Spermatogenesis is the cellular process by which spermatogonia develop into mature spermatids within seminiferous tubules, the functional unit of the mammalian testis, under the structural and nutritional support of Sertoli cells and the precise regulation of endocrine factors. As germ cells develop, they traverse the seminiferous epithelium, a process that involves restructuring of Sertoli-germ cell junctions, as well as Sertoli-Sertoli cell junctions at the blood-testis barrier. The blood-testis barrier, one of the tightest tissue barriers in the mammalian body, divides the seminiferous epithelium into 2 compartments, basal and adluminal. The blood-testis barrier is different from most other tissue barriers in that it is not only comprised of tight junctions. Instead, tight junctions coexist and cofunction with ectoplasmic specializations, desmosomes, and gap junctions to create a unique microenvironment for the completion of meiosis and the subsequent development of spermatids into spermatozoa via spermiogenesis. Studies from the past decade or so have identified the key structural, scaffolding, and signaling proteins of the blood-testis barrier. More recent studies have defined the regulatory mechanisms that underlie blood-testis barrier function. We review here the biology and regulation of the mammalian blood-testis barrier and highlight research areas that should be expanded in future studies.

  • I.

    Introduction

  • II.

    Function and Structure of the Blood-Testis Barrier

    • A.

      Function

    • B.

      Structure

  • III.

    TJ Proteins of the Blood-Testis Barrier

    • A.

      Structural proteins

    • B.

      Scaffolding proteins

    • C.

      Signaling proteins

  • IV.

    Mechanisms of Blood-Testis Barrier Restructuring

    • A.

      Phosphorylation

    • B.

      Endocytosis

  • V.

    Future Directions in the Study of the Blood-Testis Barrier and Concluding Remarks on the Status of Male Contraceptive Research

I. Introduction

Spermatogenesis is comprised of a chronological series of cellular events that result in the production of mature spermatids. It initiates on postnatal day 5 in the rat, and it occurs within seminiferous tubules, the functional unit of the mammalian testis, under the regulation of several endocrine factors that include testosterone, FSH, LH, and estrogen. This cellular process takes approximately 48–53 days in the rat (for reviews, see Refs. 14). The seminiferous epithelium contains 2 types of cells, Sertoli and germ cells. Sertoli cells are polarized epithelial cells that extend from the base of the seminiferous tubule to its lumen. They send out extensive cytoplasmic processes that contact adjacent Sertoli cells and developing germ cells, which form the basis of the specialized cell junctions in the seminiferous epithelium. Spermatogenesis begins with type A spermatogonia that either self-renew by mitosis or differentiate into type B spermatogonia. Type B spermatogonia, which are connected by cytoplasmic bridges (for a review, see Ref. 5), subsequently detach from the basement membrane and give rise to preleptotene spermatocytes, followed by leptotene, zygotene, pachytene, and diplotene spermatocytes. Thereafter, spermatocytes undergo diakinesis, which completes meiosis I, giving rise to secondary spermatocytes. Secondary spermatocytes then undergo meiosis II to produce spermatids. Thereafter, spermatids undergo spermiogenesis, a 19-step process in the rat that involves acrosome formation, tail elongation and maturation, and nuclear changes to produce elongated spermatids. Spermatogenesis ends with spermiation, the release of mature spermatids as spermatozoa from the seminiferous epithelium. Furthermore, Sertoli and germ cells are not the only cells with roles in spermatogenesis. Peritubular myoid cells, contractile cells that encircle seminiferous tubules, function in the expulsion of spermatozoa out of seminiferous tubules and into the epididymis (6, 7). On the other hand, Leydig cells residing in the interstitium secrete testosterone in the presence of LH. Testosterone is needed for the maintenance of the blood-testis barrier, spermatogenesis, and fertility (for reviews, see Refs. 8, 9), and it promotes both Sertoli-germ cell junction assembly and disassembly (10, 11; for reviews, see Refs. 8, 12). For example, testosterone withdrawal results in the detachment of step 8–19 spermatids from the seminiferous epithelium (13, 14). Under normal physiological conditions, monocytes, macrophages, dendritic cells, T cells, natural killer cells, and mast cells are also present in the interstitium. Collectively, these cells maintain spermatogenesis in mammals.

A typical cross-section of the adult rat testis shows hundreds of seminiferous tubules, each at 1 of 14 stages of the seminiferous epithelial cycle (15, 16; for reviews, see Refs. 2, 17). These 14 stages repeat consecutively along the entire length of each seminiferous tubule in the testis, and 1 cycle is comprised of stages I–XIV. Each stage is defined by a unique arrangement of Sertoli and germ cells at different stages of development so that no 2 stages mirror each other. The 14 stages can be easily discerned by the shape of the acrosome and nucleus of spermatids, as well as by the position of elongating/elongated spermatids relative to the basement membrane. For example, spermiation involves enormous changes in Sertoli cell shape as step 19 spermatids line up along the luminal edge and then detach from the seminiferous epithelium at late stage VIII as spermatozoa. The release of spermatozoa is thought to occur in synchrony with the restructuring of the blood-testis barrier at stages VIII—XI, because the 2 events occur at opposite ends of the seminiferous epithelium. Sertoli cells create the blood-testis barrier that divides the seminiferous epithelium into 2 compartments, basal and adluminal. Spermatogonia and preleptotene spermatocytes reside in the basal compartment, whereas other primary and secondary spermatocytes, round spermatids, and elongating/elongated spermatids reside in the adluminal compartment. At late stage VIII/early stage IX, preleptotene/leptotene spermatocytes initiate passage across the blood-testis barrier, eventually entering the adluminal compartment as zygotene spermatocytes at stage XI. How preleptotene/leptotene spermatocytes cross the blood-testis barrier without disrupting spermatogenesis is one of the most intriguing questions in reproductive biology.

We review here the biology and regulation of the mammalian blood-testis barrier. We begin our review by discussing the structure and function of the blood-testis barrier and emphasizing its importance in spermatogenesis. This discussion is followed by several highly informative sections, which focus on specific proteins that contribute to the “fence” (ie, structural and scaffolding proteins) and “gate” (ie, signaling) functions of the tight junction (TJ), the most important component of the blood-testis barrier and the focus of this review. Although these sections are largely meant to serve as reference guides for future studies, we also provide insights on how the blood-testis barrier can be studied. Lastly, we discuss 2 mechanisms (ie, protein phosphorylation and endocytosis) in which proteins are rapidly regulated to bring about blood-testis barrier restructuring.

II. Function and Structure of the Blood-Testis Barrier

A. Function

The blood-testis barrier, which is essential for spermatogenesis, is located near the base of the seminiferous tubule, where it divides the epithelium into 2 distinct compartments, basal and adluminal. Spermatogonia and preleptotene spermatocytes reside in the basal compartment, whereas other primary and secondary spermatocytes, round spermatids, and elongating/elongated spermatids reside in the adluminal compartment. Thus, the function of the blood-testis barrier is to sequester germ cells residing in the adluminal compartment from the circulatory and lymphatic systems, and together with local immune suppression, to provide an immunoprivileged microenvironment for the completion of meiosis (for reviews, see Refs. 1821). However, spermatogonia and preleptotene spermatocytes residing in the basal compartment are exposed to circulatory and lymphatic systems. Although a third transient compartment also exists at stages VIII–XI (2225; for reviews, see Refs. 2628), the blood-testis barrier creates 2, not 3, compartments. The intermediate compartment (Figure 1), which seals leptotene spermatocytes away from basal and adluminal compartments, is somewhat analogous to a hospital isolation room where there are 2 doors, both of which cannot be opened at the same time. As leptotene spermatocytes transit toward the tubule lumen, junction disassembly ahead of spermatocytes is coordinated with junction assembly behind these germ cells so that the 2 spermatogenic events are synchronized. In addition to Sertoli cells, peritubular myoid cells also contribute to barrier function. Interestingly, these cells prevent the entry of electron-opaque tracers (eg, lanthanum nitrate) into approximately 85% of rodent seminiferous tubules (for reviews, see Refs. 26, 29). Presently, it is not known why lanthanum nitrate penetrates some rodent seminiferous tubules and not others. Although TJs are present between peritubular myoid cells, they are not extensive and they remain poorly characterized (for a review, see Ref. 26). For example, TJ proteins that are expressed by Sertoli cells do not appear to be expressed by peritubular myoid cells, and it is not known how peritubular myoid cells contribute to barrier function. Thus, future studies should investigate whether peritubular myoid cells can enhance Sertoli cell barrier function in vitro. After the release of spermatozoa from the seminiferous epithelium, they remain isolated from the circulatory and lymphatic systems by blood-epididymis and blood-vas deferens barriers.

Figure 1

An electron micrograph of the intermediate compartment in the adult rat testis. This image shows 2 leptotene spermatocytes connected by intercellular bridges and enclosed within the intermediate compartment at stages IX–XI of the seminiferous epithelial cycle. The blood-testis barrier, which is created by Sertoli cells (SCs), is visible by the precipitation of lanthanum nitrate (arrowheads), which fails to advance beyond the TJs. Scale bar, 1 μm. (Reproduced from figure 1(e) of Mruk and Cheng [233] and used with permission.)

B. Structure

The blood-testis barrier is different from other tissue barriers in that it is composed of 4 different cell junctions. For example, TJs between capillary endothelial cells that line the cerebral vasculature form the blood-brain barrier, with astrocytes, pericytes, neurons, and the basal lamina indirectly regulating blood-brain barrier function. Capillary endothelial cells create a permeability barrier that is approximately 50–100 times tighter than the one present between peripheral endothelial cells, which is leaky, and they yield a maximal transendothelial electrical resistance measurement of 1500–2000 Ω/cm2 when cultured in vitro (for reviews, see Refs. 3033). The blood-testis barrier is another example of “one of the tightest junctions” in the mammalian body (34). The concept of the blood-testis barrier is based on early biochemical studies that analyzed testicular fluid for specific proteins, amino acids, and ions, and on subsequent morphological studies that examined the passage of iv-injected dyes and large electron-opaque tracers into seminiferous tubules (22, 23; for reviews, see Refs. 2628). Results from in vitro studies support the presence of a Sertoli cell barrier, except that its transepithelial electrical resistance measurement is 60–80 Ω/cm2 (35, 36), indicating the Sertoli cell barrier is leaky compared with the endothelial cell barrier. Although the reason(s) for this difference in Sertoli cell barrier function between in vitro and in vivo systems is not known, it may be due to the absence of peritubular myoid and germ cells in Sertoli cell cultures. As previously stated, peritubular myoid cells contribute to blood-testis barrier function in vivo, whereas germ cells regulate the Sertoli cell barrier in vitro (37), suggesting a better in vitro system is needed. Another reason for this difference between in vitro and in vivo systems may be due to the absence of a specific biological factor(s) that can enhance barrier function.

The blood-testis barrier is created by TJs, ectoplasmic specializations, desmosomes, and gap junctions that are present between Sertoli cells (for reviews, see Refs. 27, 3844) (Figure 2). In vivo, there is no blood-testis barrier between Sertoli and germ cells, as well as between germ cells. The TJ has gate and fence functions, and it is the most important component of the blood-testis barrier. The gate function prevents the passage of water, solutes, and other large molecules between the paracellular space (ie, thereby creating a barrier), whereas the fence function restricts the movement of proteins and lipids between apical and basolateral domains (ie, thereby generating cell polarity). TJs in the testis coexist and cofunction with ectoplasmic specializations (Figure 3). Desmosomes and gap junctions are present between regions of the plasma membrane that contain TJs and basal ectoplasmic specializations (Figure 3). Desmosomes are cell-cell junctions that mediate robust adhesion, whereas gap junctions are cell-cell channels that allow diffusion of metabolites, second messengers, ions, and other molecules smaller than 1 kDa (for reviews, see Refs. 27, 38, 41, 44). TJs, ectoplasmic specializations, and gap junctions attach to actin microfilaments (for reviews, see Refs. 38, 4547), whereas desmosomes attach to intermediate filaments (ie, vimentin) (48; for reviews, see Refs. 38, 41, 49) (Figure 4). Although recent studies in nontesticular cells show cross talk among actin microfilaments, intermediate filaments, and microtubules (for reviews, see Refs. 5055), this is a largely unexplored area of research in the testis.

Figure 2

An illustration of the different types of cell junctions in the seminiferous epithelium of the adult rat testis. There are 4 types of cell junctions in the testis: TJs, ectoplasmic specializations, desmosomes, and gap junctions.

Figure 3

An electron micrograph of the blood-testis barrier in the adult rat testis. The blood-testis barrier is comprised of TJs, basal ectoplasmic specializations (ESs), desmosomes (DSs), and gap junctions (data not shown). TJs are typified by “kisses,” regions of contact between Sertoli cell plasma membranes (arrowheads). The basal ES is characterized by bundles of actin microfilaments (asterisks) positioned between the Sertoli cell plasma membrane and cisternae of endoplasmic reticulum (er). DSs are typified by electron dense material between Sertoli cells. Scale bar, 0.75 μm. (Reproduced from figure 4A of Sarkar et al [267] and used with permission).

Figure 4

The localization of vimentin and γ-catenin in the adult rat testis. Testes were cryosectioned, fixed with methanol, and fluorescently immunostained for vimentin (green), a protein of the intermediate filament cytoskeleton, and γ-catenin (red), a protein of the desmosome and basal ectoplasmic specialization. This image shows a seminiferous tubule at stage IV of the seminiferous epithelial cycle. Vimentin localized to the Sertoli cell stalk, whereas plakoglobin localized at the blood-testis barrier. Cell nuclei were stained with 4′,6-diamidino-2-phenylindole. Scale bar, 10 μm.

In addition to TJs, the blood-testis barrier is also created by ectoplasmic specializations, testis-specific adhesion junctions comprised of hexagonally arranged filamentous actin microfilaments positioned between the plasma membrane and a cistern of endoplasmic reticulum. Ectoplasmic specializations between Sertoli cells are defined as basal ectoplasmic specializations, whereas those between Sertoli cells and elongating/elongated spermatids are defined as apical ectoplasmic specializations (for reviews, see Refs. 38, 45). Ectoplasmic specialization-mediated adhesion is largely constituted by the cadherin-catenin multifunctional complex (for reviews, see Refs. 56, 57). Interestingly, the blood-testis barrier is compromised and fertility declines in mice with a heart-specific deletion of N-cadherin (Cdh2) (58), indicating cadherin is essential for barrier function. Furthermore, TJs and ectoplasmic specializations are very susceptible to damage caused by environmental toxicants or heat stress. For example, an increase in the scrotal temperature results in germ cell apoptosis, thereby inducing oligospermia or azoospermia in several species (5962). When this occurs, there are reversible changes in the levels of TJ and ectoplasmic specialization proteins. Furthermore, the permeability of the blood-testis barrier is also affected, suggesting these changes are partly due to the unique organization of cell junctions within this structure.

III. TJ Proteins of the Blood-Testis Barrier

TJs constitute a specialized microdomain of continuous anastomosing fibrils that surround cells in a belt-like manner. Fibrils from adjacent cells periodically come together, which seals the paracellular space between cells and creates an impermeable barrier (Figure 3). Each fibril is comprised of an integral membrane protein that directly or indirectly associates with scaffolding and signaling proteins, as well as with cytoskeletal proteins (for a review, see Ref. 63).

A. Structural proteins

Tremendous progress has been made to identify, characterize, and study the proteins of the TJ, with the last integral membrane protein being identified in epithelial cells in 2005 (64). However, the proteins of the blood-testis barrier and their regulation remain understudied. In the next sections, we summarize results from studies on occludin, claudin, and tricellulin, the key structural proteins of TJ fibrils. We also discuss coxsackie and adenovirus receptor, because it is postulated to function in the movement of preleptotene/leptotene spermatocytes across the blood-testis barrier.

Occludin

Occludin was identified after a chick liver junction fraction was used to produce a monoclonal antibody against an approximately 65-kDa protein (Figure 5) (65). Since this time, several splice variants of occludin have also been identified. However, these splice variants have not been studied in any organ thus far. Occludin is comprised of 4 membrane-spanning domains, 2 extracellular loops, and 2 intracellular segments, and it is expressed by several cell types and organs that include the brain and liver (for reviews, see Refs. 66, 67). Occludin is also expressed by the mouse and rat testis, but not by the human and guinea pig testis (68). The C terminus of occludin, which is rich in Ser, Thr, and Tyr residues, binds several proteins (for reviews, see Refs. 6971) (Figure 5). Although occludin overexpression in epithelial cells increases transepithelial electrical resistance (72, 73), TJ fibrils are normal in occludin-deficient embryonic stem cells (74). Furthermore, zona occludens (ZO) proteins are still recruited to the TJ in occludin-deficient intestinal epithelial cells (75), revealing occludin is important, but not required, for barrier function. Occludin knockdown also affects the actin cytoskeleton, which is mediated by ras homolog family member A (76), a small GTPase (for a review, see Ref. 77). This is interesting because occludin can localize to the leading edge of migrating MDCK cells (78), and occludin overexpression induces spreading of AC2M2 cells (79). After occludin knockdown, atypical protein kinase C (PKC), par-3 family cell polarity regulator, and protein associated with Lin-7 homolog A (PALS)1-Pals-associated TJ protein (PATJ) fail to accumulate at the leading edge, and phosphoinositide-3-kinase (PI3K) fails to be activated, resulting in a reduced number of cell protrusions (78). These findings illustrate that occludin controls the localization of polarity proteins, the establishment of cell polarity, and the direction of cell movement. Occludin may have a similar role in the testis and facilitate the movement of preleptotene/leptotene spermatocytes across the blood-testis barrier.

An illustration of the main proteins of the TJ, ectoplasmic specialization, desmosome, and gap junction at the blood-testis barrier in the adult rat testis.

Occludin-null mice present with several abnormalities, ranging from inflammation and hyperplasia of the gastric epithelium to calcification of the brain and atrophy of seminiferous tubules (80), as well as germ cell exfoliation and infertility in mice older than 10 weeks of age (234). However, these pathophysiological states are not life threatening, and occludin-null mice reach adulthood. In addition, synthetic peptides corresponding to part of the second extracellular loop sequence increase paracellular permeability in A6 and Sertoli cells by disrupting loop-loop interactions (8183). The importance of this extracellular loop in TJ dynamics is further strengthened by the finding that it is required for the localization of occludin at the TJ (84). On polyacrylamide gels, occludin runs as a series of protein bands from approximately 62 to 82 kDa, which is due to protein phosphorylation, as well as other posttranslational modifications. In general, Ser/Thr-phosphorylated occludin concentrates at the TJ, whereas nonphosphorylated and nonjunctional occludin localizes within cytoplasmic vesicles (85). Taken collectively, these findings suggest that studies of TJ function, regardless of the cell type or organ in which they are performed, should include occludin as a target protein. However, other integral membrane proteins with a gate function at the TJ should be investigated as well.

