al., 1976), with three distinct parts (tripartite), with most nucleoli have two satellite chromocenters in rodent species, although three satellite structures are found occasionally in a small percentage of mice (Kushida et al., 1993; Guttenbach et al., 1996) and rat Sertoli cells (Hess & Vogl, 2015). In some species, the satellite chromo-centers form donut shapes, but these structures often are out of the plane of section. The nucleus is usually described as residing near the basement membrane (Russell et al., 1990a); however, in some species the nucleus can be located higher in the epithelium near the lumen, as is common in stages surrounding spermiation in rodents. When staining for Sertoli cell nuclear proteins, the more apical nuclei are easily recognized, as seen with the androgen receptor (Fig. 3). However, when a nuclear protein is present in both Sertoli and spermatogonial germ cells, such as the E2f family of transcription factors (El-Darwish et al., 2006), care must be taken, as stages immediately following spermiation have fewer spermatogonia and recognition of the Sertoli cells may require an evaluation of nuclear shape as well as the presence of a large nucleolus.

Major immunohistochemical markers for the Sertoli cell nucleus that are commonly used for morphology include the following: androgen receptor (AR) (Sar et al., 1990); SOX9 (SRY-box containing gene 9) (Frojdman et al., 2000); Wilms tumor protein-1 (WT1) (Sharpe et al., 2003; Wang et al., 2013); GATA-binding protein 1 (GATA1) (Chen et al., 2005); GATA-binding protein 4 (GATA4) (McClusky et al., 2009); and cyclin-dependent kinase inhibitor 1B (p27kip1) (Sharpe et al., 2003). Age-specific expression of these markers is an important consideration (Hess & Vogl, 2015), as SOX9 is strong prenatal but decreases dramatically post birth (Frojdman et al., 2000), whereas WT1, is present in the Sertoli cell nucleus throughout all developmental ages and AR shows increasing expression after the onset of puberty (Sharpe et al., 2003). GATA4 is expressed throughout development (Kyronlahti et al., 2011) and does not vary with the cycle of the seminiferous epithelium in the adult. In addition, GATA4 is not inhibited by the presence of germ cells, which is a problem with GATA1 expression (Yomogida et al., 1994).

Deep indentations or clefts of the nuclear envelope (Dym, 1973), which are signs of Sertoli cell maturation, are absent during development and often in tubules following the destruction of the germ cells and in patients with impaired fertility (Schulze et al., 1976; Hess & França, 2005). The clefts are difficult to observe in light microscopy without ultrathin sections (Hess & França, 2005) and are rarely observed with immunohistochemical staining. Little is known regarding the function of these nuclear modifications but they may provide a unique site for nuclear targeting of specific proteins (Rothbarth et al., 2001). Other unique aspects of the Sertoli cell nucleus include a high density of nuclear pores that varies depending on the stage of spermatogenesis (Cavicchia et al., 1998) and heavy vesiculation of the nucleoplasm in some species (Pawar & Wrobel, 1991).

The long cytoplasmic arms (branches as described by Sertoli) wrap the germ cells with very thin structures having widths often less than 50 nm (Hess & Vogl, 2015). These thin processes, which form cup-like areas to hold and nurture the germ cells through their differentiation (Russell, 1993a), are best illustrated by a plastic three-dimensional model built by Lonnie Russell (Russell, 1993a) and based upon the electron microscopic serial sections of a single Sertoli cell (Fig. 4). Surface area of the Sertoli cell plasma membrane is increased dramatically by the extension of these arms, up to 16,000 μm2, showing tremendous stage-dependent variation that involves the translocation of numerous organelles, the expression of hundreds of different classes of proteins for specific functions, and requiring the transport of these proteins to specific regional positions throughout the cycle of the seminiferous epithelium (Parvinen, 1982, 1993; Ritzen et al., 1982; Mather et al., 1983; Wright et al., 1983, 1989; Parvinen et al., 1986; Kaipia et al., 1991; Toppari et al., 1991; Johnston et al., 2008; Hess & Vogl, 2015; Wright, 2015). Without an understanding of such complex form, it would have been impossible to comprehend the numerous functional interactions that depend on the Sertoli cell plasmalemma, such as tight junctional complexes that comprise the blood–testis barrier, as well as sperm disengagement (spermiation) and phagocytosis of the residual body of leftover spermatid cytoplasm (Vogl et al., 2013; Hess & Vogl, 2015; Lyon et al., 2015).

Figure 4.

Figure 4

A three-dimensional drawing of a Stage V rat Sertoli cell taken from a photograph of a plastic model created from 675 micrographs of 372 serial electron microscopic sections. Cellular processes and cup-like hollows show the intimate relationship with adjacent germ cells (Wong & Russell, 1983). Reprinted with approval of the Copyright © holder John Wiley & Sons, Inc.

The intricate physical association that Sertoli cells have with germ cells begins first with the Sertoli–Sertoli tight junction (ScTj), which contributes to the blood–testis barrier (Fig. 5). The barrier is considered to be very tight (Pelletier, 2011), but differences have been found between in vivo and in vitro conditions, which were thought to be because of the peritubular myoid and germ cells contributing to an increase in transepithelial resistance (Mruk & Cheng, 2015). One of the most surprising morphological activities of the seminiferous epithelium is the passage of preleptotene and leptotene spermatocytes through the ScTj, performing a complex transit from the basal to adluminal compartment (Smith & Braun, 2012). Passage of hundreds of syncytial germ cells occurs without disrupting this important barrier, requiring a very unique coordination, similar to that of canal locks opening and closing around a ship. However, the germ cells remain connected via cytoplasmic bridges that pass through the tricellular junctions formed by three adjacent Sertoli cells (Smith & Braun, 2012). The regulation of ScTj proteins and structure is multifaceted, involving hormones such as androgens and FSH, cytokines (i.e., TNFα and TGFβ), the presence of germ cells, actin nucleating protein N-WASP (neuronal Wiskott-Aldrich syndrome protein), and phosphorylation of key proteins such as the claudins (Mruk & Cheng, 2015). In addition, a basal tubulobulbar complex has been identified morphologically as a potential component of the assembly and disassembly that is required for passage of the early spermatocytes through the ScTj (Du et al., 2013; Lyon et al., 2015). The current hypothesis is that this activity provides a remodeling of the intercellular junctions and disengagement of junctional molecules in the plasma membrane, followed by endocytosis and intracellular trafficking (Du et al., 2013; Lyon et al., 2015; Mruk, 2015).

Figure 5.

Figure 5

Electron microscopy of adjacent Sertoli cells showing the tight junctional complex (Sertoli–Sertoli tight junction) and associated basal ectoplasmic specialization. The Sertoli cell plasmalemma is seen between the two cells, which sit on the basal lamina and a peritubular myoid cell. One nucleolar chromocenters is noted in the large, euchromatic nucleus.

Lastly, another unique structural phenomenon of the physical association of Sertoli and germ cells is the positioning and transport of the elongating spermatids within the seminiferous epithelium throughout the cycle (Fig. 3). Early steps of elongated spermatids are attached in the more apical crypts (Figs 2 & 4) of the Sertoli cell (Meistrich & Hess, 2013) and lengthen perpendicular to the basement membrane. As the spermatids elongate, the germ cells are transported into deep indentations of the Sertoli cell, with their heads nearly touching the Sertoli cell nucleus (Hess, 1990). Finally, the Sertoli cell transports the late step spermatids toward the lumen where fully developed spermatozoa are released during spermiation. This dynamic mobilization of elongated spermatids is orchestrated by the Sertoli cell through the use of parallel microtubule tracts and motor proteins attached to the endoplasmic reticulum component of the ectoplasmic specialization (Vogl, 1988; Vogl et al., 1991; Beach & Vogl, 1999).

Sertoli–Sertoli cell and Sertoli–germ cell attachments are some of the most elegant and dynamic structures observed with electron microscopy. Soon after the electron microscope became popular, it was recognized that the ScTj was unique (Brökelmann, 1963; Flickinger & Fawcett, 1967; Nicander, 1967), first being called ‘junctional specialization’ by Flickinger and Fawcett. Ten years later, Lonnie Russell coined the term ‘ectoplasmic specialization’ (ES), linking the ScTj with a narrow band of actin filaments and endoplasmic reticulum on both sides of the adjoining cell membranes (Russell, 1977). This exclusive structure displayed its plasticity by also appearing as a Sertoli cell component of the Sertoli-spermatid germ cell junctional attachment site, forming apical ectoplasmic specializations facing the germ cell at the edge of the very narrow cytoplasmic arms that surrounded forming spermatid heads (Fig. 6) (Xiao et al., 2013; Gungor-Ordueri et al., 2014; Hess & Vogl, 2015; Li et al., 2015; Mruk & Cheng, 2015). The basal ES is part of the blood–testis barrier, along with the proteins and structures associated with the junctional complex (tight, gap and desmosome). Actin filament proteins are probably the most abundant and easily visualized component of the ES (Fig. 3) with an abundance of binding, adaptor, and linking proteins that coordinate this unique plasma membrane structure, whereas vimentin filaments attach to the desmosomes (Mruk & Cheng, 2015).

Figure 6.

Figure 6

Electron microscopy showing the thin arm of a Sertoli cell (light green area) containing the ectoplasmic specialization (Eps) and Sertoli cell cytoplasm (Sc) adjacent to the germ cell cytoplasm showing the manchette microtubules of spermatid 2 and the acrosome (Ac) that covers the nucleus (N) of spermatid 1.

Another fascinating structure that participates in the Sertoli–germ cell physical juncture is the tubulobulbar complex, first described by Lonnie Russell (Russell & Clermont, 1976) as an anchoring device. Tubulobulbar complexes are elongated tubular extensions of one cell into corresponding invaginations of the adjacent cell plasma membrane, terminating with a bulb that is associated with cisternae of endoplasmic reticulum (Vogl et al., 2013). These intimate structures are located at both the basal ES between adjacent Sertoli cells (as discussed with the ScTj above) and at the apical tips of Sertoli cell cytoplasm, within the concave area of the heads of late spermatids (Fig. 3; Figure S1). More recent discoveries have supported an important role for these complexes in the disengagement of mature spermatids and removal of excess germ cell cytoplasm during spermiation, as well as the recycling of junctional molecules at both locations (Young et al., 2009; Du et al., 2013; Vogl et al., 2013, 2014; Lyon et al., 2015).

The uniqueness of these specialized organelles, which support Sertoli–germ cell interactions and transport within the seminiferous epithelium, has been clearly observed morphologically over the past 50 years. However, it is the amalgamation of molecular and biochemical data with histology through immunolocalization (Fig. 3) and other imaging technologies that is now providing new explanations for the complex physiology of the Sertoli cell, which is required for the maintenance of continuous sperm production throughout life in the adult male (Vogl et al., 2013, 2014; Xiao et al., 2014a; Hess & Vogl, 2015; Lyon et al., 2015).

MOLECULAR BIOLOGY AND MECHANISMS

The original notion of the Sertoli cell as a ‘nurse cell’ was a direct result of its morphological relationships with the developing germ cells. While the morphology has been elegantly reported, even after 150 years of research with new and better technological tools the Sertoli cell has retained many of its molecular secrets. The ability to independently maintain primary Sertoli cells from rodents in relatively pure cell culture led to the first molecular studies in the 1970s and 1980s (Dorrington & Armstrong, 1975; Steinberger, 1975). This approach and the availability of specific antibodies and improvements in microscopy have revealed several gene products that are unique to the Sertoli cell.

Some of the first known products of Sertoli cells included metal transport proteins such as transferrin and ceruloplasmin (Skinner & Griswold, 1980; Skinner & Griswold, 1982; Skinner & Griswold, 1983). It was proposed that these products represented the true ‘nurse cell’ function (Fig. 5) by providing mechanisms to transport micronutrients across the blood–testis barrier to support germ cell development (Sylvester & Griswold, 1994). A model was proposed where Sertoli cells at the basal surface can take up iron bound to serum transferrin, transfer it to a newly synthesized transferrin molecule and secrete it on the apical side of the blood–testis barrier to be used by the developing germ cells. This model provided a mechanism for moving iron (Fe+3) across the blood–testis barrier for use primarily in meiotic and mitotic cell divisions in spermatogonia and spermatocytes and mitochondriogenesis in spermatids. Iron in this form is required for many cellular functions but because of its very low solubility it must be transported bound to specific transport proteins such as transferrin. Recently, with the current knowledge about additional components of the iron transport pathway and the availability of antibodies this model has been expanded (Leichtmann-Bardoogo et al., 2012). The localization and regulation of a number of proteins involved in iron transport, storage, and export were examined in mouse testes and it was determined that there is an autonomous internal iron cycle within the seminiferous tubules. The cycle consists of primary spermatocytes loading with iron from the Sertoli cells, maintaining those iron stores to support mitosis, meiosis, and mitochondriogenesis through the elongated spermatid stage and then returning the bulk of the iron to the Sertoli cells in the ingested residual bodies. The Sertoli cells then recycle the ingested iron back to the primary spermatocytes. In this model, the Sertoli cells function as ‘nurse cells’ by providing the iron required for germ cell development but also remove and recycle the potential toxic accumulation of iron in the residual bodies. The ‘nurse cell’ function in this case is equivalent to emptying the bedpans! There are likely several such mechanisms that are part of the nurse function(s) of Sertoli cells and they have been referred to as the ‘recycling and waste management’ functions (Yan et al., 2008; Leichtmann-Bardoogo et al., 2012; Young et al., 2012; Vogl et al., 2014; Yan, 2015).

The products of Sertoli cells can inhibit immune reactions (Dufour et al., 2005; Doyle et al., 2012) and provide structural features (Russell et al., 1983; Russell, 1993a) under a broad classification of ‘bioprotective and structural’ functions. Adjacent Sertoli cells synthesize the components of the tight junction complexes (Figs 3 & 5) and contribute to the basement membrane and participate in the formation of desmosomes, gap junctions and some unique forms of junctions with germ cells. The nature of spermatogenesis results in the production of enormous numbers of gametes and therefore these structural elements must be extremely dynamic and involve the spatiotemporal expression of many genes.

Sertoli cells also play key roles in signaling in the testis by serving as the targets for FSH and testosterone and by transducing those endocrine signals and other cellular cues into paracrine regulation of germ cells (Griswold, 1993a; Johnston et al., 2001). Again, both the response of gene expression to FSH and testosterone and the expression of growth factors or other signaling molecular vary with testis development and with the stage of the cycle of the seminiferous epithelium revealing the requirement for a complex and carefully controlled gene network. An example of paracrine signaling can be found in the initiation of spermatogenesis in the mouse testis. Shortly after birth the A undifferentiated spermatogonia undergo the transition to A1 differentiating spermatogonia. This transition results in a carefully timed commitment of those cells to meiosis and is absolutely dependent on retinoic acid synthesized by the Sertoli cells. In the mouse this transition occurs in patches along the tubule that lead to asynchronous entry into meiosis and the spermatogenic wave (Hogarth & Griswold, 2010; Snyder et al., 2010).

Gene expression that results in the formation of the testis in the embryo occurs within the Sertoli cells. Genes encoding transcription factors such as Sry and Sox9 expressed in the Sertoli cells alter the transcriptome such that germ cells are enclosed in seminiferous tubules resulting in the formation of the testis (for a review see DiNapoli & Capel, 2008; Svingen & Koopman, 2013). In addition, Cyp26B1 expressed in Sertoli cells breaks down any retinoic acid and prevents the germ cells from entering the pathway to meiosis (Svingen & Koopman, 2013). Recently it has been shown that loss of the DMRT1 transcription factor in mouse Sertoli cells, even in adults, activates another transcription factor, Foxl2, and reprograms Sertoli cells into granulosa cells. These studies have shown that Dmrt1 in Sertoli cells is essential for sustained mammalian testis determination (Matson et al., 2011). So, a dynamic transcriptome with precise spatiotemporal expression of genes in the Sertoli cells begins early in the embryo, has a unique program with the onset of spermatogenesis and then varies with the cycle of the seminiferous epithelium throughout the reproductive lifetime of the organism.

The production of spermatozoa by in vitro differentiation of germ cells has been reported for several species. If these reports are accepted at face value it means that the male germ cells have an autonomous program leading to sperm formation (de Winter et al., 1993; Kerkis et al., 2007; Aflatoonian et al., 2009; Sato et al., 2011b). However, even the most successful of the protocols in these reports are extremely inefficient at sperm production when compared with spermatogenesis in the testis. In 1993 and 1994, it was suggested in two separate reviews that Sertoli cells might be primarily ‘permissive’ in nature (Russell & Griswold, 1993a; Sharpe, 1994). This view would suggest that Sertoli cells provide the environment, the signaling and the structure to allow the efficient autonomous differentiation of male germ cells into spermatozoa.

Recently the use of the RiboTag mouse that allows the genetic tagging of polysomes in Sertoli cells in vivo has resulted in a relatively comprehensive description of gene expression in the postnatal Sertoli cells (Sanz et al., 2013; De Gendt et al., 2014; Evans et al., 2014). These approaches have resulted in lists of genes that are overexpressed in Sertoli cells relative to the other cell types in the testis. Results from one study showed that the genes most overexpressed in postnatal and adult Sertoli cells involved glutathione metabolism, cytochrome P450 enzymes, drug metabolism pathways, peptidase, and enzyme inhibitor pathways (De Gendt et al., 2014). Expression of these types of genes is consistent with a role in ‘recycling and waste management’. Information on which genes are expressed in Sertoli cells can lead to a couple of outcomes. If the product of the gene is a protein with a known function then the role of that protein in the function of Sertoli cells may become apparent. However, it is often the case that the gene product has an unknown function or the known function is not easily reconciled with the presumed role of Sertoli cells in spermatogenesis. Many investigators have attempted to list the various functions of Sertoli cells and generally these lists imply that Sertoli cells participate in nearly all aspects of spermatogenesis (Griswold, 1988, 1995; Griswold et al., 1988; Sharpe, 1994). In addition, the many roles of the Sertoli cells vary during development and across the cycle of the seminiferous epithelium in the adult and require active and well-controlled transcriptional programs. Very few highly differentiated cell types have a lifetime requirement for this type of plasticity in gene expression.

SERTOLI CELL AND THE SPERMATOGONIAL STEM CELL NICHE

The spermatogonial stem cell niche

Adult stem cells are essential for the maintenance, repair and regeneration of many organs, and proper regulation of their fate is therefore critical to maintain tissue homeostasis. Accumulating evidence suggests that stem cell self-renewal and differentiation depend on specialized microenvironments called niches (Spradling et al., 2001; Scadden, 2006) and that in turn stem cells influence their environment (Baraniak & McDevitt, 2010; Mosher et al., 2012; Sowa et al., 2012). In the mammalian testis, spermatogonial stem cells (SSCs) reside on the basement membrane of the seminiferous tubules and are in intimate contact with the Sertoli cells. The SSC niche is made of a complex interplay of growth factors provided by Sertoli and interstitial cells, the basement membrane and stimuli from the vascular network (Chiarini-Garcia et al., 2001; Chiarini-Garcia et al., 2003; Kanatsu-Shinohara et al., 2007; Oatley & Brinster, 2012). Because of its physical association with germ cells, the Sertoli cell is arguably the most important contributor of the niche, by providing paracrine and juxtacrine signals and secreting components of the basement membrane. In addition, experimental increase in the number of Sertoli cells leads to additional niches that can be colonized by SSCs after transplantation (Oatley et al., 2011). It is also evident that the niche must provide different sets of signals to the SSCs depending on the timing of testis development. For example, in the neonatal testis active SSC self-renewal takes place to establish the stem cell pool, and studies have demonstrated that neonatal niches are more efficient than adult niches to regenerate spermatogenesis from transplanted SSCs (Shinohara et al., 2001). In contrast, SSC self-renewal in the adult may only occur during certain stages of the seminiferous cycle (Johnston et al., 2011; Grasso et al., 2012).

Sertoli cell factors controlling SSC maintenance and self-renewal

A number of soluble factors produced by Sertoli cells have been recently discovered that are critical for maintenance of pro-spermatogonia in the fetus and self-renewal of SSCs after birth. The most extensively studied niche factor is glial cell line-derived neurotrophic factor (GDNF). Mutant mice with GDNF haploinsufficency have severe fertility defects and disrupted spermatogenesis (Meng et al., 2000). Similarly, ablation of GDNF and its receptors Ret and Gfra1 at the surface of SSCs result in loss of the stem cells and their progeny (Naughton et al., 2006). GDNF is used by SSCs throughout life; however, as its production decreases with age SSC numbers decline, which illustrates the fact that the stem cell pool is dependent upon GDNF for its maintenance (Ryu et al., 2006). Conversely, ubiquitous overexpression of GDNF in transgenic mice leads to overproduction of undifferentiated spermatogonia and sharp decrease in more differentiated germ cells (Meng et al., 2000). In vitro studies also demonstrated that GDNF is critical for SSC maintenance and self-renewal in short- and long-term cultures (Nagano et al., 2003; Kubota et al., 2004; Chen et al., 2005).

During mouse development, low levels of GDNF are already detectable in the bipotential gonad at embryonic day 11 (Beverdam & Koopman, 2006) and its expression increases steadily to reach a maximum at post-natal day 3 when the SSC population starts to expand (Tadokoro et al., 2002; Shima et al., 2004; Miles et al., 2012). In older animals, the levels of GDNF expression vary between the stages of the seminiferous epithelial cycle, but differences between species have been observed (Sato et al., 2011a; Caires et al., 2012a; Grasso et al., 2012). In the rat, the highest levels of GDNF mRNA was observed at stages XII to III, when undifferentiated spermatogonia proliferate (Johnston et al., 2011), and lowest at stages VII and VIII when most cells are quiescent and the majority of Aaligned spermatogonia transition to the differentiating A1–A4 cells. These observations indicate that GDNF levels are cyclic, and that its dosage is crucial for the regulation of perinatal germ cell fate and stage-specific proliferation of undifferentiated spermatogonia. Although the mechanisms responsible for this cyclic expression are not understood, it is evident that GDNF production must be modulated by positive or negative stimuli. For example, follicle-stimulating hormone (FSH) is a major positive regulator of GDNF expression by Sertoli cells in vivo and in vitro (Tadokoro et al., 2002). GDNF expression by cultured Sertoli cells is also stimulated by FGF2, TNFα, and IL-1β (Simon et al., 2007). Mechanisms that down-regulate the production of GDNF by Sertoli cells also exist. Such a role is fulfilled by NOTCH signaling as constitutive activation of this pathway in Sertoli cells results in sharp downregulation of GDNF, a complete loss of germ cells around birth and a Sertoli cell-only phenotype (Garcia et al., 2013). Conversely, ablation of RBPJ, a downstream effector of the NOTCH pathway, increases GDNF expression and results in a significant increase in SSCs and overall germ cell numbers (hyperplasia) (Garcia et al., 2014). Because the NOTCH ligand JAG1 is highly expressed in undifferentiated spermatogonia and drives the expression of downstream targets of NOTCH signaling in Sertoli cells, it is assumed that germ cells use JAG1 to activate the NOTCH pathway in Sertoli cells, therefore downregulating GDNF and controlling their own numbers (Fig. 7). This mechanism would ensure proper gem cell homeostasis and sperm output, and is in accord with the observations of Johnston and colleagues who suggested that spermatogenic cell density seem to limit GDNF production by Sertoli cells (Tadokoro et al., 2002; Ryu et al., 2006; Johnston et al., 2011).

