orchestration of occludins, claudins, catenins and ... · asian j androl 2007; 9 (4): 463–475...

13
Asian J Androl 2007; 9 (4): 463–475 . 463 . Orchestration of occludins, claudins, catenins and cadherins as players involved in maintenance of the blood-epididymal barrier in animals and humans Daniel G. Cyr 1,3 , Mary Gregory 1 , Évemie Dubé 1 , Julie Dufresne 1 , Peter T. K. Chan 2 , Louis Hermo 3 1 INRS-Institut Armand Frappier, Université du Québec, 245 Hymus Boulevard, Pointe Claire, QC H9R 1G6, Canada 2 Department of Urology, Royal Victoria Hospital, McGill University, Montreal, QC H3A 1A1, Canada 3 Department of Anatomy and Cell Biology, McGill University, Montreal, QC H3A 2B2, Canada Abstract Although spermatozoa are formed during spermatogenesis in the testis, testicular spermatozoa are immature and cannot swim or fertilize. These critical spermatozoal functions are acquired in the epididymis where a specific luminal environment is created by the blood-epididymal barrier; proteins secreted by epididymal principal cells bind to matur- ing spermatozoa and regulate the maturational process of the spermatozoa. In the epididymis, epithelial cell-cell interactions are mediated by adhering junctions, necessary for cell adhesion, and by tight junctions, which form the blood–epididymal barrier. The regulation of these cellular junctions is thought to represent a key determinant in the process of sperm maturation within the epididymis. Tight junctions between adjacent principal cells permit the formation of a specific microenvironment in the lumen of the epididymis that is essential for sperm maturation. Although we have made significant progress in understanding epididymal function and the blood-epididymal barrier, using animal models, there is limited information on the human epididymis. If we are to understand the normal and pathological conditions attributable to human epididymal function, we must clearly establish the physiological, cellular and molecular regulation of the human epididymis, develop tools to characterize these functions and develop clinical strategies that will use epididymal functions to improve treatment of infertility. (Asian J Androl 2007 July; 9: 463– 475) Keywords: claudins; cadherins; catenins; human; rat; mouse; tight junction; adherens junction . Review . DOI: 10.1111/j.1745-7262.2007.00308.x www.asiaandro.com © 2007, Asian Journal of Andrology, Shanghai Institute of Materia Medica, Chinese Academy of Sciences. All rights reserved. Correspondence to: Dr Daniel G. Cyr, INRS-Institut Armand Frappier, Université du Québec, 245 Hymus Boulevard, Pointe Claire, Quebec H9R 1G6, Canada. Tel: +1-514-630-8833 Fax: +1-514-630-8850 Email: [email protected] 1 Introduction The formation of apical tight junctions between adja- cent epithelial cells of the epididymis and the creation of the blood-epididymal barrier represents a key element of epididymal physiology. The formation of this cellular bar- rier is critical not only for the protection of spermatozoa from the immune system but, in cooperation with the secretory and endocytic functions of the epithelial cells, regulates the composition of the luminal environment of the epididymis that is responsible for sperm maturation. Much of our knowledge on the regulation and the cellular and molecular makeup of the blood-epididymal barrier has been acquired from animal models, which provide the basis of our understanding of this barrier. Although animal model studies are critical for understanding human function in many systems, including male reproduction, there are limi- tations regarding the applicability of observations made using animal models to studies on normal functions and pathologies of the human epididymis leading to reduced fertility. The objectives of this review are to examine what we have learned about the blood-epididymal barrier in ani- mal models and to compare it with recent data on the human epididymis by assessing the similarities and differ-

Upload: trankhanh

Post on 01-Sep-2018

213 views

Category:

Documents


0 download

TRANSCRIPT

Page 1: Orchestration of occludins, claudins, catenins and ... · Asian J Androl 2007; 9 (4): 463–475 Tel: +86-21-5492-2824; Fax: +86-21-5492-2825; Shanghai, China.463. Orchestration of

Asian J Androl 2007; 9 (4): 463–475

.463.Tel: +86-21-5492-2824; Fax: +86-21-5492-2825; Shanghai, China

Orchestration of occludins, claudins, catenins and cadherinsas players involved in maintenance of the blood-epididymalbarrier in animals and humansDaniel G. Cyr1,3, Mary Gregory1, Évemie Dubé1, Julie Dufresne1, Peter T. K. Chan2, Louis Hermo3

1INRS-Institut Armand Frappier, Université du Québec, 245 Hymus Boulevard, Pointe Claire, QC H9R 1G6, Canada2Department of Urology, Royal Victoria Hospital, McGill University, Montreal, QC H3A 1A1, Canada3Department of Anatomy and Cell Biology, McGill University, Montreal, QC H3A 2B2, Canada

Abstract

Although spermatozoa are formed during spermatogenesis in the testis, testicular spermatozoa are immature andcannot swim or fertilize. These critical spermatozoal functions are acquired in the epididymis where a specific luminalenvironment is created by the blood-epididymal barrier; proteins secreted by epididymal principal cells bind to matur-ing spermatozoa and regulate the maturational process of the spermatozoa. In the epididymis, epithelial cell-cellinteractions are mediated by adhering junctions, necessary for cell adhesion, and by tight junctions, which form theblood–epididymal barrier. The regulation of these cellular junctions is thought to represent a key determinant in theprocess of sperm maturation within the epididymis. Tight junctions between adjacent principal cells permit theformation of a specific microenvironment in the lumen of the epididymis that is essential for sperm maturation.Although we have made significant progress in understanding epididymal function and the blood-epididymal barrier,using animal models, there is limited information on the human epididymis. If we are to understand the normal andpathological conditions attributable to human epididymal function, we must clearly establish the physiological, cellularand molecular regulation of the human epididymis, develop tools to characterize these functions and develop clinicalstrategies that will use epididymal functions to improve treatment of infertility. (Asian J Androl 2007 July; 9: 463–475)

Keywords: claudins; cadherins; catenins; human; rat; mouse; tight junction; adherens junction

.Review .

DOI: 10.1111/j.1745-7262.2007.00308.xwww.asiaandro.com

© 2007, Asian Journal of Andrology, Shanghai Institute of Materia Medica, Chinese Academy of Sciences. All rights reserved.

Correspondence to: Dr Daniel G. Cyr, INRS-Institut ArmandFrappier, Université du Québec, 245 Hymus Boulevard, PointeClaire, Quebec H9R 1G6, Canada.Tel: +1-514-630-8833 Fax: +1-514-630-8850Email: [email protected]

1 Introduction

The formation of apical tight junctions between adja-cent epithelial cells of the epididymis and the creation ofthe blood-epididymal barrier represents a key element ofepididymal physiology. The formation of this cellular bar-rier is critical not only for the protection of spermatozoafrom the immune system but, in cooperation with thesecretory and endocytic functions of the epithelial cells,

regulates the composition of the luminal environment ofthe epididymis that is responsible for sperm maturation.Much of our knowledge on the regulation and the cellularand molecular makeup of the blood-epididymal barrier hasbeen acquired from animal models, which provide the basisof our understanding of this barrier. Although animal modelstudies are critical for understanding human function inmany systems, including male reproduction, there are limi-tations regarding the applicability of observations madeusing animal models to studies on normal functions andpathologies of the human epididymis leading to reducedfertility. The objectives of this review are to examine whatwe have learned about the blood-epididymal barrier in ani-mal models and to compare it with recent data on thehuman epididymis by assessing the similarities and differ-

Page 2: Orchestration of occludins, claudins, catenins and ... · Asian J Androl 2007; 9 (4): 463–475 Tel: +86-21-5492-2824; Fax: +86-21-5492-2825; Shanghai, China.463. Orchestration of

.464.

Components of the blood-epididymal barrier

http://www.asiaandro.com; [email protected]

ences between species.

2 The epididymal junctional complex

Tight junctions between epithelial principal cells areresponsible for the formation of the blood-epididymalbarrier [1, 2]. The ultrastructural anatomy of the blood-epididymal barrier was first described by Friend and Gilula[3] who reported the presence of a series of tight junc-tions between adjacent principal cells of the epididymis.Freeze fracture studies by Suzuki and Nagano [4] pro-vided further evidence of a blood-epididymal barrier byshowing the presence of tight junction mesh networksbetween principal cells. They also demonstrated thatthese junctions varied in number and complexity alongthe epididymis, being more extensive in the caputepididymidis and least extensive in the cauda epididymidis.These observations were substantiated by routine EManalyses, which showed that in the initial segment, thejunctional complex between adjacent principal cells iscomposed of an extensive tight junction that spans aconsiderable length of the adjacent plasma membranes,but contains relatively few desmosomes, whereas in theother regions of the epididymis the extent of the tightjunction is reduced and there are more desmosomes [5].We postulated that because the epididymal lumen in theinitial segment is smaller than in the other regions of theepididymis, the pressure on the epithelium might begreater and this might account for the need for a moreextensive tight junction [5].