Claudins

In mammals, the claudin family consists of at least 24 members (Figure 5) that are categorized into classical (claudins 1–10, 14, 15, 17, and 19) or nonclassical (claudins 11–13, 16, 18, and 20–24) types according to sequence similarities (86). Claudins form TJ fibrils based on a study that showed the overexpression of claudin 1 or 2 to induce TJ assembly in fibroblasts, which inherently lack TJs (87). Most cells express more than 2 claudins that together determine the overall paracellular electrical resistance and paracellular charge selectivity of TJs. In other words, a claudin molecule can associate with another claudin molecule of the same or different kind on the adjacent cell membrane (86). Similar to occludin, synthetic peptides corresponding to part of the claudin sequence disrupt claudin-claudin interactions, resulting in the mislocalization of claudin (88). In EpH4 cells, the mislocalization of claudin leads to the mislocalization of occludin (89). Claudins also mediate Ca2+-independent adhesion (90) and serve as receptors or coreceptors for the entry of different toxins and viruses such as Clostridium perfringens enterotoxin, HIV, and hepatitis C virus into cells (9195).

In general, claudins 4, 5, 8, 11, 14, and 18 increase paracellular electrical resistance, whereas claudins 2, 7, 10, 15, and 16 increase paracellular cation permeability (86). Presently, it is not known whether claudin function is restricted to TJ assembly and maintenance because claudins also participate in other events such as the epithelial-mesenchymal transition, invasion, and cell migration. For example, matrix metalloproteases 3, 7, and 8 are up-regulated several-fold in the intestine of Cldn7−/− mice, causing epithelial cell sloughing, mucosal ulcerations, and inflammation (96). Moreover, the deletion of claudin 7 in vivo decreases the expression and affects the localization of integrin α2 (96), suggesting claudins indirectly regulate extracellular matrix degradation via matrix metalloproteases. These events likely trigger the production of cytokines to bring about extracellular matrix degradation and/or cell junction disassembly, because cytokines are known to regulate TJ function in several epithelial cells, including Sertoli cells (for reviews, see Refs. 9799). Similar to occludin, there is a direct link between claudin function and cell motility. These events are mediated by neuronal Wiskott-Aldrich syndrome protein (N-WASP) (a ubiquitous protein that promotes actin nucleation) and ras homolog (RHO)-associated protein kinase (a Ser/Thr kinase that regulates the cytoskeleton), because claudin 5-deficient HECV cells cannot respond to wiskostatin (an N-WASP inhibitor) and Y-27632 (a RHO-associated protein kinase inhibitor) (100). The phosphorylation of claudins by protein kinases is also critical for TJ function (101), and nonphosphorylated claudin is rapidly internalized and degraded in lysosomes (102, 103).

In the testis, claudins 3 and 11 are highest at stages VI–VIII and V–VII, respectively, which precede the movement of preleptotene/leptotene spermatocytes across the blood-testis barrier (104). Moreover, the expression of claudin 3, but not claudin 11, decreases in Arinvflox(exl-neo)Y;Tg(Amh-Cre) mice, which present for conditional androgen sensitivity and azoospermia (105), illustrating claudin 3 is regulated by androgens. The permeability of the blood-testis barrier is also affected in these mice based on a functional assay (105), revealing other claudin molecules cannot functionally substitute for its loss in the testis. These findings also illustrate that androgens promote barrier integrity (105108). Equally important, claudin 3 is present at new TJs present behind migrating spermatocytes, but it is absent at old TJs present ahead of migrating spermatocytes (24, 105). Besides androgens, hormones (ie, FSH), cytokines (ie, TNFα, TGFβ), and germ cells also regulate claudin expression in Sertoli cells (109>–113). Furthermore, Sertoli cell polarity and TJ function are disrupted in Cldn11-null mice, resulting in Sertoli cell detachment and sloughing (114). These findings are in agreement with an earlier study that shows Cldn11−/− mice to be infertile due to the inability of germ cells to differentiate beyond the spermatocyte stage (115). Sertoli cells also proliferate in Cldn11−/− mice (114), suggesting claudin 11 inhibits postnatal Sertoli cell proliferation. Interestingly, this phenotype is similar to that observed in mice with a conditional deletion of connexin 43 (116), a protein of the gap junction that participates in blood-testis barrier function (117, 118). The detachment of Sertoli cells from the basement membrane also suggests that TJ proteins indirectly regulate hemidesmosome function. Besides claudins 3 and 11, the testis also expresses claudins 1, 4, 5, 7, and 8 (for a review, see Ref. 27). Of these, claudin 5 is expressed by Sertoli cells, preleptotene/leptotene spermatocytes, and spermatogonia (119). Presently, it is not known why germ cells express claudin 5, because they do not assemble TJs, but it is likely that claudin 5 facilitates spermatocyte movement across the blood-testis barrier (for a review, see Ref. 46).

Tricellulin and lipolysis-stimulated lipoprotein receptor (LSR)

Tricellulin is a TJ protein that localizes at both bicellular and tricellular contacts (Figure 5) (64, 120123). It is expressed by several epithelial and endothelial cells and organs that include the kidney, liver, lung, brain, and testis (37, 64; for a review, see Ref. 124). Five tricellulin isoforms (TRIC-a–TRIC-e) have been identified (125, 126), and the largest isoform (TRIC-a) is the form that is referred to when the word “tricellulin” is used. At bicellular TJs, tricellulin reduces strand discontinuities, increases paracellular electrical resistance, and decreases the permeability to ions and larger solutes. At tricellular TJs, tricellulin seals epithelial sheets and excludes the passage of macromolecules (127). Interestingly, tricellulin activates cell division cycle 42 (CDC42) via TUBA (also known as dynamin binding protein), a guanine nucleotide exchange factor that promotes actin polymerization during cell junction assembly (128). Tricellulin is also critical for hearing, because a deletion of or a mutation in tricellulin results in deafness (125, 126, 129). Tricellulin contains 4 transmembrane domains and belongs to the TJ-associated family, which is also comprised of occludin (MARVELD1) and MARVELD3 (an alternatively spliced protein that colocalizes with occludin at TJs) (130). These proteins have in common a conserved MAL and related proteins for vesicle trafficking and membrane link (MARVEL) domain, and proteins having a MARVEL domain may function in events such as the biogenesis of transport vesicles (for a review, see Ref. 131).

After tricellulin is silenced by RNA interference, bicellular and tricellular TJs are disorganized, and barrier function is compromised (64). Conversely, tricellulin overexpression excludes ions and larger solutes more effectively (127). Although tricellulin overexpression does not affect the molecular composition of TJs (127), tricellulin mislocalizes to bicellular TJs when occludin is silenced in MDCK II cells (132), suggesting occludin regulates tricellular TJ assembly by excluding tricellulin from bicellular TJs. Alternatively, tricellulin compensates for the loss of occludin (132). It is postulated that tricellulin and occludin are transported to the plasma membrane via a common pathway because both proteins localize to the same site during TJ assembly, except that tricellulin relocates to tricellular contacts after these ultrastructures are formed (133). At tricellular contacts, tricellulin links claudin-based TJ fibrils to LSR, resulting in the formation of vertical TJ fibrils. LSR, which was originally identified as a receptor for triglyceride-rich lipoproteins, functions at tricellular TJs in the liver, kidney, lung, intestine, ovaries, and testis (134). LSR also recruits tricellulin to tricellular TJs after occludin excludes tricellulin from bicellular TJs (132), illustrating LSR functions upstream of tricellulin in the formation of TJs. In addition, LSR is phosphorylated at Ser288 by c-Jun N-terminal kinase, which is necessary for LSR to localize at tricellular junctions (135). Although Lsr knockdown affects tricellular TJ assembly in vitro (136), Lsr−/− mice are embryonically lethal, except for 3 males that survive to reproductive age (137). Interestingly, these mice are infertile, with one mouse unable to develop testes altogether, revealing LSR has an important role in this organ. Interestingly, cytoplasmic bridges, which connect germ cells to form long chains (138; for a review see Ref. 139), pass through tricellular contacts. This conclusion is based on the observation that testis-expressed gene 14, a marker for cytoplasmic bridges, localizes at tricellular contacts (24). Taken collectively, these findings illustrate the importance of tricellulin and LSR, as well as occludin and claudin, in TJ dynamics in the testis. Future studies should investigate the roles of tricellulin and LSR in the movement of preleptotene/leptotene spermatocytes across the blood-testis barrier.

Coxsackievirus and adenovirus receptor (CAR)

CAR (also known as CXADR), a single-span transmembrane protein with 2 extracellular Ig domains (Figure 5), is best studied in the heart because mice deficient for Cxadr or mice with a heart-specific deletion of Cxadr before E10 are embryonically lethal due to defects in this organ. However, mice with a heart-specific deletion of Cxadr after E11 evade major cardiac abnormalities and survive into adulthood (140142), indicating CAR is essential for early embryonic heart development. Defects in several other organs that include the intestine, lung, pancreas, and thymus are also present in adult mice with a conditional deletion of Cxadr (143). At the cellular level, CAR has several functions. For example, the loss of CAR weakens cell adhesion and promotes cell migration via a mechanism that involves SRC-dependent endocytosis of E-cadherin (ie, an elevated CAR level promotes E-cadherin endocytosis) (144). CAR also interacts with occludin (145) and ZO-1 (146). However, CAR is not a component of TJ fibrils. Furthermore, barrier integrity is disrupted by soluble CAR, which is produced either by the de novo synthesis of an alternatively spliced mRNA (147) or the proteolytic cleavage of membrane-bound CAR (146). In human glioma cells, proteolytic cleavage of CAR by a disintegrin and metalloprotease 10 releases the ectodomain (∼32 kDa), whereas intramembrane proteolysis by presenilin/γ-secretase sends the intracellular domain (∼14 kDa) to the nucleus (148). Presently, it is not known whether a similar mechanism exists in the testis. Lastly, CAR functions as a receptor for type B coxsackieviruses and subgroup C adenoviruses. Although it is not completely clear how viruses gain access to CAR, which is sequestered within TJs, they likely disrupt TJ function by triggering the production of proinflammatory cytokines.

In the testis, CAR localizes to the blood-testis barrier, preleptotene/leptotene spermatocytes within the intermediate compartment, the convex side of the elongated spermatid head at stage VIII, and the acrosome (149151). After Cxadr knockdown in Sertoli cells, occludin internalizes and interacts with early endosome antigen 1 (152), which localizes to the cytosolic face of early sorting endosomes, indicating CAR regulates the presence of occludin at the cell surface via endocytosis. During inflammation/infection, CAR is 1 of several proteins involved in the migration of immune cells across the endothelium (153). Although a similar mechanism has been proposed to explain the movement of preleptotene/leptotene spermatocytes across the blood-testis barrier (for a review, see Ref. 154), cell junction dynamics, barrier integrity, germ cell movement, and fertility are not affected in Cxadr-deficient mice produced by the tamoxifen-inducible Cre/lox P system (ie, incomplete knockout) (155). Interestingly, CAR also binds and stabilizes tubulin, which is critical for cell migration (156). Likewise, it is postulated that Sertoli cell microtubules (Figure 6) facilitate the movement of elongated spermatids at stages IV–VI (157, 158). Thus, future studies should investigate whether CAR is involved. Lastly, it is not known why Cxadr knockdown affects the Sertoli cell barrier in vitro but not in vivo (152, 155).

Figure 6

The localization of tubulin in the adult rat testis. Testes were fixed with Bouin's solution and embedded in paraffin wax. Tissue blocks were sectioned and immunostained for α-tubulin, a protein of the microtubule cytoskeleton. A–E, Seminiferous tubules at different stages of the seminiferous epithelial cycle. α-Tubulin localized at the blood-testis barrier (black arrowheads) (A), as well as to the luminal edge (blue arrowheads) (C and D), the Sertoli cell stalk (red arrowheads) (D), and the acrosome of late-step spermatids (green arrowheads) (E). There was no immunoreactive signal when the primary antibody was substituted with rabbit IgG (negative control) (F). The antibody was specific for α-tubulin when tested by immunoblotting (arrowhead) (G). The approximate positions of the molecular weight markers are shown to the left of the immunoblot. Scale bars, 50 μm (A–E) and 200 μm (F).

B. Scaffolding proteins

The cytoplasmic plaque of the TJ is the area adjacent to the TJ. It is enriched in several scaffolding proteins that bring together structural proteins and link them to the cytoskeleton (Figure 5) (for a review, see Ref. 63). These scaffolding proteins also invite signaling proteins that regulate TJ function. Furthermore, many scaffolding proteins contain putative nuclear localization sequences, indicating they transit to the nucleus to turn on or off the expression of target genes (for a review, see Ref. 159). In the next section, we discuss the ZO protein family and highlight relevant findings in the testis.

Zona occludens

ZO-1 (tight junction protein 1, TJP1), ZO-2 (TJP2), and ZO-3 (TJP3) are well-studied adaptor proteins primarily used by integral membrane TJ proteins such as occludin and claudin for attachment to the actin cytoskeleton (Figure 6). ZO-1, ZO-2, and ZO-3 belong to the membrane-associated guanylate kinase (MAGUK) family, which is comprised of 10 distinct subfamilies; ZO proteins comprise 1 of these subfamilies. Several domains define this family, but only 3 of these domains (ie, postsynaptic density 95, discs large, and ZO-1 proteins [PDZ]; Src homology [SH]3; and GUK) are found within most MAGUK family members. Specifically, ZO proteins have 3 PDZ domains, 1 SH3 domain, and 1 GUK domain (for a review, see Ref. 160). These domains allow ZO proteins to interact with several proteins that support TJ function. ZO proteins also function outside of the TJ, because they are present within the nucleus and at other cell junctions (for reviews, see Refs. 161, 162). One of these functions is to regulate cell growth and proliferation (163; for reviews, see Refs. 161, 164, 165). For example, ZO-1 inhibits cell proliferation by binding ZO-1-associated nucleic acid binding protein (ZONAB), a Y-box transcription factor, which prevents the transit of ZONAB into the nucleus, thereby inhibiting its transcriptional activity (166; for a review, see Ref. 159). APG-2, a heat shock protein that controls ZONAB transcriptional activity, competes with this transcription factor for binding to ZO-1 (167, 168). However, Apg-2 knockdown does not prevent TJ assembly. Instead, a deceleration in the rate at which ZO-2, ZO-3, and occludin are recruited to the TJ is observed without any apparent effects on the actin cytoskeleton (167). These results agree with ZO-1 knockdown/knockout studies in other cells (169, 170).

In the testis, ZO-1 also coimmunoprecipitates and colocalizes with connexin 43, and it regulates gap junctional communication (Figure 6) (171). Interestingly, phosphorylation of connexin 43 at Ser373 by protein kinase B (PKB) disrupts ZO-1-connexin 43 interactions, increases gap junction size, and promotes gap junctional communication (172), indicating ZO-1 is a negative regulator of gap junctional communication. Furthermore, ZO-1 mislocalizes from Sertoli cell TJs after the endocytosis of gap junctions is triggered by γ-hexachlorocyclohexane (171). Although the mislocalization of ZO-1 may be due to its interaction with connexin 43, ZO-1 binds other integral membrane proteins such as occludin and claudin (75, 173). Interestingly, these proteins could not retain ZO-1 at the plasma membrane. Not surprisingly, both Tjp1- and Tjp2-deficient mice are embryonically lethal; Tjp1-deficient mice die midgestation (174), whereas Tjp2-deficient mice die postimplantation (175). When the lethality of Tjp2 knockout mice is rescued by injecting Tjp2−/− stem cells into wild type blastocysts to produce ZO-2 chimera, subfertility ensues (176). Although the expression of ZO-1 and ZO-3 is unaffected, blood-testis barrier function is compromised in these mice (176). By contrast, Tjp3-deficient mice show no apparent phenotype (176).

ZO-1 and ZO-2 also promote claudin polymerization, which is essential for TJ assembly (177). For example, epithelial cells polarize but fail to assemble TJs in the absence of claudin polymerization caused by the complete or partial loss of ZO-1 and ZO-2 (177). Claudins also fail to polymerize after the N terminus of ZO-1, which contains PDZ domains 1–3, is introduced into Tjp1 knockout/Tjp2 knockdown cells (177). Instead, PDZ domains 1–3, the SH3 domain, and the GUK domain are all required, indicating SH3 and GUK domains are indispensable for claudin polymerization. Other studies demonstrate that afadin, an actin-binding protein that regulates cell adhesion, is critical for TJ assembly because its knockdown prevents the recruitment of TJ proteins to sites of cell adhesion (178). In the testis, ZO-1 localizes to the blood-testis barrier, but also to the apical ectoplasmic specialization, which surrounds elongated spermatids (179; for a review, see Ref. 27). Furthermore, germ cells do not express ZO-1 (37). ZO-1 localizes to the convex side of the head of elongated spermatids at stages IV–VI of the seminiferous epithelial cycle when spermatids move toward the basement membrane and embed within Sertoli cell crypts. Thus, future studies should investigate whether ZO-1 directly or indirectly interacts with tubulin (Figure 6), tubulin-binding proteins (eg, end-binding protein, a plus-end tracking protein), and/or microtubule-dependent motor proteins (eg, dynein, a minus-end directed motor protein). The involvement of connexin 43 in spermatid movement during these stages should also be investigated because it binds ZO-1. For example, a disruption of ZO-1-connexin 43 interactions may affect spermatid movement at these stages. The use of staged seminiferous tubules provides a good starting point for future experiments.

C. Signaling proteins

Polarized epithelial cells are divided into distinct apical and basal domains that are created by the asymmetric distribution of proteins, lipids, RNA and/or cytoskeletal components (for reviews, see Refs. 180183). Cell polarity is mediated by 3 well-conserved protein modules: 1) the apical Crumbs (CRB)-PALS-PATJ complex; 2) the apicolateral partitioning-defective (PAR)3-PAR6-atypical protein kinase C (aPKC) complex; and 3) the basolateral Scribble (SCRIB)-discs-large (DLG)-lethal giant larvae (LGL) complex (for reviews, see Refs. 184187). These 3 modules function together in the formation of cell polarity, and in the assembly and maintenance of TJs. A fascinating feature of these protein modules is that they antagonize each other, and manipulating different proteins within them can alter the relative surface area of the apical and basal domains, resulting in the formation of cuboidal, columnar, or squamous cell shapes (for a review, see Ref. 188). Although these 3 modules localize to the apical/apicolateral domain in most cells, they are mostly confined to the basal/basolateral domain in Sertoli cells. In the next sections, we discuss these 3 protein modules. We will not discuss planar cell polarity, which refers to the polarity of cells within the horizontal plane.