Figure 7.

Figure 7

Model depicting the possible role of NOTCH signaling in Sertoli cells after birth. Previous studies have shown that FGF2 and FSH induce GDNF expression by Sertoli cells. Recent data suggest NOTCH signaling is a negative regulator of GDNF, which might balance the effects of FGF2 and FSH. Overactivation of NOTCH signaling suppresses the expression of GDNF and leads to sterility, whereas ablation of NOTCH signaling induces germ cell hyperplasia.

While GDNF is certainly a major component of the stem cell niche, in vitro culture experiments demonstrated that it is not the only factor needed for maintenance and long-term renewal of SSCs. Depending on the genetic background of the mice, fibroblast growth factor (FGF2) and epidermal growth factor (EGF) in addition to GDNF are critical for long-term support (Van Dissel-Emiliani et al., 1996; Kubota & Brinster, 2008; Kubota et al., 2004). Production of FGF2 by Sertoli cells has been previously demonstrated and is stimulated by FSH (Smith et al., 1989; Mullaney & Skinner, 1992). FGF2 promotes self-renewal independently of GDNF, through activation of the transcription factors ETV5 and BCL6B, but is less efficient (Ishii et al., 2012; Takashima et al., 2015).

Another Sertoli cell factor that appear to contribute to the SSCs niche is leukemia inhibitory factor (LIF) (Piquet-Pellorce et al., 2000). LIF production in Sertoli cells depends on TNFα and is widely used in cultures of primordial germ cells, prospermatogonia and SSCs from several species (Pesce et al., 1993; Nikolova et al., 1997; Kanatsu-Shinohara et al., 2003; Aponte et al., 2008). While LIF maintains SSC survival and is useful to initiate long-term cultures, it is not promoting self-renewal (Kanatsu-Shinohara et al., 2007). Another niche factor of interest is platelet-derived growth factor (PDGF). PDGF is specifically produced by Sertoli cells and induces the proliferation of prospermatogonia after birth (Loveland et al., 1995; Li et al., 1997) probably in cooperation with estrogen (Thuillier et al., 2010). Disruption of cross-talks between PDGF and estrogen-triggered signaling pathways has been suggested to take place upon exposure to xenoestrogens in the environment, which could lead to alteration of prospermatogonial behavior and preneoplastic states (Thuillier et al., 2003). Also, because WNT signaling is critical for stem cell self-renewal in a variety of tissues (Clevers et al., 2014), this pathway has been recently investigated in the testis (Golestaneh et al., 2009; Yeh et al., 2011). WNT5A is produced by Sertoli cells and promotes SSCs survival through a CTNNB1-independent mechanism that activates JNK (Yeh et al., 2011), but it does not per se induce self-renewal. Indeed, CTNNB1 ablation in germ cells leads to spermatogenesis disruption but not to SSC loss (Kerr et al., 2014; Rivas et al., 2014). Finally, although vascular endothelial growth factor A (VEGFA) family members and their receptors are all produced by germ cells, Sertoli cells, and interstitial cells (Nalbandian et al., 2003; Lu et al., 2013) only the pro-angiogenic isoform VEGFA164 promotes SSC self-renewal, as determined by the SSC transplantation assay (Caires et al., 2012b).

Niche factors controlling migration and homing

To remain in the basal part of the seminiferous epithelium, SSCs need to generate adhesion molecules that attach them to the basement membrane provided in part by Sertoli cells. The basement membrane mainly contains laminin, fibronectin and collagen IV, therefore attachment of SSCs is mediated at least in part by integrins, mainly ITGA6 and ITGB1 (Shinohara et al., 1999). However, SSCs must migrate out of the niche to differentiate, and they also move to open niches from the apical to the basal part of the seminiferous epithelium after germ cell transplantation. It has therefore been proposed that their migration depends on chemokines or other chemotactic factors. Earlier work demonstrated that Sertoli cells produce chemokines that are regulated by the transcription factor ETV5 (Chen et al., 2005) and that the chemokine CCL9, ligand of the receptor CCR1 at the surface of undifferentiated spermatogonia, is able to specifically attract these cells toward Sertoli cells (Simon et al., 2010). Another chemokine of importance is SDF1 (CXCL12) expressed in the genital ridges, which is guiding CXCR4-positive primordial germ cells toward them (Doitsidou et al., 2002; Ara et al., 2003; Molyneaux et al., 2003). In the postnatal testis SDF-1, expressed by Sertoli cells, attracts CXCR4-positive germ cells, and this activity is crucial for proper homing of SSCs after transplantation (Kanatsu-Shinohara et al., 2012; Yang et al., 2013). SDF-1 expression in Sertoli cells depends on the nuclear co-repressor Sin3a (Payne et al., 2010). However, no SDF-1 concentration gradient has been demonstrated yet in the seminiferous epithelium. In addition, because both Sertoli cells and germ cells express CXCR4 (Johnston et al., 2008; Wright, 2015), additional investigations are needed to fully understand this system.

SERTOLI CELLS AND TESTIS IMMUNE PRIVILEGE

The unique immunoregulatory status of the testis has been recognized for over two centuries; based initially on the prolonged survival of testicular tissue after transplantation into genetically disparate recipients (reviewed in Kaur et al., 2013). Sertoli cells play a central role in creation of this unique immunoregulatory environment where they provide immune protection to the developing germ cells, which if exposed to the immune system can invoke a humoral and cellular immune response. Spermatocytes and spermatids first appear after immunological self-tolerance has been established and thus express novel proteins that if detected by the immune system would result in immunologic attack. This was demonstrated by the lysis of spermatocytes and spermatids after exposure to sera collected from rodents that had been previously immunized with whole semen (O’Rand & Romrell, 1977; Tung & Fritz, 1978). The immunogenicity of the germ cells is further supported by autoimmune orchitis studies where an autoimmune reaction against the germ cells resulted in loss of spermatogenesis and infertility (Jacobo et al., 2011; Silva et al., 2014).

The blood testis or Sertoli cell barrier

Given that the autoimmunogenic germ cells described in these studies were spermatocytes and spermatids and located within the adluminal compartment of the seminiferous epithelium, historically the lack of an autoimmune response against these germ cells was attributed to their sequestration behind the blood–testis barrier (BTB) or Sertoli cell barrier (SCB). Supporting this idea, the BTB/SCB is a physical barrier formed around puberty and composed of Sertoli cell–Sertoli cell junctions and the Sertoli cell body, which surrounds the developing germ cells (Mital et al., 2011). These junctions are tight junctions (zonula occludens) comprised of occludin, claudins and junctional adhesion molecules (Mruk & Cheng, 2010). Adherens junctions, gap junctions, and desmosome-like junctions also contribute to the function of the BTB/SCB (Cheng et al., 2011). The tight junctions are located along the basal region of the Sertoli cell and separate the seminiferous epithelium into adluminal and basal compartments. This barrier separates the advanced germ cells located in the adluminal compartment from the blood supply, allowing the Sertoli cells, by expressing various transporters, to control the passage of molecules across the barrier; creating a unique microenvironment for germ cell development (Mital et al., 2011). The barrier also prevents the passage of leukocytes (immune cells) and molecules, including antibodies from crossing the seminiferous epithelium (Johnson & Setchell, 1968; Johnson, 1972; Dym & Romrell, 1975; Wang et al., 1994; Rival et al., 2006). The peritubular myoid cells form a semi-permeable barrier that also inhibits the entry of leukocytes into the seminiferous tubules (Dym & Fawcett, 1970; Fawcett et al., 1970).

The primary contributors to the BTB/SCB are occludin, claudin-3, -5, and -11, zonula occludens (ZO) 1,2 and 3 and JAM-A and -B, with occludin and claudin-11 being most important for barrier integrity. Male mice lacking claudin-11 or occludin were infertile (Gow et al., 1999; Saitou et al., 2000). However, despite the persistence of degenerating germ cells, an autoimmune reaction was not observed. In claudin-11 knockout (KO) mice degenerating germ cells are present within the lumen of the seminiferous tubules whereas spermatozoa are absent (Gow et al., 1999). Testicular autoantibodies were not detected in serum or within the adluminal compartment of the seminiferous epithelium and CD4 + T cell infiltrate was not detected within the testis. In mice treated with a mutant occludin peptide, the integrity of the BTB/SCB is disrupted and permeable to inulin, resulting in germ cell loss (Wong et al., 2007). Nevertheless, anti-sperm antibodies were not detected in serum.

Claudin-5 KO mice all died within 10 days of birth because of a defect in blood–brain barrier function (Nitta et al., 2003). Gene deletion of transcription factor ets variant 5 (ETV5) resulted in decreased claudin-5 expression in the testis and disruption of the BTB/SCB suggesting a role for claudin-5 in BTB/SCB integrity, although this has not been tested directly (Morrow et al., 2009). Claudin-3 was localized to spermatocytes and newly forming tight junctions in Sertoli cells (Smith & Braun, 2012; Chihara et al., 2013). Fifty percent reduction in claudin-3 mRNA expression after claudin-3 siRNA injection into testes of mice did not alter BTB/SCB integrity (Chihara et al., 2013) and instead delayed spermatocyte migration across barrier. Consistently, claudin-3 KO mice were fertile and had an intact barrier (Chakraborty et al., 2014).

JAMs are transmembrane proteins important for tight junction formation and cell adhesion. Deletion of JAM-A resulted in subfertility because of a defect in spermatozoa motility (Shao et al., 2008), whereas male and female mice with JAM-B gene disruption were fertile (Sakaguchi et al., 2006). Both JAM-A and –B KO mice appeared to have normal testicular morphology, however, the integrity of the BTB was not directly tested. ZOs are adaptor proteins that anchor claudins and occludin to the actin cytoskeleton. Knock-out of ZO-3 had no apparent phenotype (Xu et al., 2009), whereas knock outs of ZO-1 and -2 were embryonic lethal (Xu et al., 2009). Rescue of ZO-2 KO embryos resulted in reduced male fertility because of germ cell loss and increased permeability of the BTB to a lanthanum tracer (Xu et al., 2009). However, the effect on immune function is not known, as the immunological function was not examined in these mice.

Additional support that the BTB/SCB is involved in immunological protection of the advanced germ cells has been provided by SCARKO (Sertoli Cell Androgen Receptor Knock Out) mice, which have Sertoli cell-specific deletion of the androgen receptor (Meng et al., 2005; Meng et al., 2011). These mice have increased permeability of the BTB/SCB and evidence of an autoimmune response (Meng et al., 2005; Meng et al., 2011). Thirty minutes after injection of these mice with a biotin tracer, biotin was detected within the adluminal compartment of the seminiferous tubules. In addition, germ cell autoantibodies that recognize advanced germ cells (round and elongated spermatids) were present in the serum. An increase in the number of macrophages, neutrophils, and eosinophils within the interstitial space but not within the seminiferous tubules was also observed. SCARKO mice have decreased expression of claudin-3, which was originally attributed to these observations. However, the more recent claudin-3 KO studies did not support this conclusion and suggested that androgens in Sertoli cells are performing other additional functions related to immune regulation.

Testis immune privilege is more that the BTB/SCB

While regulating access of the immune system to germ cell autoantigens is no doubt an important aspect in controlling the immune response, immunological protection of the developing germ cells is more complex and involves Sertoli cell modulation of the immune response. As a result, the whole testis is immune privileged. Evidence that the whole testis is immune privileged is provided by several studies. For instance, when foreign tissue such as allogeneic or xenogeneic pancreatic islets, skin fragments or parathyroid grafts are transplanted into the testis, they enjoy prolonged graft survival when compared with their survival after transplantation to nonimmune privileged sites even though the cells transplanted in the testis are located in the interstitial space, outside of the BTB/SCB (Barker & Billingham, 1977; Setchell, 1990; Selawry, 1994; Mital et al., 2009).

Moreover, successful allogeneic spermatogonial stem cell transplantation, resulting in germ cell colonization of the basal compartment of the seminiferous tubules, has been performed in a wide range of species including birds, fish, goats, pigs, cattle, sheep, rodents, dogs, and cats without immunosuppression (reviewed in Kaur et al., 2013). This is consistent with an earlier study, which demonstrated that spermatogonia and preleptotene spermatocytes, located within the basal compartment of the seminiferous epithelium, were outside of the BTB/SCB, expressed auto-immunogenic antigens (Yule et al., 1988) and yet, an immune response was normally not generated against these germ cells.

In humans, fine-needle biopsies (causing local injury to the seminiferous epithelium) are performed routinely and normally do not lead to autoimmune orchitis (Mallidis & Baker, 1994). Furthermore, in seasonal breeders, during the nonbreeding cycle the BTB/SCB is disrupted. Nevertheless, meiotic spermatocytes develop normally even in the absence of a complete, impermeable BTB/SCB (Pelletier, 1986). Therefore, other mechanisms in addition to the BTB/SCB are required to create the immunologically privileged environment of the testis.

One possible downside to testis immune privilege is the potential for an increase in infections or tumors. The testis is a major site for relapse of acute lymphoblastic leukemia (ALL) (Hedger, 2015). However, overall the testis is no more susceptible to testicular tumors when compared with other tissues and infections are rare (Hedger, 2015). This has been attributed to activation of innate immunity, which could theoretically prevent infections and tumors without activating the adaptive immune response (Hedger, 2015).

Evidence for Sertoli cells in testis immune regulation

Evidence that Sertoli cells manipulate the immune response comes from transplantation studies where Sertoli cells not only survive when transplanted across immunological barriers as allografts or xenografts but also provide immune protection for co-grafted cells such as pancreatic islets to treat diabetes, adrenal chromaffin cells for neurodegenerative diseases, hepatocytes, and skin grafts (Kaur et al. 2015.). These unique immunomodulatory properties suggest that Sertoli cells are not only important for the overall protection and development of germ cells, they have therapeutic potential beyond the testis where they can protect co-grafted cells and even be engineered to express clinically relevant proteins like insulin to treat diabetes or neurotrophin-3 to treat spinal cord injury (Pelletier, 1986; Halley et al., 2010; Kaur et al., 2014b, 2015). Sertoli cells create this immune privileged environment by expressing immunoregulatory factors that actively suppress innate, humoral and cell-mediated immune responses while at the same time inducing regulatory immune cells (regulatory T cells and M2 macrophages). Sertoli cells express apoptosis inhibitors (SERPINA3N, SERPINB9), complement inhibitors (serping1, DAF or CD55, MCP or CD46, clusterin), immunomodulatory factors (IDO, galectin-1), anti-inflammatory cytokines (TGFB1), and chemokines (CCL27) that act together to modify the immune response and induce tolerance to protect the germ cells (Wang et al., 1994; Guazzone et al., 2009; Meinhardt & Hedger, 2011; Doyle et al., 2012).

Interestingly, the timing for when immune privilege first develops is not clear. The majority of Sertoli cell co-transplantation studies were performed using Sertoli cells isolated from pubertal or adult testes, indicating, as expected, that these Sertoli cells had immune protective abilities (Mital et al., 2010). The age of these Sertoli cells corresponded with the development of the autoimmunogenic germ cells. However, neonatal porcine Sertoli cells also survive when transplanted as xenografts (Dufour et al., 2003b) and testes transplanted from fetal and early postnatal (up to 15 days) rats survived better than adult testis transplants (Statter et al., 1989).

Overall Sertoli cells protect the developing auto-antigenic germ cells by forming the BTB/SCB (Fig. 3), which limits access by the immune system to the advanced germ cells, whereas at the same time modulating the immune response by secreting immunoregulatory factors that modify the immune response and induce regulatory immune cells to create a local tolerogenic environment and along with the peritubular myoid cells restricting the immune cells to the interstitial space. It is amazing that 150 years ago, when Enrico Sertoli first described Sertoli cells, he was able to intuitively suggest, based on Sertoli cell morphology, the importance of Sertoli cells in the protection of the germ cells.

COMPARATIVE SERTOLI CELL BIOLOGY

In metazoans, spermatogenesis relies on the somatic cells environment for its completion. Therefore, in order to guarantee fertility during the animal reproductive lifetime, the somatic cells named Sertoli cells in vertebrates are crucial for facilitating germ cells survival and development in such a manner that spermatozoa is usually produced in very high numbers. However, Sertoli cell structure and function in anamniotes (fish and amphibians), which present a cystic type arrangement of gametogenesis (Schulz et al., 2010), shows several important differences when compared with amniotes (mammals, birds, and reptiles) (Fig 8). These particularities may provide new insights into Sertoli cell physiology and will be addressed in this section.

Figure 8.

Figure 8

Schematic representation of the main differences between Sertoli cells (SC in the legend) in cystic and non-cystic spermatogenesis. Adiff, type A differentiated spermatogonia; Aund, type A undifferentiated spermatogonia; B, type B spermatogonia; BL, basal lamina; BV, blood vessel; EST, elongated spermatid; LE or LC, Leydig cells; MY, peritubular myoid cells; RST, round spermatid; SC, spermatocytes; SE, Sertoli cell; SG, spermatogonia. Modified from previous publications with permission of the Copyright © holder of Sertoli Cell Biology, 2nd edition, Elsevier Academic Press (Schulz et al., 2010; Francça et al., 2015).

A very important particularity is that, different from amniotes, Sertoli cells remain mitotically active in fish and amphibians after they become sexually mature and two modes of proliferation that could overlap each other are observed in fish (França et al., 2015) (Suppl. Fig. 3). In the first mode, Sertoli cell progenitors – that are regulated by FSH, thyroid hormone, estrogens, and insulin-like growth factor – proliferate to provide space for new niches that will be occupied by SSCs or single spermatogonia, forming therefore new spermatogenic cysts (Morais et al., 2013; França et al., 2015). In this instance, similar to rodents (Dovere et al., 2013), paracrine factors (i.e., GDNF) produced by Sertoli cells in fish may stimulate SSC self-renewal divisions or attract SSCs from other areas (Lacerda et al., 2013; Nakajima et al., 2014). However, in contrast to rodents (Cooke et al., 2005), thyroid hormones (thriiodothyronine-T3) in zebrafish increase the mitotic activity of Sertoli cells via Igf signaling system (igf3 gene). This is particularly true for Sertoli cells not yet associated with germ cells or in contact with type A spermatogonia. Therefore, Igf also stimulates the proliferation of undifferentiated spermatogonia in a sex steroid independent manner (Morais et al., 2013).

The second mode of Sertoli cell proliferation is under the regulation of FSH, androgens, and progestins. In this mode, Sertoli cells within the existing cysts divide to accommodate the expanding germ cell clones, according to the respective reproductive strategy and distribution of spermatogonial cells in the testis parenchyma of each species (Billard & Breton, 1978; Almeida et al., 2008; França et al., 2015). Although solid scientific evidence is still lacking for this mode, the existence of a Sertoli progenitor or stem cell population seems quite plausible and deserves careful investigation based upon the following observations: the long-term capacity of Sertoli cell division in successive reproductive cycles, the fully functional sex reversal in adults (Shibata & Hamaguchi, 1988; Kobayashi et al., 2009), and the natural sexual plasticity observed in sequentially hermaphroditic fish species (Kobayashi & Nagahama, 2009).

It seems that in anamniotes, Sertoli cells enveloping a germ cell cyst are only terminally differentiated after meiosis is complete, because this functional status correlates with the formation of tight junctions between Sertoli cells (Leal et al., 2009; França et al., 2015). Therefore, considering their proliferating activity and the establishment of tight junctions, Sertoli cells seem to behave similarly throughout vertebrates. In this regard, evaluation of an individual spermatogenic cyst in anamniotes will reveal that the number of Sertoli cells increases steadily during the mitotic phase, stabilizing upon completion of meiosis/start of spermiogenesis (Matta et al., 2002; Schulz et al., 2005; Leal et al., 2009).

Compared with mammals, the number of spermatogonial mitotic cycles in anamniotes is usually quite high, whereas much lower numbers of apoptotic germ cells (30–40% loss from the theoretically expected number) are observed in spermatogenic cysts (Vilela et al., 2003; Leal et al., 2009; França et al., 2015). Therefore, hundreds of more advanced germ cells (meiotic and post meiotic) are usually present in a cyst in association with low number of Sertoli cells. It means that, despite having little or no direct contact (junctions) with germ cells, Sertoli cells efficiency in lower vertebrate is quite high. Although this issue is very complex and deserves further evolutionary investigation, reproductive efficiency is clearly related to the number of gametes required for a particular mode of reproduction. It is at great cost to the organism that gametes are produced so it is likely that evolution carefully monitors the efficiency so that sufficient numbers of gametes are produced to ensure the continuation of the species while increasing the efficiency of fertilization and survival of the offspring. In particular, the number of spermatids per Sertoli cell, which is considered species-specific, varies greatly during vertebrate evolution and decreases strikingly from more than one-hundred in fish to less than ten in most mammalian species already investigated, reaching about four in humans (Assis et al., 2015; França et al., 2015) (Figure S4). This quite illustrative figure allows us to speculate that perhaps humans will not produce sperm in the future. As anamniote Sertoli cells present very high support capacity for germ cells, a careful and comprehensive investigation on these somatic cells may provide important clues regarding their regulatory mechanisms during evolution. An important aspect that deserves consideration is the fact that in the vast majority of fish species spermatozoa has no acrosome, requiring therefore a very high number of gametes for reproduction through external fertilization.

