SciELO - Scientific Electronic Library Online

 
vol.35 issue3Apigenin inhibits cell migration through MAPK pathways in human bladder smooth muscle cells author indexsubject indexarticles search
Home Pagealphabetic serial listing  

Services on Demand

Journal

Article

Indicators

  • Have no cited articlesCited by SciELO

Related links

Share


Biocell

Print version ISSN 0327-9545

Biocell vol.35 no.3 Mendoza Sept./Dec. 2011

 

ORIGINAL ARTICLES

The actin filament network associated to Sertoli cell ectoplasmic specializations

 

Juan Carlos Cavicchia1*, Mabel Fóscolo1, Jorge Ibañez1, Christopher Lillig2, Francisco Capani3

1. Instituto de Histología y Embriología "Dr. Mario H. Burgos" (IHEM-CONICET), Facultad de Ciencias Médicas, Universidad Nacional de Cuyo, Mendoza, Argentina.
2. Institute for Clinical Cytobiology and Cytopathology, Faculty of Medicine, Phillips-Universität Marburg, Germany.
3. Instituto de Investigaciones Cardiológicas "Prof. Dr. Alberto C Taquini" (ININCA), Facultad de Medicina, Universidad de Buenos Aires and Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina.

Address correspondence to:

Juan Carlos Cavicchia.
Instituto de Histología y Embriología (IHEM), Facultad de Ciencias Médicas, UNCuyo, Mendoza, Argentina. E-mail: jccavic@fcm.uncu.edu.ar

 


ABSTRACT:  Junctional devices in Sertoli cells conform the blood-testis barrier and play a key role in maturation and differentiation of germ cells. The spacial distribution of ectoplasmic specializations of Sertoli cells was studied by β-actin immunolabelling, using laser confocal and transmission electron microscopy. For confocal microscopy, β-actin immunolabelling of ectoplasmic specializations was studied over the background of either prosaposin or glutaredoxin immunolabelling of the Sertoli cytoplasm. Labelling was found near the basal lamina, surrounding early spermatocytes (presumably in leptotene-zygotene) or at one of two levels in the seminiferous epithelium: (1) around deep infoldings of the Sertoli cell cytoplasm, in tubular stages before spermiation, and (2) in the superficial part of the seminiferous epithelium, in tubular stages after or during spermiation. For transmission electron microscopy, β-actin immunolabelling of ectoplasmic specializations was also used. Ectoplasmic specializations were found at two different levels of the seminiferous epithelium. We also used freeze fracture to analyze the characteristics of tubulo-bulbar complexes, a known component of apical ectoplasmic specializations. Also, these different approaches allowed us to study the complex arrangement of the actin cytoskeleton of Sertoli cells branches, which surround germ cells in different stages of the spermatogenic cycle.  Our results show a consistent labelling for β-actin before, during and after the release of spermatozoa in the tubular lumen (spermiation) suggesting a significant role of the actin network in spermatic cell differentiation. In conclusion, significant interrelations among the β-actin network, the junctional complexes of the blood-testis barrier and the ectoplasmic specializations were detected at different stages of the seminiferous cycle.

Keywords: β-actin; Ectoplasmic specializations; Sertoli cell; Seminiferous epithelium.


 