The CRB-PALS-PATJ cell polarity complex

CRBs are single-span transmembrane proteins that regulate cell polarity, and the CRB-PALS-PATJ cell polarity complex is the only polarity protein module to contain a transmembrane protein (189; for reviews see Refs. 183, 186, 187, 190, 191) (Figure 5). CRBs also function outside of cell polarity; they are tumor suppressors (for reviews, see Refs. 192194), as well as regulators of cell division, survival, and growth (195199). Three CRBs exist in mammals: CRB1, CRB2, and CRB3 (200; for a review, see Ref. 186), and several organs that include the brain and testis express Crb3, the most widely expressed Crb (201, 202). The short cytoplasmic domain of CRB, which consists of a membrane proximal juxtamembrane domain and a C-terminal PDZ-binding motif, is critical for apical domain formation. In addition, the juxtamembrane domain binds proteins (eg, talin, ezrin, radixin, moesin, focal adhesion kinase [FAK]) (for a review, see Ref. 203) that contain a 4.1 protein, ezrin, radixin, and moesin binding domain (189; for reviews, see Refs. 204, 205). These results illustrate that cell polarity involves cross talk among the plasma membrane (ie, TJ), cytosol (ie, cytoskeleton), and extracellular matrix (ie, focal contact and/or hemidesmosome). Interestingly, crb overexpression in Drosophila ectodermal epithelial cells results in the expansion of the apical domain at the expense of the basal domain (206), whereas Crb3 overexpression in MDCK cells delays the assembly of TJs (207). Likewise, the loss of Crb3 disrupts TJ function (208, 209), illustrating CRB manipulation affects TJ function. In Drosophila, Crb regulates the Hippo signaling pathway (196, 210; for a review, see Ref. 211), which controls organ growth, and the Notch signaling cascade, which controls development, differentiation, and survival (for reviews, see Refs. 212, 213). Notch receptors, single-span transmembrane proteins that are activated by cell-cell contact, are cleaved by a disintegrin and metalloprotease or γ-secretase upon ligand binding. This process generates an intracellular fragment, which transits to the nucleus to turn on the expression of target genes that control growth and development (for reviews, see Refs. 214, 215). Crb inhibits the γ-secretase complex (199, 216), illustrating it is a negative regulator of the Notch signaling pathway. In the testis, aberrant Notch signaling results in defects in germ cell fate, an increase in germ cell apoptosis, and infertility (217219). However, the role of CRB in blood-testis barrier dynamics is not known.

PALS, a scaffolding protein of the MAGUK family, binds PATJ (220) and mammalian homolog of Lin-7 (also known as lin-7 homolog A) (221, 222) (Figure 5). In MDCK cells, Pals knockdown results in a decrease in Patj expression and an increase in barrier permeability. It also affects the targeting of aPKC to TJs, thereby disrupting cell polarity and TJ function (223). Interestingly, the loss of Pals1, but not Patj, disrupts the trafficking of newly synthesized E-cadherin to the plasma membrane and the assembly of adherens junctions in MDCK cells (224226), suggesting the retromer complex is involved. The retromer complex sorts and transports proteins from 1) the endosome to the trans-Golgi network, 2) the endosome to the plasma membrane, and 3) the endosome to the trans-Golgi network, and then to the plasma membrane (for a review, see Ref. 227). CRB passes through the trans-Golgi network and associates with the retromer complex (228, 229). Although PALS1 sequesters E-cadherin to the adherens junction, it is not known whether the trafficking of other integral membrane proteins is also affected. Thus, additional studies are needed to determine the roles of CRB and PALS1 in protein trafficking.

PATJ is the third member of this cell polarity complex (Figure 5) (230, 231). PATJ interacts with several proteins that include claudin 1 and ZO-3, with the latter protein sequestering PATJ to the TJ (232). Similar to the knockdown of Crb and Pals, Patj knockdown disrupts TJ function (225, 226). PATJ also binds junction enriched and associated protein, which is expressed by several organs such as the brain and testis (235). Originally identified as a protein that functions in cell movement (236238), junction enriched and associated protein removes PATJ from the plasma membrane and delivers it to the recycling endosome (239), thereby disrupting TJs. Presently, there is no detailed study on the CRB-PALS-PATJ cell polarity complex in the testis.

The PAR3-PAR6-aPKC cell polarity complex

PAR3 and PAR6 are scaffolding proteins that recruit different proteins to the TJ via their PDZ domains (Figure 5) (240, 241). They are required for the establishment and maintenance of cell polarity. PAR also participates in directional cell movement by regulating actin and microtubule cytoskeletons (242244; for reviews, see Refs. 180, 245). Both PAR3A and PAR3B partially colocalize with ZO-1 (246250), and they are also present at TJs that lack highly developed fibrils (251), suggesting they also function outside of the TJ. PAR3 is recruited to the plasma membrane by binding junctional adhesion molecule-A (JAM-A), which is present at newly assembled TJs before PAR3 (252255). This is followed by the drafting of PAR6 and aPKC (256). At the apical domain, the PAR complex facilitates the apical localization of the CRB complex and the basolateral localization of the SCRIB complex, thereby promoting cell polarity. Specifically, PAR3 interacts with CRB via PAR6 and aPKC. PAR3 also interacts with 14–3-3 (also known as PAR5) (257; for a review, see Ref. 258), but this is largely dependent on the phosphorylation state of PAR3, which is regulated by aPKC (259, 260). Interestingly, there is a delay in the delivery of proteins (ie, ezrin) to the apical domain of MDCK cells after Par3 knockdown (261), illustrating PAR3 is important for apical domain development, TJ maturation, and cytoskeletal remodeling. Likewise, Par3 knockdown in Sertoli cells disrupts the delivery of JAM-A and ZO-1 to sites of cell-cell contact after calcium switch (201). However, the significance of these findings is not clear because JAM-A is recruited to the plasma membrane independent of PAR3. Furthermore, only 1 out of 3 PAR isoforms was silenced by approximately 50%, indicating Sertoli cells still expressed Par3 after knockdown. Furthermore, the splice variant of Par3 (Par3c, it lacks residues 827–856 within the aPKC binding domain) can functionally substitute for the loss of canonical Par3 during the morphogenesis of dendritic spines (262). Thus, Par3c likely compensates for the loss of Par3 expression in Sertoli cells. Presently, additional studies are needed to determine the role of the retromer complex in PAR-mediated protein trafficking.

On the other hand, PAR6 is encoded by 3 different genes, Par6A/C, Par6B, and Par6D/G. Besides binding PAR3 and aPKC (263266), PAR6 also interacts with active CDC42 and ras-related C3 botulinum toxin (RAC) (263, 267269). This interaction allows CDC42 to activate PAR6, PAR3, and aPKC, thereby assembling a CDC42-PAR3-aPKC-PAR6 module. This protein module then associates with T-cell lymphoma invasion and metastasis 1 (a RAC-specific guanine nucleotide exchange factor that is highly expressed by the brain and testis) (270), resulting in RAC1 activation, cytoskeletal remodeling (ie, actin polymerization and microtubule stabilization) (243, 271), lamellipodia formation, and cell migration (272). In addition, PAR6C regulates cell polarity by functioning downstream of TGFβ in NMuMG cells (273). Specifically, TGFβ receptor II phosphorylates PAR6C, resulting in the recruitment of Smad ubiquitination-related factor 1 (an E3 ubiquitin ligase) (for a review, see Ref. 274), degradation of ras homolog family member A, disruption of TJs, and induction of the epithelial-mesenchymal transition (273, 275, 276). In the literature, few studies address PAR6 function. Par6 knockdown affects the morphogenesis of actin-rich dendritic spines (262) and the development of blastocysts (277). In another study, PAR6 interacts with MAPK organizer 1 (278), a scaffolding protein that mediates the binding of the PAR6-aPKC complex to CRB3 (278). Presently, it is not known whether these protein-protein interactions exist in the testis. In the testis, PAR6 interacts with fascin1, an actin cross-linking protein (for reviews, see Refs. 279, 280). However, immunoprecipitated PAR6 electrophoreses at a higher molecular weight than nonimmunoprecipitated PAR6, indicating additional biochemical experiments are needed to substantiate these findings. Furthermore, PAR6 does not colocalize with fascin1 (281).

The third component of this cell polarity complex is aPKC, a Ser/Thr kinase that is crucial for TJ assembly and maintenance (Figure 5). aPKCs are different from conventional PKCs, because they possess an N-terminal Phox and Bem1 domain, which allows them to interact with PAR6 (249; for a review, see Ref. 282). During junction assembly, aPKC forms a complex with PAR6 and LGL, followed by the phosphorylation and dissociation of LGL from the aPKC-PAR6 complex (283) and by the association of this complex with PAR3. It also allows aPKC to phosphorylate PAR3 at Ser827 (284, 285) and JAM-A at Ser285 (286), thereby facilitating TJ maturation. aPKC also regulates microtubule dynamics and vesicle trafficking (287289).

The SCRIB-DLG-LGL cell polarity complex

The third cell polarity complex, SCRIB-DLG-LGL localizes to the basolateral domain of epithelial cells (for reviews, see Refs. 183, 290292). SCRIB is a large multidomain protein that functions in cell polarity, migration, cell cycle progression, apoptosis, and protein sorting and trafficking (293299). For example, SCRIB regulates the passage of CRB through the retromer complex via aPKC-dependent and -independent mechanisms, even though the 2 cell polarity complexes antagonize each other (300). SCRIB also functions as a tumor suppressor and as a target for viral oncoproteins (301304; for reviews, see Refs. 194, 305). Although Scrib knockdown disrupts tight and adherens junction function in different cells (295, 306), it does not affect TJ function in Sertoli cells (307). However, the concurrent knockdown of Scrib, Dlg, and Lgl decreases its permeability, revealing the increase in barrier function is due to the loss of Dlg and/or Lgl (307). Furthermore, concurrent knockdown of Scrib, Dlg, and Lgl affects the orientation of step 19 spermatids within the seminiferous epithelium, but it is not known why a tighter blood-testis barrier would adversely affect spermatid orientation at the luminal edge. Nevertheless, Scrib knockout mice are embryonically lethal due to a defect in neural tube closure (308, 309), illustrating SCRIB is essential for normal morphogenesis. Besides binding and partially colocalizing with ZO-2 (310), SCRIB also binds pleckstrin homology domain leucine-rich repeat protein phosphatase (PHLPP) (311, 312), a membrane-associated Ser/Thr protein phosphatase that inactivates PKB and PKC (for reviews, see Refs. 313, 314). After Scrib knockdown, PHLPP1 transits to the cytosol, resulting in an increase in PKB phosphorylation and cell proliferation. SCRIB is unable to bind PKB in the absence of PHLPP1, and PHLPP1 is unable to bind PKB in the absence of SCRIB, indicating these 3 proteins form a heterotrimeric complex (311). Furthermore, SCRIB colocalizes with E-cadherin (315) and stabilizes E-cadherin-p120 catenin interactions (316), which maintain cell adhesion. Taken collectively, these results illustrate that SCRIB is an important regulator of tight and adherens junctions.

DLG and LGL are the other key proteins of this cell polarity complex. DLG, a member of the MAGUK family, functions in cell polarity, vesicle trafficking, cell junction dynamics, and tumor suppression (for reviews, see Refs. 290, 317, 318). Four DLGs have been identified in mammals, with DLG1 being the best studied member of this protein family. DLG1 also localizes to the adherens junction, where it binds E-cadherin and links it to PI3K (319, 320). A disruption of E-cadherin-PI3K interactions results in the mislocalization of E-cadherin, indicating DLG1 stabilizes E-cadherin. On the other hand, 2 LGLs have been identified in mammals (for reviews, see Refs. 321, 322). Interestingly, LGL phosphorylation by aPKC is critical for its function because nonphosphorylated LGL associates with the apical domain in MDCK cells (323325). Although Dlg1 and Lgl2 are expressed by the testis (307), their role in blood-testis barrier dynamics requires further study.

IV. Mechanisms of Blood-Testis Barrier Restructuring

The blood-testis barrier is a dynamic ultrastructure that restructures during the movement of preleptotene/leptotene spermatocytes at stages IX–XI of the seminiferous epithelial cycle. Sertoli cell junction disassembly is mediated by several mechanisms that generally involve changes in the levels, localizations, and interactions of structural, scaffolding, and signaling proteins. Many of these proteins were discussed in the preceding sections. For example, the opposing actions of kinases and phosphatases rapidly regulate the phosphorylation state of cell junction proteins, thereby altering their localization and interaction with other proteins. In this section, we discuss 2 key mechanisms, phosphorylation and endocytosis, that regulate blood-testis barrier dynamics.

A. Phosphorylation

The SRC kinase family is comprised of at least 8 members in the human: SRC, YES, FYN, and FGR, which constitute the SRC A subfamily; and LYN, HCK, LCK, and BLK, which constitute the SRC B subfamily. SRC, YES, and FYN are expressed by several organs, whereas the others are primarily expressed by lymphoid and myeloid tissues (for reviews, see Refs. 326, 327). Most cells and organs express more than one member of the SRC kinase family. In addition to these SRC kinases, at least 2 other members exist. FRK is expressed by the brain, breast, colon, and bladder, whereas BRK is expressed by the colon, small intestine, and prostate. SRC, YES, and FYN are the best studied kinases in the mammalian testis (328332). In the next section, we summarize key results from studies on SRC.

In general, SRC kinases consist of an N-terminal 14-carbon myristoyl group attached to an SH4 domain, followed by a unique region, an SH3 domain, an SH2 domain, an SH2 kinase linker, a kinase domain (also known as the SH1 domain), and a C-terminal domain containing a negative regulatory Tyr residue (for a review, see Ref. 327). Myristoylation, a cotranslational/posttranslational modification that involves the second amino acid residue, mediates binding of the kinase to the plasma membrane, thereby facilitating weak protein-protein and protein-lipid interactions (for reviews, see Refs. 333, 334). However, myristoylation alone is not enough for plasma membrane localization. The interaction of kinases with the plasma membrane is enhanced by the palmitoylation of an N-terminal Cys residue, which promotes their association with lipid rafts, specialized regions of the plasma membrane that serve as signaling platforms (for a review, see Ref. 335). Palmitoylation is also responsible for targeting SRC and FYN to endosomes (336), indicating these kinases participate in endocytosis. On the other hand, SH2 and SH3 domains interact with proline-rich regions and phospho-Tyr motifs, respectively, to regulate protein-protein interactions.

SRC kinase activity is regulated by intra- and intermolecular interactions that involve kinases and phosphatases (337). All SRC kinase family members contain 2 highly conserved Tyr residues that are critical for activity. Tyr416 is present within the kinase domain, and Tyr527 (both in chicken SRC) is present within the negative regulatory domain. The phosphorylation of SRC at Tyr527 by C-terminal SRC kinase or C-terminal SRC kinase homologous kinase brings this amino acid residue closer to the SH2 domain, rendering SRC catalytically inactive and unable to bind substrates. This closed conformation is further stabilized by the interaction of the SH3 domain with the proline-rich regions that link the SH2 domain with the kinase domain and by the dephosphorylation of Tyr416. Kinase activity increases upon the dissociation of Tyr527 from the SH2 domain, which yields an open conformation. As a result, Tyr527 becomes accessible to several phosphatases, the kinase domain can bind and phosphorylate substrates, and the SH2 and SH3 domains can interact with downstream proteins. Furthermore, the dephosphorylation of SRC at Tyr527 triggers the autophosphorylation of Tyr416 to yield a highly activated enzyme.

SRC kinase family members localize to the cytosol and plasma membrane, where they function in cell adhesion, migration, proliferation, differentiation, and survival. They also localize to the nucleus (mostly in neoplastic cells), endosomes, lysosomes, secretory granules/phagosomes, and the Golgi apparatus (328, 338342; for reviews, see Refs. 343, 344). In the testis, SRC localizes to the blood-testis barrier, where it is postulated to function in its restructuring (for a review, see Ref. 345). Genetic models, which are available for most SRC kinase family members, provide important insights on kinase function. For example, Src-null mice die within the first few weeks of birth if weaned. However, they survive if not weaned and fed a soft-food diet (346), indicating SRC is dispensable for embryonic development. Male homozygotes display no abnormalities in the central nervous system, where Src expression is the highest, revealing other members of the SRC kinase family can functionally substitute for its loss in the brain and spinal cord. Although female homozygotes are infertile, male homozygotes display no defects in the reproductive system (347).

FAK, a nonreceptor protein tyrosine kinase, functions in cell adhesion and migration, cytoskeletal dynamics, and cell cycle progression (for reviews, see Refs. 348350). FAK is a substrate of SRC, and this interaction between kinases is perhaps one of the most important in cell biology. In general, FAK is present at focal contacts, actin-based cell junctions that connect cells to the extracellular matrix via integrins, transmembrane receptors that facilitate inside-out and outside-in signaling (for reviews, see Refs. 351, 352). Upon ligand binding and integrin clustering, FAK is recruited to the β subunit of integrin, where it undergoes autophosphorylation at Tyr397. This phosphorylated Tyr residue becomes a docking site for SRC, which phosphorylates additional Tyr residues (ie, Tyr576 and Tyr577, which are located in the catalytic domain of FAK; Tyr861 and Tyr925, which are located in the C-terminal domain), resulting in an increase in FAK activity and the activation of other focal adhesion proteins such as paxillin, talin, and vinculin. In this way, the activated FAK-SRC complex triggers numerous signal transduction cascades to bring about changes in cell function. Integrins such as α4β1 can also activate SRC independently of FAK (353).

In the mammalian testis, the function of the FAK-SRC complex in spermatogenesis is less defined because there are no bona fide focal contacts at the base of the Sertoli cell. Only “hemidesmosome-like attachments” associating loosely with intermediate filaments have been observed by electron microscopy (23; for a review, see Ref. 45). Instead, FAK localizes to the blood-testis barrier, where it binds and colocalizes with occludin and ZO-1 (328, 354, 355). Other studies indicate that FAK-Tyr397, which localizes to the apical ectoplasmic specialization, and FAK-Tyr407, which predominantly localizes to the blood-testis barrier, exhibit opposite effects on barrier function. In Sertoli cells, FAK-Tyr397Phe overexpression prompts junction disassembly, whereas FAK-Tyr407Phe overexpression promotes junction assembly (356). Thus, it is postulated that phosphorylated FAK acts as a molecular switch during the movement of preleptotene/leptotene spermatocytes across the blood-testis barrier (357). However, it is difficult to interpret these findings, because FAK-Tyr397 is not present at the blood-testis barrier (356, 358), and FAK-Tyr407 is not stage-specific at the blood-testis barrier (356). In addition, FAK is highest at the blood-testis barrier during stages III–VI (354), which precedes the movement of preleptotene/leptotene spermatocytes across the barrier, and its expression is higher in germ cells than in Sertoli cells (356). Therefore, additional studies are needed to substantiate these findings.