Morphological evidence for the integrity of the bar-rier was provided by Hoffer and Hinton [6], who de-monstrated that although both lanthanum and inulin cancross the blood vessels and basement membrane of theepididymis, these markers cannot cross the tight junc-tions between the epithelial principal cells to enter the lu-men of the tubule. Using lanthanum exclusion experiments,Agarwal and Hoffer [7] showed that the blood-epididymalbarrier begins to form in the rat caput epididymidis by18 days of age and is complete throughout the epididymisby 21 days. Guan et al. [8] reported that in the Wistar ratthe blood–epididymal barrier is already fully formed by7 days of age. Pelletier [9] reported that the blood-epi-didymal barrier is functional during embryonic develop-ment in the mink. In the murine model, we have shownthat occludin is localized to tight junctions by embryonicday 13.5 and that an epididymal lumen is already begin-ning to form at this age. Although this observation doesnot preclude the possibility that the murine blood-epi-didymal barrier is leaky, it does suggest that a barrier isalready in place by that age. These studies point to thenecessity for re-examining the timing of the formation

of the blood-epididymal barrier at this window in time toidentify the factors regulating the formation of the blood-epididymal barrier. Horseradish peroxidase perfusionstudies have demonstrated the presence of the blood-epididymal barrier in primates, including in humans [10].

Intercellular junctions are dynamic interactions. Por-tions of the lateral plasma membrane of adjacent princi-pal cells are internalized along the entire epididymis, form-ing annular junctions [5]. In Sertoli cells of the testis,tight and gap junctions are also disposed through annularjunctions [9]. In these cells, it has been postulated that theturnover of junctions is associated with the movement ofproliferating spermatocytes from the basal compartmentto the adluminal compartment. In the epididymis, onlyblood cells, monocytes and lymphocytes appear betweenadjacent principal cells, but junctions between these twocell types are not present, nor do they penetrate the junc-tional complex under normal circumstances [11, 12].Turnover of plasma membranes in the epididymis sug-gests a mechanism whereby junctional proteins may berenewed.

3 Tight junctions and the blood-epididymal barrier

Although the data described above provide physicalevidence for a blood-epididymal barrier, the evidence ofa functional blood-tissue barrier has been suggested bythe large concentration differences between ions, inor-ganic and organic molecules within the lumen of the con-voluted tubule and the extracellular fluid [13]. Using di-rect micropuncture of the tubule following perfusion withlabeled organic molecules, the epididymis can concen-trate certain organic molecules such as carnitine and inosi-tol from 10-fold to 100-fold, while other compounds suchas inulin, glucose and serum albumin can be effectivelyexcluded [14]. This indicates that the barrier is selectiveand transports certain molecules against a concentrationgradient into the epididymal lumen.

4 Creation of a specific epididymal luminal envi-ronment

Tight junctions are essential structures for functionalepithelial physiology. They serve several functions, in-cluding a “barrier” role between cells and the paracellularspace [15, 16], as well as a boundary between the apicaland basolateral plasma membranes, which generates andmaintains cell polarity, the so-called “fence” function [17,18]. Both barrier and fence functions of tight junctions,in addition to vectorial transport across the epithelium,permit the development of a specific luminal micro-environment. In the epididymis, it has long been thought

Page 3: Orchestration of occludins, claudins, catenins and ... · Asian J Androl 2007; 9 (4): 463–475 Tel: +86-21-5492-2824; Fax: +86-21-5492-2825; Shanghai, China.463. Orchestration of

Asian J Androl 2007; 9 (4): 463–475

.465.Tel: +86-21-5492-2824; Fax: +86-21-5492-2825; Shanghai, China

that the luminal environment is responsible for spermmaturation. Although the structural components of ad-hering and tight junctions are necessary for the forma-tion and maintenance of the blood-epididymal barrier, theselective transport of ions and solutes across the epithe-lium as well as proteins secreted and endocytosed byprincipal cells, all contribute to form this specific luminalenvironment.

The movement of solutes, ions and water acrossepithelia occurs through both transcellular and paracellularroutes. The total transepithelial transport across an epi-thelium is the sum of the two distinct components:transcellular and paracellular transport. Transcellulartransport results from the regulated movement of ions,solutes and water across the apical and basolateral mem-branes by specific protein pumps, channels and trans-porter proteins. In this way, these systems maintain theproper acidic pH of the lumen, cell volume, intracellularpH and rapid movement of water across the epithelium.Transcellular transport has a very high degree of mo-lecular specificity, is tightly regulated, and is variableamong different epithelia. This type of transport is active,dependent either on hydrolysis of ATP or on the electro-osmotic gradient generated by basolaterally positionedNa+/K+-ATPase. Maintenance of these gradients is de-pendent on limiting back diffusion between cells throughthe paracellular pathway. In this way, the tightness ofthe paracellular barrier and its molecular selectivity in theepididymis contribute significantly to overall epithelialtransport characteristics and maintenance of the epid-idymal lumen to perform its function of sperm matura-tion [19, 20].

5 Adhering junctions and cadherins

The formation of intercellular junctions involves theinteractions of cell adhesion molecules between adjacentcells, followed by the addition of junctional proteins thatassemble into tight and gap junctions [21, 22]. The pro-cess of cell adhesion is a fundamental cornerstone tofurther intercellular junction formation. Adhering junc-tions are formed by cadherins, a large family of calcium-dependent cell adhesion molecules, which mediate cal-cium-dependent homotypic interactions between adjacentcells (Figure 1). Over 100 different cadherins have nowbeen identified [23]. Cadherins are single-pass trans-membrane glycoproteins that are anchored to the actin-based cortical cytoplasm via catenins [24, 25]. Cadherinshave been subdivided into several subgroups: classical/Type I cadherins; atypical/Type II cadherins; desmosomalcadherins; protocadherins; and cadherin-related proteins[26]. We will focus exclusively on classical or Type I

cadherins in this review, because they are the onlycadherins that have thus far been identified in theepididymis. The primary sequence of classical cadherinsconsists of a long extracellular region composed ofectodomains, which contain the cadherin recognition andbinding site as well as calcium-binding sites. The firstectodomain contains an HAV amino acid sequence ne-cessary for cadherin-cadherin binding [27, 28]. Theamino acids adjacent to the HAV sequence are necessaryfor the recognition specificity of cadherins. Calcium bind-ing sites are present on each subdomain of the extracel-lular region and the presence of calcium is essential forcadherin-mediated cell adhesion. The cytoplasmic do-main of cadherins forms a tight complex with severalproteins which either link cadherins to the cytoskeletonor are involved in signal transduction pathways. Theseinclude catenins, actinin, vinculin and tight junctionalprotein 1 (also known as zonula occludens-1; TJP1).The cadherin-catenin complex is essential for cadherin-mediated cell adhesion. In a variety of cell types, thelevels of cadherins appear to regulate the process of

Figure 1. Schematic representation of the epididymal tight andadherens junctional complex. Several transmembrane tight junc-tional proteins named claudins (CLDNS) have been identified inepididymal tight junctions as well as tight junctional protein 1(TJP1), which binds to the cytoplasmic tail of CLDNS and acts asboth an anchor to the cytoskeleton and a signal transduction protein.Several cadherins (CDH) have also been identified in the rat epid-idymis as well as several members of the catenin (CTNN) familythat bind to the cytoplasmic terminal of cadherins and are impli-cated in linking cadherins to the cytoskeleton and in signaltransduction.

Page 4: Orchestration of occludins, claudins, catenins and ... · Asian J Androl 2007; 9 (4): 463–475 Tel: +86-21-5492-2824; Fax: +86-21-5492-2825; Shanghai, China.463. Orchestration of

.466.

Components of the blood-epididymal barrier

http://www.asiaandro.com; [email protected]

adhesion, suggesting that the cytoplasmic binding pro-teins are present in excess [29]. Hence, one of the func-tions of cadherins might be to sequester β-catenin fromthe cytoplasm into adhering junctions. This might indi-rectly alter gene expression, because cytoplasmic β-catenin can be stabilized by signaling pathways and maythen be translocated to the nucleus where it interactswith the transcription factor LEF-1 to regulate the ex-pression of a variety of genes [30].