In fish both Sertoli and Leydig cells express receptors for FSH and LH that directly stimulate steroidogenesis. It is worth mentioning that sharks do not have steroidogenic Leydig cells in the interstitial compartment. Therefore, unlike higher vertebrates, in addition to regulating Sertoli cells activities and proliferation (Schulz et al., 2012), Fsh in fish is also a potent steroidogenic hormone (Prat et al., 1996; Campbell et al., 2003; França et al., 2015) and is associated with spermatogonial proliferation and differentiation (Skaar et al., 2011; Assis et al., 2015; Melo et al., 2015; Nóbrega et al., 2015).

New evidence from electron microscopy studies (França et al., 2015) has shown that Sertoli cells seem to be in contact with different type of germ cells clones in different phases of cystic spermatogenesis (i.e., spermatogonial and spermiogenic), an important aspect that will open new possibilities for investigating germ–Sertoli cells signaling pathways. Particularly the mechanisms related to the structural and functional aspects of Sertoli–germ cell interactions that may contribute to the strikingly anamniote Sertoli cell efficiency need to be investigated. Comprehensive studies investigating the biology of SSCs and their niche have been pivotal in this scenario (Nóbrega et al., 2010). As acting mainly on Sertoli cells, FSH plays a pivotal role in fish testis function and gametogenesis through several different growth factors (i.e., AMH, androgens, progestins, thyroid hormones, Igf3) that regulate SSCs renewal and differentiation (Nóbrega et al., 2010, 2015; Skaar et al., 2011; Chen et al., 2013; Morais et al., 2013; Assis et al., 2015; França et al., 2015; Melo et al., 2015). Through a nuclear estrogen receptor, eel Sertoli cells also regulate SSC renewal via the expression of platelet-derived endothelial cell growth factor (Pdecgf) that is considered a SSC renewal factor (Miura et al., 2003). Moreover, under the influence of progestin, trypsin expression (Miura et al., 2009), and taurine biosynthesis (Higuchi et al., 2012) were observed in eel Sertoli cells, leading to germ cells expression of a solute carrier gene (slc6a6) and their subsequent entry into meiosis (Higuchi et al., 2013).

In higher vertebrates, the derivatives of mesonephric tissue form the efferent ducts and sperm storage tissues. Considering that in most fish species spermatozoa are stored in the tubular lumen and that after the spawning season the residual spermatozoa are very efficiently phagocytized by SCs, fish represent an interesting model for investigating both the ‘recycling and waste management’ functions of Sertoli cells and spermatogonial kinetics. Finally, based on several important aspects mentioned in this section, we hope that we have convincingly demonstrated that Sertoli cells in lower vertebrates are highly dynamic and plastic cells. In contrast to mammals, Sertoli cells in fish are able to provide an adequate environment for spermatogenesis progression and sperm formation after xenogenic germ cell transplantation from phylogenetically distant species (Lacerda et al., 2013; Lacerda et al., 2014) and this is certainly another very important illustration of the amazing plasticity of anamniote Sertoli cells.

Supplementary Material

Table S1. Sertoli Cell Books and Biographies of Enrico Sertoli.

Figure S1. Key Morphological Features of Sertoli Cell are listed with representative examples illustrated.

Figure S2. Electron microscopy of the Sertoli cell nucleus (N) from a human testis showing a large nucleolus (Nu) and deep indentation (In) of the nuclear membrane (Nm).

Figure S3. Schematic representation of Sertoli cell (SC) proliferation in relation to endocrine and paracrine regulation of fish spermatogenesis.

Figure S4. Number of spermatids per Sertoli cell (SC), based on the available literature, for different vertebrate groups.

Acknowledgments

FUNDING

This work was supported in part by the following: CNPq and FAPEMIG (LRF); NIH R01 HD081244 (MCH).

Footnotes

CONFLICT OF INTEREST

The authors have no conflicts of interest to declare.

SUPPORTING INFORMATION

Additional Supporting Information may be found in the online version of this article:

References

  1. Aflatoonian B, Ruban L, Jones M, Aflatoonian R, Fazeli A, Moore HD. In vitro post-meiotic germ cell development from human embryonic stem cells. Hum Reprod. 2009;24:3150–3159. doi: 10.1093/humrep/dep334. [DOI] [PubMed] [Google Scholar]
  2. Almeida FF, Kristoffersen C, Taranger GL, Schulz RW. Spermatogenesis in Atlantic cod (Gadus morhua): a novel model of cystic germ cell development. Biol Reprod. 2008;78:27–34. doi: 10.1095/biolreprod.107.063669. [DOI] [PubMed] [Google Scholar]
  3. Andersson A-M. Inhibin B as a serum marker of spermatogenesis. In: Robaire B, Chemes H, Morales CR, editors. Andrology in the 21st Century. Medimond Publishing Company; New Jersey: 2001. pp. 123–130. [Google Scholar]
  4. Aponte PM, Soda T, Teerds KJ, Mizrak SC, van de Kant HJ, de Rooij DG. Propagation of bovine spermatogonial stem cells in vitro. Reproduction. 2008;136:543–557. doi: 10.1530/REP-07-0419. [DOI] [PubMed] [Google Scholar]
  5. Ara T, Nakamura Y, Egawa T, Sugiyama T, Abe K, Kishimoto T, Matsui Y, Nagasawa T. Impaired colonization of the gonads by primordial germ cells in mice lacking a chemokine, stromal cell-derived factor-1 (SDF-1) Proc Natl Acad Sci USA. 2003;100:5319–5323. doi: 10.1073/pnas.0730719100. [DOI] [PMC free article] [PubMed] [Google Scholar]
  6. Armstrong DT, Moon YS, Fritz IB, Dorrington JH. Synthesis of estradiol-17 beta by Sertoli cells in culture: stimulation by FSH and dibutyryl cyclic AMP. Curr Top Mol Endocrinol. 1975;2:85–96. doi: 10.1007/978-1-4613-4440-7_6. [DOI] [PubMed] [Google Scholar]
  7. Assis LH, Crespo D, Morais RD, Franca LR, Bogerd J, Schulz RW. INSL3 stimulates spermatogonial differentiation in testis of adult zebrafish (Danio rerio) Cell Tissue Res. 2015 doi: 10.1007/s00441-015-2213-9. In Press. [DOI] [PMC free article] [PubMed] [Google Scholar]
  8. Baraniak PR, McDevitt TC. Stem cell paracrine actions and tissue regeneration. Regen Med. 2010;5:121–143. doi: 10.2217/rme.09.74. [DOI] [PMC free article] [PubMed] [Google Scholar]
  9. Bardin CW, Cheng CY, Musto NA, Gunsalus GL. The Sertoli cell. In: Knobil E, Neill J, editors. The Physiology of Reproduction. Raven Press, Ltd; New York: 1988. pp. 933–974. [Google Scholar]
  10. Barker CF, Billingham RE. Immunologically privileged sites. Adv Immunol. 1977;25:1–54. [PubMed] [Google Scholar]
  11. Barrionuevo F, Burgos M, Jimenez R. Origin and function of embryonic Sertoli cells. Biomolecular Concepts. 2011;2:537–547. doi: 10.1515/BMC.2011.044. [DOI] [PubMed] [Google Scholar]
  12. Bartke A, Sinha Hikim AP, Russell LD. Sertoli cell structure and function in seasonally breeding mammals. In: Griswold MD, Russell LD, editors. The Sertoli Cell. Cache River Press; Clearwater, FL: 1993. pp. 349–364. [Google Scholar]
  13. Beach SF, Vogl AW. Spermatid translocation in the rat seminiferous epithelium: coupling membrane trafficking machinery to a junction plaque. Biol Reprod. 1999;60:1036–1046. doi: 10.1095/biolreprod60.4.1036. [DOI] [PubMed] [Google Scholar]
  14. Berruti G, Paiardi C. The dynamic of the apical ectoplasmic specialization between spermatids and Sertoli cells: the case of the small GTPase Rap1. Biomed Res Int. 2014;2014:635979. doi: 10.1155/2014/635979. [DOI] [PMC free article] [PubMed] [Google Scholar]
  15. Beverdam A, Koopman P. Expression profiling of purified mouse gonadal somatic cells during the critical time window of sex determination reveals novel candidate genes for human sexual dysgenesis syndromes. Hum Mol Genet. 2006;15:417–431. doi: 10.1093/hmg/ddi463. [DOI] [PubMed] [Google Scholar]
  16. Billard R, Breton B. Rhythm of reproduction in teleost fish. In: Thorpe JE, editor. Rhythmic Activity of Fishes. Academic Press; New York: 1978. pp. 31–53. [Google Scholar]
  17. Boekelheide K. Sertoli cell toxicants. In: Russell LD, Griswold MD, editors. The Sertoli Cell. Cache River Press; Clearwater, FL: 1993. pp. 551–576. [Google Scholar]
  18. Boekelheide K, Neely MD, Sioussat TM. The Sertoli cell cytoskeleton: a target for toxicant-induced germ cell loss. Toxicol Appl Pharmacol. 1989;101:373–389. doi: 10.1016/0041-008x(89)90188-9. [DOI] [PubMed] [Google Scholar]
  19. Brehm R, Steger K. Regulation of Sertoli cell and germ cell differentation. Adv Anat Embryol Cell Biol. 2005;181:1–93. [PubMed] [Google Scholar]
  20. Brökelmann J. Fine structure of germ cells and Sertoli cells during the cycle of the seminiferous epithelium in the rat. Z Zellforsch Mikrosk Anat. 1963;59:820–850. [PubMed] [Google Scholar]
  21. Brunocilla E, Pultrone CV, Schiavina R, Rocca C, Passaretti G, Corti B, Martorana G. Testicular sclerosing Sertoli cell tumor: an additional case and review of the literature. Anticancer Res. 2012;32:5127–5130. [PubMed] [Google Scholar]
  22. Burger HG, de Kretser D. The Testis. Raven Press; New York: 1981. p. 442. [Google Scholar]
  23. Burgos MH, Fawcett DW. Studies on the fine structure of the mammalian testis. I. Differentiation of the spermatids in the cat (Felis domestica) J Biophys Biochem Cytol. 1955;1:287–300. doi: 10.1083/jcb.1.4.287. [DOI] [PMC free article] [PubMed] [Google Scholar]
  24. Bustos-Obregon E. On Sertoli cell number and distribution in rat testis. Arch Biol (Liege) 1970;81:99–108. [PubMed] [Google Scholar]
  25. Byers S, Jegou B, MacCalman C, Blaschuk O. Sertoli cell adhesion molecules and the collective organization of the testis. In: Griswold MD, Russell LD, editors. The Sertoli Cell. Cache River Press; Clearwater, FL: 1993a. pp. 461–476. [Google Scholar]
  26. Byers S, Pelletier R-M, Suarez-Quian C. Sertoli-Sertoli cell junctions and the seminiferous epithelium barrier. In: Griswold MD, Russell LD, editors. The Sertoli Cell. Cache River Press; Clearwater, FL: 1993b. pp. 431–446. [Google Scholar]
  27. Caires KC, de Avila J, McLean DJ. Endocrine regulation of spermatogonial stem cells in the seminiferous epithelium of adult mice. Biores Open Access. 2012a;1:222–230. doi: 10.1089/biores.2012.0259. [DOI] [PMC free article] [PubMed] [Google Scholar]
  28. Caires KC, de Avila JM, Cupp AS, McLean DJ. VEGFA family isoforms regulate spermatogonial stem cell homeostasis in vivo. Endocrinology. 2012b;153:887–900. doi: 10.1210/en.2011-1323. [DOI] [PMC free article] [PubMed] [Google Scholar]
  29. Campbell B, Dickey JT, Swanson P. Endocrine changes during onset of puberty in male spring Chinook salmon, Oncorhynchus tshawytscha. Biol Reprod. 2003;69:2109–2117. doi: 10.1095/biolreprod.103.020560. [DOI] [PubMed] [Google Scholar]
  30. Cavicchia JC, Dym M. Relative volume of Sertoli cells in monkey seminiferous epithelium: a stereological analysis. Am J Anat. 1977;150:501–507. doi: 10.1002/aja.1001500309. [DOI] [PubMed] [Google Scholar]
  31. Cavicchia JC, Sacerdote FL, Morales A, Zhu BC. Sertoli cell nuclear pore number changes in some stages of the spermatogenic cycle of the rat seminiferous epithelium. Tissue Cell. 1998;30:268–273. doi: 10.1016/s0040-8166(98)80075-x. [DOI] [PubMed] [Google Scholar]
  32. Chakraborty P, William Buaas F, Sharma M, Smith BE, Greenlee AR, Eacker SM, Braun RE. Androgen-dependent sertoli cell tight junction remodeling is mediated by multiple tight junction components. Mol Endocrinol. 2014;28:1055–1072. doi: 10.1210/me.2013-1134. [DOI] [PMC free article] [PubMed] [Google Scholar]
  33. Chen C, Ouyang W, Grigura V, Zhou Q, Carnes K, Lim H, Zhao GQ, Arber S, Kurpios N, Murphy TL, Cheng AM, Hassell JA, Chandrashekar V, Hofmann MC, Hess RA, Murphy KM. ERM is required for transcriptional control of the spermatogonial stem cell niche. Nature. 2005;436:1030–1034. doi: 10.1038/nature03894. [DOI] [PMC free article] [PubMed] [Google Scholar]
  34. Chen SX, Bogerd J, Schoonen NE, Martijn J, de Waal PP, Schulz RW. A progestin (17alpha,20beta-dihydroxy-4-pregnen-3-one) stimulates early stages of spermatogenesis in zebrafish. Gen Comp Endocrinol. 2013;185:1–9. doi: 10.1016/j.ygcen.2013.01.005. [DOI] [PubMed] [Google Scholar]
  35. Cheng CY, Grima J, Lee WM, Bardin CW. The distribution of rat testibumin in the male reproductive tract. Biol Reprod. 1987;37:875–885. doi: 10.1095/biolreprod37.4.875. [DOI] [PubMed] [Google Scholar]
  36. Cheng CY, Grima J, Stahler MS, Guglielmotti A, Silvestrini B, Bardin CW. Sertoli cell synthesizes and secretes a protease inhibitor, alpha 2-macroglobulin. Biochemistry. 1990;29:1063–1068. doi: 10.1021/bi00456a031. [DOI] [PubMed] [Google Scholar]
  37. Cheng CY, Mruk DD. Cell junction dynamics in the testis: Sertoli-germ cell interactions and male contraceptive development. Physiol Rev. 2002;82:825–874. doi: 10.1152/physrev.00009.2002. [DOI] [PubMed] [Google Scholar]
  38. 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;44:245–263. doi: 10.1080/10409230903061207. [DOI] [PMC free article] [PubMed] [Google Scholar]
  39. Cheng CY, Mruk DD. A local autocrine axis in the testes that regulates spermatogenesis. Nat Rev Endocrinol. 2010;6:380–395. doi: 10.1038/nrendo.2010.71. [DOI] [PMC free article] [PubMed] [Google Scholar]
  40. Cheng CY, Mruk DD. The blood-testis barrier and its implications for male contraception. Pharmacol Rev. 2012;64:16–64. doi: 10.1124/pr.110.002790. [DOI] [PMC free article] [PubMed] [Google Scholar]
  41. Cheng CY, Mruk DD. Biochemistry of Sertoli cell/germ cell junctions, germ cell transport, and spermiation in the seminiferous epithelium. In: Griswold M, editor. Sertoli Cell Biology. Elsevier Academic Press; Oxford: 2015. pp. 333–383. [Google Scholar]
  42. Cheng CY, Wong EW, Lie PP, Li MW, Mruk DD, Yan HH, Mok KW, Mannu J, Mathur PP, Lui WY, Lee WM, Bonanomi M, Silvestrini B. Regulation of blood-testis barrier dynamics by desmosome, gap junction, hemidesmosome and polarity proteins: an unexpected turn of events. Spermatogenesis. 2011;1:105–115. doi: 10.4161/spmg.1.2.15745. [DOI] [PMC free article] [PubMed] [Google Scholar]
  43. Cheng CY, Wong EW, Yan HH, Mruk DD. Regulation of spermatogenesis in the microenvironment of the seminiferous epithelium: new insights and advances. Mol Cell Endocrinol. 2010;315:49–56. doi: 10.1016/j.mce.2009.08.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  44. Chiarini-Garcia H, Hornick JR, Griswold MD, Russell LD. Distribution of type A spermatogonia in the mouse is not random. Biol Reprod. 2001;65:1179–1185. doi: 10.1095/biolreprod65.4.1179. [DOI] [PubMed] [Google Scholar]
  45. Chiarini-Garcia H, Raymer AM, Russell LD. Non-random distribution of spermatogonia in rats: evidence of niches in the seminiferous tubules. Reproduction. 2003;126:669–680. doi: 10.1530/rep.0.1260669. [DOI] [PubMed] [Google Scholar]
  46. Chihara M, Ikebuchi R, Otsuka S, Ichii O, Hashimoto Y, Suzuki A, Saga Y, Kon Y. Mice stage-specific claudin 3 expression regulates progression of meiosis in early stage spermatocytes. Biol Reprod. 2013;89:3. doi: 10.1095/biolreprod.113.107847. [DOI] [PubMed] [Google Scholar]
  47. Chojnacka K, Mruk DD. The Src non-receptor tyrosine kinase paradigm: new insights into mammalian Sertoli cell biology. Mol Cell Endocrinol. 2015;415:133–142. doi: 10.1016/j.mce.2015.08.012. [DOI] [PubMed] [Google Scholar]
  48. Chowdhury M, Steinberger A, Steinberger E. Inhibition of de novo synthesis of FSH by the Sertoli cell factor (SCF) Endocrinology. 1978;103:644–647. doi: 10.1210/endo-103-2-644. [DOI] [PubMed] [Google Scholar]
  49. Clermont Y. Introduction to the Sertoli cell. In: Russell LD, Griswold MD, editors. The Sertoli Cell. Cache River Press; Clearwater, FL: 1993. pp. xxi–xxv. [Google Scholar]
  50. Clermont Y, Morales C, Hermo L. Endocytic activities of Sertoli cells in the rat. Ann N Y Acad Sci. 1987;513:1–15. doi: 10.1111/j.1749-6632.1987.tb24994.x. [DOI] [PubMed] [Google Scholar]
  51. Clermont Y, Morgentaler H. Quantitative study of spermatogenesis in the hypophysectomized rat. Endocrinology. 1955;57:369–382. doi: 10.1210/endo-57-3-369. [DOI] [PubMed] [Google Scholar]
  52. Clevers H, Loh KM, Nusse R. Stem cell signaling. An integral program for tissue renewal and regeneration: Wnt signaling and stem cell control. Science. 2014;346:1248012. doi: 10.1126/science.1248012. [DOI] [PubMed] [Google Scholar]
  53. Cooke PS, Holsberger DR, França LR. Thyroid hormone regulation of Sertoli cell development. In: Skinner MK, Griswold MD, editors. Sertoli Cell Biology. Elsevier Academic Press; San Diego: 2005. pp. 217–226. [Google Scholar]
  54. De Gendt K, Verhoeven G, Amieux PS, Wilkinson MF. Genome-wide identification of AR-regulated genes translated in Sertoli cells in vivo using the RiboTag approach. Mol Endocrinol. 2014;28:575–591. doi: 10.1210/me.2013-1391. [DOI] [PMC free article] [PubMed] [Google Scholar]
  55. de Kretser DM. Germ cell-Sertoli cell interactions. Reprod Fertil Dev. 1990;2:225–235. doi: 10.1071/rd9900225. [DOI] [PubMed] [Google Scholar]
  56. de Kretser DM, Loveland KL, Meehan T, O’Bryan MK, Phillips DJ, Wreford NG. Inhibins, activins and follistatin: actions on the testis. Mol Cell Endocrinol. 2001;180:87–92. doi: 10.1016/s0303-7207(01)00502-0. [DOI] [PubMed] [Google Scholar]
  57. de Rooij DG. The spermatogonial stem cell niche in mammals. In: Griswold M, editor. Sertoli Cell Biology. Elsevier Academic Press; Oxford: 2015. pp. 99–122. [Google Scholar]
  58. de Winter JP, Vanderstichele HM, Timmerman MA, Blok LJ, Themmen AP, de Jong FH. Activin is produced by rat Sertoli cells in vitro and can act as an autocrine regulator of Sertoli cell function. Endocrinology. 1993;132:975–982. doi: 10.1210/endo.132.3.7679985. [DOI] [PubMed] [Google Scholar]
  59. Demoulin A, Koulischer L, Hustin J, Hazee-Hagelstein MT, Lambotte R, Franchimont P. Organ culture of mammalian testis. III. Inhibin secretion. Horm Res. 1979;10:177–190. doi: 10.1159/000179000. [DOI] [PubMed] [Google Scholar]
  60. DiNapoli L, Capel B. SRY and the standoff in sex determination. Mol Endocrinol. 2008;22:1–9. doi: 10.1210/me.2007-0250. [DOI] [PMC free article] [PubMed] [Google Scholar]
  61. Djakiew D, Onoda M. Multichamber cell culture and directional secretion. In: Russell LD, Griswold MD, editors. The Sertoli Cell. Cache River Press; Clearwater, FL: 1993. pp. 181–194. [Google Scholar]
  62. Doitsidou M, Reichman-Fried M, Stebler J, Koprunner M, Dorries J, Meyer D, Esguerra CV, Leung T, Raz E. Guidance of primordial germ cell migration by the chemokine SDF-1. Cell. 2002;111:647–659. doi: 10.1016/s0092-8674(02)01135-2. [DOI] [PubMed] [Google Scholar]
  63. Dong WL, Tan FQ, Yang WX. Wnt signaling in testis development: unnecessary or essential? Gene. 2015;565:155–165. doi: 10.1016/j.gene.2015.04.066. [DOI] [PubMed] [Google Scholar]
  64. Dorrington JH, Armstrong DT. Follicle-stimulating hormone stimulates estradiol-17beta synthesis in cultured Sertoli cells. Proc Natl Acad Sci USA. 1975;72:2677–2681. doi: 10.1073/pnas.72.7.2677. [DOI] [PMC free article] [PubMed] [Google Scholar]
  65. Dorrington JH, Khan SA. Steroid production, metabolism, and release by Sertoli cells. In: Griswold MD, Russell LD, editors. The Sertoli Cell. Cache River Press; Clearwater, FL: 1993. pp. 537–550. [Google Scholar]
  66. Dovere L, Fera S, Grasso M, Lamberti D, Gargioli C, Muciaccia B, Lustri AM, Stefanini M, Vicini E. The niche-derived glial cell line-derived neurotrophic factor (GDNF) Induces migration of mouse spermatogonial stem/progenitor cells. PLoS ONE. 2013;8:e59431. doi: 10.1371/journal.pone.0059431. [DOI] [PMC free article] [PubMed] [Google Scholar]
  67. Doyle TJ, Kaur G, Putrevu SM, Dyson EL, Dyson M, McCunniff WT, Pasham MR, Kim KH, Dufour JM. Immunoprotective properties of primary Sertoli cells in mice: potential functional pathways that confer immune privilege. Biol Reprod. 2012;86:1–14. doi: 10.1095/biolreprod.110.089425. [DOI] [PMC free article] [PubMed] [Google Scholar]
  68. Du M, Young J, De Asis M, Cipollone J, Roskelley C, Takai Y, Nicholls PK, Stanton PG, Deng W, Finlay BB, Vogl AW. A novel subcellular machine contributes to basal junction remodeling in the seminiferous epithelium. Biol Reprod. 2013;88:60. doi: 10.1095/biolreprod.112.104851. [DOI] [PubMed] [Google Scholar]
  69. Dufour JM, Hamilton M, Rajotte RV, Korbutt GS. Neonatal porcine Sertoli cells inhibit human natural antibody-mediated lysis. Biol Reprod. 2005;72:1224–1231. doi: 10.1095/biolreprod.104.038315. [DOI] [PubMed] [Google Scholar]
  70. Dufour JM, Hemendinger R, Halberstadt CR, Gores P, Emerich DF, Korbutt GS, Rajotte RV. Genetically engineered Sertoli cells are able to survive allogeneic transplantation. Gene Ther. 2004;11:694–700. doi: 10.1038/sj.gt.3302218. [DOI] [PubMed] [Google Scholar]
  71. Dufour JM, Rajotte RV, Korbutt GS, Emerich DF. Harnessing the immunomodulatory properties of Sertoli cells to enable xenotransplantation in type I diabetes. Immunol Invest. 2003a;32:275–297. doi: 10.1081/imm-120025106. [DOI] [PubMed] [Google Scholar]
  72. Dufour JM, Rajotte RV, Seeberger K, Kin T, Korbutt GS. Long-term survival of neonatal porcine Sertoli cells in non-immunosuppressed rats. Xenotransplantation. 2003b;10:577–586. doi: 10.1034/j.1399-3089.2003.00059.x. [DOI] [PubMed] [Google Scholar]
  73. Dym M. The fine structure of the monkey (Macaca) Sertoli cell and its role in maintaining the blood-testis barrier. Anat Rec. 1973;175:639–656. doi: 10.1002/ar.1091750402. [DOI] [PubMed] [Google Scholar]
  74. Dym M. Basement membrane regulation of Sertoli cells. Endocr Rev. 1994;15:102–115. doi: 10.1210/edrv-15-1-102. [DOI] [PubMed] [Google Scholar]
  75. Dym M, Fawcett DW. The blood-testis barrier in the rat and the physiological compartmentation of the seminiferous epithelium. Biol Reprod. 1970;3:308–326. doi: 10.1093/biolreprod/3.3.308. [DOI] [PubMed] [Google Scholar]
  76. Dym M, Romrell LJ. Intraepithelial lymphocytes in the male reproductive tract of rats and rhesus monkeys. J Reprod Fertil. 1975;42:1–7. doi: 10.1530/jrf.0.0420001. [DOI] [PubMed] [Google Scholar]
  77. Ebner V. Zur Spermatogenese bei den Säugethieren. Arch Mikr Anat. 1888;31:236–291. [Google Scholar]
  78. El-Darwish KS, Parvinen M, Toppari J. Differential expression of members of the E2F family of transcription factors in rodent testes. Reprod Biol Endocrinol. 2006;4:63. doi: 10.1186/1477-7827-4-63. [DOI] [PMC free article] [PubMed] [Google Scholar]
  79. Elftman H. The Sertoli cell cycle in the mouse. Anat Rec. 1950;106:381–392. doi: 10.1002/ar.1091060306. [DOI] [PubMed] [Google Scholar]
  80. Elftman H. Sertoli cells and testis structure. Am J Anat. 1963;113:25–33. doi: 10.1002/aja.1001130104. [DOI] [PubMed] [Google Scholar]
  81. Escott GM, da Rosa LA, Loss Eda S. Mechanisms of hormonal regulation of sertoli cell development and proliferation: a key process for spermatogenesis. Curr Mol Pharmacol. 2014;7:96–108. doi: 10.2174/1874467208666150126155032. [DOI] [PubMed] [Google Scholar]
  82. Evans E, Hogarth C, Mitchell D, Griswold M. Riding the spermatogenic wave: profiling gene expression within neonatal germ and sertoli cells during a synchronized initial wave of spermatogenesis in mice. Biol Reprod. 2014;90:108. doi: 10.1095/biolreprod.114.118034. [DOI] [PMC free article] [PubMed] [Google Scholar]
  83. Ewing LL, Robaire B. Regulation of Testicular Function: Signaling Molecules and Cell-Cell Communication. The New York Academy of Science; New York: 1989. p. 305. [PubMed] [Google Scholar]
  84. Fawcett DW. Ultrastructure and function of the Sertoli cell. In: Hamilton DW, Greep RO, editors. Handbook of Physiology. Vol. 5. American Physiology Society; Washington, DC: 1975. pp. 21–55. [Google Scholar]
  85. Fawcett DW, Burgos M. The fine structure of the Sertoli cells in the human testis. Anat Rec. 1956;124:401–402. [Google Scholar]
  86. Fawcett DW, Leak LV, Heidger PM., Jr Electron microscopic observations on the structural components of the blood-testis barrier. J Reprod Fertil Suppl. 1970;10:105–122. [PubMed] [Google Scholar]
  87. Fawcett DW, Phillips DM. Observations on the release of spermatozoa and on changes in the head during passage through the epididymis. J Reprod Fert Suppl. 1969;6:405–418. [Google Scholar]
  88. Flickinger C, Fawcett DW. The junctional specializations of Sertoli cells in the seminiferous epithelium. Anat Rec. 1967;158:207–221. doi: 10.1002/ar.1091580210. [DOI] [PubMed] [Google Scholar]
  89. Fok KL, Chen H, Ruan YC, Chan HC. Novel regulators of spermatogenesis. Semin Cell Dev Biol. 2014;29:31–42. doi: 10.1016/j.semcdb.2014.02.008. [DOI] [PubMed] [Google Scholar]
  90. Franca LR, Auharek SA, Hess RA, Dufour JM, Hinton BT. Blood-tissue barriers: morphofunctional and immunological aspects of the blood-testis and blood-epididymal barriers. Adv Exp Med Biol. 2012;763:237–259. [PubMed] [Google Scholar]
  91. França LR, Nóbrega RH, Morais RDVS, Assis LHC, Schulz RW. Sertoli cell structure and function in anamniote vertebrates. In: Griswold M, editor. Sertoli Cell Biology. Elsevier Academic Press; San Diego: 2015. pp. 385–407. [Google Scholar]
  92. Fritz IB. Comparison of granulosa and sertoli cells at various stages of maturation: similarities and differences. Adv Exp Med Biol. 1982;147:357–384. doi: 10.1007/978-1-4615-9278-5_21. [DOI] [PubMed] [Google Scholar]
  93. Fritz IB, Griswold MD, Louis BG, Dorrington JH. Similarity of responses of cultured Sertoli cells to cholera toxin and FSH. Mol Cell Endocrinol. 1976;5:289–294. doi: 10.1016/0303-7207(76)90090-3. [DOI] [PubMed] [Google Scholar]