B. Endocytosis

Endocytosis is the internalization of cargo, usually an integral membrane protein, together with a short stretch of the plasma membrane and fluid via vesicles. The key vesicles involved in this process are early, late, and recycling endosomes. Early endosomes are highly efficient sorting organelles and the site of bifurcation (ie, protein recycling or degradation) (for reviews, see Refs. 359361). Late endosomes receive proteins, lipids, and possibly other molecules en route to lysosomes for degradation by hydrolytic enzymes, whereas recycling endosomes receive proteins and lipids en route to the plasma membrane. In the fast recycling pathway, proteins are retrieved directly from early endosomes and recycled back to the plasma membrane within approximately 2–3 minutes vs approximately 10 minutes in the slow recycling pathway, which involves recycling endosomes. In general, endocytosis occurs via dynamin dependent (ie, clathrin- and caveolin-mediated) and independent (ie, macropinocytosis and phagocytosis) pathways. Most cells, including Sertoli cells, internalize cargo by the clathrin-mediated pathway, which involves clathrin, a 3-legged protein that assembles into a curved lattice around the budding vesicle (also known as the clathrin-coated pit), as well as other clathrin-associated proteins. Clathrin-coated pits, which comprise approximately 2% of the surface area of the plasma membrane in most cells, internalize plasma membrane components, growth factors, cytokines, and hormones within minutes because clathrin-coated pits have a very short half-life (∼1 min). By contrast, caveolae are cave-like invaginations present on terminally differentiated cells such as adipocytes, fibroblasts, endothelial cells, and smooth muscle cells. They are detergent-resistant and rich in cholesterol and sphingolipids (for reviews, see Refs. 361363). Caveolae are defined by the presence of caveolin, a cholesterol-binding protein that oligomerizes to form caveolin-rich domains that bud from the plasma membrane (364). Thus, there are apparent differences in the involvement of proteins and lipids, and in the formation of vesicles between endocytic pathways.

The cytoskeleton is essential for clathrin- and caveolin-mediated endocytosis, and actin microfilaments and actin-associated proteins participate in endosome maturation, membrane scission, and cargo sorting (for reviews, see Refs. 365, 366). For example, clathrin-mediated endocytosis has been shown to involve cortactin, which upon phosphorylation by SRC promotes actin polymerization and remodeling, resulting in the recruitment of a 7-protein complex known as the actin-related protein 2/3 complex (367, 368; for reviews, see Refs. 369, 370). The actin-related protein 2/3 complex, when activated by N-WASP (371, 372), nucleates the formation of new actin microfilaments on preexisting actin microfilaments (373), illustrating actin polymerization is important for the internalization of cargo. On the other hand, caveolin-mediated endocytosis involves filamin (374, 375), an F-actin cross-linking protein that promotes orthogonal branching of microfilaments (for reviews, see Refs. 376, 377). Caveolin is also a substrate of SRC (378, 379), suggesting phosphorylation induces the assembly of caveolin-rich domains and the internalization of caveolae. Although the actin cytoskeleton is involved in early endocytic events, the microtubule cytoskeleton is involved in late endocytic events (for a review, see Ref. 380). Microtubules are highly dynamic ultrastructures that exhibit phases of growth at plus-ends and shrinkage at minus-ends in a phenomenon known as dynamic instability (381; for reviews, see Refs. 382384). They are essential for vesicle trafficking and intracellular organization (Figure 6) (for reviews, see Refs. 385388).

Endocytosis is known to regulate the gate and fence functions of TJs. For example, different cells internalize occludin and claudin, usually in response to an external stimulus, via clathrin- and caveolin-mediated pathways, resulting in a disruption of TJ function (389, 390; for reviews, see Refs. 391393). Specifically, caveolin-1 regulates Ras-related protein Rab-5A (RAB5), a small GTPase that localizes to TJs (394) and controls the localization of occludin (395). Claudin uses a distinct mechanism of internalization, with adjoining claudin molecules internalizing as an entity into one cell (396; for a review, see Ref. 393). Connexin 43 also employs this mechanism (397, 398; for reviews, see Refs. 399, 400). In Sertoli cells, testosterone promotes the recycling of endocytosed occludin back to the plasma membrane via the slow recycling pathway, which supports TJ function (401). Testosterone also increases the levels of clathrin and caveolin-1 (402), suggesting androgens facilitate endocytosis. It is postulated that testosterone facilitates transcytosis, the transport of cargo across a polarized cell (for a review, see Ref. 403), during the movement of preleptotene/leptotene spermatocytes across the blood-testis barrier. However, there is no in vitro model of germ cell movement across the Sertoli cell barrier and little evidence that TJ proteins are transcytosed from in front of migrating spermatocytes to behind these germ cells. By contrast, cytokines such as TGFβ target TJ proteins to late endosomes for degradation (401, 402). These events are partly mediated by kinases because Src knockdown affects the endocytosis of CAR and JAM-A by decelerating their kinetic profiles and decreasing their internalization levels (328). Furthermore, endocytosed proteins are trafficked to late endosomes for degradation, resulting in a disruption of TJ function. Src knockdown also affects the actin cytoskeleton, which can also affect protein internalization and trafficking (328). Thus, it is not known whether SRC directly functions in endocytosis. Nevertheless, clathrin knockdown disrupts the delivery of proteins to the basolateral, but not the apical, domain in MDCK cells (404). Similarly, the loss of caveolin-1 affects cell polarity and directional cell movement (405, 406), illustrating endocytosis also regulates the fence function of TJs.

Interestingly, the testis uses a unique mechanism of internalization defined as the “junction internalization hypothesis” to bring about the movement of preleptotene/leptotene spermatocytes across the blood-testis barrier. It states tubulobulbar complexes internalize intact junctions into one cell via the clathrin-mediated pathway (407, 408; for a review, see Ref. 409), similar to the way in which claudin and connexin are endocytosed in epithelial cells. Tubulobulbar complexes are testis-specific, actin-rich ultrastructures that consist of a long tube and a large bulb near the end of the tube. Tubulobulbar complexes are capped at the distal end by a clathrin-coated pit that eventually buds from the ultrastructure and enters the endocytic pathway. Tubulobulbar complexes between Sertoli cells are defined as basal tubulobulbar complexes, and they are present in areas of the plasma membrane that are occupied by ectoplasmic specializations. Tight and gap junctions are interspersed between basal tubulobulbar complexes (407, 410; for a review, see Ref. 409), indicating tubulobulbar complexes internalize these junctions. Indeed, claudin 11, connexin 43, and nectin-2, as well as with RAB5 and early endosome antigen 1, associate with basal tubulobulbar complexes (407, 411). Cortactin also associates with the basal tubulobulbar complex (324). It is postulated that basal tubulobulbar complexes reduce junction size and number at stages IV and V to prepare for the movement of preleptotene/leptotene spermatocytes across the blood-testis barrier (407). Presently, it is not known whether Sertoli cells internalize proteins via other mechanisms.

V. Future Directions in the Study of the Blood-Testis Barrier and Concluding Remarks on the Status of Male Contraceptive Research

We have reviewed here the biology and regulation of the mammalian blood-tests barrier and discussed studies that may trigger new questions, foster new ideas, and drive the field forward. Throughout the review, we have emphasized the uniqueness of the blood-testis barrier, providing examples of how it is similar and dissimilar from other blood-tissue barriers. Lastly, we have highlighted specific research areas that require additional studies. For example, it is not known whether basal tubulobulbar complexes internalize the proteins of the tricellular TJ. Although pioneering observations on the blood-testis barrier were made more than 5 decades ago, the most important questions we ask today are: why study the blood-testis barrier and why is it important? An intact blood-testis barrier is critical for spermatogenesis, and subfertility or infertility can ensue if its function is perturbed. Fertility can also be affected if Sertoli-germ cell interactions are disrupted because undeveloped germ cells cannot fertilize an egg. Many investigators, including us, are purposefully affecting cell-cell interactions in the testis to determine whether this approach can result in the development of a safe and reversible nonhormonal male contraceptive. Thus, it is important to understand the biology of the mammalian blood-testis barrier so that this information can be applied to male reproduction and contraceptive development. We must also be assured that nonhormonal male contraceptives will be desired by men in the long term.

A recent study conducted in Pakistan by our organization, with the support of the World Bank through the Bank-Netherlands Partnership Program, shows a concrete change in the attitude of men toward family planning. This study reveals that men are enthusiastic in receiving more information on contraception and in playing a greater role in family planning. This gradual change in attitude is likely due to the enormous financial difficulties that men face in raising large families in Pakistan. However, a lack of affordable contraceptive methods and family planning services impede men from playing a greater role. In light of these findings, there is a great need to introduce male contraceptives that are safe, effective, reversible, and affordable. However, no compound has met the strict guidelines of the United States Food and Drug Administration and reached the consumer market. Healthy individuals would use male contraceptives for extended lengths of time. Thus, there can be no/few short- and long-term side effects caused by their use, no/minimal adverse interactions with other drugs that treat serious and life-threatening diseases, and absolutely no deaths. For example, liver enzymes such as cytochrome P-450 eliminate from the body most cholesterol-lowering drugs (ie, statins), which are taken by many men in their mid/late forties and early fifties. Any drug that blocks the action of these liver enzymes will increase the level of the statin in the blood, which may cause severe skeletal muscle damage and other serious conditions. Other contraindications may exist with male contraceptive use as well, and additional studies are needed once the candidate compound shows promise in clinical trials.

In addition, there are other issues that need to be addressed, because they will partly determine marketability, profit, and overall patient satisfaction. For example, will men from different ethnic backgrounds respond equally to a given male contraceptive? How will contraceptive efficacy (ie, the period of complete to reversible infertility) be established in different individuals without having to visit a urologist or clinic, especially for nonhormonal agents? How will long-term infertility affect Sertoli cell and sperm function should a man decide that he wants to father a child/children? Would reversibility be immediate and complete? Finally, will successful antifertility tests in animals translate to successful antifertility tests in humans? The United States Food and Drug Administration reports that approximately 92% of drugs approved for testing in humans never receive approval for human use. Of those that are approved, more than half are later recalled due to serious or lethal effects in humans. For example, the rheumatoid arthritis drug, rofecoxib (Vioxx; Merck), was safe in animal studies and approved in 1999 for human use, but it was eventually withdrawn from the consumer market in 2004, because it doubled the risk of myocardial infarction and stroke, resulting in the death of more than 60 000 individuals in the United States alone. Thus, any male contraceptive that is approved for human use will require stringent and careful testing.

Abbreviations

     
  • aPKC

    atypical protein kinase C

  •  
  • CAR

    coxsackievirus and adenovirus receptor

  •  
  • CDC42

    cell division cycle 42

  •  
  • CRB

    Crumbs

  •  
  • DLG

    discs-large

  •  
  • ES

    ectoplasmic specialization

  •  
  • FAK

    focal adhesion kinase

  •  
  • JAM-A

    junctional adhesion molecule-A

  •  
  • LGL

    lethal giant larvae

  •  
  • LIN-7

    lin-7 homolog A

  •  
  • LSR

    lipolysis-stimulated lipoprotein receptor

  •  
  • MAGUK

    membrane-associated guanylate kinase

  •  
  • MARVEL

    MAL and related proteins for vesicle trafficking and membrane link

  •  
  • N-WASP

    neuronal Wiskott-Aldrich syndrome protein

  •  
  • PALS

    protein associated with Lin-7

  •  
  • PAR

    partitioning defective

  •  
  • PATJ

    Pals-associated TJ protein

  •  
  • PDZ

    postsynaptic density 95, discs large, and ZO-1 proteins

  •  
  • PHLPP

    pleckstrin homology domain leucine-rich repeat protein phosphatase

  •  
  • PI3K

    phosphoinositide-3-kinase

  •  
  • PKB

    protein kinase B

  •  
  • PKC

    protein kinase C

  •  
  • RAB5

    Ras-related protein Rab-5A

  •  
  • RAC

    ras-related C3 botulinum toxin

  •  
  • RHO

    ras homolog

  •  
  • SCRIB

    Scribble

  •  
  • SH

    Src homology

  •  
  • TJ

    tight junction

  •  
  • TJP

    tight junction protein

  •  
  • ZO

    zona occludens

  •  
  • ZONAB

    ZO-1-associated nucleic acid binding protein.

Acknowledgments

This work was supported by grants from the National Institutes of Health (NICHD; 2R01 HD056034 and U54 HD029990 Project 5).

Disclosure Summary: The authors have nothing to disclose.

Reference

1

Clermont
Y
.
Kinetics of spermatogenesis in mammals: seminiferous epithelium cycle and spermatogonial renewal
.
Physiol Rev
.
1972
;
52
:
198
236
.

2

Hess
RA
,
Renato de Franca
L
.
Spermatogenesis and cycle of the seminiferous epithelium
.
Adv Exp Med Biol
.
2008
;
636
:
1
15
.

3

O'Donnell
L
,
Meachem
SJ
,
Stanton
PG
,
McLachlan
RI
.
Endocrine regulation of spermatogenesis
. In:
Neill
JD
, ed.
Knobil and Neill's Physiology of Reproduction
.
New York, NY
:
Elsevier
,
2006
;
1017
1069
.

4

de Kretser
DM
,
Kerr
JB
.
The cytology of the testis
. In:
Knobil
E
,
Neill
JB
,
Ewing
LL
,
Greenwald
GS
,
Markert
CL
,
Pfaff
DW
, eds.
The Physiology of Reproduction
.
New York, NY
:
Raven Press
,
1988
;
837
932
.

5

Greenbaum
MP
,
Iwamori
T
,
Buchold
GM
,
Matzuk
MM
.
Germ cell intercellular bridges
.
Cold Spring Harb Perspect Biol
.
2011
;
3
:
a005850
.

6

Clermont
Y
.
Contractile elements in the limiting membrane of the seminiferous tubules of the rat
.
Exp Cell Res
.
1958
;
15
:
438
440
.

7

Niemi
M
,
Kormano
M
.
Contractility of the seminiferous tubules of the postnatal rat testis and its response to oxytocin
.
Am Med Exp Fenn
.
1965
;
13
:
40
42
.

8

Smith
LB
,
Walker
WH
.
The regulation of spermatogenesis by androgens
.
Semin Cell Dev Biol
.
2014
;
30
:
2
13
.

9

Wang
RS
,
Yeh
S
,
Tzeng
CR
,
Chang
C
.
Androgen receptor roles in spermatogenesis and fertility: lessons from testicular cell-specific androgen receptor knockout mice
.
Endocr Rev
.
2009
;
30
:
119
132
.

10

Cheng
J
,
Watkins
SC
,
Walker
WH
.
Testosterone activates mitogen-activated protein kinase via Src kinase and the epidermal growth factor receptor in Sertoli cells
.
Endocrinology
.
2007
;
148
:
2066
2074
.

11

Shupe
J
,
Cheng
J
,
Puri
P
,
Kostereva
N
,
Walker
WH
.
Regulation of Sertoli-germ cell adhesion and sperm release by FSH and nonclassical testosterone signaling
.
Mol Endocrinol
.
2011
;
25
:
238
252
.

12

Walker
WH
.
Non-classical actions of testosterone and spermatogenesis
.
Philos Trans R Soc Lond B Biol Sci
.
2010
;
365
:
1557
1569
.

13

O'Donnell
L
,
McLachlan
RI
,
Wreford
NG
,
de Kretser
DM
,
Robertson
DM
.
Testosterone withdrawal promotes stage-specific detachment of round spermatids from the rat seminiferous epithelium
.
Biol Reprod
.
1996
;
55
:
895
901
.

14

Wong
CH
,
Xia
W
,
Lee
NP
,
Mruk
DD
,
Lee
WM
,
Cheng
CY
.
Regulation of ectoplasmic specialization dynamics in the seminiferous epithelium by focal adhesion-associated proteins in testosterone-suppressed rat testes
.
Endocrinology
.
2005
;
146
:
1192
1204
.

15

LeBlond
CP
,
Clermont
Y
.
Definition of the stages of the cycle of the seminiferous epithelium in the rat
.
Ann NY Acad Sci
.
1952
;
55
:
548
573
.

16

Hess
RA
.
Quantitative and qualitative characteristics of the stages and transitions in the cycle of the rat seminiferous epithelium: light microscopic observations of perfusion-fixed and plastic-embedded testes
.
Biol Reprod
.
1990
;
43
:
525
542
.

17

Russell
LD
,
Ettlin
RA
,
Hikim
APS
,
Clegg
ED
, eds.
Histological and Histopathological Evaluation of the Testis
.
St. Louis, MO
:
Cache River Press
,
1990
.

18

Fijak
M
,
Meinhardt
A
.
The testis in immune privilege
.
Immunol Rev
.
2006
;
213
:
66
81
.

19

Hedger
MP
,
Hales
DB
.
Immunophysiology of the male reproductive tract
. In:
Neill
JD
, ed.
Knobil and Neill's Physiology of Reproduction
.
New York, NY
:
Elsevier
,
2006
;
1195
1286
.

20

O'Bryan
MK
,
Hedger
MP
.
Inflammatory networks in the control of spermatogenesis. Chronic inflammation in an immunologically privileged tissue?
In:
Cheng
CY
, ed.
Molecular Mechanisms in Spermatogenesis
.
Autin, TX
:
Landes Bioscience and Springer Science+Business Media
,
2008
;
92
114
.

21

Li
N
,
Wang
T
,
Han
D
.
Structural, cellular and molecular aspects of immune privilege in the testis
.
Front Immunol
.
2012
;
3
:
152
.

22

Setchell
BP
,
Voglmayr
JK
,
Waites
GM
.
A blood-testis barrier restricting passage from blood into rete testis fluid but not into lymph
.
J Physiol
.
1969
;
200
:
73
85
.

23

Russell
L
.
Movement of spermatocytes from the basal to the adluminal compartment of the rat testis
.
Am J Anat
.
1977
;
148
:
313
328
.

24

Smith
BE
,
Braun
RE
.
Germ cell migration across Sertoli cell tight junctions
.
Science
.
2012
;
338
:
798
802
.

25

Yazama
F
.
Continual maintenance of the blood-testis barrier during spermatogenesis: the intermediate compartment theory revisited
.
J Reprod Dev
.
2008
;
54
:
299
305
.

26

Dym
M
,
Fawcett
DW
.
The blood-testis barrier in the rat and the physiological compartmentation of the seminiferous epithelium
.
Biol Reprod
.
1970
;
3
:
308
326
.

27

Mruk
DD
,
Cheng
CY
.
Sertoli-Sertoli and Sertoli-germ cell interactions and their significance in germ cell movement in the seminiferous epithelium during spermatogenesis
.
Endocr Rev
.
2004
;
25
:
747
806
.

28

Setchell
BP
,
Waites
GMB
.
The blood-testis barrier
. In:
Hamilton
DW
,
Greep
RO
, eds.
The Handbook of Physiology
.
Baltimore, MD
:
Williams and Wilkens
,
1975
;
143
172
.

29

Setchell
BP
.
Blood-testis barrier, junctional and transport proteins and spermatogenesis
. In:
Cheng
CY
, ed.
Molecular Mechanisms in Spermatogenesis
.
Austin, TX
:
Landes Bioscience and Springer Science+Business Media
,
2008
;
212
233
.

30

Rubin
LL
,
Staddon
JM
.
The cell biology of the blood-brain barrier
.
Annu Rev Neurosci
.
1999
;
22
:
11
28
.

31

Goncalves
A
,
Ambrosio
AF
,
Fernandes
R
.
Regulation of claudins in blood-tissue barriers under physiological and pathological states
.
Tissue Barriers
.
2013
;
1
:
e24782
.

32

Weiss
N
,
Miller
F
,
Cazaubon
S
,
Couraud
PO
.
The blood-brain barrier in brain homeostasis and neurological disease
.
Biochim Biophys Acta
.
2009
;
149
:
6198
6206
.