The identification of catenins binding to the cyto-plasmic domain of cadherins has been shown to be im-portant not only for linking cadherins to the cell cytosk-eleton but also for intracellular signaling [31–34]. Bothβ-catenin and γ-catenin, or plakoglobulin, have a highdegree of homology with the Armadillo family of proteins,which encodes components of a signal transduction path-way in Drosophila [35].

There has been considerable interest in signal trans-duction pathways and cadherins. Tyrosine kinases havebeen identified in adherens junctions and their phospho-tyrosine activity can be induced [36]. Increased tyrosinephosphorylation by transfection of cells with the Src andRas oncogenes is correlated with a loss of intercellularadhesion [36]. Tyrosine phosphatase inhibitors have beenreported to increase intercellular adhesion. Proteins suchas p100/p120 are also members of the Armadillo familyand have been localized to adhering junctions. Activa-tion of protein kinase C (PKC), a serine/threonine kinase,results in a dephosphorylation of p100/p120 by perturb-ing the phosphorylation cycle, leading to increased per-meability across epithelial cell monolayers [37]. Hence,the regulation of cadherins, either directly or indirectly,is an important factor contributing to the regulation ofintercellular junctions [38]. Small GTPases (Rac, Rhoand Cdc42), have also been implicated in cadherin-medi-ated adhesion [39]. These proteins appear to be involvedin regulating actin-membrane interactions. Although thephysiological role of these proteins is unclear, they ap-pear to be involved primarily in the assembly and/or dis-assembly of adherens junctions [33, 39]. Nectins andintegrins have also been implicated in regulation ofcadherins [40].

E-Cadherin (CDH1) and P-Cadherin (CDH3) arepresent in the rat epididymis [41, 42]. CDH3 mRNAlevels are maximally expressed in the first 7 days postna-tally, whereas CDH1 mRNA peaks at the time of puberty.Immunogold labeling at the electron microscope levelindicates that CDH1 is localized between the lateral plasmamembranes of adjacent principal cells at the level ofapically located junctional complexes in the adult ratepididymis, as well as in the deeper underlying regionsof the extracellular space between the lateral plasma

membranes [43, 44]. Similar observations were alsonoted in the human and murine epididymis [45-47].

As tissues differentiate, they switch from onecadherin to another to form new cell contacts. This es-sential characteristic of cell adhesion molecules in devel-oping tissues leads to the conclusion that they must beregulated either by endocrine, autocrine and/or paracrinefactors, which allow cells to coordinate the expression oftheir cell adhesion molecules to establish new contactswith each other. In the caput-corpus epididymidis, age-dependent changes in CDH1 mRNA concentrations dur-ing postnatal development increase to peak at 42 days ofage [43]. These changes in CDH1 mRNA levels in thecaput-corpus epididymidis are correlated with develop-mental changes in 5α-reductase activity [48]. This en-zyme catalyzes the conversion of testosterone to its ac-tive metabolite dihydrotestosterone. Interestingly, CDH1mRNA concentrations in the cauda epididymidis are notregulated in a similar fashion [43]; rather, CDH1 mRNAlevels do not increase with rising serum levels of andro-gens that occur during postnatal development. However,in adult orchidectomized rats whose testosterone levelsare maintained, Cyr et al. [42] demonstrate that in allsegments of the epididymis, including the cauda, CDH1mRNA levels are androgen-dependent. Hence, the regu-lation of CDH1 in the cauda epididymidis might reflectmultifactorial regulation. CDH1 immunostaining in thecorpus epididymidis decreased from an intenseimmunoreaction in 3-month-old rats to below the levelsof detection in 24-month-old aging Brown-Norway rats.The blood-epididymal barrier in these older rats was com-promised and immunostaining for both occludin and TJP1was absent. In contrast, in the initial segment, CDH1immunostaining increased as a function of age, and inthis region the blood-epididymal barrier remained func-tional [44].

Alpha-Catenin (CTNNA) was localized along the lat-eral plasma membranes of adjacent principal cells as wellas between principal and both clear and basal cells.Immunostaining was more intense in the distal corpusand cauda regions of the epididymis compared with otherregions. A postnatal developmental analysis of CTNNAdistribution revealed that in the epididymis, immuno-staining was already intense by day 7 between the adja-cent epithelial cells in all regions except the cauda, andby day 28 staining was present throughout the epididymis.Because androgen levels are still low at these ages, thedata suggest that CTNNA is not regulated by androgens[49]. Similar observations have also been made forβ-catenin [49]. Furthermore, p120ctn, a catenin that bindscadherins and can alter cell adhesion, has also been iden-tified in the epididymis by immunostaining [49].

Page 5: Orchestration of occludins, claudins, catenins and ... · Asian J Androl 2007; 9 (4): 463–475 Tel: +86-21-5492-2824; Fax: +86-21-5492-2825; Shanghai, China.463. Orchestration of

Asian J Androl 2007; 9 (4): 463–475

.467.Tel: +86-21-5492-2824; Fax: +86-21-5492-2825; Shanghai, China

Interestingly, in orchidectomized rats, there is an increasein cytoplasmic staining for both α-catenin and β-cateninimmunostaining. This suggests that in the absence ofandrogens there might be a degradation or disruption ofthe adhering junction [49]. These data indicate that thecadherin-catenin complex in the epididymis resemblesthat of other epithelial cells, but appears to be regulated,at least in part, by testicular androgens.

6 Occludin

Occludin was the first transmembrane protein iden-tified in tight junctional strands. It is a phosphoproteinof approximately 65 kDa containing four membrane span-ning domains and two extracellular loops [50]. It has along extracellular carboxy-terminal and a short intracel-lular amino terminal and two extracellular loops. Oneoccludin binds to an occludin molecule from an adjacentcell and has been shown to be a functional component oftight junctions. In Xenopus, the expression of truncatedoccludin increases paracellular leakage, whereas in cul-tured epithelial cells, synthetic peptides corresponding tothe second extracellular loop of occludin decreasetransepithelial resistance [51]. Occludin has been identi-fied in a variety of tissues and is associated with thecytoskeleton through a direct interaction with variousproteins [50, 52].

Tight junctions are formed in the murine epididymisas early as embryonic day 12, as shown by freeze frac-ture electron microscopy [54]. At this age, a mesh net-work of tight junctions surrounds the entire circumferenceof the epithelial epididymal cells at the juxtaluminal posi-tion [54]. Occludin is expressed in the murine epididymisas early as embryonic day 13.5, although at this age thereis a strong cytoplasmic reaction, suggesting that the for-mation of tight junctions at this age might still beincomplete. Although it has been reported that tight junc-tions increase in the epididymis during embryonic devel-opment [54], we could not assess by immunofluores-cence whether or not occludin levels were also increas-ing [52]. The expression of occludin in the area of thetight junctions in the developing epididymis (embryonicday 18.5) occurs much earlier than noted in the semini-ferous epithelium (postnatal day 14). Occludin is loca-lized to the apical cell surface of the mouse epididymisby embryonic day 18.5, which coincides with peak an-drogen levels, suggesting that androgens might be im-portant in the regulation of embryonic epididymal tightjunctions. Clearly, the results of these experiments sug-gest that the regulation of occludin expression in testicu-lar tight junctions differs from that in epididymal tightjunctions.

In the adult, occludin has been noted apically betweenadjacent principal cells except in the proximal initial segment.In this region, occludin has been seen only in associationwith narrow cells, this despite the fact that there is anextensive tight junction in the initial segment. These datasuggest that other tight junctional proteins must be presentbetween principal cells in this region, as tight junctionshave been shown to exist between these cells [5].

7 Claudins (CLDNS)

The discovery of a second family of transmembranetight junctional proteins named claudins (CLDNS) dem-onstrates the complexity of tight junctions. To date, 20claudins have been identified, and their tissue distribu-tion is widespread and variable. CLDNS can co-localizewith occludin; however, in the absence of occludin,claudins are still recruited to tight junctions, suggesting acrucial role in tight junction formation [53, 55].

We reported that Cldn1 mRNA transcripts and pro-tein are present in all epididymal segments in rat, as earlyas postnatal day 7 [56]. Additionally, immunohistochemi-cal studies show that CLDN1 is not exclusively localizedto tight junctions, but is also located along the lateralplasma membranes of epithelial principal cells, and be-tween principal and basal cells, in all regions of theepididymis. This suggests that although CLDN1 mightbe involved in the formation and maintenance of epididy-mal tight junctions, it might also be involved in othercell–cell interactions. It has been suggested that CLDNSalong the lateral plasma membrane might represent re-serves of CLDNS prior to their incorporation into thetight junctions.