Introduction

The concept of a blood-testis barrier originated from the physiological studies of Setchell (1969) and Setchell and Waites (1975) in the mammalian testis. They showed a differential transport of ions and metabolites between the basal area of the seminiferous tubules, the basal compartment, in wide contact with bodily fluids, and the apical sector of the seminiferous epithelium, the adluminal compartment, close to the tubular lumen and isolated by the barrier. The morphological correlate of such a barrier was found to be located in specialized junctional devices found between adjacent Sertoli cells, integrated by occluding or tight junctions that seal each inter-Sertoli space near the base of the seminiferous epithelium (Dym and Fawcett, 1970; Dym, 1973). Related to the last mentioned topic is the long-time controversy on the existence of a third compartment in the seminiferous epithelium (see Yazama, 2008, for review).
The early interpretations of Russell (1977a, b, 1978) on the rat blood-testis barrier were superseded by our observations on monkeys (Dym and Cavicchia, 1977, 1978) as we showed that  the impermeable junctions assemble and disassemble simultaneously without forming an actual compartment. Later, in the rat, using intratubular administration of intercellular tracers we arrived to similar conclusions (Cavicchia and Sacerdote, 1988).
The second junctional device found in the seminiferous epithelium is the zonula adherens,  mainly made by a dense circumferential belt of actin filaments near the Sertoli cell membranes and located (1) at the level of the blood-testis barrier, i.e., in the Sertoli cytoplasm close to tight junctions and surrounding a smooth cistern of the endoplasmic reticulum (Dym and Fawcett, 1970) and (2) at a higher level in the epithelium, in close apposition to germ cells in progressive steps of differentiation (round and elongated spermatids) (Gumbiner et al., 1988). These two junctional devices , as a whole, were termed ectoplasmic specializations of the Sertoli cell by Russell (1977b).
Another junctional device found in ectoplasmic specializations at both levels are the so-called tubulo-bulbar complexes (Russell, 1979; Russell and Griswold, 1993). At the higher place of Sertoli-germ cell contacts, they are elongate processes developed from the spermatid plasma membrane and surrounded by a deep invagination of the Sertoli cell membrane and a specialized layer of Sertoli cell cytoplasm. This layer contains abundant β-actin filaments bundles surrounding the whole invagination. These tubulo-bulbar complexes have been found to develop in the regions previously occupied by the ectoplasmic specializations and have been proposed to internalize intact junctions during spermiation (Guttman et al., 2004). Thus, these complexes are likely significant participants in the mechanism of junctional turnover in the seminiferous epithelium (Young et al., 2009), which is related to the well-known endocytic activity of Sertoli cells, residual body formation, their phagocytic activity during spermiation and Sertoli lysosome types including multivesicular bodies ("multivesicular endosomes"; Gruenberg and Stenmark, 2004) and secondary lysosomes (reviewed by Morales and Clermont, 1993). Significant progress has been made in the understanding of the mechanisms that regulate multivesicular transport and the degradation pathway that leads to lysosome formation since the classical study that observed a maximum number of lysosomes marked by acid phosphatase in stages V-VIII of the seminiferous tubules, close to spermiation (Chemes, 1986). To review the stages of the seminiferous tubules see Russell et al. (1990).
Recently, experimental evidence has been accumulating on the regulation and functional significance of Sertoli ectoplasmic specializations (Wong et al., 2004, 2008; reviewed by Cheng and Mruk, 2010). The simultaneous occurrence of transit of new germ cells through the blood-testis barrier and the process of spermiation has been put into a theoretical scheme in which these two cellular events are coordinated by the release of definite protein products, and the presence of an "apical ectoplasmic specializations- blood-testis barrier-hemidesmosome axis" in Sertoli cell has been put forward as a crucial element in the coordination of these events (reviewed in Cheng and Mruk, 2010). Although the actin network was previously described in Sertoli junctional devices (Morales and Clermont, 1993) its role in the subcellular organization of blood-testis barrier and ectoplasmic specializations at different stages of the seminiferous cycle have not been thoroughly investigated, and its significance in current schemes of spermatogenesis is not well understood.
Thus, the main objective of this paper is to report our findings about the cellular and fine structural organization of β-actin rich intercellular junctions, and its possible role in germ cell maturation, differentiation and migration towards the lumen of theseminiferous tubule.