  94. Fritz IB, Tung PS, Ailenberg M. Proteases and antiproteases in the seminiferous tubule. In: Griswold MD, Russell LD, editors. The Sertoli Cell. Cache River Press; Clearwater, FL: 1993. pp. 217–236. [Google Scholar]
  95. Frojdman K, Harley VR, Pelliniemi LJ. Sox9 protein in rat sertoli cells is age and stage dependent. Histochem Cell Biol. 2000;113:31–36. doi: 10.1007/s004180050004. [DOI] [PubMed] [Google Scholar]
  96. Gao F, Maiti S, Alam N, Zhang Z, Deng JM, Behringer RR, Lecureuil C, Guillou F, Huff V. The Wilms tumor gene, Wt1, is required for Sox9 expression and maintenance of tubular architecture in the developing testis. Proc Natl Acad Sci USA. 2006;103:11987–11992. doi: 10.1073/pnas.0600994103. [DOI] [PMC free article] [PubMed] [Google Scholar]
  97. Gao Y, Mruk DD, Cheng CY. Sertoli cells are the target of environmental toxicants in the testis - a mechanistic and therapeutic insight. Expert Opin Ther Targets. 2015;19:1073–1090. doi: 10.1517/14728222.2015.1039513. [DOI] [PMC free article] [PubMed] [Google Scholar]
  98. Garcia TX, DeFalco T, Capel B, Hofmann MC. Constitutive activation of NOTCH1 signaling in Sertoli cells causes gonocyte exit from quiescence. Dev Biol. 2013;377:188–201. doi: 10.1016/j.ydbio.2013.01.031. [DOI] [PMC free article] [PubMed] [Google Scholar]
  99. Garcia TX, Farmaha JK, Kow S, Hofmann MC. RBPJ in mouse Sertoli cells is required for proper regulation of the testis stem cell niche. Development. 2014;141:4468–4478. doi: 10.1242/dev.113969. [DOI] [PMC free article] [PubMed] [Google Scholar]
  100. Garcia TX, Hofmann MC. NOTCH signaling in Sertoli cells regulates gonocyte fate. Cell Cycle. 2013;12:2538–2545. doi: 10.4161/cc.25627. [DOI] [PMC free article] [PubMed] [Google Scholar]
  101. Gassei K, Schlatt S. Testicular morphogenesis: comparison of in vivo and in vitro models to study male gonadal development. Ann N Y Acad Sci. 2007;1120:152–167. doi: 10.1196/annals.1411.015. [DOI] [PubMed] [Google Scholar]
  102. Golestaneh N, Beauchamp E, Fallen S, Kokkinaki M, Uren A, Dym M. Wnt signaling promotes proliferation and stemness regulation of spermatogonial stem/progenitor cells. Reproduction. 2009;138:151–162. doi: 10.1530/REP-08-0510. [DOI] [PubMed] [Google Scholar]
  103. Gondos B, Berndston WE. Postnatal and pubertal development. In: Russell LD, Griswold MD, editors. The Sertoli Cell. Cache River Press; Clearwater, FL: 1993. pp. 115–154. [Google Scholar]
  104. Gow A, Southwood CM, Li JS, Pariali M, Riordan GP, Brodie SE, Danias J, Bronstein JM, Kachar B, Lazzarini RA. CNS myelin and sertoli cell tight junction strands are absent in Osp/claudin-11 null mice. Cell. 1999;99:649–659. doi: 10.1016/s0092-8674(00)81553-6. [DOI] [PubMed] [Google Scholar]
  105. Grasso M, Fuso A, Dovere L, de Rooij DG, Stefanini M, Boitani C, Vicini E. Distribution of GFRA1-expressing spermatogonia in adult mouse testis. Reproduction. 2012;143:325–332. doi: 10.1530/REP-11-0385. [DOI] [PubMed] [Google Scholar]
  106. Grier HJ. Comparative organization of Sertoli cells including the Sertoli cell barrier. In: Griswold MD, Russell LD, editors. The Sertoli Cell. Cache River Press; Clearwater, FL: 1993. pp. 703–740. [Google Scholar]
  107. Griswold MD. Protein secretions of Sertoli cells. Int Rev Cytol. 1988;110:133–156. doi: 10.1016/s0074-7696(08)61849-5. [DOI] [PubMed] [Google Scholar]
  108. Griswold MD. Action of FSH on mammalian Sertoli cells. In: Griswold MD, Russell LD, editors. The Sertoli Cell. Cache River Press; Clearwater, FL: 1993a. pp. 493–508. [Google Scholar]
  109. Griswold MD. Protein secretion by Sertoli cells: general considerations. In: Griswold MD, Russell LD, editors. The Sertoli Cell. Cache River Press; Clearwater, FL: 1993b. pp. 195–200. [Google Scholar]
  110. Griswold MD. Unique aspects of the biochemistry and metabolism of Sertoli cells. In: Griswold MD, Russell LD, editors. The Sertoli Cell. Cache River Press; Clearwater, FL: 1993c. pp. 485–492. [Google Scholar]
  111. Griswold MD. Interactions between germ cells and Sertoli cells in the testis. Biol Reprod. 1995;52:211–216. doi: 10.1095/biolreprod52.2.211. [DOI] [PubMed] [Google Scholar]
  112. Griswold MD. The central role of Sertoli cells in spermatogenesis. Semin Cell Dev Biol. 1998;9:411–416. doi: 10.1006/scdb.1998.0203. [DOI] [PubMed] [Google Scholar]
  113. Griswold MD. Sertoli Cell Biology(ed MD Griswold. Elsevier Academic Press; Oxford; 2015a. The initiation of spermatogenesis and the cycle of the seminiferous epithelium; pp. 233–245. [Google Scholar]
  114. Griswold MD. Sertoli Cell Biology. Elsevier Academic Press; Oxford: 2015b. p. 469. [Google Scholar]
  115. Griswold MD. Spermatogenesis: the commitment to meiosis. Physiol Rev. 2016;96:1–17. doi: 10.1152/physrev.00013.2015. [DOI] [PMC free article] [PubMed] [Google Scholar]
  116. Griswold MD, McLean D. Sertoli cell gene expression and protein secretion. In: Skinner M, Griswold M, editors. Sertoli Cell Biology. Academic Press; San Diego: 2005. pp. 95–106. [Google Scholar]
  117. Griswold MD, Morales C, Sylvester SR. Molecular biology of the Sertoli cell. Oxf Rev Reprod Biol. 1988;10:124–161. [PubMed] [Google Scholar]
  118. Guazzone VA, Jacobo P, Theas MS, Lustig L. Cytokines and chemokines in testicular inflammation: a brief review. Microsc Res Tech. 2009;72:620–628. doi: 10.1002/jemt.20704. [DOI] [PubMed] [Google Scholar]
  119. Gungor-Ordueri NE, Celik-Ozenci C, Cheng CY. Fascin 1 is an actin filament bundling protein that regulates ectoplasmic specialization dynamics in the rat testis. Am J Physiol Endocrinol Metab. 2014;307:E738–753. doi: 10.1152/ajpendo.00113.2014. [DOI] [PMC free article] [PubMed] [Google Scholar]
  120. Guraya SS. The comparative cell biology of accessory somatic (or Sertoli) cells in the animal testis. Int Rev Cytol. 1995;160:163–220. doi: 10.1016/s0074-7696(08)61555-7. [DOI] [PubMed] [Google Scholar]
  121. Guttenbach M, Martinez-Exposito MJ, Engel W, Schmid M. Interphase chromosome arrangement in Sertoli cells of adult mice. Biol Reprod. 1996;54:980–986. doi: 10.1095/biolreprod54.5.980. [DOI] [PubMed] [Google Scholar]
  122. Hai Y, Hou J, Liu Y, Liu Y, Yang H, Li Z, He Z. The roles and regulation of Sertoli cells in fate determinations of spermatogonial stem cells and spermatogenesis. Semin Cell Dev Biol. 2014;29:66–75. doi: 10.1016/j.semcdb.2014.04.007. [DOI] [PubMed] [Google Scholar]
  123. Halley K, Dyson EL, Kaur G, Mital P, Uong PM, Dass B, Crowell SN, Dufour JM. Delivery of a therapeutic protein by immune-privileged Sertoli cells. Cell Transplant. 2010;19:1645–1657. doi: 10.3727/096368910X516628. [DOI] [PMC free article] [PubMed] [Google Scholar]
  124. Heckert L, Griswold MD. Expression of the FSH receptor in the testis. Recent Prog Horm Res. 1993;48:61–77. doi: 10.1016/b978-0-12-571148-7.50006-3. [DOI] [PubMed] [Google Scholar]
  125. Heckert LL, Agbor VA. DMRT1 and the road to masculinity. In: Griswold M, editor. Sertoli Cell Biology. Elsevier Academic Press; Oxford; 2015. pp. 123–174. [Google Scholar]
  126. Hedger MP. The Immunophysiology of Male Reproduction. Academic Press; New York: 2015. pp. 805–889. [Google Scholar]
  127. Heller CG, Maddock WO, et al. The Sertoli cell. J Clin Invest. 1948;27:540. [PubMed] [Google Scholar]
  128. Hermo L, Pelletier RM, Cyr DG, Smith CE. Surfing the wave, cycle, life history, and genes/proteins expressed by testicular germ cells. Part 5: intercellular junctions and contacts between germs cells and Sertoli cells and their regulatory interactions, testicular cholesterol, and genes/proteins associated with more than one germ cell generation. Microsc Res Tech. 2010;73:409–494. doi: 10.1002/jemt.20786. [DOI] [PubMed] [Google Scholar]
  129. Hess R, França LR. Structure of the Sertoli cell. In: Griswold M, Skinner M, editors. Sertoli Cell Biology. Elsevier Academic Press; San Diego: 2005. pp. 19–40. [Google Scholar]
  130. Hess RA. Quantitative and qualitative characteristics of the stages and transitions in the cycle of the rat seminiferous epithelium: light microscopic observations of perfusion-fixed and plastic-embedded testes. Biol Reprod. 1990;43:525–542. doi: 10.1095/biolreprod43.3.525. [DOI] [PubMed] [Google Scholar]
  131. Hess RA, Cooke PS, Bunick D, Kirby JD. Adult testicular enlargement induced by neonatal hypothyroidism is accompanied by increased Sertoli and germ cell numbers. Endocrinology. 1993;132:2607–2613. doi: 10.1210/endo.132.6.8504761. [DOI] [PubMed] [Google Scholar]
  132. Hess RA, Vogl AW. Sertoli cell anatomy and cytoskeleton. In: Griswold M, editor. Sertoli Cell Biology. Elsevier Academic Press; Oxford; 2015. pp. 1–55. [Google Scholar]
  133. Higuchi M, Celino FT, Tamai A, Miura C, Miura T. The synthesis and role of taurine in the Japanese eel testis. Amino Acids. 2012;43:773–781. doi: 10.1007/s00726-011-1128-3. [DOI] [PubMed] [Google Scholar]
  134. Higuchi M, Miura C, Iwai T, Miura T. Trypsin regulates meiotic initiation in the Japanese eel (Anguilla japonica) by promoting the uptake of taurine into germ cells during spermatogenesis. Biol Reprod. 2013;89:58. doi: 10.1095/biolreprod.113.109777. [DOI] [PubMed] [Google Scholar]
  135. Hinsch GW. Comparative organization and cytology of Sertoli cells in invertebrates. In: Griswold MD, Russell LD, editors. The Sertoli Cell. Cache River Press; Clearwater, FL: 1993. pp. 659–684. [Google Scholar]
  136. Hinton BT, Setchell BP. Fluid secretion and movement. In: Russell LD, Griswold MD, editors. The Sertoli Cell. Cache River Press; Clearwater, FL: 1993. pp. 249–267. [Google Scholar]
  137. Hogarth C. Retinoic acid metabolism, signaling, and function in the adult testis. In: Griswold M, editor. Sertoli Cell Biology. Elsevier Academic Press; Oxford; 2015. pp. 247–272. [Google Scholar]
  138. Hogarth CA, Griswold MD. The key role of vitamin A in spermatogenesis. J Clin Invest. 2010;120:956–962. doi: 10.1172/JCI41303. [DOI] [PMC free article] [PubMed] [Google Scholar]
  139. Hogarth CA, Griswold MD. Immunohistochemical approaches for the study of spermatogenesis. Methods Mol Biol. 2013;927:309–320. doi: 10.1007/978-1-62703-038-0_28. [DOI] [PubMed] [Google Scholar]
  140. Innes JRM. Neoplastic diseases of the testis in animals. J Path Bact. 1942;54:485–498. [Google Scholar]
  141. Ishii K, Kanatsu-Shinohara M, Toyokuni S, Shinohara T. FGF2 mediates mouse spermatogonial stem cell self-renewal via upregulation of Etv5 and Bcl6b through MAP2K1 activation. Development. 2012;139:1734–1743. doi: 10.1242/dev.076539. [DOI] [PubMed] [Google Scholar]
  142. Jacobo P, Guazzone VA, Theas MS, Lustig L. Testicular autoimmunity. Autoimmun Rev. 2011;10:201–204. doi: 10.1016/j.autrev.2010.09.026. [DOI] [PubMed] [Google Scholar]
  143. Jakob S, Lovell-Badge R. Sex determination and the control of Sox9 expression in mammals. FEBS J. 2011;278:1002–1009. doi: 10.1111/j.1742-4658.2011.08029.x. [DOI] [PubMed] [Google Scholar]
  144. Jean P, Hartung M, Mirre C, Stahl A. Association of centromeric heterochromatin with the nucleolus in mouse Sertoli cells. Anat Rec. 1983;205:375–380. doi: 10.1002/ar.1092050402. [DOI] [PubMed] [Google Scholar]
  145. Jegou B. Spermatids are regulators of Sertoli cell function. Ann N Y Acad Sci. 1991;637:340–353. doi: 10.1111/j.1749-6632.1991.tb27321.x. [DOI] [PubMed] [Google Scholar]
  146. Jegou B. The Sertoli cell. Baillieres Clin Endocrinol Metab. 1992a;6:273–311. doi: 10.1016/s0950-351x(05)80151-x. [DOI] [PubMed] [Google Scholar]
  147. Jegou B. The Sertoli cell in vivo and in vitro. Cell Biol Toxicol. 1992b;8:49–54. doi: 10.1007/BF00130510. [DOI] [PubMed] [Google Scholar]
  148. Jegou B. The Sertoli-germ cell communication network in mammals. Int Rev Cytol. 1993;147:25–96. [PubMed] [Google Scholar]
  149. Jegou B, Stephan JP, Cudicini C, Gomez E, Bauche F, Piquet-Pellorce C, Touzalin AM. The Sertoli cell-germ cell interactions and the seminiferous tubule interleukin-1 and interleukin-6 system. Results Probl Cell Differ. 2000;28:53–68. doi: 10.1007/978-3-540-48461-5_3. [DOI] [PubMed] [Google Scholar]
  150. Jiang XH, Bukhari I, Zheng W, Yin S, Wang Z, Cooke HJ, Shi QH. Blood-testis barrier and spermatogenesis: lessons from genetically-modified mice. Asian J Androl. 2014;16:572–580. doi: 10.4103/1008-682X.125401. [DOI] [PMC free article] [PubMed] [Google Scholar]
  151. Johnson KJ. Testicular histopathology associated with disruption of the Sertoli cell cytoskeleton. Spermatogenesis. 2014;4:e979106. doi: 10.4161/21565562.2014.979106. [DOI] [PMC free article] [PubMed] [Google Scholar]
  152. Johnson L, Thompson DL., Jr Age-related and seasonal variation in the Sertoli cell population, daily sperm production and serum concentrations of follicle-stimulating hormone, luteinizing hormone and testosterone in stallions. Biol Reprod. 1983;29:777–789. doi: 10.1095/biolreprod29.3.777. [DOI] [PubMed] [Google Scholar]
  153. Johnson L, Thompson DL, Jr, Varner DD. Role of Sertoli cell number and function on regulation of spermatogenesis. Anim Reprod Sci. 2008;105:23–51. doi: 10.1016/j.anireprosci.2007.11.029. [DOI] [PubMed] [Google Scholar]
  154. Johnson L, Zane RS, Petty CS, Neaves WB. Quantification of the human Sertoli cell population: its distribution, relation to germ cell numbers, and age-related decline. Biol Reprod. 1984;31:785–795. doi: 10.1095/biolreprod31.4.785. [DOI] [PubMed] [Google Scholar]
  155. Johnson MH. The distribution of immunoglobulin and spermatozoal autoantigen in the genital tract of the male guinea pig: its relationship to autoallergic orchitis. Fertil Steril. 1972;23:383–392. doi: 10.1016/s0015-0282(16)39003-3. [DOI] [PubMed] [Google Scholar]
  156. Johnson MH, Setchell BP. Protein and immunoglobulin content of rete testis fluid of rams. J Reprod Fertil. 1968;17:403–406. doi: 10.1530/jrf.0.0170403. [DOI] [PubMed] [Google Scholar]
  157. 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;85:763–769. doi: 10.1095/biolreprod.110.087676. [DOI] [PMC free article] [PubMed] [Google Scholar]
  158. Johnston DS, Russell LD, Friel PJ, Griswold MD. Murine germ cells do not require functional androgen receptors to complete spermatogenesis following spermatogonial stem cell transplantation. Endocrinology. 2001;142:2405–2408. doi: 10.1210/endo.142.6.8317. [DOI] [PubMed] [Google Scholar]
  159. 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 USA. 2008;105:8315–8320. doi: 10.1073/pnas.0709854105. [DOI] [PMC free article] [PubMed] [Google Scholar]
  160. Josso N. Anti-mullerian hormone and Sertoli cell function. Horm Res. 1992;38(Suppl 2):72–76. doi: 10.1159/000182602. [DOI] [PubMed] [Google Scholar]
  161. Kaipia A, Parvinen M, Shimasaki S, Ling N, Toppari J. Stage-specific cellular regulation of inhibin alpha-subunit mRNA expression in the rat seminiferous epithelium. Mol Cell Endocrinol. 1991;82:165–173. doi: 10.1016/0303-7207(91)90028-q. [DOI] [PubMed] [Google Scholar]
  162. Kanatsu-Shinohara M, Inoue K, Ogonuki N, Miki H, Yoshida S, Toyokuni S, Lee J, Ogura A, Shinohara T. Leukemia inhibitory factor enhances formation of germ cell colonies in neonatal mouse testis culture. Biol Reprod. 2007;76:55–62. doi: 10.1095/biolreprod.106.055863. [DOI] [PubMed] [Google Scholar]
  163. Kanatsu-Shinohara M, Inoue K, Takashima S, Takehashi M, Ogonuki N, Morimoto H, Nagasawa T, Ogura A, Shinohara T. Reconstitution of mouse spermatogonial stem cell niches in culture. Cell Stem Cell. 2012;11:567–578. doi: 10.1016/j.stem.2012.06.011. [DOI] [PubMed] [Google Scholar]
  164. Kanatsu-Shinohara M, Ogonuki N, Inoue K, Miki H, Ogura A, Toyokuni S, Shinohara T. Long-term proliferation in culture and germline transmission of mouse male germline stem cells. Biol Reprod. 2003;69:612–616. doi: 10.1095/biolreprod.103.017012. [DOI] [PubMed] [Google Scholar]
  165. Kaur G, Long CR, Dufour JM. Genetically engineered immune privileged Sertoli cells: a new road to cell based gene therapy. Spermatogenesis. 2012;2:23–31. doi: 10.4161/spmg.19119. [DOI] [PMC free article] [PubMed] [Google Scholar]
  166. Kaur G, Mital P, Dufour JM. Testisimmune privilege -assumptions facts. Anim Reprod. 2013;10:3–15. [PMC free article] [PubMed] [Google Scholar]
  167. Kaur G, Thompson LA, Dufour JM. Therapeutic potential of immune privileged Sertoli cells. Anim Reprod. 2015;12:105–117. [Google Scholar]
  168. Kaur G, Thompson LA, Dufour JM. Sertoli cells – immunological sentinels of spermatogenesis. Semin Cell Dev Biol. 2014a;30:36–44. doi: 10.1016/j.semcdb.2014.02.011. [DOI] [PMC free article] [PubMed] [Google Scholar]
  169. Kaur G, Thompson LA, Pasham M, Tessanne K, Long CR, Dufour JM. Sustained expression of insulin by a genetically engineered Sertoli cell line after allotransplantation in diabetic BALB/c mice. Biol Reprod. 2014b;90:109. doi: 10.1095/biolreprod.113.115600. [DOI] [PMC free article] [PubMed] [Google Scholar]
  170. Kent J, Wheatley SC, Andrews JE, Sinclair AH, Koopman P. A male-specific role for SOX9 in vertebrate sex determination. Development. 1996;122:2813–2822. doi: 10.1242/dev.122.9.2813. [DOI] [PubMed] [Google Scholar]
  171. Kerkis A, Fonseca SA, Serafim RC, Lavagnolli TM, Abdelmassih S, Abdelmassih R, Kerkis I. In vitro differentiation of male mouse embryonic stem cells into both presumptive sperm cells and oocytes. Cloning Stem Cells. 2007;9:535–548. doi: 10.1089/clo.2007.0031. [DOI] [PubMed] [Google Scholar]
  172. Kerr GE, Young JC, Horvay K, Abud HE, Loveland KL. Regulated Wnt/beta-catenin signaling sustains adult spermatogenesis in mice. Biol Reprod. 2014;90:3. doi: 10.1095/biolreprod.112.105809. [DOI] [PubMed] [Google Scholar]
  173. Kerr JB. A light microscopic and morphometric analysis of the Sertoli cell during the spermatogenic cycle of the rat. Anat Embryol (Berl) 1988a;177:341–348. doi: 10.1007/BF00315842. [DOI] [PubMed] [Google Scholar]
  174. Kerr JB. An ultrastructural and morphometric analysis of the Sertoli cell during the spermatogenic cycle of the rat. Anat Embryol (Berl) 1988b;179:191–203. doi: 10.1007/BF00304701. [DOI] [PubMed] [Google Scholar]
  175. Kerr JB. Macro, micro, and molecular research on spermatogenesis: the quest to understand its control. Microsc Res Tech. 1995;32:364–384. doi: 10.1002/jemt.1070320503. [DOI] [PubMed] [Google Scholar]
  176. Kerr JB, De Kretser DM. Cyclic variations in Sertoli cell lipid content throughout the spermatogenic cycle in the rat. J Reprod Fertil. 1975;43:1–8. doi: 10.1530/jrf.0.0430001. [DOI] [PubMed] [Google Scholar]
  177. Kerr JB, Loveland KL, O’Bryan MK, de Kretser DM. Cytology of the testis and intrinsic control mechanisms. In: Neill JD, editor. Kobil and Neill’s Physiology of Reproduction. Vol. 1. Elsevier St; Louis, MO: 2006. pp. 827–947. [Google Scholar]
  178. Ketola I, Rahman N, Toppari J, Bielinska M, Porter-Tinge SB, Tapanainen JS, Huhtaniemi IT, Wilson DB, Heikinheimo M. Expression and regulation of transcription factors GATA-4 and GATA-6 in developing mouse testis. Endocrinology. 1999;140:1470–1480. doi: 10.1210/endo.140.3.6587. [DOI] [PubMed] [Google Scholar]
  179. Kierszenbaum AL. Effect of trenimon on the ultrastructure of Sertoli cells in the mouse. Virchows Arch B Cell Pathol. 1970;5:1–12. doi: 10.1007/BF02893547. [DOI] [PubMed] [Google Scholar]
  180. Kim KH, Wang Z. Action of vitamin A on the testis: role of the Sertoli cell. In: Griswold MD, Russell LD, editors. The Sertoli Cell. Cache River Press; Clearwater, FL: 1993. pp. 517–536. [Google Scholar]
  181. Kobayashi T, Nagahama Y. Molecular aspects of gonadal differentiation in a teleost fish, the nile tilapia. Sex Dev. 2009;3:108–117. doi: 10.1159/000223076. [DOI] [PubMed] [Google Scholar]
  182. Kobayashi Y, Nakamura M, Sunobe T, Usami T, Kobayashi T, Manabe H, Paul-Prasanth B, Suzuki N, Nagahama Y. Sex change in the Gobiid fish is mediated through rapid switching of gonadotropin receptors from ovarian to testicular portion or vice versa. Endocrinology. 2009;150:1503–1511. doi: 10.1210/en.2008-0569. [DOI] [PubMed] [Google Scholar]
  183. Koopman P, Munsterberg A, Capel B, Vivian N, Lovell-Badge R. Expression of a candidate sex-determining gene during mouse testis differentiation. Nature. 1990;348:450–452. doi: 10.1038/348450a0. [DOI] [PubMed] [Google Scholar]
  184. Kopera IA, Bilinska B, Cheng CY, Mruk DD. Sertoli-germ cell junctions in the testis: a review of recent data. Philos Trans R Soc Lond B Biol Sci. 2010;365:1593–1605. doi: 10.1098/rstb.2009.0251. [DOI] [PMC free article] [PubMed] [Google Scholar]
  185. Kubota H, Avarbock MR, Brinster RL. Growth factors essential for self-renewal and expansion of mouse spermatogonial stem cells. Proc Natl Acad Sci USA. 2004;101:16489–16494. doi: 10.1073/pnas.0407063101. [DOI] [PMC free article] [PubMed] [Google Scholar]
  186. Kubota H, Brinster RL. Culture of rodent spermatogonial stem cells, male germline stem cells of the postnatal animal. Methods Cell Biol. 2008;86:59–84. doi: 10.1016/S0091-679X(08)00004-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  187. Kushida T, Iijima H, Nagato Y, Kushida H. Studies on thick sections of the nucleus of mouse Sertoli cells using an electron microscope operating at 300 kV. Okajimas Folia Anat Jpn. 1993;70:41–50. doi: 10.2535/ofaj1936.70.2-3_41. [DOI] [PubMed] [Google Scholar]