Tight junctions are comprised of multiple CLDNS.Reports in the rat epididymis indicate that tight junctionsare comprised of CLDN1, 2, 3, 4, 5, 6, 7, 8, 9, 10 and11. However, CLDN5 was localized only to the endot-helial cells and although Cldn2 transcripts are present inthe epididymis, these are not translated into proteins. Thesignificance of such a large number of proteins that com-prise epididymal tight junctions is unknown, as is theexact nature of the relationship between these proteins inthe regulation of the blood-epididymal barrier. In MDCKcells, certain CLDNS, such as CLDN2, might increaseparacellular permeability without altering the number oftight junctional strands [57]. Turksen and Troy [58]reported epidermal barrier dysfunction in transgenic micethat overexpressed CLDN6. These animals lived only ashort time after birth, putatively as a consequence ofincreased water loss due to a defective epidermal barrier.The authors suggested that CLDN6 was involved in bothepithelial differentiation as well as development of the

Page 6: Orchestration of occludins, claudins, catenins and ... · Asian J Androl 2007; 9 (4): 463–475 Tel: +86-21-5492-2824; Fax: +86-21-5492-2825; Shanghai, China.463. Orchestration of

.468.

Components of the blood-epididymal barrier

http://www.asiaandro.com; [email protected]

epidermal barrier. These findings are similar to thosereported by Furuse et al. [59], who observed rapid waterloss due to lack of an intact epidermal barrier, and deathshortly thereafter, in Cldn1 deficient (knockout) mice.Both studies were among the first to demonstrate theimportance of CLDNS in tight junctions of stratified epi-thelia (such as epidermis), as well as in simple epithelia.It appears that certain CLDNS might play specific rolesin regulating paracellular permeability within the tight junc-tional complex.

The paracellular component of transport is a passiveprocess, resulting from paracellular dissipation of theelectro-osmotic gradients established by transcellulartransport. It also lacks the vast functional diversity andmolecular discrimination of the transcellular component.Paracellular transport has two basic characteristics: (i)permeability, the magnitude of the barrier; and (ii)selectivity, the ability to discriminate molecular size andionic charge. In the kidney, CLDN16 has been shownto be important for Mg2+ transport between cells [60].Numerous claudins have been implicated in regulation ofparacellular barriers in a variety of epithelia [15].

Although these functions have been demonstrated inthe epididymis, changes in ionic composition along theepididymis suggest that paracellular transport is likely torepresent a critical component in the regulation of lumi-nal ionic homeostasis. This might explain why there aredifferences in the expression of CLDNS along theepididymis.

8 Other tight junction proteins

Tight junctions are also composed of integral trans-membrane proteins that are linked to membrane-associ-ated proteins, such as junction-associated membraneproteins and peripheral membrane proteins, including themembrane-associated guanylate kinase (MAGUKhomologues, such as TJP proteins) family and non-MAGUK proteins; namely, symplekin, cingulin and 7H6antigen.

Byers et al. [46] reported the presence of both TJP1and cingulin both in vivo and in cultured epithelial princi-pal cells. In the adult rat epididymis, TJP1 is localizedapically between adjacent principal cells [44, 49]. Im-munoprecipitation studies suggest that TJP1 associateswith components of the adhering junction [49]. BecauseTJP1 is a signaling protein, it is possible that it is in-volved in signaling pathways between adhering and tightjunctions. To determine if β-catenin is associated withTJP1, proteins were isolated from 7, 18 and 91-day-oldrats and immunoprecipitated with either β-catenin or TJP1to compare differences in protein binding. These studies

showed that TJP1 immunoprecipitates contain β-cateninat all ages but that they are associated in greater quantitiesin younger rats as compared to adults. These findingssuggest that TJP1 and β-catenin interact extensively dur-ing the formation of the epididymal tight junctional strands,and much less so after the formation of the tight junc-tional complex appears to stabilize (Figure 2) [49].

9 Regulation of epididymal tight junctions

The factors regulating the formation and maintenanceof the blood-epididymal barrier are unknown. Suzukiand Nagano [4] have reported that orchidectomy of adultmice results in a decrease in the number of tight junc-tional strands in the epididymis. This suggests that main-tenance of epididymal tight junctions is regulated by oneor more testicular factors. In the mink, a seasonalbreeder, Pelletier [9] has reported that the blood-testis

Figure 2. Schematic representation of the assembly of tight junc-tions in the epididymis. (A): Tight junctions are composed ofoccludin, transmembrane tight junctional proteins named claudins(CLDNS) and tight junctional protein (TJP) as well as severalother cytoplasmic proteins (not shown). Adherens junctions arecomposed of cadherins and catenins. It has been proposed in thecalcium switch model, in which calcium induces the formation oftight junctions, that cadherin-cadherin interaction (B) activates se-veral small G-proteins, such as Rho. The activation of G-proteinsand the breakdown of membrane phospholipids, such as phos-phoinositol triphosphate and diacylglycerol, stimulate Ca2+

uptake.Protein kinase C (PKC) is activated and phosphorylates TJP1. Inthe epididymis, TJP1 associates with catenin of the adhering junc-tion during the formation of epididymal tight junctions. This asso-ciation is thought to be important to localize TJP1 to the apicalregion of the cell. TJP1 is then available to associate with CLDNSand occludin in formation of tight junctions.

Page 7: Orchestration of occludins, claudins, catenins and ... · Asian J Androl 2007; 9 (4): 463–475 Tel: +86-21-5492-2824; Fax: +86-21-5492-2825; Shanghai, China.463. Orchestration of

Asian J Androl 2007; 9 (4): 463–475

.469.Tel: +86-21-5492-2824; Fax: +86-21-5492-2825; Shanghai, China

barrier is compromised at the end of the reproductiveseason when serum testosterone levels decrease.However, unlike the blood-testis barrier, the blood-epid-idymal barrier remains intact throughout the year, sug-gesting that it is regulated differently from the blood-testis barrier. Based on the ultrastructure of the blood-epididymal barrier during embryonic and postnataldevelopment, it would appear that at least two factorsregulate epididymal tight junctions. The first occursduring embryonic development, when tight junctions areformed and the second, postnatally, when the number ofjunctional strands increases to form the barrier.

The regulation of epididymal tight junctions and theblood–epididymal barrier is poorly understood. In theadult, the integrity of the barrier is unaltered by experi-mental treatments with either estradiol or vasectomy. Theonly evidence for manipulation of the barrier is the mor-phological observation that the tight junctions betweenprincipal cells 3 days following orchidectomy appearedleaky [4], suggesting that testosterone or other testicularfactors might regulate the integrity of the blood-epididy-mal barrier. Such an effect might be a result of a dedif-ferentiation of cells, which might occur following theremoval of androgens [4].

We have shown that orchidectomy in rats resulted inchanges to the staining pattern of CLDN1 in the initialsegment of the epididymis. In this region of the epididymis,there was an absence of CLDN1 immunostaining in thearea of the epididymal tight junction 14 days afterorchidectomy. Testosterone replacement maintained ex-pression apically between the lateral plasma membranesof adjacent principal cells. No changes to CLDN1 ex-pression were noted in any other regions of the epididymis.Therefore, androgens appear to regulate CLDN1 expres-sion in epididymal tight junctions in a segment-specificmanner. Postnatal developmental studies indicate that fac-tors other than androgens regulate CLDN1 expression inthe initial segment and other epididymal regions, becauseCLDN1 is present in the epithelium as early as day 7, at atime when androgen levels are below detection [48].

Thyroid hormones also appear to play a role in theregulation of epididymal CLDN1. In studies in whichrats were made hypothyroid during pubertal developmentby the administration of n-propylthiouracil (PTU), CLDN1expression does not occur in the initial segment of theepididymis (Figure 3). CLDN1 expression was unalteredin other regions of the epididymis.