33

Stamatovic
SM
,
Keep
RF
,
Andjelkovic
AV
.
Brain endothelial cell-cell junctions: how to “open” the blood-brain barrier
.
Curr Neuropharmacol
.
2008
;
6
:
179
192
.

34

Gilula
NB
,
Fawcett
DW
.
The Sertoli cell occluding junctions and gap junctions in mature and developing mammalian testis
.
Dev Biol
.
1976
;
59
:
142
168
.

35

Chung
NP
,
Cheng
CY
.
Is cadmium chloride-induced inter-Sertoli tight junction permeability barrier disruption a suitable in vitro model to study the events of junction disassembly during spermatogenesis in the rat testis?
Endocrinology
.
2001
;
142
:
1878
1888
.

36

Janecki
A
,
Jakubowiak
A
,
Steinberger
A
.
Regulation of transepithelial electrical resistance in two-compartment Sertoli cell cultures: in vitro model of the blood-testis barrier
.
Endocrinology
.
1991
;
129
:
1489
1496
.

37

Su
L
,
Kopera-Sobota
IA
,
Bilinska
B
,
Cheng
CY
,
Mruk
DD
.
Germ cells contribute to the function of the Sertoli cell barrier: an in vitro study
.
Spermatogenesis
.
2013
;
3
:
e26460
.

38

Vogl
AW
,
Vaid
KS
,
Guttman
JA
.
The Sertoli cell cytoskeleton
. In:
Cheng
CY
, ed.
Molecular Mechanisms in Spermatogenesis
.
Austin, TX
:
Landes Bioscience and Springer Science+Business Media
,
2008
;
186
211
.

39

Mruk
DD
,
Silvestrini
B
,
Cheng
CY
.
Anchoring junctions as drug targets: role in contraceptive development
.
Pharmacol Rev
.
2008
;
60
:
146
180
.

40

Mruk
DD
,
Cheng
CY
.
Tight junctions in the testis: new perspectives
.
Philos Trans R Soc Lond B Biol Sci
.
2010
;
365
:
1621
1635
.

41

Lie
PP
,
Cheng
CY
,
Mruk
DD
.
The biology of the desmosome-like junction: a versatile anchoring junction and signal transducer in the seminiferous epithelium
.
Int Rev Cell Mol Biol
.
2011
;
286
:
223
269
.

42

Pointis
G
,
Segretain
D
.
Role of connexin-based gap junction channels in testis
.
Trends Endocrinol Metab
.
2005
;
16
:
300
306
.

43

Pointis
G
,
Fiorini
C
,
Defamie
N
,
Segretain
D
.
Gap junctional communication in the male reproductive system
.
Biochim Biophys Acta
.
2005
;
1719
:
102
116
.

44

Pointis
G
,
Gilleron
J
,
Carette
D
,
Segretain
D
.
Physiological and physiopathological aspects of connexins and communicating gap junctions in spermatogenesis
.
Philos Trans R Soc Lond B Biol Sci
.
2010
;
365
:
1607
1620
.

45

Vogl
AW
,
Pfeiffer
DC
,
Redenbach
DM
,
Grove
BD
.
Sertoli cell cytoskeleton
. In:
Russell
LD
,
Griswold
MD
, eds.
The Sertoli Cell
.
Clearwater, FL
:
Cache River Press
,
1993
;
39
86
.

46

Russell
LD
,
Peterson
RN
.
Sertoli cell junctions: morphological and functional correlates
.
Int Rev Cytol
.
1985
;
94
:
177
211
.

47

Rodgers
LS
,
Fanning
AS
.
Regulation of epithelial permeability by the actin cytoskeleton
.
Cytoskeleton
.
2011
;
68
:
653
660
.

48

Amlani
S
,
Vogl
AW
.
Changes in the distribution of microtubules and intermediate filaments in mammalian Sertoli cells during spermatogenesis
.
Anat Rec
.
1988
;
220
:
143
160
.

49

Delva
E
,
Tucker
DK
,
Kowalczyk
AP
.
The desmosome
.
Cold Spring Harb Perspect Biol
.
2009
;
1
:
a002543
.

50

Akhshi
TK
,
Wernike
D
,
Piekny
A
.
Microtubules and actin crosstalk in cell migration and division
.
Cytoskeleton (Hoboken)
.
2014
;
71
:
1
23
.

51

Kaverina
I
,
Straube
A
.
Regulation of cell migration by dynamic microtubules
.
Semin Cell Dev Biol
.
2011
;
22
:
968
974
.

52

Chang
L
,
Goldman
RD
.
Intermediate filaments mediate cytoskeletal crosstalk
.
Nat Rev Mol Cell Biol
.
2004
;
5
:
601
613
.

53

Etienne-Manneville
S
.
Actin and microtubules in cell motility: which one is in control?
Traffic
.
2004
;
5
:
470
477
.

54

Lai
CH
,
Kuo
KH
,
Leo
JM
.
Critical role of actin in modulating BBB permeability
.
Brain Res Brain Res Rev
.
2005
;
50
:
7
13
.

55

Huber
F
,
Boire
A
,
López
MP
,
Koenderink
GH
.
Cytoskeletal crosstalk: when three different personalities team up
.
Curr Opin Cell Biol
.
2015
;
32
:
39
47
.

56

Collins
C
,
Nelson
WJ
.
Running with neighbors: coordinating cell migration and cell-cell adhesion
.
Curr Opin Cell Biol
.
2015
;
17
:
62
70
.

57

Padmanabhan
A
,
Rao
MV
,
Wu
Y
,
Zaidel-Bar
R
.
Jack of all trades: functional modularity in the adherens junction
.
Curr Opin Cell Biol
.
2015
;
36
:
32
40
.

58

Jiang
X
,
Ma
TY
,
Zhang
Y
, et al. .
Specific deletion of Cdh2 in Sertoli cells leads to altered meiotic progression and subfertility of mice
.
Biol Reprod
.
2015
;
92
:
79
.

59

Wettemann
RP
,
Bazer
FW
.
Influence of environmental temperature on prolificacy of pigs
.
J Reprod Fertil Suppl
.
1985
;
33
:
199
208
.

60

Paul
C
,
Melton
DW
,
Saunders
PT
.
Do heat stress and deficits in DNA repair pathways have a negative impact on male fertility?
Mol Hum Reprod
.
2008
;
14
:
1
8
.

61

Liu
YX
.
Temperature control of spermatogenesis and prospect of male contraception
.
Front Biosci (Schol Ed)
.
2010
;
2
:
730
755
.

62

Cai
H
,
Ren
Y
,
Li
XX
, et al. .
Scrotal heat stress causes a transient alteration in tight junctions and induction of TGF-β expression
.
Int J Androl
.
2011
;
34
:
352
362
.

63

Cereijido
M
,
Anderson
J
.
Tight Junctions
.
New York, NY
:
CRC Press
;
2001
.

64

Ikenouchi
J
,
Furuse
M
,
Furuse
K
,
Sasaki
H
,
Tsukita
S
,
Tsukita
S
.
Tricellulin constitutes a novel barrier at tricellular contacts of epithelial cells
.
J Cell Biol
.
2005
;
171
:
939
945
.

65

Furuse
M
,
Hirase
T
,
Itoh
M
, et al. .
Occludin: a novel integral membrane protein localizing at tight junctions
.
J Cell Biol
.
1993
;
123
:
1777
1788
.

66

Feldman
GJ
,
Mullin
JM
,
Ryan
MP
.
Occludin: structure, function and regulation
.
Adv Drug Deliv Rev
.
2005
;
57
:
883
917
.

67

Morrow
CM
,
Mruk
D
,
Cheng
CY
,
Hess
RA
.
Claudin and occludin expression and function in the seminiferous epithelium
.
Philos Trans R Soc Lond B Biol Sci
.
2010
;
365
:
1679
1696
.

68

Moroi
S
,
Saitou
M
,
Fujimoto
K
, et al. .
Occludin is concentrated at tight junctions of mouse/rat but not human/guinea pig Sertoli cell in testes
.
Am J Physiol
.
1998
;
274
:
C1708
C1717
.

69

Van Itallie
CM
,
Anderson
JM
.
Architecture of tight junctions and principles of molecular composition
.
Semin Cell Dev Biol
.
2014
;
36
:
157
165
.

70

González -Mariscal
L
,
Betanzos
A
,
Nava
P
,
Jaramillo
BE
.
Tight junction proteins
.
Prog Biophys Mol Biol
.
2003
;
81
:
1
44
.

71

Schneeberger
EE
,
Lynch
RD
.
The tight junction: a multifunctional complex
.
Am J Physiol Cell Physiol
.
2004
;
286
:
C1213
C1228
.

72

Mankertz
J
,
Waller
JS
,
Hillenbrand
B
, et al. .
Gene expression of the tight junction protein occludin includes differential splicing and alternative promoter usage
.
Biochem Biophys Res Commun
.
2002
;
298
:
657
666
.

73

Balda
MS
,
Whitney
JA
,
Flores
C
,
González
S
,
Cereijido
M
,
Matter
K
.
Functional dissociation of paracellular permeability and transepithelial electrical resistance and disruption of the apical-basolateral intramembrane diffusion barrier by expression of a mutant tight junction membrane protein
.
J Cell Biol
.
1996
;
134
:
1031
1049
.

74

Saitou
M
,
Fujimoto
K
,
Doi
Y
, et al. .
Occludin-deficient embryonic stem cells can differentiate into polarized epithelial cells bearing tight junctions
.
J Cell Biol
.
1998
;
141
:
397
408
.

75

Itoh
M
,
Furuse
M
,
Morita
K
,
Kubota
K
,
Saitou
M
,
Tsukita
S
.
Direct binding of three tight junction-associated MAGUKs, ZO-1, ZO-2, and ZO-3, with the COOH termini of claudins
.
J Cell Biol
.
1999
;
147
:
1351
1363
.

76

Yu
AS
,
McCarthy
KM
,
Francis
SA
, et al. .
Knockdown of occludin expression leads to diverse phenotypic alterations in epithelial cells
.
Am J Physiol Cell Physiol
.
2005
;
288
:
C1231
C1241
.

77

Sit
ST
,
Manser
E
.
Rho GTPases and their role in organizing the actin cytoskeleton
.
J Cell Sci
.
2011
;
124
:
679
683
.

78

Du
D
,
Xu
F
,
Yu
L
, et al. .
The tight junction protein, occludin, regulates the directional migration of epithelial cells
.
Dev Cell
.
2010
;
18
:
52
63
.

79

Osanai
M
,
Murata
M
,
Nishikiori
N
,
Chiba
H
,
Kojima
T
,
Sawada
N
.
Occludin-mediated premature senescence is a fail-safe mechanism against tumorigenesis in breast carcinoma cells
.
Cancer Sci
.
2007
;
98
:
1027
1034
.

80

Saitou
M
,
Furuse
M
,
Sasaki
H
, et al. .
Complex phenotype of mice lacking occludin, a component of tight junction strands
.
Mol Biol Cell
.
2000
;
11
:
4131
4142
.

81

Lacaz-Vieira
F
,
Jaeger
MM
,
Farshori
P
,
Kachar
B
.
Small synthetic peptides homologous to segments of the first external loop of occludin impair tight junction resealing
.
J Membr Biol
.
1999
;
168
:
289
297
.

82

Wong
V
,
Gumbiner
BM
.
A synthetic peptide corresponding to the extracellular domain of occludin perturbs the tight junction permeability barrier
.
J Cell Biol
.
1997
;
136
:
399
409
.

83

Chung
NP
,
Mruk
D
,
Mo
MY
,
Lee
WM
,
Cheng
CY
.
A 22-amino acid synthetic peptide corresponding to the second extracellular loop of rat occludin perturbs the blood-testis barrier and disrupts spermatogenesis reversibly in vivo
.
Biol Reprod
.
2001
;
65
:
1340
1351
.

84

Medina
R
,
Rahner
C
,
Mitic
LL
,
Anderson
JM
,
Van Itallie
CM
.
Occludin localization at the tight junction requires the second extracellular loop
.
J Membr Biol
.
2000
;
178
:
235
247
.

85

Sakakibara
A
,
Furuse
M
,
Saitou
M
,
Ando-Akatsuka
Y
,
Tsukita
S
.
Possible involvement of phosphorylation of occludin in tight junction formation
.
J Cell Biol
.
1997
;
137
:
1393
1401
.

86

Lapointe
TK
,
Buret
AG
.
Interleukin-18 facilitates neutrophil transmigration via myosin light chain kinase-dependent disruption of occludin, without altering epithelial permeability
.
Am J Physiol Gastrointest Liver Physiol
.
2012
;
302
:
G343
G351
.

87

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

88

Baumgartner
HK
,
Beeman
N
,
Hodges
RS
,
Neville
MC
.
A D-peptide analog of the second extracellular loop of claudin-3 and -4 leads to mislocalized claudin and cellular apoptosis in mammary epithelial cells
.
Chem Biol Drug Des
.
2011
;
77
:
124
136
.

89

Beeman
N
,
Webb
PG
,
Baumgartner
HK
.
Occludin is required for apoptosis when claudin-claudin interactions are disrupted
.
Cell Death Dis
.
2012
;
3
:
e273
.

90

Kubota
K
,
Furuse
M
,
Sasaki
H
, et al. .
Ca(2+)-independent cell adhesion activity of claudins, a family of integral membrane proteins localized at tight junctions
.
Curr Biol
.
1999
;
9
:
1035
1038
.

91

Evans
MJ
,
von Hahn
T
,
Tscherne
DM
, et al. .
Claudin-1 is a hepatitis C virus co-receptor required for a late step in entry
.
Nature
.
2007
;
446
:
801
805
.

92

Meertens
L
,
Bertaux
C
,
Cukierman
L
, et al. .
The tight junction proteins claudin-1, -6, and -9 are entry cofactors for hepatitis C virus
.
J Virol
.
2008
;
82
:
3555
3560
.

93

Katahira
J
,
Sugiyama
H
,
Inoue
N
,
Horiguchi
Y
,
Matsuda
M
,
Sugimoto
N
.
Clostridium perfringens enterotoxin utilizes two structurally related membrane proteins as functional receptors in vivo
.
J Biol Chem
.
1997
;
272
:
26652
26658
.

94

Fujita
K
,
Katahira
J
,
Horiguchi
Y
,
Sonoda
N
,
Furuse
M
,
Tsukita
S
.
Clostridium perfringens enterotoxin binds to the second extracellular loop of claudin-3, a tight junction integral membrane protein
.
FEBS Lett
.
2000
;
476
:
258
261
.

95

Sonoda
N
,
Furuse
M
,
Sasaki
H
, et al. .
Clostridium perfringens enterotoxin fragment removes specific claudins from tight junction strands: evidence for direct involvement of claudins in tight junction barrier
.
J Cell Biol
.
1999
;
147
:
195
204
.

96

Ding
L
,
Lu
Z
,
Foreman
O
, et al. .
Inflammation and disruption of the mucosal architecture in claudin-7-deficient mice
.
Gastroenterology
.
2012
;
305
315
.

97

Walsh
SV
,
Hopkins
AM
,
Nusrat
A
.
Modulation of tight junction structure and function by cytokines
.
Adv Drug Deliv Rev
.
2000
;
41
:
303
313
.

98

Capaldo
CT
,
Nusrat
A
.
Cytokine regulation of tight junctions
.
Biochim Biophys Acta
.
2009
;
1788
:
864
871
.

99

Xia
W
,
Mruk
DD
,
Lee
WM
,
Cheng
CY
.
Cytokines and junction restructuring during spermatogenesis–a lesson to learn from the testis
.
Cytokine Growth Factor Rev
.
2005
;
16
:
469
493
.

100

Escudero-Esparza
A
,
Jiang
WG
,
Martin
TA
.
Claudin-5 participates in the regulation of endothelial cell motility
.
Mol Cell Biochem
.
2012
;
362
:
71
85
.

101

Stamatovic
SM
,
Dimitrijevic
OB
,
Keep
RF
,
Andjelkovic
AV
.
Protein kinase Cα-RhoA cross-talk in CCL2-induced alterations in brain endothelial permeability
.
J Biol Chem
.
2006
;
281
:
8379
8388
.

102

Van Itallie
CM
,
Tietgens
AJ
,
LoGrande
K
,
Aponte
A
,
Gucek
M
,
Anderson
JM
.
Phosphorylation of claudin-2 on serine 208 promotes membrane retention and reduces trafficking to lysosomes
.
J Cell Sci
.
2012
;
125
:
4902
4912
.

103

Ikari
A
,
Matsumoto
S
,
Harada
H
, et al. .
Phosphorylation of paracellin-1 at Ser217 by protein kinase A is essential for localization in tight junctions
.
J Cell Sci
.
2006
;
119
:
1781
1789
.

104

Chihara
M
,
Otsuka
S
,
Ichii
O
,
Hashimoto
Y
,
Kon
Y
.
Molecular dynamics of the blood-testis barrier components during murine spermatogenesis
.
Mol Reprod Dev
.
2010
;
77
:
630
639
.

105

Meng
J
,
Holdcraft
RW
,
Shima
JE
,
Griswold
MD
,
Braun
RE
.
Androgens regulate the permeability of the blood-testis barrier
.
Proc Natl Acad Sci USA
.
2005
;
102
:
16696
16700
.

106

Wang
RS
,
Yeh
S
,
Chen
LM
, et al. .
Androgen receptor in Sertoli cell is essential for germ cell nursery and junctional complex formation in mouse testes
.
Endocrinology
.
2006
;
147
:
5624
5633
.

107

de Gendt
K
,
Swinnen
JV
,
Saunders
PT
,
Schoonjans
L
, et al. .
A Sertoli cell-selective knockout of the androgen receptor causes spermatogenic arrest in meiosis
.
Proc Natl Acad Sci USA
.
2004
;
101
:
6876
6881
.

108

Meng
J
,
Greenlee
AR
,
Taub
CJ
,
Braun
RE
.
Sertoli cell-specific deletion of the androgen receptor compromises testicular immune privilege in mice
.
Biol Reprod
.
2011
;
85
:
254
260
.

109

Hellani
A
,
Ji
J
,
Mauduit
C
,
Deschildre
C
,
Tabone
E
,
Benahmed
M
.
Developmental and hormonal regulation of the expression of oligodendrocyte-specific protein/claudin 11 in mouse testis
.
Endocrinology
.
2000
;
141
:
3012
3019
.

110

Lui
WY
,
Lee
WM
,
Cheng
CY
.
Transforming growth factor-β3 perturbs the inter-Sertoli tight junction permeability barrier in vitro possibly mediated via its effects on occludin, zonula occludens-1, and claudin-11
.
Endocrinology
.
2001
;
142
:
1865
1877
.

111

Kaitu'u-Lino
TJ
,
Sluka
P
,
Foo
CF
,
Stanton
PG
.
Claudin-11 expression and localisation is regulated by androgens in rat Sertoli cells in vitro
.
Reproduction
.
2007
;
133
:
1169
1179
.

112

Florin
A
,
Maire
M
,
Bozec
A
, et al. .
Androgens and postmeiotic germ cells regulate claudin-11 expression in rat Sertoli cells
.
Endocrinology
.
2005
;
146
:
1532
1540
.