Materials and Methods

Animals

All studies involving experimental animals were conducted in accordance with the the UFAW Handbook on the Care and Management of Laboratory Animals (Pole, 1999) and were approved by the Ethical Board of the School of Medicine of the National University of Cuyo, Mendoza, Argentina. Young male Wistar rats bred in our laboratory, being 90-100 days old and weighing 300 ± 50 g, were used throughout this protocol. They were housed under controlled light (lights-on from 07:00 to 19:00 h) and temperature (24 ± 2ºC) conditions, and water and food were provided ad libitum. Rats were anesthetized with ether under a bell and perfused through the spermatic artery according to the technique of Dym (1973) with 4% paraformaldehyde in 0.1M phosphate buffer, pH 7.4 (for light microscopy studies) or with 4% paraformaldehyde in 0.1 M cacodylate buffer, pH 7.4, containing 0.1% glutaraldehyde for electron microscopy studies. Testes were dissected and post-fixed in the same solution for 2 additional hours. Forty µm sections were first obtained in an Oxford Vibratome and were then embedded in either paraffin or epoxy resin.

Immunochemistry

Paraffin sections

Sections (5-10 µm thick) were mounted on sylanized slices (Saraceno et al., 2010) and were incubated overnight in β-actin antibody (1:500 dilution) (Sigma, USA) and prosaposin (1:500 dilution) (Morales, et al., 1998) or glutaredoxin antibodies (1:500) (Lönn et al., 2008; Godoy et al., 2010) for colocalization studies. Sections were washed several times with phosphate buffer-saline and incubated with the proper secondary antibody (Vector Laboratories Inc., Burlingame, CA, USA), followed by incubation with a biotin streptavidin complex (HRP Histo Mark, Caramillo, CA USA). After several washings in buffer, sections were developed with AEC substrate kit (Invitrogen Gauttersburg, MD, USA) until staining was optimal. Finally, samples were examined in a Spectral Confocal Olympus FV-1000. Collected pictures were analyzed and compiled using Adobe Photoshop 8.0 CS3.

Epoxy resin sections

Sections were incubated overnight with a monoclonal anti-β-actin antibody (Sigma, USA; diluted 1:500 in phosphate buffer-saline). Following several washings with buffer, sections were incubated with biotinylated anti-mouse secondary antibody for 1 h (Amersham) and followed by incubation with ABC complex solution (Vectastain ABC kits, Vector Laboratories Inc., Burlingame, CA, USA) for another 1 h. After washing in buffer, sections were developed with diaminobenzidine solution until staining was optimal as examined by light microscopy. Sections were post-fixed in 1% osmium tetroxide in 0.1 M cacodylate buffer, rinsed in distilled water, and stained with 1% aqueous uranyl acetate overnight. Tissue sections were dehydrated in increasing series of ethanol concentrations followed by dry acetone, and embedded in epoxy resin (Durcupan ACM, Fluka, Buchs, Switzerland). Thin 1µm sections were stained with toluidine blue-sodium borate for light microscopy. Ultrathin sections less than 0.1µm (silver-gold interference colour) were stained with uranyl acetate and lead citrate and examined in a Zeiss EM 900 electron microscope (Carl Zeiss Inc., Berlin, Germany).

Freeze-Fracture

The method of Cavicchia et al. (1998) was used. In brief, rats were anesthetized and perfused as indicated above for immunohistochemistry, and the fixative used was 5% glutaraldehyde in 0.1M sodiumcacodylate buffer, pH 7.2-7.4. After perfusion, testes were removed, cut in small piecesand immersed in the same fixative for 2 h, placed for additional 2 h in 30% glycerol atroom temperature (18°C), rapidly frozen in liquid Freon 22 and stored in liquid nitrogen. The material was fractured in a Balzers BAF 301 apparatus at -105°C and shadowed with platinum followed by carbon. Replicas were cleaned with sodium hypochlorite, mounted on copper grids and examined under the electron microscope.