The recent development of epididymal cell lines indifferent species, including the rat, which expressCLDN1, has provided long awaited tools for studyingepididymal gene expression [61–64]. We investigatedthe transcriptional regulation of the Cldn1 gene in the rat

epididymis. A 1.8-kb sequence of the 5’ flanking regionof the rat Cldn1 gene was cloned. The transcriptionalstart site was identified as an adenine located at the –198position relative to the first codon and 26 bp downstreamof the putative TATA box. It is the only start site for theCldn1 gene transcription in the rat epididymis. The Cldn1promoter was inserted into a luciferase gene expressionvector and transfected into a rat caput epididymidal cellline (RCE-1) [64]. Sequential deletion analysis revealedthat minimal promoter activity was achieved with theconstruct containing –61 bp to +164 bp of the promoter.This sequence contained a TATA box and two consen-sus SP1 binding sites. Electrophoretic mobility shift andsupershift assays confirmed that SP1 and SP3 werepresent in RCE-1 cell and epididymal nuclear extracts,and that they bind to the 5' SP1 binding motif of thepromoter. Site directed mutagenesis of the 5' SP1 bind-ing site resulted in a 4-fold decrease in transactivation ofthe minimal promoter sequence [64]. These findingsindicate that SP1 binds to the Cldn1 promoter region andthat this interaction influences the expression of Cldn1

Figure 3. Thyroid hormones regulate the expression of transmem-brane tight junctional proteins named claudin-1 (CLDN1) in theinitial segment of the epididymis. Rats were treated with the anti-thyroid drug n-propylthiouracil (0.01%; PTU) in their drinking wa-ter from days 0 to 28. Controls received water alone. At day 30,CLDN1 (arrows) is localized along the lateral plasma membrane andat the region of the tight junction in the initial segment (A) and caput(C) of the epididymis. A similar pattern of immunostaining wasobserved in the corpus and cauda epididymidis (not shown). InPTU-treated rats, CLDN1 immunostaining was absent from the ini-tial segment (B) but its expression in the caput epididymidis (D) andother regions of the epididymis was unaltered (not shown). (400 ×).L, Lumen; IT, interstitium.

Page 8: Orchestration of occludins, claudins, catenins and ... · Asian J Androl 2007; 9 (4): 463–475 Tel: +86-21-5492-2824; Fax: +86-21-5492-2825; Shanghai, China.463. Orchestration of

.470.

Components of the blood-epididymal barrier

http://www.asiaandro.com; [email protected]

in the rat epididymis. Protein binding to the 5' regulatoryregion of the Cldn1 gene was also demonstrated, althoughthe binding factors have yet to be identified.

10 The human epididymis

The human epididymal epithelium is a pseudostratifiedcolumnar epithelium, which presents different cell typesincluding principal and basal and clear cells. Principalcells, the predominant secretory cells of the epididymis,are characterized by the presence of a secretory appara-tus (endoplasmic reticulum [ER], Golgi apparatus, secre-tory granules) and an endocytic apparatus (coated pits,endosomes, multivesicular bodies and lysosomes). Basalcells, which are hemispherical in appearance, adhere tothe basement membrane and do not have direct accessto the lumen of the duct [49]. Different secretory andendocytic organelles could be observed in the cytoplasmof the principal cells as well as apical tight junctionalcomplexes between adjacent principal cells (Figure 4).As in the rodent model, the human tight junctional com-plex is localized between adjacent epithelial cells that sur-round the lumen of the epididymis. The ultrastructureof the tight junction appears similar between the differ-ent regions of the epididymis.

11 Human pathologies and the blood-epididymalbarrier

Almost 12% of couples experience fertility problemsand male infertility accounts for approximately half ofthese cases [65]. Male infertility can be classified intothree major categories: pre-testicular dysfunction(endocrinological and genetic disorders), intra-testiculardisorders (e.g. Sertoli cell only syndrome, spermatoge-nic defects, gonadotoxin exposure, impaired sperm DNAintegrity and cryptorchidism), and post-testicular disor-

ders (obstruction, infection, immune, ejaculatory andcoital disorders). In patients with normal or high spermnumbers and sperm motility who are diagnosed with id-iopathic infertility, it is generally thought that post-tes-ticular factors might contribute to their infertility, as theproblem may be associated with the process of spermmaturation [66, 67]. Although male gametes are formedin the seminiferous epithelium of the testis throughspermatogenesis, testicular spermatozoa do not have theability to fertilize or swim. These functions are acquiredin the epididymis [12]. Therefore, post-testicular infer-tility might be the result of pathological dysfunction inthe epididymis, which results in incomplete or dysfunc-tional sperm maturation. There are several lines of evi-dence for a direct role of epididymal dysfunction in maleinfertility. Boué et al. [68] identify a protein, P34H, se-creted by the epididymis and which binds to spermato-zoa and is required for binding of the sperm to the zonapellucida of the egg. This protein accumulates on thesperm during epididymal transit and has been proposedas a marker of sperm maturation. In fertile patients,P34H is detectable in all patients by western blot analyses.However, in infertile patients, 40% of patients have un-detectable P34H levels, suggesting that epididymal spermmaturation was incomplete [68].

A second line of evidence for epididymal dysfunc-tion in infertile patients is related to the presence of sperm-reactive antibodies. These antibodies in seminal fluid orbound to spermatozoa have been reported in 6–11% ofinfertile patients and are usually absent or present in lowtiters in men with no fertility problems [69, 70]. Theseantibodies are usually not present until the time of puberty.Sperm are antigenic and are protected from the immunesystem in the testis during the first wave of spermatoge-nesis by the formation of tight junctions between Sertolicells [71]. These tight junctions are necessary for sper-matogenesis and, therefore, because these patients have

Figure 4. Electron micrograph of apical tight junctions in the human epididymis. Apical tight junctions in the caput (A), the corpus (B) andcauda (C) epididymidis. D, desmosome; Lu. Lumen; PM, lateral plasma membrane; TJ, tight junction. Scale bar = 0.5 μm.

Page 9: Orchestration of occludins, claudins, catenins and ... · Asian J Androl 2007; 9 (4): 463–475 Tel: +86-21-5492-2824; Fax: +86-21-5492-2825; Shanghai, China.463. Orchestration of

Asian J Androl 2007; 9 (4): 463–475

.471.Tel: +86-21-5492-2824; Fax: +86-21-5492-2825; Shanghai, China

normal sperm counts, sperm-reactive antibodies are un-likely to arise as a result of testicular dysfunction. In theepididymis, tight junctions between the epithelial cells thatline the lumen of the tubule are responsible for formingthe blood-epididymal barrier [1]. One of the roles of thisbarrier is to protect sperm from the immune system.Diekman et al. [72] analyze a range of potential antigensassociated with sperm-reactive antibodies. They reportthat these antibodies are directed against an epididymalglycoprotein that shares homology with a CD52 lym-phocyte surface molecule, suggesting that the blood-epi-didymal barrier is compromised and that the immunesystem had been activated. In addition there are manyother causes associated with the development of sperm-reactive antibodies, including obstructive epididymalazoospermia, acute epididymitis, infections, scrotal/tes-ticular trauma and post-vasectomy [73, 74]. Becauseinflammatory cytokines are recognized as repressors oftight junctional proteins, it is conceivable that these in-flammatory processes are associated with the disruptionof the blood-epididymal barrier.

12 Proteins implicated in human epididymal tightjunction

There is no information on the proteins that com-prise human epididymal intercellular junctions. To un-derstand the regulation and functions of epididymal tightjunctions, based on our previous studies in the rat, it isessential to establish the components of both adherensand tight junctions that are expressed in the epididymis.To this end, we used a genomics approach to study theexpression of epididymal genes. Epididymides from fourfertile patients undergoing radical orchidectomy for tes-ticular cancer confined within the testicular tunica al-buginea were obtained and subdivided into the caput,corpus and cauda epididymidis. All patients had normalhormone levels, showed intact normal gross anatomy oftheir epididymides, and by light and electron microscopythe ultrastructural features of the epithelium had a nor-mal morphological appearance.

Using oligonucleotide microarrays, we identified2 980 genes that were differentially expressed by at least2-fold throughout the epididymis. Genes that were ex-pressed included several encoding for adhesion proteins(cadherins and catenins) and tight junctional proteins(claudins and tight junction proteins). The human epi-didymis expresses a large number of both cadherins andprotocadherins (Tables 1 and 2). CDH1 was among thehighest expressed cadherin, which is also the case forthe rat epididymis [42]. Interestingly, CDH22 (alsoknown as PB-cadherin) and CDH24 are also highly ex-

pressed in the human epididymis. It has been reported inthe published literature that both CDH22 and CDH24 havetwo transcripts. CDH22 has been reported to be ex-pressed in gonocytes of the testis of young rats [75]. Allthree of these cadherins are expressed at similar levelsthroughout the epididymis. Only one cadherin, CDH16(also known as Ksp-cadherin), showed large differencesin its expression pattern along the epididymis, with highexpression in the corpus and cauda epididymidis. CDH16was first identified in the kidney and has been shown tobe expressed in the murine epididymis during embryonicdevelopment [76]. This cadherin does not have a cha-racteristic HAV sequence in its binding domain and lacksan extensive cytoplasmic region that is present in classi-cal cadherins [76]. Several catenins were also expressedin the epididymis, including three members of the α-catenin family (CTNNA1, CTNNA2 and CTNNA3), β-catenin (CTNNB1) and two p120 catenins (CTNND1

Table 1. Relative expression of cadherins in the human epididymis.The results were obtained by microarrays (n = 4). –, not expressed;+, very weakly expressed (0–30); ++, weakly expressed (30–100);+++, moderately expressed (100–200); ++++, highly expressed(200 and more).