  188. Kyronlahti A, Euler R, Bielinska M, Schoeller EL, Moley KH, Toppari J, Heikinheimo M, Wilson DB. GATA4 regulates Sertoli cell function and fertility in adult male mice. Mol Cell Endocrinol. 2011;333:85–95. doi: 10.1016/j.mce.2010.12.019. [DOI] [PMC free article] [PubMed] [Google Scholar]
  189. Labrie F, Legace L, Ferland L, Kelly PA, Drouin J, Massicotte J, Bonne C, Raynaud JP, Dorrington JH. Interactions between LHRH, sex steroids and “Inhibin” in the control of LH and FSH secretion. Int J Androl. 1978;1:81–89. [Google Scholar]
  190. Lacerda SM, Costa GM, Campos-Junior PH, Segatelli TM, Yazawa R, Takeuchi Y, Morita T, Yoshizaki G, Franca LR. Germ cell transplantation as a potential biotechnological approach to fish reproduction. Fish Physiol Biochem. 2013;39:3–11. doi: 10.1007/s10695-012-9606-4. [DOI] [PubMed] [Google Scholar]
  191. Lacerda SM, Costa GM, de Franca LR. Biology and identity of fish spermatogonial stem cell. Gen Comp Endocrinol. 2014;207:56–65. doi: 10.1016/j.ygcen.2014.06.018. [DOI] [PubMed] [Google Scholar]
  192. Lacroix M, Parvinen M, Fritz IB. Localization of testicular plasminogen activator in discrete portions (stage VII and VIII) of the seminiferous tubule. Biol Reprod. 1981;25:143–146. doi: 10.1095/biolreprod25.1.143. [DOI] [PubMed] [Google Scholar]
  193. Leal MC, Cardoso ER, Nobrega RH, Batlouni SR, Bogerd J, Franca LR, Schulz RW. Histological and stereological evaluation of zebrafish (Danio rerio) spermatogenesis with an emphasis on spermatogonial generations. Biol Reprod. 2009;81:177–187. doi: 10.1095/biolreprod.109.076299. [DOI] [PubMed] [Google Scholar]
  194. Leblond CP, Clermont Y. Definition of the stages of the cycle of the seminiferous epithelium in the rat. Ann N Y Acad Sci. 1952;55:548–573. doi: 10.1111/j.1749-6632.1952.tb26576.x. [DOI] [PubMed] [Google Scholar]
  195. Leichtmann-Bardoogo Y, Cohen LA, Weiss A, Marohn B, Schubert S, Meinhardt A, Meyron-Holtz EG. Compartmentalization and regulation of iron metabolism proteins protect male germ cells from iron overload. Am J Physiol Endocrinol Metab. 2012;302:E1519–E1530. doi: 10.1152/ajpendo.00007.2012. [DOI] [PubMed] [Google Scholar]
  196. Li H, Papadopoulos V, Vidic B, Dym M, Culty M. Regulation of rat testis gonocyte proliferation by platelet-derived growth factor and estradiol: identification of signaling mechanisms involved. Endocrinology. 1997;138:1289–1298. doi: 10.1210/endo.138.3.5021. [DOI] [PubMed] [Google Scholar]
  197. Li N, Mruk DD, Cheng CY. Actin binding proteins in blood-testis barrier function. Curr Opin Endocrinol Diabetes Obes. 2015;22:238–247. doi: 10.1097/MED.0000000000000155. [DOI] [PMC free article] [PubMed] [Google Scholar]
  198. Lie PP, Cheng CY, Mruk DD. Signalling pathways regulating the blood-testis barrier. Int J Biochem Cell Biol. 2013;45:621–625. doi: 10.1016/j.biocel.2012.12.009. [DOI] [PMC free article] [PubMed] [Google Scholar]
  199. Lie PP, Mruk DD, Lee WM, Cheng CY. Cytoskeletal dynamics and spermatogenesis. Philos Trans R Soc Lond B Biol Sci. 2010;365:1581–1592. doi: 10.1098/rstb.2009.0261. [DOI] [PMC free article] [PubMed] [Google Scholar]
  200. Loveland KL, Hedger MP. Activins and inhibins in Sertoli cell biology: implications for testis development and function. In: Griswold M, editor. Sertoli Cell Biology. Elsevier Academic Press; Oxford; 2015. pp. 201–232. [Google Scholar]
  201. Loveland KL, Zlatic K, Stein-Oakley A, Risbridger G, deKretser DM. Platelet-derived growth factor ligand and receptor subunit mRNA in the Sertoli and Leydig cells of the rat testis. Mol Cell Endocrinol. 1995;108:155–159. doi: 10.1016/0303-7207(94)03471-5. [DOI] [PubMed] [Google Scholar]
  202. Lu N, Sargent KM, Clopton DT, Pohlmeier WE, Brauer VM, McFee RM, Weber JS, Ferrara N, Silversides DW, Cupp AS. Loss of vascular endothelial growth factor A (VEGFA) isoforms in the testes of male mice causes subfertility, reduces sperm numbers, and alters expression of genes that regulate undifferentiated spermatogonia. Endocrinology. 2013;154:4790–4802. doi: 10.1210/en.2013-1363. [DOI] [PMC free article] [PubMed] [Google Scholar]
  203. Lucas TF, Pimenta MT, Pisolato R, Lazari MF, Porto CS. 17beta-estradiol signaling and regulation of Sertoli cell function. Spermatogenesis. 2011;1:318–324. doi: 10.4161/spmg.1.4.18903. [DOI] [PMC free article] [PubMed] [Google Scholar]
  204. Lui WY, Cheng CY. Transcription regulation in spermatogenesis. In: Cheng CY, editor. Molecular Mechanisms in Spermatogenesis. Landes Bioscience and Springer Science+Business Media; Texas; 2008. [Google Scholar]
  205. Lui WY, Cheng CY. Transcriptional regulation of cell adhesion at the blood-testis barrier and spermatogenesis in the testis. Adv Exp Med Biol. 2012;763:281–294. doi: 10.1007/978-1-4614-4711-5_14. [DOI] [PMC free article] [PubMed] [Google Scholar]
  206. Lui WY, Mruk D, Lee WM, Cheng CY. Sertoli cell tight junction dynamics: their regulation during spermatogenesis. Biol Reprod. 2003;68:1087–1097. doi: 10.1095/biolreprod.102.010371. [DOI] [PubMed] [Google Scholar]
  207. Lyon KRP, Bosseboeuf E, Vogl AW. An alternative model of tubulobulbar complex internalization during junction remodeling in the seminiferous epithelium of the rat testis. Biol Reprod. 2015;93:1–11. doi: 10.1095/biolreprod.115.128942. [DOI] [PubMed] [Google Scholar]
  208. Magrek S, Jost A. Sertoli cells and testicular differentiation in the rat fetus. J Electron Microsc Tech. 1991;19:172–188. doi: 10.1002/jemt.1060190205. [DOI] [PubMed] [Google Scholar]
  209. Mallidis C, Baker HW. Fine needle tissue aspiration biopsy of the testis. Fertil Steril. 1994;61:367–375. [PubMed] [Google Scholar]
  210. Mather JP, Gunsalus GL, Musto NA, Cheng CY, Parvinen M, Wright W, Perez-Infante V, Margioris A, Liotta A, Becker R, Krieger DT, Bardin CW. The hormonal and cellular control of Sertoli cell secretion. J Steroid Biochem. 1983;19:41–51. [PubMed] [Google Scholar]
  211. Matson CK, Murphy MW, Sarver AL, Griswold MD, Bardwell VJ, Zarkower D. DMRT1 prevents female reprogramming in the postnatal mammalian testis. Nature. 2011;476:101–104. doi: 10.1038/nature10239. [DOI] [PMC free article] [PubMed] [Google Scholar]
  212. Matta SL, Vilela DA, Godinho HP, Franca LR. The goitrogen 6-n-propyl-2-thiouracil (PTU) given during testis development increases Sertoli and germ cell numbers per cyst in fish: the tilapia (Oreochromis niloticus) model. Endocrinology. 2002;143:970–978. doi: 10.1210/endo.143.3.8666. [DOI] [PubMed] [Google Scholar]
  213. McClusky LM, Patrick S, Barnhoorn IE, van Dyk JC, de Jager C, Bornman MS. Immunohistochemical study of nuclear changes associated with male germ cell death and spermiogenesis. J Mol Histol. 2009;40:287–299. doi: 10.1007/s10735-009-9240-3. [DOI] [PubMed] [Google Scholar]
  214. McGuinness MP, Griswold MD. Interactions between Sertoli cells and germ cells in the testis. Semin Dev Biol. 1994;5:61–66. [Google Scholar]
  215. McKinnell C, Brackenbury ET, Qureshi SJ, Hargreave TB, Sharpe RM. Comparative analysis of proteins secreted in vitro by isolated seminiferous tubules from man and the rat. Int J Androl. 1995;18:103–111. doi: 10.1111/j.1365-2605.1995.tb00394.x. [DOI] [PubMed] [Google Scholar]
  216. Means AR, Dedman JR, Tash JS, Tindall DJ, van Sickle M, Welsh MJ. Regulation of the testis sertoli cell by follicle stimulating hormone. Annu Rev Physiol. 1980;42:59–70. doi: 10.1146/annurev.ph.42.030180.000423. [DOI] [PubMed] [Google Scholar]
  217. Means AR, Fakunding JL, Huckins C, Tindall DJ, Vitale R. Follicle-stimulating hormone, the Sertoli cell, and spermatogenesis. Recent Prog Horm Res. 1976;32:477–527. doi: 10.1016/b978-0-12-571132-6.50027-0. [DOI] [PubMed] [Google Scholar]
  218. Meinhardt A, Hedger MP. Immunological, paracrine and endocrine aspects of testicular immune privilege. Mol Cell Endocrinol. 2011;335:60–68. doi: 10.1016/j.mce.2010.03.022. [DOI] [PubMed] [Google Scholar]
  219. Meistrich ML, Hess RA. Assessment of spermatogenesis through staging of seminiferous tubules. Methods Mol Biol. 2013;927:299–307. doi: 10.1007/978-1-62703-038-0_27. [DOI] [PubMed] [Google Scholar]
  220. Melo MC, van Dijk P, Andersson E, Nilsen TO, Fjelldal PG, Male R, Nijenhuis W, Bogerd J, de Franca LR, Taranger GL, Schulz RW. Androgens directly stimulate spermatogonial differentiation in juvenile Atlantic salmon (Salmo salar) Gen Comp Endocrinol. 2015;211:52–61. doi: 10.1016/j.ygcen.2014.11.015. [DOI] [PubMed] [Google Scholar]
  221. Meng J, Greenlee AR, Taub CJ, Braun RE. Sertoli cell-specific deletion of the androgen receptor compromises testicular immune privilege in mice. Biol Reprod. 2011;85:254–260. doi: 10.1095/biolreprod.110.090621. [DOI] [PMC free article] [PubMed] [Google Scholar]
  222. Meng J, Holdcraft RW, Shima JE, Griswold MD, Braun RE. Androgens regulate the permeability of the blood-testis barrier. Proc Natl Acad Sci USA. 2005;102:16696–16700. doi: 10.1073/pnas.0506084102. [DOI] [PMC free article] [PubMed] [Google Scholar]
  223. Meng X, Lindahl M, Hyvonen ME, Parvinen M, de Rooij DG, Hess MW, Raatikainen-Ahokas A, Sainio K, Rauvala H, Lakso M, Pichel JG, Westphal H, Saarma M, Sariola H. Regulation of cell fate decision of undifferentiated spermatogonia by GDNF. Science. 2000;287:1489–1493. doi: 10.1126/science.287.5457.1489. [DOI] [PubMed] [Google Scholar]
  224. Miles DC, van den Bergen JA, Wakeling SI, Anderson RB, Sinclair AH, Western PS. The proto-oncogene Ret is required for male foetal germ cell survival. Dev Biol. 2012;365:101–109. doi: 10.1016/j.ydbio.2012.02.014. [DOI] [PubMed] [Google Scholar]
  225. Mital P, Hinton BT, Dufour JM. The blood-testis and blood-epididymis barriers are more than just their tight junctions. Biol Reprod. 2011;84:851–858. doi: 10.1095/biolreprod.110.087452. [DOI] [PMC free article] [PubMed] [Google Scholar]
  226. Mital P, Kaur G, Dufour J. Immunoprotective Sertoli cells: making allogeneic and xenogeneic transplantation feasible. Reproduction. 2010;139:495–504. doi: 10.1530/REP-09-0384. [DOI] [PubMed] [Google Scholar]
  227. Mital P, Kaur G, Dufour JM. Immunoprotective sertoli cells: making allogeneic and xenogeneic transplantation feasible. Reproduction. 2010;139:495–504. doi: 10.1530/REP-09-0384. [DOI] [PubMed] [Google Scholar]
  228. Miura C, Ohta T, Ozaki Y, Tanaka H, Miura T. Trypsin is a multifunctional factor in spermatogenesis. Proc Natl Acad Sci USA. 2009;106:20972–20977. doi: 10.1073/pnas.0907631106. [DOI] [PMC free article] [PubMed] [Google Scholar]
  229. Miura T, Ohta T, Miura CI, Yamauchi K. Complementary deoxyribonucleic acid cloning of spermatogonial stem cell renewal factor. Endocrinology. 2003;144:5504–5510. doi: 10.1210/en.2003-0800. [DOI] [PubMed] [Google Scholar]
  230. Molyneaux KA, Zinszner H, Kunwar PS, Schaible K, Stebler J, Sunshine MJ, O’Brien W, Raz E, Littman D, Wylie C, Lehmann R. The chemokine SDF1/CXCL12 and its receptor CXCR4 regulate mouse germ cell migration and survival. Development. 2003;130:4279–4286. doi: 10.1242/dev.00640. [DOI] [PubMed] [Google Scholar]
  231. Monesi V. Synthetic activities during spermatogenesis in the mouse RNA and protein. Exp Cell Res. 1965;39:197–224. doi: 10.1016/0014-4827(65)90023-6. [DOI] [PubMed] [Google Scholar]
  232. Montgomery JTH. Differentiation of the human cells of Sertoli. Biol Bull. 1911;21:367–388. [Google Scholar]
  233. Morais RD, Nóbrega RH, Gómez-Gónzalez NE, Schmidt R, Bogerd J, Franca LR, Schulz RW. Thyroid hormone stimulates the proliferation of Sertoli cells and single type A spermatogonia in adult zebrafish (Danio rerio) testis. Endocrinology. 2013;154:4365–4376. doi: 10.1210/en.2013-1308. [DOI] [PubMed] [Google Scholar]
  234. Morales C, Clermont Y. Structural changes of the Sertoli cell during the cycle of the seminiferous epithelium. In: Russell LD, Griswold MD, editors. The Sertoli Cell. Cache River Press; Clearwater, FL: 1993. pp. 305–329. [Google Scholar]
  235. Morales C, Griswold MD. Retinol-induced stage synchronization in seminiferous tubules of the rat. Endocrinology. 1987;121:432–434. doi: 10.1210/endo-121-1-432. [DOI] [PubMed] [Google Scholar]
  236. Morrow CM, Tyagi G, Simon L, Carnes K, Murphy KM, Cooke PS, Hofmann MC, Hess RA. Claudin 5 expression in mouse seminiferous epithelium is dependent upon the transcription factor ets variant 5 and contributes to blood-testis barrier function. Biol Reprod. 2009;81:871–879. doi: 10.1095/biolreprod.109.077040. [DOI] [PMC free article] [PubMed] [Google Scholar]
  237. Mosher KI, Andres RH, Fukuhara T, Bieri G, Hasegawa-Moriyama M, He Y, Guzman R, Wyss-Coray T. Neural progenitor cells regulate microglia functions and activity. Nat Neurosci. 2012;15:1485–1487. doi: 10.1038/nn.3233. [DOI] [PMC free article] [PubMed] [Google Scholar]
  238. Mruk DD. Emergent roles for intercellular adhesion molecule-1 in the restructuring of the blood-testis barrier during spermatogenesis in the mammal. Histol Histopathol. 2015;31:159–166. doi: 10.14670/HH-11-672. [DOI] [PubMed] [Google Scholar]
  239. Mruk DD, Cheng CY. Cell-cell interactions at the ectoplasmic specialization in the testis. Trends Endocrinol Metab. 2004a;15:439–447. doi: 10.1016/j.tem.2004.09.009. [DOI] [PubMed] [Google Scholar]
  240. 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. 2004b;25:747–806. doi: 10.1210/er.2003-0022. [DOI] [PubMed] [Google Scholar]
  241. Mruk DD, Cheng CY. Tight junctions in the testis: new perspectives. Philos Trans R Soc Lond B Biol Sci. 2010;365:1621–1635. doi: 10.1098/rstb.2010.0010. [DOI] [PMC free article] [PubMed] [Google Scholar]
  242. Mruk DD, Cheng CY. The mammalian blood-testis barrier: its biology and regulation. Endocr Rev. 2015;36:564–591. doi: 10.1210/er.2014-1101. [DOI] [PMC free article] [PubMed] [Google Scholar]
  243. Mulholland DJ, Dedhar S, Vogl AW. Rat seminiferous epithelium contains a unique junction (Ectoplasmic specialization) with signaling properties both of cell/cell and cell/matrix junctions. Biol Reprod. 2001;64:396–407. doi: 10.1095/biolreprod64.1.396. [DOI] [PubMed] [Google Scholar]
  244. Mullaney BP, Skinner MK. Growth factors as mediators of testicular cell-cell interactions. Baillieres Clin Endocrinol Metab. 1991;5:771–790. doi: 10.1016/s0950-351x(10)80014-x. [DOI] [PubMed] [Google Scholar]
  245. Mullaney BP, Skinner MK. Basic fibroblast growth factor (bFGF) gene expression and protein production during pubertal development of the seminiferous tubule: follicle-stimulating hormone-induced Sertoli cell bFGF expression. Endocrinology. 1992;131:2928–2934. doi: 10.1210/endo.131.6.1446630. [DOI] [PubMed] [Google Scholar]
  246. Murphy CJ, Richburg JH. Implications of Sertoli cell induced germ cell apoptosis to testicular pathology. Spermatogenesis. 2014;4:e979110. doi: 10.4161/21565562.2014.979110. [DOI] [PMC free article] [PubMed] [Google Scholar]
  247. Nagano M, Ryu BY, Brinster CJ, Avarbock MR, Brinster RL. Maintenance of mouse male germ line stem cells in vitro. Biol Reprod. 2003;68:2207–2214. doi: 10.1095/biolreprod.102.014050. [DOI] [PubMed] [Google Scholar]
  248. Nakajima S, Hayashi M, Kouguchi T, Yamaguchi K, Miwa M, Yoshizaki G. Expression patterns of gdnf and gfralpha1 in rainbow trout testis. Gene Expr Patterns. 2014;14:111–120. doi: 10.1016/j.gep.2014.01.006. [DOI] [PubMed] [Google Scholar]
  249. Nalbandian A, Dettin L, Dym M, Ravindranath N. Expression of vascular endothelial growth factor receptors during male germ cell differentiation in the mouse. Biol Reprod. 2003;69:985–994. doi: 10.1095/biolreprod.102.013581. [DOI] [PubMed] [Google Scholar]
  250. Naughton CK, Jain S, Strickland AM, Gupta A, Milbrandt J. Glial cell-line derived neurotrophic factor-mediated RET signaling regulates spermatogonial stem cell fate. Biol Reprod. 2006;74:314–321. doi: 10.1095/biolreprod.105.047365. [DOI] [PubMed] [Google Scholar]
  251. Negrini F, Pugliese A, Ferrari A, Sertoli E. Onoranze al Prof. Enrico Sertoli. La Clinica Veterinaria. 1908;31:49–62. [Google Scholar]
  252. Nicander L. An electron microscopical study of cell contacts in the seminiferous tubules of some mammals. Z Zellforsch Mikrosk Anat. 1967;83:375–397. doi: 10.1007/BF00336866. [DOI] [PubMed] [Google Scholar]
  253. Nicholls PK, Stanton PG, Chen JL, Olcorn JS, Haverfield JT, Qian H, Walton KL, Gregorevic P, Harrison CA. Activin signaling regulates Sertoli cell differentiation and function. Endocrinology. 2012;153:6065–6077. doi: 10.1210/en.2012-1821. [DOI] [PubMed] [Google Scholar]
  254. Nikolova DB, Martinova YS, Seidensticker M, Bellve AR. Leukaemia inhibitory factor stimulates proliferation of prospermatogonial stem cells. Reprod Fertil Dev. 1997;9:717–721. doi: 10.1071/r96116. [DOI] [PubMed] [Google Scholar]
  255. Nitta T, Hata M, Gotoh S, Seo Y, Sasaki H, Hashimoto N, Furuse M, Tsukita S. Size-selective loosening of the blood-brain barrier in claudin-5-deficient mice. J Cell Biol. 2003;161:653–660. doi: 10.1083/jcb.200302070. [DOI] [PMC free article] [PubMed] [Google Scholar]
  256. Nóbrega RH, de Souza Morais RD, Crespo D, de Waal PP, de França LR, Schulz RW, Bogerd J. Fsh stimulates spermatogonial proliferation and differentiation in zebrafish via Igf3. Endocrinology. 2015;156:3804–3817. doi: 10.1210/en.2015-1157. [DOI] [PubMed] [Google Scholar]
  257. Nóbrega RH, Greebe CD, van de Kant H, Bogerd J, de França LR, Schulz RW. Spermatogonial stem cell niche and spermatogonial stem cell transplantation in zebrafish. PLoS ONE. 2010;5(9) doi: 10.1371/journal.pone.0012808. [DOI] [PMC free article] [PubMed] [Google Scholar]
  258. O’Donnell L, Nicholls PK, O’Bryan MK, McLachlan RI, Stanton PG. Spermiation: the process of sperm release. Spermatogenesis. 2011;1:14–35. doi: 10.4161/spmg.1.1.14525. [DOI] [PMC free article] [PubMed] [Google Scholar]
  259. O’Donnell L, O’Bryan MK. Microtubules and spermatogenesis. Semin Cell Dev Biol. 2014;30:45–54. doi: 10.1016/j.semcdb.2014.01.003. [DOI] [PubMed] [Google Scholar]
  260. O’Rand MG, Romrell LJ. Appearance of cell surface auto- and isoantigens during spermatogenesis in the rabbit. Dev Biol. 1977;55:347–358. doi: 10.1016/0012-1606(77)90178-6. [DOI] [PubMed] [Google Scholar]
  261. Oatley JM, Brinster RL. The germline stem cell niche unit in mammalian testes. Physiol Rev. 2012;92:577–595. doi: 10.1152/physrev.00025.2011. [DOI] [PMC free article] [PubMed] [Google Scholar]
  262. Oatley MJ, Racicot KE, Oatley JM. Sertoli cells dictate spermatogonial stem cell niches in the mouse testis. Biol Reprod. 2011;84:639–645. doi: 10.1095/biolreprod.110.087320. [DOI] [PMC free article] [PubMed] [Google Scholar]
  263. Oliveira PF, Martins AD, Moreira AC, Cheng CY, Alves MG. The Warburg effect revisited–lesson from the Sertoli cell. Med Res Rev. 2015;35:126–151. doi: 10.1002/med.21325. [DOI] [PMC free article] [PubMed] [Google Scholar]
  264. Orth J. Proliferation of Sertoli cells in fetal and postnatal rats: a quantitative autoradiographic study. Anat Rec. 1982;203:485–492. doi: 10.1002/ar.1092030408. [DOI] [PubMed] [Google Scholar]
  265. Orth JM. The role of follicle-stimulating hormone in controlling Sertoli cell proliferation in testes of fetal rats. Endocrinology. 1984;115:1248–1255. doi: 10.1210/endo-115-4-1248. [DOI] [PubMed] [Google Scholar]
  266. Orth JM, Jester WF, Li LH, Laslett AL. Gonocyte-Sertoli cell interactions during development of the neonatal rodent testis. Curr Top Dev Biol. 2000;50:103–124. doi: 10.1016/s0070-2153(00)50006-4. [DOI] [PubMed] [Google Scholar]
  267. Parvinen M. Regulation of the seminiferous epithelium. Endocr Rev. 1982;3:404–417. doi: 10.1210/edrv-3-4-404. [DOI] [PubMed] [Google Scholar]
  268. Parvinen M. Cyclic function of Sertoli cells. In: Russell LD, Griswold MD, editors. The Sertoli Cell. Cache River Press; Clearwater, FL: 1993. pp. 39–86. [Google Scholar]
  269. Parvinen M, Vanha-Perttula T. Identification and enzyme quantitation of the stages of the seminiferous epithelial wave in the rat. Anat Rec. 1972;174:435–449. doi: 10.1002/ar.1091740404. [DOI] [PubMed] [Google Scholar]
  270. Parvinen M, Vihko KK, Toppari J. Cell interactions during the seminiferous epithelial cycle. Int Rev Cytol. 1986;104:115–151. doi: 10.1016/s0074-7696(08)61925-7. [DOI] [PubMed] [Google Scholar]
  271. Pawar HS, Wrobel KH. Quantitative aspects of water buffalo (Bubalus bubalis) spermatogenesis. Arch Histol Cytol. 1991;54:491–509. doi: 10.1679/aohc.54.491. [DOI] [PubMed] [Google Scholar]
  272. Payne CJ, Gallagher SJ, Foreman O, Dannenberg JH, Depinho RA, Braun RE. Sin3a is required by sertoli cells to establish a niche for undifferentiated spermatogonia, germ cell tumors, and spermatid elongation. Stem Cells. 2010;28:1424–1434. doi: 10.1002/stem.464. [DOI] [PMC free article] [PubMed] [Google Scholar]
  273. Pelletier J, Schalling M, Buckler AJ, Rogers A, Haber DA, Housman D. Expression of the Wilms’ tumor gene WT1 in the murine urogenital system. Genes Dev. 1991;5:1345–1356. doi: 10.1101/gad.5.8.1345. [DOI] [PubMed] [Google Scholar]
  274. Pelletier RM. Cyclic formation and decay of the blood-testis barrier in the mink (Mustela vison), a seasonal breeder. Am J Anat. 1986;175:91–117. doi: 10.1002/aja.1001750109. [DOI] [PubMed] [Google Scholar]
  275. Pelletier RM. The blood-testis barrier: the junctional permeability, the proteins and the lipids. Prog Histochem Cytochem. 2011;46:49–127. doi: 10.1016/j.proghi.2011.05.001. [DOI] [PubMed] [Google Scholar]