113

Gye
MC
.
Changes in the expression of claudins and transepithelial electrical resistance of mouse Sertoli cells by Leydig cell coculture
.
Int J Androl
.
2003
;
26
:
271
278
.

114

Mazaud-Guittot
S
,
Meugnier
E
,
Pesenti
S
, et al. .
Claudin 11 deficiency in mice results in loss of the Sertoli cell epithelial phenotype in the testis
.
Biol Reprod
.
2010
;
82
:
202
213
.

115

Gow
A
,
Southwood
CM
,
Li
JS
, et al. .
CNS myelin and Sertoli cell tight junction strands are absent in Osp/claudin-11 null mice
.
Cell
.
1999
;
99
:
649
659
.

116

Sridharan
S
,
Simon
L
,
Meling
DD
, et al. .
Proliferation of adult Sertoli cells following conditional knockout of the gap junctional protein GJA1 (connexin 43) in mice
.
Biol Reprod
.
2007
;
76
:
804
812
.

117

Li
MW
,
Mruk
DD
,
Lee
WM
,
Cheng
CY
.
Connexin 43 and plakophilin-2 as a protein complex that regulates blood-testis barrier dynamics
.
Proc Natl Acad Sci USA
.
2009
;
106
:
10213
10218
.

118

Li
MW
,
Mruk
DD
,
Lee
WM
,
Cheng
CY
.
Connexin 43 is critical to maintain the homeostasis of the blood-testis barrier via its effects on tight junction reassembly
.
Proc Natl Acad Sci USA
.
2010
;
107
:
17998
18003
.

119

Morrow
CM
,
Tyagi
G
,
Simon
L
, et al. .
Claudin 5 expression in mouse seminiferous epithelium is dependent upon the transcription factor ets variant 5 and contributes to blood-testis barrier function
.
Biol Reprod
.
2009
;
81
:
871
879
.

120

Ohkuni
T
,
Kojima
T
,
Ogasawara
N
, et al. .
Expression and localization of tricellulin in human nasal epithelial cells in vivo and in vitro
.
Med Mol Morphol
.
2009
;
42
:
204
211
.

121

Beese
M
,
Wyss
K
,
Haubitz
T
,
Kirsch
T
.
Effect of cAMP derivates on assembly and maintenance of tight junctions in human umbilical vein endothelial cells
.
BMC Cell Biol
.
2010
;
11
:
68
.

122

Mariano
C
,
Silva
SL
,
Pereira
P
,
Fernandes
A
,
Brites
D
,
Brito
MA
.
Evidence of tricellulin expression by immune cells, particularly microglia
.
Biochem Biophys Res Commun
.
2011
;
409
:
799
802
.

123

Raleigh
DR
,
Marchiando
AM
,
Zhang
Y
, et al. .
Tight junction-associated MARVEL proteins marvelD3, tricellulin, and occludin have distinct but overlapping functions
.
Mol Biol Cell
.
2010
;
21
:
1200
1213
.

124

Furuse
M
,
Izumi
Y
,
Oda
Y
,
Higashi
T
,
Iwamoto
N
.
Molecular organization of tricellular tight junctions
.
Tissue Barriers
.
2014
;
2
:
e28755
.

125

Riazuddin
S
,
Ahmed
ZM
,
Fanning
AS
, et al. .
Tricellulin is a tight-junction protein necessary for hearing
.
Am J Hum Genet
.
2006
;
79
:
1040
1051
.

126

Nayak
G
,
Lee
SI
,
Yousaf
R
, et al. .
Tricellulin deficiency affects tight junction architecture and cochlear hair cells
.
J Clin Invest
.
2013
;
123
:
4036
4049
.

127

Krug
SM
,
Amasheh
S
,
Richter
JF
, et al. .
Tricellulin forms a barrier to macromolecules in tricellular tight junctions without affecting ion permeability
.
Mol Biol Cell
.
2009
;
20
:
3713
3724
.

128

Oda
Y
,
Otani
T
,
Ikenouchi
J
,
Furuse
M
.
Tricellulin regulates junctional tension of epithelial cells at tricellular contacts via Cdc42
.
J Cell Sci
.
2014
;
127
:
4201
4212
.

129

Chishti
MS
,
Bhatti
A
,
Tamim
S
, et al. .
Splice-site mutations in the TRIC gene underlie autosomal recessive nonsyndromic hearing impairment in Pakistani families
.
J Hum Genet
.
2008
;
53
:
101
105
.

130

Steed
E
,
Rodrigues
NT
,
Balda
MS
,
Matter
K
.
Identification of MarvelD3 as a tight junction-associated transmembrane protein of the occludin family
.
BMC Cell Biol
.
2009
;
10
:
95
.

131

Sanchez-Pulido
L
,
Martín-Belmonte
F
,
Valencia
A
,
Alonso
MA
.
MARVEL: a conserved domain involved in membrane apposition events
.
Trends Biochem Sci
.
2002
;
27
:
599
601
.

132

Ikenouchi
J
,
Sasaki
H
,
Tsukita
S
,
Furuse
M
,
Tsukita
S
.
Loss of occludin affects tricellular localization of tricellulin
.
Mol Biol Cell
.
2008
;
19
:
4687
4693
.

133

Westphal
JK
,
Dörfel
MJ
,
Krug
SM
, et al. .
Tricellulin forms homomeric and heteromeric tight junctional complexes
.
Cell Mol Life Sci
.
2010
;
67
:
2057
2068
.

134

Yen
FT
,
Masson
M
,
Clossais-Besnard
N
, et al. .
Molecular cloning of a lipolysis-stimulated remnant receptor expressed in the liver
.
J Biol Chem
.
1999
;
274
:
13390
13398
.

135

Nakatsu
D
,
Kano
F
,
Taguchi
Y
, et al. .
JNK1/2-dependent phosphorylation of angulin-1/LSR is required for the exclusive localization of angulin-1/LSR and tricellulin at tricellular contacts in EpH4 epithelial sheet
.
Genes Cells
.
2014
;
19
:
565
581
.

136

Masuda
S
,
Oda
Y
,
Sasaki
H
, et al. .
LSR defines cell corners for tricellular tight junction formation in epithelial cells
.
J Cell Sci
.
2011
;
124
:
548
555
.

137

Mesli
S
,
Javorschi
S
,
Bérard
AM
, et al. .
Distribution of the lipolysis stimulated receptor in adult and embryonic murine tissues and lethality of LSR−/− embryos at 12.5 to 14.5 days of gestation
.
Eur J Biochem
.
2004
;
271
:
3103
3114
.

138

Weber
JE
,
Russell
LD
.
A study of intercellular bridges during spermatogenesis in the rat
.
Am J Anat
.
1987
;
180
:
1
24
.

139

Greenbaum
MP
,
Yan
W
,
Wu
MH
, et al. .
TEX14 is essential for intercellular bridges and fertility in male mice
.
Proc Natl Acad Sci USA
.
2006
;
103
:
4982
4987
.

140

Chen
JW
,
Zhou
B
,
Yu
QC
, et al. .
Cardiomyocyte-specific deletion of the coxsackievirus and adenovirus receptor results in hyperplasia of the embryonic left ventricle and abnormalities of sinuatrial valves
.
Circ Res
.
2006
;
98
:
923
930
.

141

Asher
DR
,
Cerny
AM
,
Weiler
SR
, et al. .
Coxsackievirus and adenovirus receptor is essential for cardiomyocyte development
.
Genesis
.
2005
;
42
:
77
85
.

142

Dorner
AA
,
Wegmann
F
,
Butz
S
, et al. .
Coxsackievirus-adenovirus receptor (CAR) is essential for early embryonic cardiac development
.
J Cell Sci
.
2005
;
118
:
3509
3521
.

143

Pazirandeh
A
,
Sultana
T
,
Mirza
MD
, et al. .
Multiple phenotypes in adult mice following inactivation of the coxsackievirus and adenovirus receptor (Car) gene
.
PLoS One
.
2011
;
6
:
e20203
.

144

Morton
PE
,
Hicks
A
,
Nastos
T
,
Santis
G
,
Parsons
M
.
CAR regulates epithelial cell junction stability through control of E-cadherin trafficking
.
Sci Rep
.
2013
;
3
:
2889
.

145

Raschperger
E
,
Thyberg
J
,
Pettersson
S
,
Philipson
L
,
Fuxe
J
,
Pettersson
RF
.
The coxsackie- and adenovirus receptor (CAR) is an in vivo marker for epithelial tight junctions, with a potential role in regulating permeability and tissue homeostasis
.
Exp Cell Res
.
2006
;
312
:
1566
1580
.

146

Cohen
CJ
,
Shieh
JT
,
Pickles
RJ
,
Okegawa
T
,
Hsieh
JT
,
Bergelson
JM
.
The coxsackievirus and adenovirus receptor is a transmembrane component of the tight junction
.
Proc Natl Acad Sci USA
.
2001
;
98
:
15191
15196
.

147

Dörner
A
,
Xiong
D
,
Couch
K
,
Yajima
T
,
Knowlton
KU
.
Alternatively spliced soluble coxsackie-adenovirus receptors inhibit coxsackievirus infection
.
J Biol Chem
.
2004
;
279
:
18497
18503
.

148

Houri
N
,
Huang
KC
,
Nalbantoglu
J
.
The coxsackievirus and adenovirus receptor (CAR) undergoes ectodomain shedding and regulated intramembrane proteolysis (RIP)
.
PLoS One
.
2013
;
8
:
e73296
.

149

Fok
PT
,
Huang
KC
,
Holland
PC
,
Nalbantoglu
J
.
The coxsackie and adenovirus receptor binds microtubules and plays a role in cell migration
.
J Biol Chem
.
2007
;
282
:
7512
7521
.

150

Mirza
M
,
Petersen
C
,
Nordqvist
K
,
Sollerbrant
K
.
Coxsackievirus and adenovirus receptor is up-regulated in migratory germ cells during passage of the blood-testis barrier
.
Endocrinology
.
2007
;
148
:
5459
5469
.

151

Mirza
M
,
Hreinsson
J
,
Strand
ML
, et al. .
Coxsackievirus and adenovirus receptor (CAR) is expressed in male germ cells and forms a complex with the differentiation factor JAM-C in mouse testis
.
Exp Cell Res
.
2006
;
312
:
817
830
.

152

Su
L
,
Mruk
DD
,
Cheng
CY
.
Regulation of the blood-testis barrier by coxsackievirus and adenovirus receptor
.
Am J Physiol Cell Physiol
.
2012
;
303
:
C843
C853
.

153

Zen
K
,
Liu
Y
,
McCall
IC
, et al. .
Neutrophil migration across tight junctions is mediated by adhesive interactions between epithelial coxsackie and adenovirus receptor and a junctional adhesion molecule-like protein on neutrophils
.
Mol Biol Cell
.
2005
;
16
:
2694
2703
.

154

Wang
CQ
,
Cheng
CY
.
A seamless trespass: germ cell migration across the seminiferous epithelium during spermatogenesis
.
J Cell Biol
.
2007
;
178
:
549
556
.

155

Sultana
T
,
Hou
M
,
Stukenborg
JB
, et al. .
Mice depleted of the coxsackievirus and adenovirus receptor display normal spermatogenesis and an intact blood-testis barrier
.
Reproduction
.
2014
;
147
:
875
883
.

156

Bazzoni
G
,
Tonetti
P
,
Manzi
L
,
Cera
MR
,
Balconi
G
,
Dejana
E
.
Expression of junctional adhesion molecule-A prevents spontaneous and random motility
.
J Cell Sci
.
2005
;
118
:
623
632
.

157

Vogl
AW
.
Spatially dynamic intercellular adhesion junction is coupled to a microtubule-based motility system: evidence from an in vitro binding assay
.
Cell Motil Cytoskeleton
.
1996
;
34
:
1
12
.

158

Beach
SF
,
Vogl
AW
.
Spermatid translocation in the rat seminiferous epithelium: coupling membrane trafficking machinery to a junction plaque
.
Biol Reprod
.
1999
;
60
:
1036
1046
.

159

Matter
K
,
Balda
MS
.
Epithelial tight junctions, gene expression and nucleo-junctional interplay
.
J Cell Sci
.
2007
;
120
:
1505
1511
.

160

González-Mariscal
L
,
Betanzos
A
,
Avila-Flores
A
.
MAGUK proteins: structure and role in the tight junction
.
Semin Cell Dev Biol
.
2000
;
11
:
315
324
.

161

Traweger
A
,
Toepfer
S
,
Wagner
RN
, et al. .
Beyond cell-cell adhesion: emerging roles of the tight junction scaffold ZO-2
.
Tissue Barriers
.
2013
;
1
:
e25039
.

162

Hervé
JC
,
Derangeon
M
,
Sarrouilhe
D
,
Bourmeyster
N
.
Influence of the scaffolding protein zonula occludens (ZOs) on membrane channels
.
Biochim Biophys Acta
.
2014
;
1838
:
595
604
.

163

Gonzalez-Mariscal
L
,
Tapia
R
,
Huerta
M
,
Lopez-Bayghen
E
.
The tight junction protein ZO-2 blocks cell cycle progression and inhibits cyclin D1 expression
.
Ann NY Acad Sci
.
2009
;
1165
:
121
125
.

164

Bauer
H
,
Zweimueller-Mayer
J
,
Steinbacher
P
,
Lametschwandtner
A
,
Bauer
HC
.
The dual role of zonula occludens (ZO) proteins
.
J Biomed Biotechnol
.
2010
;
2010
:
402593
.

165

Spadaro
D
,
Tapia
R
,
Pulimeno
P
,
Citi
S
.
The control of gene expression and cell proliferation by the epithelial apical junctional complex
.
Essays Biochem
.
2012
;
53
:
83
93
.

166

Balda
MS
,
Matter
K
.
The tight junction protein ZO-1 and an interacting transcription factor regulate ErbB-2 expression
.
EMBO J
.
2000
;
19
:
2024
2033
.

167

Aijaz
S
,
Sanchez-Heras
E
,
Balda
MS
,
Matter
K
.
Regulation of tight junction assembly and epithelial morphogenesis by the heat shock protein Apg-2
.
BMC Cell Biol
.
2007
;
8
:
49
.

168

Tsapara
A
,
Matter
K
,
Balda
MS
.
The heat-shock protein Apg-2 binds to the tight junction protein ZO-1 and regulates transcriptional activity of ZONAB
.
Mol Biol Cell
.
2006
;
17
:
1322
1330
.

169

Umeda
K
,
Matsui
T
,
Nakayama
M
, et al. .
Establishment and characterization of cultured epithelial cells lacking expression of ZO-1
.
J Biol Chem
.
2004
;
279
:
44785
44794
.

170

McNeil
E
,
Capaldo
CT
,
Macara
IG
.
Zonula occludens-1 function in the assembly of tight junctions in Madin-Darby canine kidney epithelial cells
.
Mol Biol Cell
.
2006
;
17
:
1922
1932
.

171

Segretain
D
,
Fiorini
C
,
Decrouy
X
,
Defamie
N
,
Prat
JR
,
Pointis
G
.
A proposed role for ZO-1 in targeting connexin 43 gap junctions to the endocytic pathway
.
Biochimie
.
2004
;
86
:
241
244
.

172

Dunn
CA
,
Lampe
PD
.
Injury-triggered Akt phosphorylation of Cx43: a ZO-1-driven molecular switch that regulates gap junction size
.
J Cell Sci
.
2014
;
127
:
455
464
.

173

Furuse
M
,
Itoh
M
,
Hirase
T
, et al. .
Direct association of occludin with ZO-1 and its possible involvement in the localization of occludin at tight junctions
.
J Cell Biol
.
1994
;
127
:
1617
1626
.

174

Katsuno
T
,
Umeda
K
,
Matsui
T
, et al. .
Deficiency of zonula occludens-1 causes embryonic lethal phenotype associated with defected yolk sac angiogenesis and apoptosis of embryonic cells
.
Mol Biol Cell
.
2008
;
19
:
2465
2475
.

175

Xu
J
,
Kausalya
PJ
,
Phua
DC
,
Ali
SM
,
Hossain
Z
,
Hunziker
W
.
Early embryonic lethality of mice lacking ZO-2, but not ZO-3, reveals critical and nonredundant roles for individual zonula occludens proteins in mammalian development
.
Mol Cell Biol
.
2008
;
28
:
1669
1678
.

176

Xu
J
,
Anuar
F
,
Ali
SM
, et al. .
Zona occludens-2 is critical for blood-testis barrier integrity and male fertility
.
Mol Biol Cell
.
2009
;
20
:
4268
4277
.

177

Umeda
K
,
Ikenouchi
J
,
Katahira-Tayama
S
, et al. .
ZO-1 and ZO-2 independently determine where claudins are polymerized in tight-junction strand formation
.
Cell
.
2006
;
126
:
741
754
.

178

Sato
T
,
Fujita
N
,
Yamada
A
, et al. .
Regulation of the assembly and adhesion activity of E-cadherin by nectin and afadin for the formation of adherens junctions in Madin-Darby canine kidney cells
.
J Biol Chem
.
2006
;
281
:
5288
5299
.

179

Byers
S
,
Graham
R
,
Dai
HN
,
Hoxter
B
.
Development of Sertoli cell junctional specializations and the distribution of the tight-junction-associated protein ZO-1 in the mouse testis
.
Am J Anat
.
1991
;
191
:
35
47
.

180

Iden
S
,
Collard
JG
.
Crosstalk between small GTPases and polarity proteins in cell polarization
.
Nat Rev Mol Cell Biol
.
2008
;
9
:
846
859
.

181

Cereijido
M
,
Valdés
J
,
Shoshani
L
,
Contreras
RG
.
Role of tight junctions in establishing and maintaining cell polarity
.
Annu Rev Physiol
.
1998
;
60
:
161
177
.

182

Yeaman
C
,
Grindstaff
KK
,
Hansen
MD
,
Nelson
WJ
.
Cell polarity: versatile scaffolds keep things in place
.
Curr Biol
.
1999
;
9
:
R515
R517
.

183

Assémat
E
,
Bazellières
E
,
Pallesi-Pocachard
E
,
Le Bivic
A
,
Massey-Harroche
D
.
Polarity complex proteins
.
Biochim Biophys Acta
.
2008
;
1778
:
614
630
.

184

Margolis
B
,
Borg
JP
.
Apicobasal polarity complexes
.
J Cell Sci
.
2005
;
118
:
5157
5159
.

185

Suzuki
A
,
Ohno
S
.
The PAR-aPKC system: lessons in polarity
.
J Cell Sci
.
2006
;
119
:
979
987
.

186

Shin
K
,
Fogg
VC
,
Margolis
B
.
Tight junctions and cell polarity
.
Annu Rev Cell Dev Biol
.
2006
;
22
:
207
235
.

187

Pieczynski
J
,
Margolis
B
.
Protein complexes that control renal epithelial polarity
.
Am J Physiol Renal Physiol
.
2011
;
300
:
F589
F601
.