Gene name Relative intensityCaput Corpus Cauda

CDH1 ++++ ++++ ++++CDH2 +++ ++ +CDH3 +++ +++ +++CDH4 ++ ++ ++CDH5 + – +CDH6 ++ ++ ++CDH7 + (n = 1) – + (n = 1)CDH8 + + (n = 1) +CDH9 + + +CDH10 + – +CDH11 ++ ++ ++CDH12 ++ ++ +CDH13 ++ ++ ++CDH15 ++ ++ ++CDH16 ++ ++++ ++++CDH17 – – –CDH18 + (n = 1) – + (n = 1)CDH19 – – –CDH20 – – –CDH22 ++++ ++++ ++++CDH23 ++ ++ +CDH24 ++++ ++++ ++++CDH26 + (n = 1) – + (n = 1)

Page 10: Orchestration of occludins, claudins, catenins and ... · Asian J Androl 2007; 9 (4): 463–475 Tel: +86-21-5492-2824; Fax: +86-21-5492-2825; Shanghai, China.463. Orchestration of

.472.

Components of the blood-epididymal barrier

http://www.asiaandro.com; [email protected]

and CTNND2 [Table 3]). Interestingly, the epididymiscontained high expression levels of CTNNAL1 (alsoknown as α-catulin). This member of the catenin fam-ily is involved in the signal transduction pathway of smallG-proteins, such as Rho [77]. The expression of highlevels of Rho in the human epididymis suggests that theRho signaling pathways might play an important role inepididymal function and the regulation of cadherin medi-ated intracellular signaling.

As in the rat model, the human epididymis also expresseda large number of CLDNS (Table 4). CLDN4, CLDN5 and

CLDN7 appeared to be expressed at highest levels in theepididymis. We have shown in the rat that CLDN4 is ex-pressed at high levels and CLDN5 was localized toendothelial cells. CLDN7 has been reported to be local-ized in the basolateral region of the distal nephron of the

Table 3. Relative expression of catenins in the human epididymis.The results were obtained by microarrays (n = 4). –, not expressed;+, very weakly expressed (0–30); ++, weakly expressed (30–100);+++, moderately expressed (100–200); ++++, highly expressed(200 and more).

Gene name Relative intensityCaput Corpus Cauda

CTNNA1 ++++ ++++ ++++CTNNA2 +++ ++ +++CTNNA3 ++ ++ +CTNNAL1 ++++ ++++ ++++CTNNB1 ++ ++ ++CTNND1 ++ ++ ++CTNND2 ++ ++ ++

Table 2. Relative expression of protocadherins in the humanepididymis. The results were obtained by microarrays (n = 4). –,not expressed; +, very weakly expressed (0–30); ++, weakly ex-pressed (30–100); +++, moderately expressed (100–200); ++++,highly expressed (200 and more).

Gene name Relative intensityCaput Corpus Cauda

PCDH1 ++ ++ ++PCDH7 + (n = 1) – + (n = 1)PCDH8 + + +PCDH10 – – –PCDH12 ++ + ++PCDH15 – – –PCDH17 ++ ++ ++PCDH18 +++ +++ +++PCDH19 – – –PCDH20 – – –PCDH21 + – +PCDHB1 + (n = 1) – + (n = 1)PCDHB3 – – –PCDHB4 ++ ++ ++PCDHB5 ++ ++ ++PCDHB6 ++ (n = 1) – ++ (n = 1)PCDHB7 ++ ++ ++PCDHB8PCDHB9 + – +PCDHB10 + (n = 1) – + (n = 1)PCDHB11 ++ ++ ++PCDHB12 ++ ++ ++ (n = 1)PCDHB13 +++ +++ +++PCDHB14 ++ ++ +PCDHB15 + (n = 1) – + (n = 1)PCDHB16 ++ ++ ++PCDHAC2 +++ (n = 1) – +++ (n = 1)PCDHGA8 ++ ++ ++PCDH11Y – – –PC-LKC ++ ++ ++

Table 4. Relative expression of transmembrane tight junctional pro-teins named claudins (CLDNS) in the human epididymis. The re-sults were obtained by microarrays (n = 4). –, not expressed; +,very weakly expressed (0–50); ++, weakly expressed (50–200);+++, moderately expressed (200–600); ++++, highly expressed(600 and more).

Gene name Relative intensityCaput Corpus Cauda

CLDN1 ++ ++ ++CLDN2 ++ +++ ++++CLDN3 ++ ++ ++CLDN4 ++++ ++++ ++++CLDN5 +++ ++++ ++++CLDN6 + + +CLDN7 ++++ ++++ ++++CLDN8 ++ + +CLDN9 ++ ++ ++CLDN10 ++ ++++ ++++CLDN11 ++ ++ ++CLDN12 ++ ++ ++CLDN14 + + +CLDN15 + + +CLDN16 ++ – ++CLDN17 + + +CLDN18 + + +CLDN19 ++ ++ ++CLDN22 – – –CLDN23 ++ + +

Page 11: Orchestration of occludins, claudins, catenins and ... · Asian J Androl 2007; 9 (4): 463–475 Tel: +86-21-5492-2824; Fax: +86-21-5492-2825; Shanghai, China.463. Orchestration of

Asian J Androl 2007; 9 (4): 463–475

.473.Tel: +86-21-5492-2824; Fax: +86-21-5492-2825; Shanghai, China

kidney and along with CLDN8 have been reported bepart of the cation barrier of the nephron [78]. The ex-pression of most CLDNS was equivalent along theepididymis. However, both CLDN8 and CLDN10showed segment-specific expression levels that wereopposite from one another. CLDN8 was more highlyexpressed in the caput and less so in the corpus andcauda, which is consistent with a role as part of the cat-ion barrier. CLDN10 was more strongly expressed inthe corpus and cauda epididymidis and is also consid-ered part of the cation barrier.

Immunolocalization of CLDN1, 3, 4, 8 and 10 re-vealed that the localization of CLDNS also differed alongthe epididymis. CLDN1, 3 and 4 were localized to tightjunctions, along the lateral margins of principal cells andbetween basal and principal cells in all three segments ofthe human epididymis. CLDN8, in the caput, was loca-lized to the lateral margins of principal cells and to tightjunctions whereas, in the corpus, CLDN8 was localizedto tight junctions and between principal and basal cells.In the cauda, CLDN8 was exclusively localized to tightjunctions. CLDN10, TJP1 and occludin were exclusivelylocalized to tight junctions in all three segments of theepididymis. CLDN10 immunoreaction was much moreintense in the cauda epididymidis than in other regions ofthe epididymis. This is in contrast with studies on therat in which CLDN10 was expressed primarily in theinitial segment [8].

These data suggest that the proteins that compriseepididymal adherens and tight junctions appear to be con-served between the rodent animal models and human.However, there are differences in either expression pat-tern or localization of proteins that comprise these inter-cellular junctions. At the moment, it is unclear why thesedifferences exist or if there are redundancies in the ex-pression of certain proteins that might functionally com-pensate for differences. Clearly, however, as we beginto attempt to understand the regulation of human epi-didymal protein to assess there function in fertility, newtools will need to be developed that will permit more ex-tensive studies on the regulation of human epididymalproteins implicated in the formation and maintenance ofthe human blood–epididymal barrier.

13 Conclusion

Cellular interactions in the epididymis involve com-plex interactions between large multimember families ofproteins. In the epididymis, these interactions are fur-ther complicated by the influence of testicular factorsthat regulate the expression of epididymal genes and cel-lular targeting of proteins. Although these influences in-

crease the complexity of cell-cell interactions, they alsoprovide a unique model in which it is possible to modu-late cellular interactions in vivo. Demonstrating how thevarious cellular interactions in the epididymis functionwith respect to one another and their significance to epi-didymal physiology and sperm maturation remains a majorbut crucial challenge for our understanding of theepididymis.