Results

The combined immunocytochemistry for β-actin, prosaposin and glutaredoxin allowed us to observe significant details of the staining degree, the localization of the blood-testis barrier in relation to spermatocytes and to spermatids before, during and after spermiation (Figs. 1 to 5). Both prosaposin and glutaredoxin are proteins localized in the Sertoli cytoplasm (see Lönn et al., 2008; Godoy et al., 2010). In Figs. 2 and 5, and in the inset A of Fig. 6, β-actin condensation appeared surrounding early spermatocytes (probably leptotene-zygotene stages) at the level of the blood-testis barrier. When tubules at a later stage were observed (Fig.2) β-actin marked spermatids were observed above the blood-testis barrier, probably indicating β-actin marking of Sertoli junctional devices at this level. In an even later stage of development (Fig. 3) β-actin marking was associated to spermatid-Sertoli ectoplasmic specializations and lysosomes close to the tubular lumen.


Figure 1. Immunostaining for both β-actin (orange-yellow) and prosaposin (green) showing cross sections of two seminiferous tubules in diametrically opposite stages. The upper tubule (pre-spermiation stage) displays elongated spermatids orange-marked for b-actin and which are located in deep infoldings of Sertoli cells, i.e., close to the basal lamina and thus to the blood-testis barrier. The lower tubule (post-spermiation stage) exhibits elongated spermatids with their flagella (F) protruding into the tubular lumen (L). Arrows indicate the basal lamina. Bar = 25 µm.


Figure 2. Spermatids (SP) located close to the blood-testis barrier, which is shown as a seemingly continuous band of β-actin condensation and by condensations around early spermatocytes (asterisks). Bar = 25 µm.


Figure 3. Immunostaining for both β-actin (orange-yellow) and prosaposin (green) showing a pre-spermiation stage in which spermatids are located higher than in the inset of Fig. 1. The thinly marked blood-testis barrier (arrow) is located above a spermatogonium (G). BL, basal lamina. Bar = 25 µm.


Figure 4. Immunostaining for both β-actin (orange-yellow) and prosaposin (green) showing a tubular section at a pre-spermiation stage (VII or VIII, i.e., near spermiation). Spermatids are located higher than in the lower tubule in Fig.1. Note the intense actin immunostaining surrounding the spermatid heads and numerous spermatid flagella (F) that are protruding into the lumen (L). Residual bodies (RB) are also shown. Numerous small round β-actin condensations probably correspond to multivesicular endosomes. Bar = 25 µm.


Figure 5. Immunostaining for both β-actin (orange-yellow) and glutaredoxin (dark red) showing adjacent basal portions of two seminiferous tubules which are close to each other and to the basal lamina (BL). The immune stain for glutaredoxin is observed in red, the immune stain for β-actin in yellow. In this particular stage, β-actin staining appears partly above and below early spermatocytes (asterisks), which are then probably in leptotene-zygotene stages. Also, intense β-actin staining (probably corresponding to ectoplasmic specializations) is surrounding elongated spermatid heads (SP) located deeply into the seminiferous epithelium (but above the blood-testis barrier). BL, basal lamina. Bar = 25 µm.


Figure 6. Low magnification) of 40 µm epoxy resin sections showing two adjacent seminiferous tubules in different spermatogenic stages. The left one exhibits spermatid heads (SP) in deep infoldings of the Sertoli cell (as in Fig. 2). Numerous dense bodies marked for β-actin are located high, close to lumen (L) and some are indicated with arrows. They may correspond to the multivesicular endosomes.The seminiferous tubule to the right shows spermatid heads located high in the epithelium and with their flagella protruding into the lumen (L). In this particular seminiferous stage numerous electron dense bodies of β-actin reaction (arrows) are located close to the seminiferous basal lamina. They also seem to correspond to secondary endosomes. Bar = 25 µm. Inset A: It shows at higher magnification β-actin labelled spermatids (SP) located close to the blood-testis barrier. Note that β-actin labelling surrounds incompletely early spermatocytes, which appear to be in the leptotene-zygotene stage. Bar = 25 µm. Inset B: It depicts two electron dense bodies containing β-actin reaction (visible as small dots). The one to the right corresponds to a multivesicular endosomes. Notice that the reaction is mainly located in the matrix, while vesicles are almost devoid of the reaction. The image to the left shows the β-actin reaction homogeneously distributed inside the body. It may correspond to secondary lysosomes. Bar = 0.5 µm.