188

St Johnston
D
,
Sanson
B
.
Epithelial polarity and morphogenesis
.
Curr Opin Cell Biol
.
2011
;
23
:
540
546
.

189

Tepass
U
,
Theres
C
,
Knust
E
.
Crumbs encodes an EGF-like protein expressed on apical membranes of Drosophila epithelial cells and required for organization of epithelia
.
Cell
.
1990
;
61
:
787
799
.

190

Pocha
SM
,
Knust
E
.
Complexities of Crumbs function and regulation in tissue morphogenesis
.
Curr Biol
.
2013
;
23
:
R289
R293
.

191

Tepass
U
.
The apical polarity protein network in Drosophila epithelial cells: regulation of polarity, junctions, morphogenesis, cell growth, and survival
.
Annu Rev Cell Dev Biol
.
2012
;
28
:
655
685
.

192

Ellenbroek
SI
,
Iden
S
,
Collard
JG
.
Cell polarity proteins and cancer
.
Semin Cancer Biol
.
2012
;
22
:
208
215
.

193

Martin-Belmonte
F
,
Perez-Moreno
M
.
Epithelial cell polarity, stem cells and cancer
.
Nat Rev Cancer
.
2012
;
12
:
23
38
.

194

Halaoui
R
,
McCaffrey
L
.
Rewiring cell polarity signaling in cancer
.
Oncogene
.
2015
;
34
:
939
950
.

195

Grzeschik
NA
,
Parsons
LM
,
Allott
ML
,
Harvey
KF
,
Richardson
HE
.
Lgl, aPKC, and Crumbs regulate the Salvador/Warts/Hippo pathway through two distinct mechanisms
.
Curr Biol
.
2010
;
20
:
573
581
.

196

Chen
CL
,
Gajewski
KM
,
Hamaratoglu
F
, et al. .
The apical-basal cell polarity determinant Crumbs regulates Hippo signaling in Drosophila
.
Proc Natl Acad Sci USA
.
2010
;
107
:
15810
15815
.

197

Hafezi
Y
,
Bosch
JA
,
Hariharan
IK
.
Differences in levels of the transmembrane protein Crumbs can influence cell survival at clonal boundaries
.
Dev Biol
.
2012
;
368
:
358
369
.

198

Ling
C
,
Zheng
Y
,
Yin
F
, et al. .
The apical transmembrane protein Crumbs functions as a tumor suppressor that regulates Hippo signaling by binding to Expanded
.
Proc Natl Acad Sci USA
.
2010
;
107
:
10532
10537
.

199

Richardson
EC
,
Pichaud
F
.
Crumbs is required to achieve proper organ size control during Drosophila head development
.
Development
.
2010
;
137
:
641
650
.

200

Fan
S
,
Fogg
V
,
Wang
Q
,
Chen
XW
,
Liu
CJ
,
Margolis
B
.
A novel Crumbs3 isoform regulates cell division and ciliogenesis via importin β interactions
.
J Cell Biol
.
2007
;
178
:
387
398
.

201

Wong
EW
,
Mruk
DD
,
Lee
WM
,
Cheng
CY
.
Par3/Par6 polarity complex coordinates apical ectoplasmic specialization and blood-testis barrier restructuring during spermatogenesis
.
Proc Natl Acad Sci USA
.
2008
;
105
:
9657
9662
.

202

den Hollander
AI
,
Ghiani
M
,
de Kok
YJ
, et al. .
Isolation of Crb1, a mouse homologue of Drosophila Crumbs, and analysis of its expression pattern in eye and brain
.
Mech Dev
.
2002
;
110
:
203
207
.

203

Frame
MC
,
Patel
H
,
Serrels
B
,
Lietha
D
,
Eck
MJ
.
The FERM domain: organizing the structure and function of FAK
.
Nat Rev Mol Cell Biol
.
2010
;
11
:
802
814
.

204

Chishti
AH
,
Kim
AC
,
Marfatia
SM
, et al. .
The FERM domain: a unique module involved in the linkage of cytoplasmic proteins to the membrane
.
Trends Biochem Sci
.
1998
;
23
:
281
282
.

205

Tepass
U
.
FERM proteins in animal morphogenesis
.
Curr Opin Gen Dev
.
2009
;
19
:
357
367
.

206

Wodarz
A
,
Hinz
U
,
Engelbert
M
,
Knust
E
.
Expression of crumbs confers apical character on plasma membrane domains of ectodermal epithelia of Drosophila
.
Cell
.
1995
;
82
:
67
76
.

207

Roh
MH
,
Fan
S
,
Liu
CJ
,
Margolis
B
.
The Crumbs3-Pals1 complex participates in the establishment of polarity in mammalian epithelial cells
.
J Cell Sci
.
2003
;
116
:
2895
2906
.

208

Fogg
VC
,
Liu
CJ
,
Margolis
B
.
Multiple regions of Crumbs3 are required for tight junction formation in MCF10A cells
.
J Cell Sci
.
2005
;
118
:
2859
2869
.

209

Whiteman
EL
,
Liu
CJ
,
Fearon
ER
,
Margolis
B
.
The transcription factor snail represses Crumbs3 expression and disrupts apico-basal polarity complexes
.
Oncogene
.
2008
;
27
:
3875
3879
.

210

Robinson
BS
,
Huang
J
,
Hong
Y
,
Moberg
KH
.
Crumbs regulates Salvador/Warts/Hippo signaling in Drosophila via the FERM-domain protein expanded
.
Curr Biol
.
2010
;
20
:
582
590
.

211

Parsons
LM
,
Grzeschik
NA
,
Allott
ML
,
Richardson
HE
.
Lgl/aPKC and Crb regulate the Salvador/Warts/Hippo pathway
.
Fly (Austin)
.
2010
;
4
:
288
293
.

212

Wang
MM
.
Notch signaling and Notch signaling modifiers
.
Int J Biochem Cell Biol
.
2011
;
43
:
1550
1562
.

213

Nichols
JT
,
Miyamoto
A
,
Weinmaster
G
.
Notch signaling–constantly on the move
.
Traffic
.
2007
;
8
:
959
969
.

214

Takebe
N
,
Harris
PJ
,
Warren
RQ
,
Ivy
SP
.
Targeting cancer stem cells by inhibiting Wnt, Notch, and Hedgehog pathways
.
Nat Rev Clin Oncol
.
2011
;
8
:
97
106
.

215

Zolkiewska
A
.
ADAM proteases: ligand processing and modulation of the Notch pathway
.
Cell Mol Life Sci
.
2008
;
65
:
2056
2068
.

216

Herranz
H
,
Stamataki
E
,
Feiguin
F
,
Milán
M
.
Self-refinement of Notch activity through the transmembrane protein Crumbs: modulation of γ-secretase activity
.
EMBO Rep
.
2006
;
7
:
297
302
.

217

Hayashi
T
,
Yamada
T
,
Kageyama
Y
,
Negishi
T
,
Kihara
K
.
Expression failure of the Notch signaling system is associated with the pathogenesis of maturation arrest in male infertility patients
.
Fertil Steril
.
2004
;
81
:
697
699
.

218

Hayashi
T
,
Kageyama
Y
,
Ishizaka
K
,
Xia
G
,
Kihara
K
,
Oshima
H
.
Requirement of Notch 1 and its ligand jagged 2 expressions for spermatogenesis in rat and human testes
.
J Androl
.
2001
;
22
:
999
1011
.

219

Murta
D
,
Batista
M
,
Trindade
A
, et al. .
In vivo Notch signaling blockade induces abnormal spermatogenesis in the mouse
.
PLoS One
.
2014
;
9
:
e113365
.

220

Roh
MH
,
Makarova
O
,
Liu
CJ
, et al. .
The MAGUK protein, Pals1, functions as an adapter, linking mammalian homologues of Crumbs and Discs Lost
.
J Cell Biol
.
2002
;
157
:
161
172
.

221

Kamberov
E
,
Makarova
O
,
Roh
M
, et al. .
Molecular cloning and characterization of Pals, proteins associated with mLin-7
.
J Biol Chem
.
2000
;
275
:
11425
11431
.

222

Zhang
J
,
Yang
X
,
Wang
Z
, et al. .
Structure of an L27 domain heterotrimer from cell polarity complex Patj/Pals1/Mals2 reveals mutually independent L27 domain assembly mode
.
J Biol Chem
.
2012
;
287
:
11132
11140
.

223

Straight
SW
,
Shin
K
,
Fogg
VC
, et al. .
Loss of PALS1 expression leads to tight junction and polarity defects
.
Mol Biol Cell
.
2004
;
15
:
1981
1990
.

224

Wang
Q
,
Chen
XW
,
Margolis
B
.
PALS1 regulates E-cadherin trafficking in mammalian epithelial cells
.
Mol Biol Cell
.
2007
;
18
:
874
885
.

225

Shin
K
,
Straight
S
,
Margolis
B
.
PATJ regulates tight junction formation and polarity in mammalian epithelial cells
.
J Cell Biol
.
2005
;
168
:
705
711
.

226

Michel
D
,
Arsanto
JP
,
Massey-Harroche
D
, et al. .
PATJ connects and stabilizes apical and lateral components of tight junctions in human intestinal cells
.
J Cell Sci
.
2005
;
118
:
4049
4057
.

227

Seaman
MN
.
The retromer complex - endosomal protein recycling and beyond
.
J Cell Sci
.
2012
;
125
:
4693
4702
.

228

Pocha
SM
,
Wassmer
T
,
Niehage
C
,
Hoflack
B
,
Knust
E
.
Retromer controls epithelial cell polarity by trafficking the apical determinant Crumbs
.
Curr Biol
.
2011
;
21
:
1111
1117
.

229

Zhou
B
,
Wu
Y
,
Lin
X
.
Retromer regulates apical-basal polarity through recycling crumbs
.
Dev Biol
.
2011
;
360
:
87
95
.

230

Lemmers
C
,
Médina
E
,
Delgrossi
MH
,
Michel
D
,
Arsanto
JP
,
Le Bivic
A
.
hINADl/PATJ, a homolog of discs lost, interacts with crumbs and localizes to tight junctions in human epithelial cells
.
J Biol Chem
.
2002
;
277
:
25408
25415
.

231

Philipp
S
,
Flockerzi
V
.
Molecular characterization of a novel human PDZ domain protein with homology to INAD from Drosophila melanogaster
.
FEBS Lett
.
1997
;
413
:
243
248
.

232

Roh
MH
,
Liu
CJ
,
Laurinec
S
,
Margolis
B
.
The carboxyl terminus of zona occludens-3 binds and recruits a mammalian homolog of discs lost to tight junctions
.
J Biol Chem
.
2002
;
277
:
27501
27509
.

233

Mruk
DD
,
Cheng
CY
.
Delivering non-hormonal contraceptives to men: advances and obstacles
.
Trends Biotechnol
.
2008
;
26
:
90
99
.

234

Takehashi
M
,
Kanatsu-Shinohara
M
,
Miki
H
, et al. .
Production of knockout mice by gene targeting in multipotent germline stem cells
.
Dev Biol
.
2007
;
312
:
344
352
.

235

Bratt
A
,
Wilson
WJ
,
Troyanovsky
B
, et al. .
Angiomotin belongs to a novel protein family with conserved coiled-coil and PDZ binding domains
.
Gene
.
2002
;
298
:
69
77
.

236

Troyanovsky
B
,
Levchenko
T
,
Månsson
G
,
Matvijenko
O
,
Holmgren
L
.
Angiomotin: an angiostatin binding protein that regulates endothelial cell migration and tube formation
.
J Cell Biol
.
2001
;
152
:
1247
1254
.

237

Ernkvist
M
,
Luna Persson
N
,
Audebert
S
, et al. .
The Amot/Patj/Syx signaling complex spatially controls RhoA GTPase activity in migrating endothelial cells
.
Blood
.
2009
;
113
:
244
253
.

238

Aase
K
,
Ernkvist
M
,
Ebarasi
L
, et al. .
Angiomotin regulates endothelial cell migration during embryonic angiogenesis
.
Genes Dev
.
2007
;
21
:
2055
2068
.

239

Heller
B
,
Adu-Gyamfi
E
,
Smith-Kinnaman
W
, et al. .
Amot recognizes a juxtanuclear endocytic recycling compartment via a novel lipid binding domain
.
J Biol Chem
.
2010
;
285
:
12308
12320
.

240

Hung
TJ
,
Kemphues
KJ
.
PAR-6 is a conserved PDZ domain-containing protein that colocalizes with PAR-3 in Caenorhabditis elegans embryos
.
Development
.
1999
;
126
:
127
135
.

241

Etemad-Moghadam
B
,
Guo
S
,
Kemphues
KJ
.
Asymmetrically distributed PAR-3 protein contributes to cell polarity and spindle alignment in early C. elegans embryos
.
Cell
.
1995
;
83
:
743
752
.

242

Crespo
CL
,
Vernieri
C
,
Keller
PJ
, et al. .
The PAR complex controls the spatiotemporal dynamics of F-actin and the MTOC in directionally migrating leukocytes
.
J Cell Sci
.
2014
;
127
:
4381
4395
.

243

Pegtel
DM
,
Ellenbroek
SI
,
Mertens
AE
,
van der Kammen
RA
,
de Rooij
J
,
Collard
JG
.
The Par-Tiam1 complex controls persistent migration by stabilizing microtubule-dependent front-rear polarity
.
Curr Biol
.
2007
;
17
:
1623
1634
.

244

Hidalgo-Carcedo
C
,
Hooper
S
,
Chaudhry
SI
, et al. .
Collective cell migration requires suppression of actomyosin at cell-cell contacts mediated by DDR1 and the cell polarity regulators Par3 and Par6
.
Nat Cell Biol
.
2011
;
13
:
49
58
.

245

Woodham
EF
,
Machesky
LM
.
Polarised cell migration: intrinsic and extrinsic drivers
.
Curr Opin Cell Biol
.
2014
;
30
:
25
32
.

246

Johansson
A
,
Driessens
M
,
Aspenström
P
.
The mammalian homologue of the Caenorhabditis elegans polarity protein PAR-6 is a binding partner for the Rho GTPases Cdc42 and Rac1
.
J Cell Sci
.
2000
;
113
:
3267
3275
.

247

Kohjima
M
,
Noda
Y
,
Takeya
R
,
Saito
N
,
Takeuchi
K
,
Sumimoto
H
.
PAR3β, a novel homologue of the cell polarity protein PAR3, localizes to tight junctions
.
Biochem Biophys Res Commun
.
2002
;
299
:
641
646
.

248

Gao
L
,
Macara
IG
,
Joberty
G
.
Multiple splice variants of Par3 and of a novel related gene, Par3L, produce proteins with different binding properties
.
Gene
.
2002
;
294
:
99
107
.

249

Izumi
Y
,
Hirose
T
,
Tamai
Y
, et al. .
An atypical PKC directly associates and colocalizes at the epithelial tight junction with ASIP, a mammalian homologue of Caenorhabditis elegans polarity protein PAR-3
.
J Cell Biol
.
1998
;
143
:
95
106
.

250

Bultje
RS
,
Castaneda-Castellanos
DR
,
Jan
LY
,
Jan
YN
,
Kriegstein
AR
,
Shi
SH
.
Mammalian Par3 regulates progenitor cell asymmetric division via notch signaling in the developing neocortex
.
Neuron
.
2009
;
63
:
189
202
.

251

Hirose
T
,
Izumi
Y
,
Nagashima
Y
, et al. .
Involvement of ASIP/PAR-3 in the promotion of epithelial tight junction formation
.
J Cell Sci
.
2002
;
115
:
2485
2495
.

252

Mizuno
K
,
Suzuki
A
,
Hirose
T
, et al. .
Self-association of PAR-3-mediated by the conserved N-terminal domain contributes to the development of epithelial tight junctions
.
J Biol Chem
.
2003
;
278
:
31240
31250
.

253

Itoh
M
,
Sasaki
H
,
Furuse
M
,
Ozaki
H
,
Kita
T
,
Tsukita
S
.
Junctional adhesion molecule (JAM) binds to PAR-3: a possible mechanism for the recruitment of PAR-3 to tight junctions
.
J Cell Biol
.
2001
;
154
:
491
497
.

254

Ebnet
K
,
Aurrand-Lions
M
,
Kuhn
A
, et al. .
The junctional adhesion molecule (JAM) family members JAM-2 and JAM-3 associate with the cell polarity protein PAR-3: a possible role for JAMs in endothelial cell polarity
.
J Cell Sci
.
2003
;
116
:
3879
3891
.

255

Ebnet
K
,
Suzuki
A
,
Horikoshi
Y
, et al. .
The cell polarity protein ASIP/PAR-3 directly associates with junctional adhesion molecule (JAM)
.
EMBO J
.
2001
;
20
:
3738
3748
.

256

Suzuki
A
,
Yamanaka
T
,
Hirose
T
, et al. .
Atypical protein kinase C is involved in the evolutionarily conserved PAR protein complex and plays a critical role in establishing epithelia-specific junctional structures
.
J Cell Biol
.
2001
;
152
:
1183
1196
.

257

Ling
C
,
Zuo
D
,
Xue
B
,
Muthuswamy
S
,
Muller
WJ
.
A novel role for 14–3-3σ in regulating epithelial cell polarity
.
Genes Dev
.
2010
;
24
:
947
956
.

258

Fu
H
,
Subramanian
RR
,
Masters
SC
.
14-3-3 proteins: structure, function, and regulation
.
Annu Rev Pharmacol Toxicol
.
2000
;
40
:
617
647
.

259

Hurd
TW
,
Fan
S
,
Liu
CJ
,
Kweon
HK
,
Hakansson
K
,
Margolis
B
.
Phosphorylation-dependent binding of 14-3-3 to the polarity protein Par3 regulates cell polarity in mammalian epithelia
.
Curr Biol
.
2003
;
13
:
2082
2090
.

260

Wang
Y
,
Du
D
,
Fang
L
, et al. .
Tyrosine phosphorylated Par3 regulates epithelial tight junction assembly promoted by EGFR signaling
.
EMBO J
.
2006
;
25
:
5058
5070
.

261

Horikoshi
Y
,
Suzuki
A
,
Yamanaka
T
, et al. .
Interaction between PAR3 and the aPKC-PAR-6 complex is indispensable for apical domain development of epithelial cells
.
J Cell Sci
.
2009
;
122
:
1595
1606
.

262

Zhang
H
,
Macara
IG
.
The polarity protein PAR-3 and TIAM1 cooperate in dendritic spine morphogenesis
.
Nat Cell Biol
.
2006
;
8
:
227
237
.

263

Joberty
G
,
Petersen
C
,
Gao
L
,
Macara
IG
.
The cell-polarity protein Par6 links Par3 and atypical protein kinase C to Cdc42
.
Nat Cell Biol
.
2000
;
2
:
531
539
.

264

Lemmers
C
,
Michel
D
,
Lane-Guermonprez
L
, et al. .
CRB3 binds directly to Par6 and regulates the morphogenesis of the tight junctions in mammalian epithelial cells
.
Mol Biol Cell
.
2004
;
15
:
1324
1333
.