  276. Pelletier RM, Byers SW. The blood-testis barrier and Sertoli cell junctions: structural considerations. Microsc Res Tech. 1992;20:3–33. doi: 10.1002/jemt.1070200104. [DOI] [PubMed] [Google Scholar]
  277. Pelliniemi LJ, KFrojdman K, Paranko J. Embryological and prenatal development and function of Sertoli cells. In: Russell LD, Griswold MD, editors. The Sertoli Cell. Cache River Press; Clearwater, FL: 1993. pp. 87–113. [Google Scholar]
  278. Pesce M, Farrace MG, Piacentini M, Dolci S, De Felici M. Stem cell factor and leukemia inhibitory factor promote primordial germ cell survival by suppressing programmed cell death (apoptosis) Development. 1993;118:1089–1094. doi: 10.1242/dev.118.4.1089. [DOI] [PubMed] [Google Scholar]
  279. Picard JY, Josso N. Purification of testicular anti-Mullerian hormone allowing direct visualization of the pure glycoprotein and determination of yield and purification factor. Mol Cell Endocrinol. 1984;34:23–29. doi: 10.1016/0303-7207(84)90155-2. [DOI] [PubMed] [Google Scholar]
  280. Piquet-Pellorce C, Dorval-Coiffec I, Pham MD, Jegou B. Leukemia inhibitory factor expression and regulation within the testis. Endocrinology. 2000;141:1136–1141. doi: 10.1210/endo.141.3.7399. [DOI] [PubMed] [Google Scholar]
  281. Pitetti JL, Calvel P, Zimmermann C, Conne B, Papaioannou MD, Aubry F, Cederroth CR, Urner F, Fumel B, Crausaz M, Docquier M, Herrera PL, Pralong F, Germond M, Guillou F, Jegou B, Nef S. An essential role for insulin and IGF1 receptors in regulating sertoli cell proliferation, testis size, and FSH action in mice. Mol Endocrinol. 2013;27:814–827. doi: 10.1210/me.2012-1258. [DOI] [PMC free article] [PubMed] [Google Scholar]
  282. Prat F, Sumpter JP, Tyler CR. Validation of radioimmunoassays for two salmon gonadotropins (GTH I and GTH II) and their plasma concentrations throughout the reproductivecycle in male and female rainbow trout (Oncorhynchus mykiss) Biol Reprod. 1996;54:1375–1382. doi: 10.1095/biolreprod54.6.1375. [DOI] [PubMed] [Google Scholar]
  283. Pudney J. Comparative cytology of the non-mammalian Sertoli cell. In: Griswold MD, Russell LD, editors. The Sertoli Cell. Cache River Press; Clearwater, FL: 1993. pp. 611–658. [Google Scholar]
  284. Qian X, Mruk DD, Cheng YH, Tang EI, Han D, Lee WM, Wong EW, Cheng CY. Actin binding proteins, spermatid transport and spermiation. Semin Cell Dev Biol. 2014;30:75–85. doi: 10.1016/j.semcdb.2014.04.018. [DOI] [PMC free article] [PubMed] [Google Scholar]
  285. Ramaiah M, Wilkinson MF. MicroRNAs and Sertoli cells. In: Griswold M, editor. Sertoli Cell Biology. Elsevier Academic Press; Oxford: 2015. pp. 307–332. [Google Scholar]
  286. Rambourg A, Clermont Y, Hermo L. Three-dimensional architecture of the golgi apparatus in Sertoli cells of the rat. Am J Anat. 1979;154:455–476. doi: 10.1002/aja.1001540402. [DOI] [PubMed] [Google Scholar]
  287. Rato L, Socorro S, Cavaco JE, Oliveira PF. Tubular fluid secretion in the seminiferous epithelium: ion transporters and aquaporins in Sertoli cells. J Membr Biol. 2010;236:215–224. doi: 10.1007/s00232-010-9294-x. [DOI] [PubMed] [Google Scholar]
  288. Raymond CS, Kettlewell JR, Hirsch B, Bardwell VJ, Zarkower D. Expression of Dmrt1 in the genital ridge of mouse and chicken embryos suggests a role in vertebrate sexual development. Dev Biol. 1999;215:208–220. doi: 10.1006/dbio.1999.9461. [DOI] [PubMed] [Google Scholar]
  289. Raymond CS, Shamu CE, Shen MM, Seifert KJ, Hirsch B, Hodgkin J, Zarkower D. Evidence for evolutionary conservation of sex-determining genes. Nature. 1998;391:691–695. doi: 10.1038/35618. [DOI] [PubMed] [Google Scholar]
  290. Regaud C. Sur la morphologie de la cellule de Sertoli et sur son role dans la spermatogénese chez les mammiféres. Comptes rendus de l’Association des anatomistes. 1899;1:21–31. [Google Scholar]
  291. Regaud CL. Études sur la structure des tubes séminifères et sur la spermatogénèse chez les mammifères. Archives d’Anatomie Microscopique. 1901;4:101–156. [Google Scholar]
  292. Reis MM, Moreira AC, Sousa M, Mathur PP, Oliveira PF, Alves MG. Sertoli cell as a model in male reproductive toxicology: advantages and disadvantages. J Appl Toxicol. 2015;35:870–883. doi: 10.1002/jat.3122. [DOI] [PubMed] [Google Scholar]
  293. Rich KA, de Kretser DM. Spermatogenesis and the Sertoli cell. In: de Kretser DM, Burger HG, Hudson B, editors. The Pituitary and Testis: Clinical and Experimental Studies. Springer-Verlag; New York: 1983. pp. 84–105. [Google Scholar]
  294. Ritzen EM, Boitani C, Parvinen M, French FC, Feldman M. Stage-dependent secretion of ABP by rat seminiferous tubules. Mol Cell Endocrinol. 1982;25:25–33. doi: 10.1016/0303-7207(82)90166-6. [DOI] [PubMed] [Google Scholar]
  295. Ritzén EM, Hansson V, French FS. The Sertoli cell. In: Burger HG, de Kretser D, editors. The Testis. Raven Press; New York: 1981. pp. 171–194. [Google Scholar]
  296. Rival C, Lustig L, Iosub R, Guazzone VA, Schneider E, Meinhardt A, Fijak M. Identification of a dendritic cell population in normal testis and in chronically inflamed testis of rats with autoimmune orchitis. Cell Tissue Res. 2006;324:311–318. doi: 10.1007/s00441-005-0129-5. [DOI] [PubMed] [Google Scholar]
  297. Rivas B, Huang Z, Agoulnik AI. Normal fertility in male mice with deletion of beta-catenin gene in germ cells. Genesis. 2014;52:328–332. doi: 10.1002/dvg.22742. [DOI] [PubMed] [Google Scholar]
  298. Robaire B, Bayly SF. Testicular signaling: incoming and outgoing messages. Ann N Y Acad Sci. 1989;564:250–260. doi: 10.1111/j.1749-6632.1989.tb25902.x. [DOI] [PubMed] [Google Scholar]
  299. Roosen-Runge EC. Quantitative studies on spermatogenesis in the albino rat. III. Volume changes in the cells of the seminiferous tubules. Anat Rec. 1955;123:385–398. doi: 10.1002/ar.1091230402. [DOI] [PubMed] [Google Scholar]
  300. Rothbarth K, Kempf T, Juodka B, Glaser T, Stammer H, Werner D. Intracellular location and nuclear targeting of the Spi-1, Spi-2 and Spi-3 gene-derived serine protease inhibitors in non-secretory cells. Eur J Cell Biol. 2001;80:341–348. doi: 10.1078/0171-9335-00170. [DOI] [PubMed] [Google Scholar]
  301. Russell L. Observations on rat Sertoli ectoplasmic (‘junctional’) specializations in their association with germ cells of the rat testis. Tissue Cell. 1977;9:475–498. doi: 10.1016/0040-8166(77)90007-6. [DOI] [PubMed] [Google Scholar]
  302. Russell L, Clermont Y. Anchoring device between Sertoli cells and late spermatids in rat seminiferous tubules. Anat Rec. 1976;185:259–278. doi: 10.1002/ar.1091850302. [DOI] [PubMed] [Google Scholar]
  303. Russell L, Myers P, Ostenburg J, Malone J. Sertoli ectoplasmic specializations during spermatogenesis. Ann N Y Acad Sci. 1987;513:55–64. [Google Scholar]
  304. Russell LD. Sertoli-germ cell interactions: a review. Gamete Res. 1980;3:179–202. [Google Scholar]
  305. Russell LD. Spermiation – the sperm release process: ultrastructural observations and unresolved problems. In: Van Blerkom J, Motta PM, editors. Electron Microscopy in Biology and Medicine. Vol. 2. Martinus Nijhoff; Boston, MA: 1984. pp. 46–66. [Google Scholar]
  306. Russell LD. Form, dimensions, and cytology of mammalian Sertoli cells. In: Russell LD, Griswold MD, editors. The Sertoli Cell. Cache River Press; Clearwater, FL: 1993a. pp. 1–37. [Google Scholar]
  307. Russell LD. Morphological and functional evidence for Sertoli-germ cell relationships. In: Russell LD, Griswold MD, editors. The Sertoli Cell. Cache River Press; Clearwater, FL: 1993b. pp. 365–390. [Google Scholar]
  308. Russell LD. Role in spermiation. In: Russell LD, Griswold MD, editors. The Sertoli Cell. Cache River Press; Clearwater, FL: 1993c. pp. 269–304. [Google Scholar]
  309. Russell LD, Clermont Y. Degeneration of germ cells in normal, hypophysectomized and hormone treated hypophysectomized rats. Anat Rec. 1977;187:347–366. doi: 10.1002/ar.1091870307. [DOI] [PubMed] [Google Scholar]
  310. Russell LD, Ettlin RA, Sinha Hikim AP, Clegg ED. Histological and Histopathological Evaluation of the Testis. Cache River Press; Clearwater: 1990a. p. 286. [Google Scholar]
  311. Russell LD, Gardner RJ, Weber JE. Reconstruction of a type-B configuration monkey Sertoli cell: size, shape, and configurational and specialized cell-to-cell relationships. Am J Anat. 1986;175:73–90. doi: 10.1002/aja.1001750108. [DOI] [PubMed] [Google Scholar]
  312. Russell LD, Griswold MD. Preface. In: Russell LD, Griswold MD, editors. The Sertoli Cell. Cache River Press; Clearwater, FL: 1993a. pp. ix–xi. [Google Scholar]
  313. Russell LD, Griswold MD. The Sertoli Cell. Cache River Press; Clearwater, FL: 1993b. p. 801. [Google Scholar]
  314. Russell LD, Myer P, Ostenburg J, Malone J. Sertoli ectoplasmic specializations during spermatogenesis. In: Steinberger A, Steinberger E, editors. Testicular Development, Structure, and Function. Raven Press; New York: 1980. pp. 55–63. [Google Scholar]
  315. Russell LD, Peterson RN. Sertoli cell junctions: morphological and functional correlates. Int Rev Cytol. 1985;94:177–211. doi: 10.1016/s0074-7696(08)60397-6. [DOI] [PubMed] [Google Scholar]
  316. Russell LD, Ren HP, Sinha Hikim I, Schulze W, Sinha Hikim AP. A comparative study in twelve mammalian species of volume densities, volumes, and numerical densities of selected testis components, emphasizing those related to the Sertoli cell. Am J Anat. 1990b;188:21–30. doi: 10.1002/aja.1001880104. [DOI] [PubMed] [Google Scholar]
  317. Russell LD, Saxena NK, Turner TT. Cytoskeletal involvement in spermiation and sperm transport. Tissue Cell. 1989;21:361–379. doi: 10.1016/0040-8166(89)90051-7. [DOI] [PubMed] [Google Scholar]
  318. Russell LD, Steinberger A. Sertoli cells in culture: views from the perspectives of an in vivoist and an in vitroist. Biol Reprod. 1989;41:571–577. doi: 10.1095/biolreprod41.4.571. [DOI] [PubMed] [Google Scholar]
  319. Russell LD, Tallon-Doran M, Weber JE, Wong V, Peterson RN. Three-dimensional reconstruction of a rat stage V Sertoli cell: III. A study of specific cellular relationships. Am J Anat. 1983;167:181–192. doi: 10.1002/aja.1001670204. [DOI] [PubMed] [Google Scholar]
  320. Ryu BY, Orwig KE, Oatley JM, Avarbock MR, Brinster RL. Effects of aging and niche microenvironment on spermatogonial stem cell self-renewal. Stem Cells. 2006;24:1505–1511. doi: 10.1634/stemcells.2005-0580. [DOI] [PMC free article] [PubMed] [Google Scholar]
  321. Saitou M, Furuse M, Sasaki H, Schulzke JD, Fromm M, Takano H, Noda T, Tsukita S. Complex phenotype of mice lacking occludin, a component of tight junction strands. Mol Biol Cell. 2000;11:4131–4142. doi: 10.1091/mbc.11.12.4131. [DOI] [PMC free article] [PubMed] [Google Scholar]
  322. Sakaguchi T, Nishimoto M, Miyagi S, Iwama A, Morita Y, Iwamori N, Nakauchi H, Kiyonari H, Muramatsu M, Okuda A. Putative “stemness” gene jam-B is not required for maintenance of stem cell state in embryonic, neural, or hematopoietic stem cells. Mol Cell Biol. 2006;26:6557–6570. doi: 10.1128/MCB.00729-06. [DOI] [PMC free article] [PubMed] [Google Scholar]
  323. Sanborn BM, Caston LA, Buzek SW, Ussuf KK. Hormonal regulation of Sertoli cell function. Adv Exp Med Biol. 1987;219:561–588. doi: 10.1007/978-1-4684-5395-9_25. [DOI] [PubMed] [Google Scholar]
  324. Sanborn BM, Elkington ISH, Steinberger A, Steinberger E, Meistrich ML. Androgen binding in the testis: in vitro production of androgen binding protein (ABP) by Sertoli cell cultures and measurement of nuclear bound androgen by a nuclear exhange assay. In: French F, Hansson V, Ritzen E, Nayfeh S, editors. Hormonal Regulation of Spermatogenesis. Plenum; New York: 1975. pp. 293–310. [DOI] [PubMed] [Google Scholar]
  325. Sanz E, Evanoff R, Quintana A, Evans E, Miller JA, Ko C, Amieux PS, Griswold MD, McKnight GS. RiboTag analysis of actively translated mRNAs in Sertoli and Leydig cells in vivo. PLoS ONE. 2013;8:e66179. doi: 10.1371/journal.pone.0066179. [DOI] [PMC free article] [PubMed] [Google Scholar]
  326. Sar M, Hall SH, Wilson EM, French FS. Androgen regulation of Sertoli cells. In: Griswold MD, Russell LD, editors. The Sertoli Cell. Cache River Press; Clearwater, FL: 1993. pp. 509–516. [Google Scholar]
  327. Sar M, Lubahn DB, French FS, Wilson EM. Immunohistochemical localization of the androgen receptor in rat and human tissues. Endocrinology. 1990;127:3180–3186. doi: 10.1210/endo-127-6-3180. [DOI] [PubMed] [Google Scholar]
  328. Sato T, Aiyama Y, Ishii-Inagaki M, Hara K, Tsunekawa N, Harikae K, et al. Cyclical and patch-like GDNF distribution along the basal surface of Sertoli cells in mouse and hamster testes. PLoS ONE. 2011a;6:e28367. doi: 10.1371/journal.pone.0028367. [DOI] [PMC free article] [PubMed] [Google Scholar]
  329. Sato T, Katagiri K, Gohbara A, Inoue K, Ogonuki N, Ogura A, Kubota Y, Ogawa T. In vitro production of functional sperm in cultured neonatal mouse testes. Nature. 2011b;471:504–507. doi: 10.1038/nature09850. [DOI] [PubMed] [Google Scholar]
  330. Scadden DT. The stem-cell niche as an entity of action. Nature. 2006;441:1075–1079. doi: 10.1038/nature04957. [DOI] [PubMed] [Google Scholar]
  331. Schulz RW, de Franca LR, Lareyre JJ, Le Gac F, Chiarini-Garcia H, Nobrega RH, Miura T. Spermatogenesis in fish. Gen Comp Endocrinol. 2010;165:390–411. doi: 10.1016/j.ygcen.2009.02.013. [DOI] [PubMed] [Google Scholar]
  332. Schulz RW, Menting S, Bogerd J, Franca LR, Vilela DA, Godinho HP. Sertoli cell proliferation in the adult testis–evidence from two fish species belonging to different orders. Biol Reprod. 2005;73:891–898. doi: 10.1095/biolreprod.105.039891. [DOI] [PubMed] [Google Scholar]
  333. Schulz RW, van Dijk W, Chaves-Pozo E, Garcia-Lopez A, de Franca LR, Bogerd J. Sertoli cell proliferation in the adult testis is induced by unilateral gonadectomy in African catfish. Gen Comp Endocrinol. 2012;177:160–167. doi: 10.1016/j.ygcen.2012.03.001. [DOI] [PubMed] [Google Scholar]
  334. Schulze C, Holstein AF. Human Sertoli cell structure. In: Griswold MD, Russell LD, editors. The Sertoli Cell. Cache River Press; Clearwater, FL: 1993a. pp. 685–702. [Google Scholar]
  335. Schulze C, Holstein AF. Human Sertoli cells under pathological conditions. In: Griswold MD, Russell LD, editors. The Sertoli Cell. Cache River Press; Clearwater, FL: 1993b. pp. 597–610. [Google Scholar]
  336. Schulze C, Holstein AF, Schirren C, Korner F. On the morphology of the human Sertoli cells under normal conditions and in patients with impaired fertility. Andrologia. 1976;8:167–178. doi: 10.1111/j.1439-0272.1976.tb02128.x. [DOI] [PubMed] [Google Scholar]
  337. Sekido R, Lovell-Badge R. Sex determination involves synergistic action of SRY and SF1 on a specific Sox9 enhancer. Nature. 2008;453:930–934. doi: 10.1038/nature06944. [DOI] [PubMed] [Google Scholar]
  338. Selawry HP. Islet transplantation to immuneprivilged sites. In: Lanza RP, Chick WL, editors. Pancreatic Islet Transplantation: Immunomodulation of Pancreatic Islets. Landes/CRC Press; Austin, TX: 1994. pp. 75–86. [Google Scholar]
  339. Selawry HP, Cameron DF. Sertoli cell-enriched fractions in successful islet cell transplantation. Cell Transplant. 1993;2:123–129. doi: 10.1177/096368979300200206. [DOI] [PubMed] [Google Scholar]
  340. Sertoli E. Dell’esistenza di particolari cellule ramificate nei canalicoli seminiferi del testicolo umano. Morgagni. 1865;7:31–40. [Google Scholar]
  341. Setchell BP. The blood-testicular fluid barrier in sheep. J Physiol. 1967;189:63–65. [PubMed] [Google Scholar]
  342. Setchell BP. The secretion of fluid by the testes of rats, rams and goats with some observations on the effect of age, cryptorchidism and hypophysectomy. J Reprod Fert. 1970;23:79–85. doi: 10.1530/jrf.0.0230079. [DOI] [PubMed] [Google Scholar]
  343. Setchell BP. Secretions of the testis and epididymis. J Reprod Fertil. 1974;37:165–177. doi: 10.1530/jrf.0.0370165. [DOI] [PubMed] [Google Scholar]
  344. Setchell BP. The testis and tissue transplantation: historical aspects. J Reprod Immunol. 1990;18:1–8. doi: 10.1016/0165-0378(90)90020-7. [DOI] [PubMed] [Google Scholar]
  345. Setchell BP, Davies RV, Gladwell RT, Hinton BT, Main SJ, Pilsworth L, Waites GMH. The movement of fluid in the seminiferous tubules and rete testis. Ann Biol Anim biochem Biophys. 1978;18:623–632. [Google Scholar]
  346. Setchell BP, Dawson RM, White RW. The high concentration of free myo-inositol in rete testis fluid from rams. J Reprod Fertil. 1968;17:219–220. doi: 10.1530/jrf.0.0170219. [DOI] [PubMed] [Google Scholar]
  347. Setchell BP, Main SJ. The blood - testis barrier and steroids. Curr Top Mol Endocrinol. 1975;2:223–233. doi: 10.1007/978-1-4613-4440-7_16. [DOI] [PubMed] [Google Scholar]
  348. Shao M, Ghosh A, Cooke VG, Naik UP, Martin-DeLeon PA. JAM-A is present in mammalian spermatozoa where it is essential for normal motility. Dev Biol. 2008;313:246–255. doi: 10.1016/j.ydbio.2007.10.013. [DOI] [PMC free article] [PubMed] [Google Scholar]
  349. Sharpe R. Experimental evidence for Sertoli-germ cell and Sertoli-Leydig cell interactions. In: Russell LD, Griswold MD, editors. The Sertoli Cell. Cache River Press; Clearwater, FL: 1993. pp. 391–418. [Google Scholar]
  350. Sharpe R. Regulation of spermatogenesis. In: Knobil E, Neill J, editors. The Physiology of Reproduction. Vol. 2. Raven Press; New York: 1994. pp. 1363–1434. [Google Scholar]
  351. Sharpe RM. Bidirectional secretion by the Sertoli cell. Int J Androl. 1988;11:87–91. doi: 10.1111/j.1365-2605.1988.tb00983.x. [DOI] [PubMed] [Google Scholar]
  352. Sharpe RM. Monitoring of spermatogenesis in man–measurement of Sertoli cell- or germ cell-secreted proteins in semen or blood [editorial] Int J Androl. 1992;15:201–210. doi: 10.1111/j.1365-2605.1992.tb01340.x. [DOI] [PubMed] [Google Scholar]
  353. Sharpe RM, McKinnell C, Kivlin C, Fisher JS. Proliferation and functional maturation of Sertoli cells, and their relevance to disorders of testis function in adulthood. Reproduction. 2003;125:769–784. doi: 10.1530/rep.0.1250769. [DOI] [PubMed] [Google Scholar]
  354. Shibata N, Hamaguchi S. Evidence for the sexual bipotentiality of spermatogonia in the fish, Oryzias latipes. J Exp Zool. 1988;245:71–77. doi: 10.1002/jez.1402450111. [DOI] [PubMed] [Google Scholar]
  355. Shima JE, McLean DJ, McCarrey JR, Griswold MD. The murine testicular transcriptome: characterizing gene expression in the testis during the progression of spermatogenesis. Biol Reprod. 2004;71:319–330. doi: 10.1095/biolreprod.103.026880. [DOI] [PubMed] [Google Scholar]
  356. Shinohara T, Avarbock MR, Brinster RL. Beta1- and alpha6-integrin are surface markers on mouse spermatogonial stem cells. Proc Natl Acad Sci USA. 1999;96:5504–5509. doi: 10.1073/pnas.96.10.5504. [DOI] [PMC free article] [PubMed] [Google Scholar]
  357. Shinohara T, Orwig KE, Avarbock MR, Brinster RL. Remodeling of the postnatal mouse testis is accompanied by dramatic changes in stem cell number and niche accessibility. Proc Natl Acad Sci USA. 2001;98:6186–6191. doi: 10.1073/pnas.111158198. [DOI] [PMC free article] [PubMed] [Google Scholar]
  358. Silva CA, Cocuzza M, Carvalho JF, Bonfa E. Diagnosis and classification of autoimmune orchitis. Autoimmun Rev. 2014;13:431–434. doi: 10.1016/j.autrev.2014.01.024. [DOI] [PubMed] [Google Scholar]
  359. Silva FR, Leite LD, Wassermann GF. Rapid signal transduction in Sertoli cells. Eur J Endocrinol. 2002;147:425–433. doi: 10.1530/eje.0.1470425. [DOI] [PubMed] [Google Scholar]
  360. Simon L, Ekman GC, Garcia T, Carnes K, Zhang Z, Murphy T, Murphy KM, Hess RA, Cooke PS, Hofmann MC. ETV5 regulates sertoli cell chemokines involved in mouse stem/progenitor spermatogonia maintenance. Stem Cells. 2010;28:1882–1892. doi: 10.1002/stem.508. [DOI] [PMC free article] [PubMed] [Google Scholar]
  361. Simon L, Ekman GC, Tyagi G, Hess RA, Murphy KM, Cooke PS. Common and distinct factors regulate expression of mRNA for ETV5 and GDNF, Sertoli cell proteins essential for spermatogonial stem cell maintenance. Exp Cell Res. 2007;313:3090–3099. doi: 10.1016/j.yexcr.2007.05.002. [DOI] [PubMed] [Google Scholar]
  362. Sinha Hikim A, Amador A, Klemcke H, Bartke A, Russell L. Correlative morphology and endocrinology of Sertoli cells in hamster testes in active and inactive states of spermatogenesis. Endocrinology. 1989;125:1829–1843. doi: 10.1210/endo-125-4-1829. [DOI] [PubMed] [Google Scholar]
  363. Siu MK, Cheng CY. Extracellular matrix and its role in spermatogenesis. Adv Exp Med Biol. 2009;636:74–91. doi: 10.1007/978-0-387-09597-4_5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  364. Skaar KS, Nobrega RH, Magaraki A, Olsen LC, Schulz RW, Male R. Proteolytically activated, recombinant anti-mullerian hormone inhibits androgen secretion, proliferation, and differentiation of spermatogonia in adult zebrafish testis organ cultures. Endocrinology. 2011;152:3527–3540. doi: 10.1210/en.2010-1469. [DOI] [PubMed] [Google Scholar]
  365. Skinner MK. Cell-cell interactions in the testis. Ann N Y Acad Sci. 1987;513:158–171. doi: 10.1111/j.1749-6632.1987.tb25006.x. [DOI] [PubMed] [Google Scholar]
  366. Skinner MK. Secretion of growth factors and other regulatory factors. In: Griswold MD, Russell LD, editors. The Sertoli Cell. Cache River Press; Clearwater, FL: 1993a. pp. 237–248. [Google Scholar]