Immunolabelling of β-actin observed under the transmission electron microscope showed spermatid heads located either in deep infoldings of the Sertoli cell membrane (pre-spermiation stages; Fig. 6, left) or located higher in the epithelium and with their tails protruding into the tubular lumen (post-spermiation stage; Fig. 6, right). Fig. 6 (inset A) shows at higher magnification a tubule in a pre-spermiation stage, in which β-actin condensations are also seen partially surrounding early spermatocytes, probably in leptotene-zygotene stages. Also, numerous immunolabelled β-actin bodies were found near the tubular lumen before spermiation (and probably corresponded to multivesicular endosomes; Fig. 6, left), while much larger and less numerous electron dense bodies, were seen close to the basal lamina (they probably corresponded to secondary lysosomes, i.e., multivesicular endosomes in later stages of development, Fig. 6, right). In these bodies, β-actin immunolabelling  appeared located in the intervesicular matrix of  multivesicular endosomes (Fig. 6, inset B, right), while it was uniformly distributed within secondary lysosomes (Fig.  6, inset B, left).
When tubulo-bulbar complexes are favourably freeze fractured they exhibited their characteristic outline (Fig. 7) similar to that described by scanning electron microscopy (Russell and Griswold, 1993). When we compare freeze fracture of this structure with electron microscopy and the β-actin marking, it appears that the label is seen on the Sertoli cell cytoplasm surrounding the tubulo-bulbar processes, and is concentrated between it and the smooth reticulum cistern (Fig. 8).


Figure 7. It depicts the freeze fracture image of a tubulo-bulbar process in longitudinal view. The line that crosses the tubular area approximates the region shown in the next figure under transmission electron microscopy. Bar = 0.5 µm.


Figure 8. Transmission electron microscopy of a thin sections immunolabelled for β-actin showing a cross section of a tubulo-bulbar process (asterisk), which is sandwiched between the tubulo-bulbar process and smooth cistern of the endoplasmic reticulum (arrow). Bar = 0.5 µm.