Acknowledgments

We wish to thank S. DeBellefeuille, N. St. Pierre, N.Egenberger, B. Moosavi and J. Mui for their contributions.Financial support for our studies from the Canadian In-stitutes for Health Research and the Natural Sciencesand Engineering Research Council of Canada is grate-fully acknowledged.

References

1 Cyr DG, Finnson K, Dufresne J, Gregory M. Cellular interac-tions and the blood–epididymal barrier. In: Robaire B, HintonB, editors. The Epididymis: From Molecules to ClinicalPractice. New York: Plenum Press; 2002: 103–18.

2 Cyr DG, Dufresne J, Gregory M. Cellular communicationand the regulation of gap junctions in the epididymis. In:Hinton BT, Turner TT, editors. The third International Con-ference on the Epididymis. Charlottesville: The Van DorenCompany; 2003: 50–9.

3 Friend DS, Gilula NB. Variations in tight junctions and gapjunctions in mammalian tissues. J Cell Biol 1972; 53: 758–76.

4 Suzuki F, Nagano T. Development of tight junctions in thecaput epididymal epithelium of the mouse. Dev Biol 1978;63: 321–34.

5 Cyr DG, Robaire B, Hermo L. Structure and turnover ofjunctional complexes between principal cells of the ratepididymis. Microsc Res Tech 1995; 30: 54–66.

6 Hoffer AP, Hinton BT. Morphological evidence for a bloodepididymis barrier and the effects of gossypol on its integrity.Biol Reprod 1984; 30: 991–1004.

7 Agarwal A, Hoffer AP. Ultrastructural studies on the devel-opment of the blood-epididymal barrier in immature rats. JAndrol 1989; 10: 425–31.

8 Guan X, Inai T, Shibata Y. Segment-specific expression oftight junction proteins, claudin-2 and -10, in the rat epididy-mal epithelium. Arch Histol Cytol 2005; 68: 213–25.

9 Pelletier RM. Cyclic modulation of Sertoli junctional com-plexes in a seasonal breeder: the mink (Mustela vison). AmerJ Anat 1988; 183: 68–102.

10 Yeung CH, Cooper TG, Weinbauer GF, Bergmann M,Kleinhans G, Schulze H, et al. Fluid-phase transcytosis in theprimate epididymis in vitro and in vivo. Int J Androl 1989; 12:384–94.

11 Robaire B, Hermo L. Efferent ducts, epididymis, and vasdeferens: structure, functions, and their regulation. In: Knobil

Page 12: Orchestration of occludins, claudins, catenins and ... · Asian J Androl 2007; 9 (4): 463–475 Tel: +86-21-5492-2824; Fax: +86-21-5492-2825; Shanghai, China.463. Orchestration of

.474.

Components of the blood-epididymal barrier

http://www.asiaandro.com; [email protected]

cell adhesion and signal transduction. Ann NY Acad Sci 1999;886: 37–47.

31 Yap AS, Mullin JM, Stevenson BR. Molecular analysis oftight junction physiology: insights and paradoxes. J MembrBiol 1998; 163: 159–67.

32 Thoreson MA, Anastasiadis PZ, Daniel JM, Ireton RC,Wheelock MJ, Johnson KR, et al. Selective uncoupling ofp120(ctn) from E-cadherin disrupts strong adhesion. J CellBiol 2000; 148: 189–202.

33 Gumbiner BM. Regulation of cadherin adhesive activity. JCell Biol 2000; 148: 399–404.

34 Yamada S, Pokutta S, Drees F, Weis WI, Nelson WJ.Deconstructing the cadherin-catenin-actin complex. Cell 2005;123: 889–901.

35 Peifer M, Pai LM, Casey M. Phosphorylation of the Droso-phila adherens junction protein Armadillo: roles for winglesssignal and zeste-white 3 kinase. Dev Biol 1994;166: 543–56.

36 Owens DW, McLean GW, Wyke AW, Paraskeva C, ParkinsonEK, Frame MC, et al. The catalytic activity of the Src familykinases is required to disrupt cadherin-dependent cell-cellcontacts. Mol Biol Cell 2000; 11: 51–64.

37 RatcliffeMJ, Rubin LL, Staddon JM. Dephosphorylation ofthe cadherin-associated p100/p120 proteins in response toactivation of protein kinase C in epithelial cells. J Biol Chem1997; 272: 31894–901.

38 Xiao K, Oas RG, Chiasson CM, Kowalcyzk AP. Role ofp120-catenin in cadherin trafficking. Biochim Biophys Acta2007; 1773: 8–16.

39 Anastasiadis PZ. P120-ctn: A nexus for contextual signallingvia Rho GTPases. Biochim Biophys Acta 2007; 1773: 34–46.

40 Chen X, Gumbiner BM. Crosstalk between different adhe-sion molecules. Curr Opin Cell Biol 2006; 18: 572–8.

41 Cyr DG, Robaire B. Developmental regulation of epithelialand placental-cadherin mRNA in the rat epididymis. Ann NYAcad Sci 1991; 637: 399–408.

42 Cyr DG, Hermo L, Blaschuk OW, Robaire B. Distributionand regulation of epithelial-cadherin messenger ribonucleic acidand immunocytochemical localization of epithelial cadherin inthe rat epididymis. Endocrinology 1992; 130: 353–63.

43 Cyr DG, Hermo L, Robaire B. Developmental changes in epithe-lial cadherin messenger ribonucleic acid and immunocytochemicallocalization of epithelial cadherin during postnatal epididymaldevelopment in the rat. Endocrinology 1993; 132: 1115–24.

44 Levy S, Robaire B. Segment-specific changes with age in the ex-pression of junctional proteins and the permeability of the blood-epididymis barrier in rats. Biol Reprod 1999; 60: 1392–401.

45 Andersson AM, Edvardsen K, Skakkebaek NE. Expressionand localization of N- and E-cadherin in the human testis andepididymis. Int J Androl 1994; 17: 174–80.

46 Byers SW, Citi S, Anderson JM, Hoxter B. Polarized func-tions and permeability properties of rat epididymal epithelialcells in vitro. J Reprod Fertil 1992; 95: 385–96.

47 Byers S, Jegou B, MacCalman C, Blaschuk O. Sertoli celladhesion molecules and the collective organization of the testis.In: Russell LD, Griswold MD, editors. The Sertoli Cell.Clearwater: Cache River Press; 1993: 461–76.

48 Scheer H, Robaire B. Steroid delta 4-5 alpha-reductase and 3

E, Neill J, editors. The Physiology of Reproduction. NewYork: Raven Press; 1988, 999–1080.

12 Robaire B, Hinton B, Orgebin-Crist MC. The epididymis. In:Knobil E, Neill J, editors. Physiology of Reproduction, 3rdedn. New York: Elsevier; 2006: 1071–148.

13 Hinton BT, Palladino MA. Epididymal epithelium: its contri-bution to the formation of a luminal fluid microenvironment.Microsc Res Tech 1995; 30: 67–81.

14 Hinton BT, Howards SS. Permeability characteristics of theepithelium in the rat caput epididymidis. J Reprod Fertil1981; 63: 95–9.

15 Van Itallie CM, Anderson JM. Claudins and epithelialparacellular transport. Ann Rev Physiol 2006; 68: 403–29.

16 Spring K. Routes and mechanisms of fluid transport byepithelia. Ann Rev Physiol 1998; 60: 105–19.

17 Yap AS, Niessen CM, Gumbiner BM. The juxtamembraneregion of the cadherin cytoplasmic tail supports lateralclustering, adhesive strengthening, and interaction with p120ctn.J Cell Biol 1998;141: 779–89.

18 Cereijido M, Valdes J, Shoshani L, Contreras RG. Role oftight junctions in establishing and maintaining cell polarity.Ann Rev Physiol 1998; 60: 161–77.

19 Hermo L, Robaire B. Epididymal cell types and their functions. In:Robaire B, Hinton B, editors. The Epididymis: from Molecules toClinical Practice. New York: Plenum Press; 2002: 81–102.

20 Breton S. Luminal acidification in the epididymis and vasdeferens. In: Hinton BT, Turner TT, editors. The third Inter-national Conference on the Epididymis. Charlottesville: TheVan Doren Company; 2003: 60–72.

21 Mège RM, Gavard J, Lambert M. Regulation of cell-cell junc-tions by the cytoskeleton. Curr Opin Cell Biol 2006; 18: 541–8.