  367. Skinner MK. Setoli cell-peritubular myoid cell interactions. In: Griswold MD, Russell LD, editors. The Sertoli Cell. Cache River Press; Clearwater, FL: 1993b. pp. 477–484. [Google Scholar]
  368. Skinner MK, Griswold MD. Sertoli cells synthesize and secrete transferrin-like protein. J Biol Chem. 1980;255:9523–9525. [PubMed] [Google Scholar]
  369. Skinner MK, Griswold MD. Secretion of testicular transferrin by cultured Sertoli cells is regulated by hormones and retinoids. Biol Reprod. 1982;27:211–221. doi: 10.1095/biolreprod27.1.211. [DOI] [PubMed] [Google Scholar]
  370. Skinner MK, Griswold MD. Sertoli cells synthesize and secrete a ceruloplasmin-like protein. Biol Reprod. 1983;28:1225–1229. doi: 10.1095/biolreprod28.5.1225. [DOI] [PubMed] [Google Scholar]
  371. Skinner MK, Griswold MD. Sertoli Cell Biology. Elsevier Academic Press; San Diego: 2005. p. 494. [Google Scholar]
  372. Skinner MK, Takacs K, Coffey RJ. Transforming growth factor-alpha gene expression and action in the seminiferous tubule: peritubular cell-Sertoli cell interactions. Endocrinology. 1989;124:845–854. doi: 10.1210/endo-124-2-845. [DOI] [PubMed] [Google Scholar]
  373. Smith BE, Braun RE. Germ cell migration across Sertoli cell tight junctions. Science. 2012;338:798–802. doi: 10.1126/science.1219969. [DOI] [PMC free article] [PubMed] [Google Scholar]
  374. Smith EP, Hall SH, Monaco L, French FS, Wilson EM, Conti M. A rat Sertoli cell factor similar to basic fibroblast growth factor increases c-fos messenger ribonucleic acid in cultured Sertoli cells. Mol Endocrinol. 1989;3:954–961. doi: 10.1210/mend-3-6-954. [DOI] [PubMed] [Google Scholar]
  375. Smith LB, Walker WH. The regulation of spermatogenesis by androgens. Semin Cell Dev Biol. 2014;30:2–13. doi: 10.1016/j.semcdb.2014.02.012. [DOI] [PMC free article] [PubMed] [Google Scholar]
  376. Smith LB, Walker WH, O’Donnell L. Hormonal regulation of spermatogenesis through Sertoli cells by androgens and estrogens. In: Griswold M, editor. Sertoli Cell Biology. Elsevier Academic Press; Oxford; 2015. pp. 175–200. [Google Scholar]
  377. Snyder EM, Small C, Griswold MD. Retinoic acid availability drives the asynchronous initiation of spermatogonial differentiation in the mouse. Biol Reprod. 2010;83:783–790. doi: 10.1095/biolreprod.110.085811. [DOI] [PMC free article] [PubMed] [Google Scholar]
  378. Sowa Y, Imura T, Numajiri T, Nishino K, Fushiki S. Adipose-derived stem cells produce factors enhancing peripheral nerve regeneration: influence of age and anatomic site of origin. Stem Cells Dev. 2012;21:1852–1862. doi: 10.1089/scd.2011.0403. [DOI] [PubMed] [Google Scholar]
  379. Spradling A, Drummond-Barbosa D, Kai T. Stem cells find their niche. Nature. 2001;414:98–104. doi: 10.1038/35102160. [DOI] [PubMed] [Google Scholar]
  380. Statter MB, Fahrner KJ, Barksdale EM, Jr, Parks DE, Flavell RA, Donahoe PK. Correlation of fetal kidney and testis congenic graft survival with reduced major histocompatibility complex burden. Transplantation. 1989;47:651–660. doi: 10.1097/00007890-198904000-00017. [DOI] [PubMed] [Google Scholar]
  381. Steinberger A. Inhibin production by Sertoli cells in culture. J Reprod Fertil Suppl. 1979;26:31–45. [PubMed] [Google Scholar]
  382. Steinberger A, Jakubowiak A. Sertoli cell culture: historical perspective and review of methods. In: Russell LD, Griswold MD, editors. The Sertoli Cell. Cache River Press; Clearwater, FL: 1993. pp. 155–180. [Google Scholar]
  383. Steinberger A, Steinberger E. Tissue culture of male mammalian gonads. In Vitro. 1970;5:17–27. doi: 10.1007/BF02618371. [DOI] [PubMed] [Google Scholar]
  384. Steinberger A, Steinberger E. Testicular Development, Structure, and Function. Raven Press; New York: 1987. p. 536. [Google Scholar]
  385. Steinberger A, Steinberger E, Perloff WH. Mammalian testes in organ culture. Exp Cell Res. 1964;36:19–27. doi: 10.1016/0014-4827(64)90156-9. [DOI] [PubMed] [Google Scholar]
  386. Steinberger E. Hormonal regulation of the seminiferous tubule function. Curr Top Mol Endocrinol. 1975;2:337–352. doi: 10.1007/978-1-4613-4440-7_24. [DOI] [PubMed] [Google Scholar]
  387. Su W, Mruk DD, Cheng CY. Regulation of actin dynamics and protein trafficking during spermatogenesis–insights into a complex process. Crit Rev Biochem Mol Biol. 2013;48:153–172. doi: 10.3109/10409238.2012.758084. [DOI] [PMC free article] [PubMed] [Google Scholar]
  388. Svingen T, Koopman P. Building the mammalian testis: origins, differentiation, and assembly of the component cell populations. Genes Dev. 2013;27:2409–2426. doi: 10.1101/gad.228080.113. [DOI] [PMC free article] [PubMed] [Google Scholar]
  389. Sylvester SR. Secretion of transport and binding proteins. In: Russell LD, Griswold MD, editors. The Sertoli Cell. Cache River Press; Clearwater, FL: 1993. pp. 201–216. [Google Scholar]
  390. Sylvester SR, Griswold MD. The testicular iron shuttle: a “nurse” function of the Sertoli cells. J Androl. 1994;15:381–385. [PubMed] [Google Scholar]
  391. 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;113:29–39. doi: 10.1016/s0925-4773(02)00004-7. [DOI] [PubMed] [Google Scholar]
  392. Takashima S, Kanatsu-Shinohara M, Tanaka T, Morimoto H, Inoue K, Ogonuki N, Jijiwa M, Takahashi M, Ogura A, Shinohara T. Functional differences between GDNF-dependent and FGF2-dependent mouse spermatogonial stem cell self-renewal. Stem Cell Reports. 2015;4:489–502. doi: 10.1016/j.stemcr.2015.01.010. [DOI] [PMC free article] [PubMed] [Google Scholar]
  393. Tarulli GA, Stanton PG, Meachem SJ. Is the adult Sertoli cell terminally differentiated? Biol Reprod. 2012;87:11–11. doi: 10.1095/biolreprod.111.095091. [DOI] [PubMed] [Google Scholar]
  394. Teilum G. Estrogen-producing Sertoli cell tumors (androblastoma tubulare lipoides) of the human testis and ovary; homologous ovarian and testicular tumors. J Clin Endocrinol Metab. 1949;9:301–318. doi: 10.1210/jcem-9-4-301. [DOI] [PubMed] [Google Scholar]
  395. Thuillier R, Mazer M, Manku G, Boisvert A, Wang Y, Culty M. Interdependence of platelet-derived growth factor and estrogen-signaling pathways in inducing neonatal rat testicular gonocytes proliferation. Biol Reprod. 2010;82:825–836. doi: 10.1095/biolreprod.109.081729. [DOI] [PMC free article] [PubMed] [Google Scholar]
  396. Thuillier R, Wang Y, Culty M. Prenatal exposure to estrogenic compounds alters the expression pattern of platelet-derived growth factor receptors alpha and beta in neonatal rat testis: identification of gonocytes as targets of estrogen exposure. Biol Reprod. 2003;68:867–880. doi: 10.1095/biolreprod.102.009605. [DOI] [PubMed] [Google Scholar]
  397. Tindall DJ, Rowley DR, Lipshultz LI. Sertoli cell structure and function in vivo and in vitro. In: Lipshultz LI, Howards SS, editors. Infertility in the Male. Churchill Livingstone; New York: 1983. pp. 71–98. [Google Scholar]
  398. Tindall DJ, Rowley DR, Murthy L, Lipshultz LI, Chang CH. Structure and biochemistry of the Sertoli cell. Int Rev Cytol. 1985;94:127–149. doi: 10.1016/s0074-7696(08)60395-2. [DOI] [PubMed] [Google Scholar]
  399. Tindall DJ, Vitale R, Means AR. Androgen binding protein as a biochemical marker of formation of the blood-testis barrier. Endocrinology. 1975;97:636–648. doi: 10.1210/endo-97-3-636. [DOI] [PubMed] [Google Scholar]
  400. Toppari J, Kangasniemi M, Kaipia A, Mali P, Huhtaniemi I, Parvinen M. Stage- and cell-specific gene expression and hormone regulation of the seminiferous epithelium. J Electron Microsc Tech. 1991;19:203–214. doi: 10.1002/jemt.1060190207. [DOI] [PubMed] [Google Scholar]
  401. Toyama Y, Maekawa M, Yuasa S. Ectoplasmic specializations in the Sertoli cell: new vistas based on genetic defects and testicular toxicology. Anat Sci Int. 2003;78:1–16. doi: 10.1046/j.0022-7722.2003.00034.x. [DOI] [PubMed] [Google Scholar]
  402. Tung PS, Dorrington IH, Fritz IB. Responsiveness of cultured Sertoli cells to FSH. Proc Natl Acad Sci USA. 1975;72:1838–1842. doi: 10.1073/pnas.72.5.1838. [DOI] [PMC free article] [PubMed] [Google Scholar]
  403. Tung PS, Fritz IB. Specific surface antigens on rat pachytene spermatocytes and successive classes of germinal cells. Dev Biol. 1978;64:297–315. doi: 10.1016/0012-1606(78)90080-5. [DOI] [PubMed] [Google Scholar]
  404. Tung PS, Skinner MK, Fritz IB. Cooperativity between Sertoli cells and peritubular myoid cells in the formation of the basal lamina in the seminiferous tubule. Ann N Y Acad Sci. 1984;438:435–446. doi: 10.1111/j.1749-6632.1984.tb38304.x. [DOI] [PubMed] [Google Scholar]
  405. Ueno H, Mori H. Morphometrical analysis of Sertoli cell ultrastructure during the seminiferous epithelial cycle in rats. Biol Reprod. 1990;43:769–776. doi: 10.1095/biolreprod43.5.769. [DOI] [PubMed] [Google Scholar]
  406. Van Dissel-Emiliani FM, De Boer-Brouwer M, De Rooij DG. Effect of fibroblast growth factor-2 on Sertoli cells and gonocytes in coculture during the perinatal period. Endocrinology. 1996;137:647–654. doi: 10.1210/endo.137.2.8593814. [DOI] [PubMed] [Google Scholar]
  407. van Haaster LH, de Jong FH, Docter R, de Rooij DG. High neonatal triiodothyronine levels reduce the period of Sertoli cell proliferation and accelerate tubular lumen formation in the rat testis, and increase serum inhibin levels. Endocrinology. 1993;133:755–760. doi: 10.1210/endo.133.2.8344214. [DOI] [PubMed] [Google Scholar]
  408. Viger RS, Mertineit C, Trasler JM, Nemer M. Transcription factor GATA-4 is expressed in a sexually dimorphic pattern during mouse gonadal development and is a potent activator of the Mullerian inhibiting substance promoter. Development. 1998;125:2665–2675. doi: 10.1242/dev.125.14.2665. [DOI] [PubMed] [Google Scholar]
  409. Vilela DAR, Silva SGB, Peixoto MTD, Godinho HP, França LR. Spermatogenesis in teleost; insights from the Nile tilapia (Oreochromis niloticus) model. Fish Physiol Biochem. 2003;28:187–190. [Google Scholar]
  410. Vogl AW. Changes in the distribution of microtubules in rat Sertoli cells during spermatogenesis. Anat Rec. 1988;222:34–41. doi: 10.1002/ar.1092220107. [DOI] [PubMed] [Google Scholar]
  411. Vogl AW. Distribution and function of organized concentrations of actin filaments in mammalian spermatogenic cells and Sertoli cells. Int Rev Cytol. 1989;119:1–56. doi: 10.1016/s0074-7696(08)60648-8. [DOI] [PubMed] [Google Scholar]
  412. Vogl AW, Du M, Wang XY, Young JS. Novel clathrin/actin-based endocytic machinery associated with junction turnover in the seminiferous epithelium. Semin Cell Dev Biol. 2014;30:55–64. doi: 10.1016/j.semcdb.2013.11.002. [DOI] [PubMed] [Google Scholar]
  413. Vogl AW, Lin YC, Dym M, Fawcett DW. Sertoli cells of the golden-mantled ground squirrel (Spermophilus lateralis): a model system for the study of shape change. Am J Anat. 1983a;168:83–98. doi: 10.1002/aja.1001680109. [DOI] [PubMed] [Google Scholar]
  414. Vogl AW, Linck RW, Dym M. Colchicine-induced changes in the cytoskeleton of the golden-mantled ground squirrel (Spermophilus lateralis) Sertoli cells. Am J Anat. 1983b;168:99–108. doi: 10.1002/aja.1001680110. [DOI] [PubMed] [Google Scholar]
  415. Vogl AW, Pfeiffer DC, Mulholland D, Kimel G, Guttman J. Unique and multifunctional adhesion junctions in the testis: ectoplasmic specializations. Arch Histol Cytol. 2000;63:1–15. doi: 10.1679/aohc.63.1. [DOI] [PubMed] [Google Scholar]
  416. Vogl AW, Pfeiffer DC, Redenbach DM. Ectoplasmic (“junctional”) specializations in mammalian Sertoli cells: influence on spermatogenic cells. Ann N Y Acad Sci. 1991;637:175–202. doi: 10.1111/j.1749-6632.1991.tb27310.x. [DOI] [PubMed] [Google Scholar]
  417. Vogl AW, Pfeiffer DC, Redenbach DM, Grove BD. Sertoli cell cytoskeleton. In: Russell LD, Griswold MD, editors. The Sertoli Cell. Cache River Press; Clearwater, FL: 1993. pp. 39–86. [Google Scholar]
  418. Vogl AW, Soucy LJ. Arrangement and possible function of actin filament bundles in ectoplasmic specializations of ground squirrel Sertoli cells. J Cell Biol. 1985;100:814–825. doi: 10.1083/jcb.100.3.814. [DOI] [PMC free article] [PubMed] [Google Scholar]
  419. Vogl AW, Vaid KS, Guttman JA. The Sertoli cell cytoskeleton. Adv Exp Med Biol. 2008;636:186–211. doi: 10.1007/978-0-387-09597-4_11. [DOI] [PubMed] [Google Scholar]
  420. Vogl AW, Weis M, Pfeiffer DC. The perinuclear centriole-containing centrosome is not the major microtubule organizing center in Sertoli cells. Eur J Cell Biol. 1995;66:165–179. [PubMed] [Google Scholar]
  421. Vogl AW, Young JS, Du M. New insights into roles of tubulobulbar complexes in sperm release and turnover of blood-testis barrier. Int Rev Cell Mol Biol. 2013;303:319–355. doi: 10.1016/B978-0-12-407697-6.00008-8. [DOI] [PubMed] [Google Scholar]
  422. Walker C, Embleton A. On the origin of the Sertoli or foot-cells of the testis. Proc Royal Soc London, Series B. 1906;78:50–52. [Google Scholar]
  423. Walker WH. Molecular mechanisms controlling Sertoli cell proliferation and differentiation. Endocrinology. 2003a;144:3719–3721. doi: 10.1210/en.2003-0765. [DOI] [PubMed] [Google Scholar]
  424. Walker WH. Nongenomic actions of androgen in Sertoli cells. Curr Top Dev Biol. 2003b;56:25–53. doi: 10.1016/s0070-2153(03)01006-8. [DOI] [PubMed] [Google Scholar]
  425. Walker WH, Cheng J. FSH and testosterone signaling in Sertoli cells. Reproduction. 2005;130:15–28. doi: 10.1530/rep.1.00358. [DOI] [PubMed] [Google Scholar]
  426. Wang J, Wreford NG, Lan HY, Atkins R, Hedger MP. Leukocyte populations of the adult rat testis following removal of the Leydig cells by treatment with ethane dimethane sulfonate and subcutaneous testosterone implants. Biol Reprod. 1994;51:551–561. doi: 10.1095/biolreprod51.3.551. [DOI] [PubMed] [Google Scholar]
  427. 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:e1003645. doi: 10.1371/journal.pgen.1003645. [DOI] [PMC free article] [PubMed] [Google Scholar]
  428. Weber JE, Russell LD, Wong V, Peterson RN. Three-dimensional reconstruction of a rat stage V Sertoli cell: II. Morphometry of Sertoli-Sertoli and Sertoli–germ-cell relationships. Am J Anat. 1983;167:163–179. doi: 10.1002/aja.1001670203. [DOI] [PubMed] [Google Scholar]
  429. Wilson RM, Griswold MD. Secreted proteins from rat Sertoli cells. Exp Cell Res. 1979;123:127–135. doi: 10.1016/0014-4827(79)90429-4. [DOI] [PubMed] [Google Scholar]
  430. Wong CH, Cheng CY. The blood-testis barrier: its biology, regulation, and physiological role in spermatogenesis. Curr Top Dev Biol. 2005;71:263–296. doi: 10.1016/S0070-2153(05)71008-5. [DOI] [PubMed] [Google Scholar]
  431. Wong CH, Mruk DD, Lee WM, Cheng CY. Targeted and reversible disruption of the blood-testis barrier by an FSH mutant-occludin peptide conjugate. Faseb J. 2007;21:438–448. doi: 10.1096/fj.05-4144com. [DOI] [PMC free article] [PubMed] [Google Scholar]
  432. Wong EW, Cheng CY. Polarity proteins and cell-cell interactions in the testis. Int Rev Cell Mol Biol. 2009;278:309–353. doi: 10.1016/S1937-6448(09)78007-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  433. Wong EW, Cheng CY. Impacts of environmental toxicants on male reproductive dysfunction. Trends Pharmacol Sci. 2011;32:290–299. doi: 10.1016/j.tips.2011.01.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  434. Wong V, Russell LD. Three-dimensional reconstruction of a rat stage V Sertoli cell: I. Methods, basic configuration, and dimensions. Am J Anat. 1983;167:143–161. doi: 10.1002/aja.1001670202. [DOI] [PubMed] [Google Scholar]
  435. Wright WW. Stage-specific gene expression by Sertoli cells. In: Griswold M, editor. Sertoli Cell Biology. Elsevier Academic Press; Oxford; 2015. pp. 273–306. [Google Scholar]
  436. Wright WW, Luzarraga ML. Isolation of cyclic protein-2 from rat seminiferous tubule fluid and Sertoli cell culture medium. Biol Reprod. 1986;35:761–772. doi: 10.1095/biolreprod35.3.761. [DOI] [PubMed] [Google Scholar]
  437. Wright WW, Parvinen M, Musto NA, Gunsalus GL, Phillips DM, Mather JP, Bardin CW. Identification of stage-specific proteins synthesized by rat seminiferous tubules. Biol Reprod. 1983;29:257–270. doi: 10.1095/biolreprod29.1.257. [DOI] [PubMed] [Google Scholar]
  438. Wright WW, Zabludoff SD, Erickson-Lawrence M, Karzai AW. Germ cell-Sertoli cell interactions. Studies of cyclic protein-2 in the seminiferous tubule. Ann N Y Acad Sci. 1989;564:173–185. doi: 10.1111/j.1749-6632.1989.tb25896.x. [DOI] [PubMed] [Google Scholar]
  439. Wrobel KH, Schimmel M. Morphology of the bovine Sertoli cell during the spermatogenetic cycle. Cell Tissue Res. 1989;257:93–103. doi: 10.1007/BF00221638. [DOI] [PubMed] [Google Scholar]
  440. Xiao X, Cheng CY, Mruk DD. Intercellular adhesion molecule-2 is involved in apical ectoplasmic specialization dynamics during spermatogenesis in the rat. J Endocrinol. 2013;216:73–86. doi: 10.1530/JOE-12-0434. [DOI] [PMC free article] [PubMed] [Google Scholar]
  441. Xiao X, Mruk DD, Wong CK, Cheng CY. Germ cell transport across the seminiferous epithelium during spermatogenesis. Physiology (Bethesda) 2014a;29:286–298. doi: 10.1152/physiol.00001.2014. [DOI] [PMC free article] [PubMed] [Google Scholar]
  442. Xiao X, Wong EW, Lie PP, Mruk DD, Wong CK, Cheng CY. Cytokines, polarity proteins, and endosomal protein trafficking and signaling-the sertoli cell blood-testis barrier system in vitro as a study model. Methods Enzymol. 2014b;534:181–194. doi: 10.1016/B978-0-12-397926-1.00010-X. [DOI] [PMC free article] [PubMed] [Google Scholar]
  443. Xu J, Anuar F, Ali SM, Ng MY, Phua DC, Hunziker W. Zona occludens-2 is critical for blood-testis barrier integrity and male fertility. Mol Biol Cell. 2009;20:4268–4277. doi: 10.1091/mbc.E08-12-1236. [DOI] [PMC free article] [PubMed] [Google Scholar]
  444. Yan HH, Mruk DD, Lee WM, Cheng CY. Blood-testis barrier dynamics are regulated by testosterone and cytokines via their differential effects on the kinetics of protein endocytosis and recycling in Sertoli cells. FASEB J. 2008;22:1945–1959. doi: 10.1096/fj.06-070342. [DOI] [PMC free article] [PubMed] [Google Scholar]
  445. Yan W. Gene knockouts that affect Sertoli cell function. In: Griswold M, editor. Sertoli Cell Biology. Elsevier Academic Press; Oxford; 2015. pp. 437–469. [Google Scholar]
  446. Yang Q-E, Oatley JM. Early postnatal interactions between Sertoli and germ cells. In: Griswold M, editor. Sertoli Cell Biology. Elsevier Academic Press; Oxford; 2015. pp. 81–98. [Google Scholar]
  447. 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;126:1009–1020. doi: 10.1242/jcs.119826. [DOI] [PMC free article] [PubMed] [Google Scholar]
  448. Yao HH, Ungewitter E, Franco H, Capel B. Establishment of fetal Sertoli cells and their role in testis morphogenesis. In: Griswold M, editor. Sertoli Cell Biology. Elsevier Academic Press; Oxford; 2015. pp. 57–79. [Google Scholar]
  449. 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;124:2357–2366. doi: 10.1242/jcs.080903. [DOI] [PubMed] [Google Scholar]
  450. Yomogida K, Ohtani H, Harigae H, Ito E, Nishimune Y, Engel JD, Yamamoto M. Developmental stage- and spermatogenic cycle-specific expression of transcription factor GATA-1 in mouse Sertoli cells. Development. 1994;120:1759–1766. doi: 10.1242/dev.120.7.1759. [DOI] [PubMed] [Google Scholar]
  451. Young JC, Wakitani S, Loveland KL. TGF-beta superfamily signaling in testis formation and early male germline development. Semin Cell Dev Biol. 2015;45:94–103. doi: 10.1016/j.semcdb.2015.10.029. [DOI] [PubMed] [Google Scholar]
  452. Young JS, Guttman JA, Vaid KS, Vogl AW. Tubulobulbar complexes are intercellular podosome-like structures that internalize intact intercellular junctions during epithelial remodeling events in the rat testis. Biol Reprod. 2009;80:162–174. doi: 10.1095/biolreprod.108.070623. [DOI] [PubMed] [Google Scholar]
  453. Young JS, Takai Y, Kojic KL, Vogl AW. Internalization of adhesion junction proteins and their association with recycling endosome marker proteins in rat seminiferous epithelium. Reproduction. 2012;143:347–357. doi: 10.1530/REP-11-0317. [DOI] [PubMed] [Google Scholar]
  454. Yule TD, Montoya GD, Russell LD, Williams TM, Tung KS. Autoantigenic germ cells exist outside the blood testis barrier. J Immunol. 1988;141:1161–1167. [PubMed] [Google Scholar]
  455. Zebrun W, Mollenhauer HH. Electron microscopic observations on mitochondria of rat testes fixed in potassium permanganate. J Biophys Biochem Cytol. 1960;7:311–314. doi: 10.1083/jcb.7.2.311. [DOI] [PMC free article] [PubMed] [Google Scholar]

Associated Data

This section collects any data citations, data availability statements, or supplementary materials included in this article.

Supplementary Materials

Table S1. Sertoli Cell Books and Biographies of Enrico Sertoli.

Figure S1. Key Morphological Features of Sertoli Cell are listed with representative examples illustrated.

Figure S2. Electron microscopy of the Sertoli cell nucleus (N) from a human testis showing a large nucleolus (Nu) and deep indentation (In) of the nuclear membrane (Nm).

Figure S3. Schematic representation of Sertoli cell (SC) proliferation in relation to endocrine and paracrine regulation of fish spermatogenesis.

Figure S4. Number of spermatids per Sertoli cell (SC), based on the available literature, for different vertebrate groups.

RESOURCES