Discussion

Mammalian spermatogenesis is probably the most exquisite known example of a spatially organized sequence of cellular associations, involving continuous synchronized cell proliferation, differentiation, meiosis, apoptosis, germ translocation, migration and morphogenesis. Spermatogenesis relies on a population of germ cells capable of self renewal, migration towards the lumen of the seminiferous tubules and differentiation into the male gamete (for review see Sharpe, 1994). To maintain close interaction with the surrounding Sertoli cells, both cell lineages possess a well-organized but still poorly understood system of intercellular communications, some of which were explored in the present paper.
The presence of actin in the ectoplasmic specializations in Sertoli cells originally raised the question about its involvement in cell movements related to spermiation, mostly the retraction of Sertoli cell apical cytoplasm. However, accumulated evidence has weakened such a hypothetical role for actin in Sertoli cells. Vogl and Soucy (1985) found no evidence of contraction in a glycerinated model of squirrel seminiferous epithelium. Then, Grove and Vogl (1989) postulated that actin could have a more structural than mechanical or contractile function in these locations since no myosin-based system was detected by immunohistochemistry.
In these two locations, actin consists of the β isoform, which forms filament bundles (Pollard and Earnshaw, 2008). Actin filaments in each bundle are unipolar and hexagonally packed. The bundles are coupled to the adjacent Sertoli membrane and to each other.
A different function for the actin bundles at ectoplasmic specializations has emerged from works aimed to elucidate the dynamic state of intercellular junctions, while many other studies have been directed towards the identification of adhesion molecules in spermatogenesis and on the turnover of the elements of cellular junctions that have provided evidence about a support role of actin in these junctions (reviewed by Cheng and Mruk, 2010). The tubulo-bulbar complexes are known to be the structures following the ectoplasmic specializations both in the apical locations and in the basal location at the blood-testis barrier (Guttman et al., 2004). An immunohistochemical and ultrastructural study has shown that these tubule-bulbar complexes contain the integral membrane adhesion molecules nectin 2 (in Sertoli cells) and nectin 3 (in spermatids) as well as the actin-binding protein espin (Guttman et al., 2004). Furthermore, these molecules (as well as other associated with ectoplasmic specializations) are enriched at the ends of the tubule-bulbar complexes and in the vesicles associated with these endings, which are also labelled with antibodies for lysosomal and endosomal markers (Guttman et al., 2004). These observations have led to the proposal that tubulo-bulbar complexes are structures that internalize intercellular junction materials, both at the apical and at the basal ectoplasmic specializations (Young et al., 2009).
Tubulo-bulbar devices have not been observed before using freeze fracture replicas, which permit a detailed observation of the cell membranes involved (Fig. 6). Moreover, using immune electron microscopy we detected β-actin surrounding the tubule-bulbar complex, extending previous observations by Morales and Clermont (1993) who reported the presence of actin but not of its specific β-isoform in this Sertoli specialization.
Our results show a significant correlation between the special organization of the β-actin filament network and the stages of the spermatogenic cycle of the seminiferous epithelium. Moreover, β-actin condensation appears modulated according to the degree of germ cell maturation and differentiation. We also observed a clear correlation between the localization of the β-actin network in the spermiation process and that of the germ cells that appear to ascend from the basal to the adluminal compartment.
Related to the last mentioned topic is the long standing controversy on the existence of a third compartment in the seminiferous epithelium. As we mentioned in the Introduction,  Russell (1977a, 1978) was of the opinion that late leptotene spermatocytes in stages X-XI of the cycle of the seminiferous epithelium are the cells which cross the blood-testis barrier in the rat and penetrate into a third or intermediate compartment, which is limited, both below and above, by non-permeable inter-Sertoli junctions as seen by electron-opaque tracers. Simultaneously, in the monkey we observed a similar phenomenon but our interpretation was that these impermeable junctions are assembled and disassembled simultaneously without forming a real compartment. (Dym and Cavicchia, 1977, 1978). Later, using intratubular administration of intercellular tracers in the rat, we arrived to a similar conclusion (Cavicchia and Sacerdote, 1988). The current results also stand against the existence of the so-called third compartment, since we never observed leptotene-zygotene spermatocytes totally surrounded by β-actin. Thus, it appears that the dynamic changes of β-actin polymerization/depolymerization cycle contribute to a fast ascent of spermatocytes avoiding the formation of the postulated intermediate compartment.
Numerous multivesicular bodies were especially located in the upper regions of the seminiferous epithelium (for example, see Fig. 3 and Fig 5 inset B). Early electron microscopy studies (Trump et al., 1965) indicated the existence of multivesicular bodies, suggesting that they were related somehow to lysosomes and that contained characteristic accumulations of small vesicles in their lumen or matrix.
We also observed β-actin in the matrix of multivesicular bodies and in secondary lysosomes near the basal lamina of the seminiferous epithelium (Figs. 3 and 5, and Fig. 5, inset B). In MVB, it could be related to the fact that tubulo-bulbar complexes appear to internalize apical ectoplasmic specializations once that spermatids are released into the lumen of the seminiferous epithelium (Morales and Clermont, 1993; Young et al., 2009). Secondary lysosomes (Fig. 5, right tubule) containing β-actin could have a similar function. In relation to our results, we remark that Young et al.
(2009) postulate that tubulobulbar complexes are intercellular podosome-like structures that internalize intact intercellular junctions during epithelial remodelling events in the rat testis. Thus, our observations support the claim that tubulo bulbar complexes are actin-related, double-membrane projections that form at intercellular junctions in the seminiferous epithelium, and that intact junctions are internalized during sperm release and during spermatocyte translocation through basal junctional complexes between neighboring Sertoli cells.