22 Citi S. The molecular organization of tight junctions. J CellBiol 1993; 121: 485–9.

23 Takeichi M. The cadherin superfamily in neuronal connec-tions and interactions. Nature Rev Neurosci 2007; 8: 11–20.

24 Kowalczyk AP, Reynolds AB. Protecting your tail: regula-tion of cadherin degradation by p120-catenin. Curr Opin CellBiol 2004; 16: 522–7.

25 Fujimoto K, Nagafuchi A, Tsukita S, Kuraoka A, Ohokuma A,Shibata Y. Dynamics of connexins, E-cadherin and alphacatenin on cell membranes during gap junction formation. JCell Sci 1997; 110: 311–22.

26 Nollet F, Kools P, van Roy F. Phylogenetic analysis of thecadherin superfamily allows identification of six major sub-families besides several solitary member. J Mol Biol 2000;299: 551–72.

27 Blaschuk OW, Sullivan R, David S, Pouliot Y. Identificationof a cadherin cell adhesion recognition sequence. Dev Biol1990; 139: 227–29.

28 Ozawa M, Ringwald M, Kemler R. Uvomorulin-catenin com-plex formation is regulated by a specific domain in the cyto-plasmic region of the cell adhesion molecule. Proc Natl AcadSci USA 1990; 87: 4246–50.

29 Guger KA, Gumbiner BM. β-catenin has Wnt-like activityand mimics the Nieuwkoop signaling center in Xenopus dor-sal-ventral patterning. Dev Biol 1995; 172: 115–25.

30 Ben-Ze’ev A. The dual role of cytoskeletal anchor proteins in

Page 13: Orchestration of occludins, claudins, catenins and ... · Asian J Androl 2007; 9 (4): 463–475 Tel: +86-21-5492-2824; Fax: +86-21-5492-2825; Shanghai, China.463. Orchestration of

Asian J Androl 2007; 9 (4): 463–475

.475.Tel: +86-21-5492-2824; Fax: +86-21-5492-2825; Shanghai, China

alpha-hydroxysteroid dehydrogenase in the rat epididymisduring development. Endocrinology 1980; 107: 948–53.

49 DeBellefeuille S, Hermo L, Gregory M, Dufresne J, Cyr DG.Catenins in the rat epididymis: their expression and regulationin adulthood and during postnatal development. Endocrino-logy 2003; 144: 5040–9.

50 Tsukita S. Furuse M. Occludin and claudins in tight junctionstrands: leading or supporting players? Trends Cell Biol 1999;9: 268–73.

51 Wong V, Gumbiner BM. A synthetic peptide correspondingto the extracellular domain of occludin perturbs the tight jun-ction permeability barrier. J Cell Biol 1997; 136: 399–409.

52 Cyr DG, Hermo L, Egenberger N, Mertineit C, Trasler JM,Laird DW. Cellular immunolocalization of occludin duringembryonic and postnatal development of the mouse testis andepididymis. Endocrinology 1999; 140: 3815–25.

53 Saitou M, Furuse M, Sasaki H, Schulzke JD, Fromm M,Takano H, et al. Complex phenotype of mice lacking occludin,a component of tight junction strands. Mol Biol Cell 2000;11: 4131–42.

54 Suzuki F, Nagano T. Changes in occluding tight junctions ofepididymal epithelium in the developing and gonadectomizedmammals. J Cell Biol 1976; 70: 101A.

55 Furuse M, Sasaki H, Fujimoto K, Tsukita S. A single geneproduct, claudin-1 or -2, reconstitutes tight junction strandsand recruits occludin in fibroblasts. J Cell Biol 1998; 143:391–401.

56 Gregory M, Dufresne J, Hermo L, Cyr DG. Claudin-1 is notrestricted to tight junctions in the rat epididymis. Endocrino-logy 2001; 142: 854–63.

57 Furuse, M, Furuse K, Sasaki H, Tsukita S. Conversion ofzonulae occludentes from tight to leaky strand type by intro-ducing claudin-2 into Madin-Darby canine kidney I cells. JCell Biol 2001; 153: 263–72.

58 Turksen K, Troy TC. Permeability barrier dysfunction intransgenic mice overexpressing claudin-6. Development 2002;129: 1775–84.

59 Furuse M, Hata M, Furuse K, Yoshida Y, Haratake A, SugitaniY, et al. Claudin-based tight junctions are crucial for the mam-malian epidermal barrier: a lesson from claudin-1-deficient mice.J Cell Biol 2002; 156:1099–111.

60 Ikari A, Hirai N, Harad H, Sakai H, Hayashi H, Suzuki Y, et al.Association of paracellin-1 with ZO-1 augments the reab-sorption of divalent cations in renal epithelial cells. J BiolChem 2004; 279: 522–7.

61 Telgmann R, Brosens JJ, Kappler-Hanno K, Ivell R, KirchhoffC. Epididymal epithelium immortalized by simian virus 40large T antigen: a model to study epididymal gene expression.Mol Hum Reprod 2001; 7: 935–45.

62 Araki Y, Suzuki K, Matusik RJ, Obinata M, Orgebin-CristMC. Immortalized epididymal cell lines from transgenic miceoverexpressing temperature-sensitive simian virus 40 large T-antigen gene. J Androl 2002; 23: 854–69.

63 Sipila P, Shariatmadari R, Huhtaniemi IT, Poutanen M. Im-mortalization of epididymal epithelium in transgenic mice ex-pressing simian virus 40 T antigen: characterization of celllines and regulation of the polyoma enhancer activator 3. En-docrinology 2004; 145: 437–46.

64 Dufresne J, Cyr DG. The role of Sp-1 as a transcriptionalregulator of Claudin-1 expression in the rat epididymis. BiolReprod 2007 (in press).

65 McLachlan RI. Basis, diagnosis and treatment of immunologi-cal infertility in men. J Reprod Immunol 2002; 57: 35–45.

66 Sullivan R. Male fertility markers, myth or reality. AnimReprod Sci 2004; 82–83: 341–7.

67 Sandlow JI. Evaluation of male reproductive disorders. In: ChanP, Goldstein M, Rosenwaks Z, editorss. Reproductive MedicineSecrets. Philadelphia: Hanley and Belfus; 2004: 23–30.

68 Boué F, Sullivan R. Cases of human infertility are associatedwith the absence of P34H an epididymal sperm antigen. BiolReprod 1996; 54: 1018–24.

69 Baker HW, Clark GN, Hudson B, McBain JC, McGowanMP, et al. Treatment of sperm autoimmunity in men. ClinReprod Fertil 1983; 2: 55–71.

70 Pattinson HA, Mortimer D. Prevalence of sperm surface anti-bodies in the male partners of infertile couples as determined byimmunobead screening. Fertil Steril 1987; 48: 466–9.

71 Pelletier RM. The tight junctions in the testis, epididymis,and vas deferens. In: Cereijido M, Anderson J, editors. TightJunctions, 2nd edn. Boa Raton: CRC Press; 2001: 599–628.

72 Diekman AB, Norton EJ, Westbrook VA, Klotz KL, Naaby-Hansen S, Herr JC. Anti-sperm antibodies from infertile pa-tients and their cognate sperm antigens: a review. Identitybetween SAGA-1, the H6-3C4 antigen, and CD52. Am JReprod Immunol 2000; 43: 134–43.

73 Chan PTK, Schlegal P. Epididymitis and other inflammatoryconditions of the male. In: Robaire B, Hinton B, editors. TheEpididymis: From Molecules to Clinical Practice. New York:Plenum Press; 2002: 533–55.

74 Schoysman R. Management of epididymal dysfunction: Cor-relation with basic physiology. In: Robaire B, Hinton B,editors. The Epididymis: From Molecules to Clinical Practice.New York: Plenum Press; 2002, 473–82.

75 Wu J, Jester WF, Laslett AL, Meinhardt A, Orth JM. Expres-sion of a novel factor, short-type PB-cadherin, in Sertoli cellsand spermatogenic stem cell of the neonatal rat testis. JEndocrinol 2003; 176: 381–91.

76 Thompson MA, Ward DC, Ouaggin SE, Igarashi P, AronsonPS. cDNA cloning and chromosomal localization of the hu-man and mouse isoforms of Ksp-cadherin. Genomics 1998;51: 445–51.

77 Braga VM. Cell-cell adhesion and signalling. Curr Opin CellBiol 2002; 14: 546–56.

78 Li WY, Huey CL, Yu AS. Expression of claudin-7 and -8 alongthe mouse nephron. Am J Physiol Renal Physiol 2004; 286:F1063–71.