Acknowledgments

The authors thanks the valuable technical assistance of Mrs. Juanita Dávila. This investigation was supported by the grants from Secretaría de Ciencia, Técnica y Postgrado, Universidad Nacional de Cuyo, Argentina (06/J13) and the National Research Council of Argentina (CONICET, PIP 6486). The prosaposin antibody was a generous gift of Prof. Carlos Morales, Anatomy and Cell Biology Department, Mc Gill University, Montreal, Canada.

References

1. Cavicchia JC, Sacerdote FL (1988). Topography of the rat bloodtestis barrier after intratubular administration of intercellular tracers. Tissue & Cell 20: 577-586.         [ Links ]

2. Cavicchia JC, Sacerdote FL, Morales A, Zhu BC (1998). Sertoli cell nuclear pore number changes in two stage sets of the rat spermatogenic cycle. Tissue & Cell 30: 268-273.         [ Links ]

3. Chemes H (1986). The phagocytic function of Sertoli cells: a morphological,biochemical, and endocrinological study of lysosomes and acid phosphatase localization in the rat testis. Endocrinology 119: 1673-1681.         [ Links ]

4. Cheng CY, Mruk DD (2010). A local autocrine axis in the testes that regulates spermatogenesis. Nature Reviews Endocrinology 6: 380-395.         [ Links ]

5. Dym M, Fawcett DW(1970). The blood-testis barrier in the rat and the physiological compartmentation of the seminiferous epithelium. Biology of Reproduction 3: 308-326.         [ Links ]

6. Dym M (1973). The fine structure of the monkey (Macaca) Sertoli cell and its role in maintaining the blood-testis barrier. Anatomical Record 175: 636-656.         [ Links ]

7. Dym M, Cavicchia JC (1977). Further observations on the bloodtestis barrier in monkeys. Biology of Reproduction 17: 390-403.         [ Links ]

8. Dym M, Cavicchia JC (1978). Functional Morphology of the testis. Biology of Reproduction 18: 1-15.         [ Links ]

9. Godoy JR, Funke M, Ackermann W, Haunhorst P, Oesteritz S, Capani F, Elsässer HP, Lillig CH (2010). Redox atlas of the mouse. Immunohistochemical detection of glutaredoxin-, peroxiredoxin-,and thioredoxin-family proteins in various tissues of the laboratory mouse. Biochimica et Biophysica acta 1810: 2-92.         [ Links ]

10. Grove BD, Vogl AW (1989). Sertoli cell ectoplasmic specialization: a type of actin associated adhesion junction? Journal of Cell Science 93: 309-324.         [ Links ]

11. Gruenberg J, Stenmark H (2004). The biogenesis of multivesicular endosomes. National Review & Molecular Cell Biology 5: 317-323.         [ Links ]

12. Gumbiner B, Stevenson B, Grimaldi A (1988). The role of the cell adhesion molecule uvomorulin in the formation and maintenance of the epithelial junctional complex. The Journal of Cell Biology 107: 1575-1587.         [ Links ]

13. Guttman JA, Takai Y, Vogl AW (2004). Evidence that tubulobulbar complexes in the seminiferous epithelium are involved with internalization of adhesion junctions. Biology of Reproduction 71: 548-559.         [ Links ]

14. Lönn ME, Hudemann C, Carsten B, Cherkasov V, Capani F, Holmgren A, Zeng J, Lillig CH (2008). Expression pattern of human glutaredoxin 2 isoforms: identication and characterization of two testis/cancer cell-specific isoforms. Antioxidants & Redox Signaling 10: 547-557.         [ Links ]

15. Morales C, Clermont Y (1993). Structural changes of the Sertoli cell during the cycle of the seminiferous epithelium. In: Russell LD, Griswold MD, editors. The Sertoli cell. Clearwater FL: Cache River Press. p 305-330.         [ Links ]

16. Morales CR, Hay N, El-Alfy M, Zhao Q (1998). Distribution of mouse sulfated glycoprotein-1 (prosaposin) in the testis and other tissues. Journal of Andrology 19: 156-164.         [ Links ]

17. Pole T (1999). UFAW Handbook on the care and management of laboratory animals. Blackwell Science Ltd. Vol. 2        [ Links ]

18. Pollard TD, Earnshaw WC (2008). Actin and actin-binding Proteins. Saunders Elsevier. Philadelphia. Cell Biology 33(Second edition).         [ Links ]

19. Russell LD (1977a). Movement of spermatocytes from the basal to the adluminal compartment of the rat testis. American Journal of Anatomy 149: 313-328.         [ Links ]

20. Russell LD (1977b). Observations on rat Sertoli ectoplasmic ("junctional") specializations in their association with germ cells of the rat testis. Tissue & Cell 9: 475-498.         [ Links ]

21. Russell LD (1978). The blood-testis barrier and its formation relative to spermatocyte maturation in the adult rat: a lanthanum tracer study. Anatomical Record 190: 99-112.         [ Links ]

22. Russell LD (1979). Spermatid-Sertoli tubulo-bulbar complexes as devices for elimination of cytoplasm from the head region of late spermatids of the rat. Anatomical Record 194: 233-246.         [ Links ]

23. Russell LD, Ettlin RA, Hikim APS, Clegg ED (1990). Histological and histopathological evaluation of the testis. Clearwater, Fl: Cache River Press.         [ Links ]

24. Russell LD, Griswold MD (1993). The Sertoli Cell. Clearwater FL: Cache River Press.         [ Links ]

25. Saraceno GE, Bertolino ML, Galeano P, Romero JI, Garcia-Segura LM, Capani F (2010). Estradiol therapy in adulthood reverses glial and neuronal alterations caused by perinatal asphyxia. Experimental Neurology 223: 615-622.         [ Links ]

26. Setchell BP (1969). Do Sertoli cells secrete fluid into the seminiferous tubules? Journal of Reproduction & Fertility 19: 391-392.         [ Links ]

27. Setchell BP, Waites GMH (1975). The blood-testis barrier. In: Handbook of Physiology, Section 7: Endocrinology, Vol. V. Male reproductive System. (D.W. Hamilton and R.O.Greep, eds). American Physiology Society, Washington pp. 143-172.         [ Links ]

28. Sharpe RM (1994). Regulation of spermatogenesis. In: Knobil E, Neill J D, editors. The physiology of reproduction. New York: Raven Press p 1363-1434.         [ Links ]

29. Trump BF, Goldblatt PJ, Stowell RE (1965). Studies of necrosis in vitro of mouse hepatic parenchymal cells. Ultrastructural and cytochemical alterations of cytosomes, cytosegresomes, multivesicular bodies, and microbodies and their relation to the lysosome concept. Laboratory Investigation 14: 1946-1968.         [ Links ]

30. Vogl AW, Soucy LJ (1985). Arrangement and possible function of actin filament bundles in ectoplasmic specializations of ground squirrel Sertoli cells. Journal of Cell Biology 100: 814-825.         [ Links ]

31. Wong CH, Mruk DD, Lui WY, Cheng CY (2004). Regulation of blood-testis barrier dymamics: an in vivo study. Journal of Cell Science 117: 383-798.         [ Links ]

32. Wong EW, Mruk DD, Cheng CY (2008). Biology and regulation of ectoplasmic specialization, an atypical adherens junction type, in the testis. Biochimica et Biophysica Acta 1778: 692-708.         [ Links ]

33. Yazama F (2008). Continual maintenance of the blood-testis barrier during spermatogenesis: the intermediate compartment theory revised. The Journal of Reproduction and Development 54: 299-305.         [ Links ]

34. Young JS, Guttman JA, Vaid KS, Vogl AW (2009). Tubulobulbar complexes are intercellular podosome-like structures that internalize intact intercellular junctions during epithelial remodelling events in the rat testis. Biology of Reproduction 80: 162-174.         [ Links ]

Received: March 31, 2011.
Revised version received:
November 30, 2011.
Accepted:
November 29, 2011.

Creative Commons License All the contents of this journal, except where otherwise noted, is licensed under a Creative Commons Attribution License