265

Lin
D
,
Edwards
AS
,
Fawcett
JP
,
Mbamalu
G
,
Scott
JD
,
Pawson
T
.
A mammalian PAR-3-PAR-6 complex implicated in Cdc42/Rac1 and aPKC signalling and cell polarity
.
Nat Cell Biol
.
2000
;
2
:
540
547
.

266

Hung
AY
,
Sheng
M
.
PDZ domains: structural modules for protein complex assembly
.
J Biol Chem
.
2002
;
277
:
5699
5702
.

267

Sarkar
O
,
Mathur
PP
,
Cheng
CY
,
Mruk
DD
.
Interleukin alpha (1L1A) is a novel regulator of the blood-testis barrier in the rat
.
Biol Reprod
.
2008
;
78
:
445
454
.

268

Hutterer
A
,
Betschinger
J
,
Petronczki
M
,
Knoblich
JA
.
Sequential roles of Cdc42, Par-6, aPKC, and Lgl in the establishment of epithelial polarity during Drosophila embryogenesis
.
Dev Cell
.
2004
;
6
:
845
854
.

269

Noda
Y
,
Takeya
R
,
Ohno
S
,
Naito
S
,
Ito
T
,
Sumimoto
H
.
Human homologues of the Caenorhabditis elegans cell polarity protein PAR6 as an adaptor that links the small GTPases Rac and Cdc42 to atypical protein kinase C
.
Genes Cells
.
2001
;
6
:
107
119
.

270

Habets
GG
,
van der Kammen
RA
,
Stam
JC
,
Michiels
F
,
Collard
JG
.
Sequence of the human invasion-inducing TIAM1 gene, its conservation in evolution and its expression in tumor cell lines of different tissue origin
.
Oncogene
.
1995
;
10
:
1371
1376
.

271

Ellenbroek
SI
,
Iden
S
,
Collard
JG
.
The Rac activator Tiam1 is required for polarized protrusional outgrowth of primary astrocytes by affecting the organization of the microtubule network
.
Small GTPases
.
2012
;
3
:
4
14
.

272

Nishimura
T
,
Yamaguchi
T
,
Kato
K
, et al. .
PAR-6-PAR-3 mediates Cdc42-induced Rac activation through the Rac GEFs STEF/Tiam1
.
Nat Cell Biol
.
2005
;
7
:
270
277
.

273

Ozdamar
B
,
Bose
R
,
Barrios-Rodiles
M
,
Wang
HR
,
Zhang
Y
,
Wrana
JL
.
Regulation of the polarity protein Par6 by TGFβ receptors controls epithelial cell plasticity
.
Science
.
2005
;
307
:
1603
1609
.

274

Huang
C
.
Roles of E3 ubiquitin ligases in cell adhesion and migration
.
Cell Adh Migr
.
2010
;
4
:
1
10
.

275

Wang
HR
,
Ogunjimi
AA
,
Zhang
Y
,
Ozdamar
B
,
Bose
R
,
Wrana
JL
.
Degradation of RhoA by Smurf1 ubiquitin ligase
.
Methods Enzymol
.
2006
;
406
:
437
447
.

276

Sánchez
NS
,
Barnett
JV
.
TGFβ and BMP-2 regulate epicardial cell invasion via TGFβR3 activation of the Par6/Smurf1/RhoA pathway
.
Cell Signal
.
2012
;
24
:
539
548
.

277

Alarcon
VB
.
Cell polarity regulator PARD6B is essential for trophectoderm formation in the preimplantation mouse embryo
.
Biol Reprod
.
2010
;
83
:
347
358
.

278

Hayase
J
,
Kamakura
S
,
Iwakiri
Y
, et al. .
The WD40 protein Morg1 facilitates Par6-aPKC binding to Crb3 for apical identity in epithelial cells
.
J Cell Biol
.
2013
;
200
:
635
650
.

279

Edwards
RA
,
Bryan
J
.
Fascins, a family of actin bundling proteins
.
Cell Motil Cytoskeleton
.
1995
;
32
:
1
9
.

280

Hashimoto
Y
,
Kim
DJ
,
Adams
JC
.
The roles of fascins in health and disease
.
J Pathol
.
2011
;
224
:
289
300
.

281

Gungor-Ordueri
NE
,
Celik-Ozenci
C
,
Cheng
CY
.
Fascin 1 is an actin filament-bundling protein that regulates ectoplasmic specialization dynamics in the rat testis
.
Am J Physiol Endocrinol Metab
.
2014
;
307
:
E738
E753
.

282

Moscat
J
,
Diaz-Meco
MT
,
Wooten
MW
.
Of the atypical PKCs, Par-4 and p62: recent understandings of the biology and pathology of a PB1-dominated complex
.
Cell Death Differ
.
2009
;
16
:
1426
1437
.

283

Yamanaka
T
,
Horikoshi
Y
,
Sugiyama
Y
, et al. .
Mammalian Lgl forms a protein complex with PAR-6 and aPKC independently of PAR-3 to regulate epithelial cell polarity
.
Curr Biol
.
2003
;
13
:
734
743
.

284

Nagai-Tamai
Y
,
Mizuno
K
,
Hirose
T
,
Suzuki
A
,
Ohno
S
.
Regulated protein-protein interaction between aPKC and PAR-3 plays an essential role in the polarization of epithelial cells
.
Genes Cells
.
2002
;
7
:
1161
1171
.

285

Suzuki
A
,
Ishiyama
C
,
Hashiba
K
,
Shimizu
M
,
Ebnet
K
,
Ohno
S
.
aPKC kinase activity is required for the asymmetric differentiation of the premature junctional complex during epithelial cell polarization
.
J Cell Sci
.
2002
;
115
:
3565
3573
.

286

Iden
S
,
Misselwitz
S
,
Peddibhotla
SS
, et al. .
aPKC phosphorylates JAM-A at Ser285 to promote cell contact maturation and tight junction formation
.
J Cell Biol
.
2012
;
196
:
623
639
.

287

Harris
TJ
,
Peifer
M
.
aPKC controls microtubule organization to balance adherens junction symmetry and planar polarity during development
.
Dev Cell
.
2007
;
12
:
727
738
.

288

Harris
KP
,
Tepass
U
.
Cdc42 and Par proteins stabilize dynamic adherens junctions in the Drosophila neuroectoderm through regulation of apical endocytosis
.
J Cell Biol
.
2008
;
183
:
1129
1143
.

289

Georgiou
M
,
Marinari
E
,
Burden
J
,
Baum
B
.
Cdc42, Par6, and aPKC regulate Arp2/3-mediated endocytosis to control local adherens junction stability
.
Curr Biol
.
2008
;
18
:
1631
1638
.

290

Yamanaka
T
,
Ohno
S
.
Role of Lgl/Dlg/Scribble in the regulation of epithelial junction, polarity and growth
.
Front Biosci
.
2008
;
13
:
6693
6707
.

291

Humbert
P
,
Russell
S
,
Richardson
H
.
Dlg, Scribble and Lgl in cell polarity, cell proliferation and cancer
.
BioEssays
.
2003
;
25
:
542
553
.

292

Dow
LE
,
Humbert
PO
.
Polarity regulators and the control of epithelial architecture, cell migration, and tumorigenesis
.
Int Rev Cytol
.
2007
;
262
:
253
302
.

293

Dow
LE
,
Kauffman
JS
,
Caddy
J
, et al. .
The tumour-suppressor Scribble dictates cell polarity during directed epithelial migration: regulation of Rho GTPase recruitment to the leading edge
.
Oncogene
.
2007
;
26
:
2272
2282
.

294

Nola
S
,
Sebbagh
M
,
Marchetto
S
, et al. .
Scrib regulates PAK activity during the cell migration process
.
Hum Mol Genet
.
2008
;
17
:
3552
3565
.

295

Qin
Y
,
Capaldo
C
,
Gumbiner
BM
,
Macara
IG
.
The mammalian Scribble polarity protein regulates epithelial cell adhesion and migration through E-cadherin
.
J Cell Biol
.
2005
;
171
:
1061
1071
.

296

Osmani
N
,
Vitale
N
,
Borg
JP
,
Etienne-Manneville
S
.
Scrib controls Cdc42 localization and activity to promote cell polarization during astrocyte migration
.
Curr Biol
.
2006
;
16
:
2395
2405
.

297

Sone
K
,
Nakagawa
S
,
Nakagawa
K
, et al. .
hScrib, a human homologue of Drosophila neoplastic tumor suppressor, is a novel death substrate targeted by caspase during the process of apoptosis
.
Genes Cells
.
2008
;
13
:
771
785
.

298

Nagasaka
K
,
Nakagawa
S
,
Yano
T
, et al. .
Human homolog of Drosophila tumor suppressor Scribble negatively regulates cell-cycle progression from G1 to S phase by localizing at the basolateral membrane in epithelial cells
.
Cancer Sci
.
2006
;
97
:
1217
1225
.

299

Norman
M
,
Wisniewska
KA
,
Lawrenson
K
, et al. .
Loss of Scribble causes cell competition in mammalian cells
.
J Cell Sci
.
2012
;
125
:
59
66
.

300

de Vreede
G
,
Schoenfeld
JD
,
Windler
SL
,
Morrison
H
,
Lu
H
,
Bilder
D
.
The Scribble module regulates retromer-dependent endocytic trafficking during epithelial polarization
.
Development
.
2014
;
141
:
2796
2802
.

301

Zhan
L
,
Rosenberg
A
,
Bergami
KC
, et al. .
Deregulation of scribble promotes mammary tumorigenesis and reveals a role for cell polarity in carcinoma
.
Cell
.
2008
;
135
:
865
878
.

302

Pearson
HB
,
Perez-Mancera
PA
,
Dow
LE
, et al. .
SCRIB expression is deregulated in human prostate cancer, and its deficiency in mice promotes prostate neoplasia
.
J Clin Invest
.
2011
;
121
:
4257
4267
.

303

Takizawa
S
,
Nagasaka
K
,
Nakagawa
S
, et al. .
Human scribble, a novel tumor suppressor identified as a target of high-risk HPV E6 for ubiquitin-mediated degradation, interacts with adenomatous polyposis coli
.
Genes Cells
.
2006
;
11
:
453
464
.

304

Javier
RT
,
Rice
AP
.
Emerging theme: cellular PDZ proteins as common targets of pathogenic viruses
.
J Virol
.
2011
;
85
:
11544
11556
.

305

Bilder
D
.
Epithelial polarity and proliferation control: links from the Drosophila neoplastic tumor suppressors
.
Genes Dev
.
2004
;
18
:
1909
1925
.

306

Ivanov
AI
,
Young
C
,
Den Beste
K
, et al. .
Tumor suppressor scribble regulates assembly of tight junctions in the intestinal epithelium
.
Am J Pathol
.
2010
;
176
:
134
145
.

307

Su
W
,
Wong
EW
,
Mruk
DD
,
Cheng
CY
.
The Scribble/Lgl/Dlg polarity protein complex is a regulator of blood-testis barrier dynamics and spermatid polarity during spermatogenesis
.
Endocrinology
.
2012
;
153
:
6041
6053
.

308

Murdoch
JN
,
Henderson
DJ
,
Doudney
K
, et al. .
Disruption of scribble (Scrb1) causes severe neural tube defects in the circletail mouse
.
Hum Mol Genet
.
2003
;
12
:
87
98
.

309

Zarbalis
K
,
May
SR
,
Shen
Y
,
Ekker
M
,
Rubenstein
JL
,
Peterson
AS
.
A focused and efficient genetic screening strategy in the mouse: identification of mutations that disrupt cortical development
.
PLoS Biol
.
2004
;
2
:
E219
.

310

Métais
JY
,
Navarro
C
,
Santoni
MJ
,
Audebert
S
,
Borg
JP
.
hScrib interacts with ZO-2 at the cell-cell junctions of epithelial cells
.
FEBS Lett
.
2005
;
579
:
3725
3730
.

311

Li
X
,
Yang
H
,
Liu
J
,
Schmidt
MD
,
Gao
T
.
Scribble-mediated membrane targeting of PHLPP1 is required for its negative regulation of Akt
.
EMBO Rep
.
2011
;
12
:
818
824
.

312

Gao
T
,
Furnari
F
,
Newton
AC
.
PHLPP: a phosphatase that directly dephosphorylates Akt, promotes apoptosis, and suppresses tumor growth
.
Mol Cell
.
2005
;
18
:
13
24
.

313

Brognard
J
,
Newton
AC
.
PHLiPPing the switch on Akt and protein kinase C signaling
.
Trends Endocrinol Metab
.
2008
;
19
:
223
230
.

314

Newton
AC
,
Trotman
LC
.
Turning off AKT: PHLPP as a drug target
.
Annu Rev Pharmacol Toxicol
.
2014
;
54
:
537
558
.

315

Navarro
C
,
Nola
S
,
Audebert
S
, et al. .
Junctional recruitment of mammalian Scribble relies on E-cadherin engagement
.
Oncogene
.
2005
;
24
:
4330
4339
.

316

Lohia
M
,
Qin
Y
,
Macara
IG
.
The Scribble polarity protein stabilizes E-cadherin/p120-catenin binding and blocks retrieval of E-cadherin to the Golgi
.
PLoS One
.
2012
;
7
:
e51130
.

317

Roberts
S
,
Delury
C
,
Marsh
E
.
The PDZ protein discs-large (DLG): the ‘Jekyll and Hyde’ of the epithelial polarity proteins
.
FEBS J
.
2012
;
279
:
3549
3558
.

318

Walch
L
.
Emerging role of the scaffolding protein Dlg1 in vesicle trafficking
.
Traffic
.
2013
;
14
:
964
973
.

319

Reuver
SM
,
Garner
CC
.
E-cadherin mediated cell adhesion recruits SAP97 into the cortical cytoskeleton
.
J Cell Sci
.
1998
;
111
:
1071
1080
.

320

Laprise
P
,
Viel
A
,
Rivard
N
.
Human homolog of disc-large is required for adherens junction assembly and differentiation of human intestinal epithelial cells
.
J Biol Chem
.
2004
;
279
:
10157
10166
.

321

Vasioukhin
V
.
Lethal giant puzzle of Lgl
.
Dev Neurosci
.
2006
;
28
:
13
24
.

322

Grifoni
D
,
Froldi
F
,
Pession
A
.
Connecting epithelial polarity, proliferation and cancer in Drosophila: the many faces of lgl loss and function
.
Int J Dev Biol
.
2013
;
57
:
677
687
.

323

Müsch
A
,
Cohen
D
,
Yeaman
C
,
Nelson
WJ
,
Rodriguez-Boulan
E
,
Brennwald
PJ
.
Mammalian homolog of Drosophila tumor suppressor lethal (2) giant larvae interacts with basolateral exocytic machinery in Madin-Darby canine kidney cells
.
Mol Biol Cell
.
2002
;
13
:
158
168
.

324

Young
JS
,
Guttman
JA
,
Vaid
KS
,
Shahinian
H
,
Vogl
AW
.
Cortactin (CTTN), N-WASP (WASL) and clathrin (CLTC) are present at podosome-like tubulobulbar complexes in the rat testis
.
Biol Reprod
.
2009
;
80
:
153
161
.

325

Plant
PJ
,
Fawcett
JP
,
Lin
DC
, et al. .
A polarity complex of mPar-6 and atypical PKC binds, phosphorylates and regulates mammalian Lgl
.
Nat Cell Biol
.
2003
;
5
:
301
308
.

326

Weller
SG
,
Capitani
M
,
Cao
H
, et al. .
Src kinase regulates the integrity and function of the Golgi apparatus via activation of dynamin 2
.
Proc Natl Acad Sci USA
.
2010
;
107
:
5863
5868
.

327

Thomas
SM
,
Brugge
JS
.
Cellular functions regulated by Src family kinases
.
Annu Rev Cell Dev Biol
.
1997
;
13
:
513
609
.

328

Xiao
X
,
Mruk
DD
,
Wong
EW
, et al. .
Differential effects of c-Src and c-Yes on the endocytic vesicle-mediated trafficking events at the Sertoli cell blood-testis barrier: an in vitro study
.
Am J Physiol Endocrinol Metab
.
2014
;
307
:
E553
E562
.

329

Xiao
X
,
Mruk
DD
,
Lee
WM
,
Cheng
CY
.
c-Yes regulates cell adhesion at the blood-testis barrier and the apical ectoplasmic specialization in the seminiferous epithelium of rat testes
.
Int J Biochem Cell Biol
.
2011
;
43
:
651
665
.

330

Xiao
X
,
Mruk
DD
,
Cheng
CY
.
c-Yes regulates cell adhesion at the apical ectoplasmic specialization-blood-testis barrier axis via its effects on protein recruitment and distribution
.
Am J Physiol Endocrinol Metab
.
2013
;
304
:
E145
E159
.

331

Maekawa
M
,
Toyama
Y
,
Yasuda
M
,
Yagi
T
,
Yuasa
S
.
Fyn tyrosine kinase in Sertoli cells is involved in mouse spermatogenesis
.
Biol Reprod
.
2002
;
66
:
211
221
.

332

Luo
J
,
Gupta
V
,
Kern
B
, et al. .
Role of FYN kinase in spermatogenesis: defects characteristic of Fyn-null sperm in mice
.
Biol Reprod
.
2012
;
86
:
1
8
.

333

Taniguchi
H
.
Protein myristoylation in protein-lipid and protein-protein interactions
.
Biophys Chem
.
1999
;
82
:
129
137
.

334

Farazi
TA
,
Waksman
G
,
Gordon
JI
.
The biology and enzymology of protein N-myristoylation
.
J Biol Chem
.
2001
;
276
:
39501
39504
.

335

Simons
K
,
Sampaio
JL
.
Membrane organization and lipid rafts
.
Cold Spring Harb Perspect Biol
.
2011
;
3
:
a004697
.

336

Sandilands
E
,
Cans
C
,
Fincham
VJ
, et al. .
RhoB and actin polymerization coordinate Src activation with endosome-mediated delivery to the membrane
.
Dev Cell
.
2004
;
7
:
855
869
.

337

Ingley
E
.
Src family kinases: regulation of their activities, levels and identification of new pathways
.
Biochim Biophys Acta
.
2008
;
1784
:
56
65
.

338

Kaplan
KB
,
Swedlow
JR
,
Varmus
HE
,
Morgan
DO
.
Association of p60c-src with endosomal membranes in mammalian fibroblasts
.
J Cell Biol
.
1992
;
118
:
321
333
.

339

Möhn
H
,
Le Cabec
V
,
Fischer
S
,
Maridonneau-Parini
I
.
The src-family protein-tyrosine kinase p59hck is located on the secretory granules in human neutrophils and translocates towards the phagosome during cell activation
.
Biochem J
.
1995
;
309
:
657
665
.

340

Matsuda
D
,
Nakayama
Y
,
Horimoto
S
, et al. .
Involvement of Golgi-associated Lyn tyrosine kinase in the translocation of annexin II to the endoplasmic reticulum under oxidative stress
.
Exp Cell Res
.
2006
;
312
:
1205
1217
.

341

Kasahara
K
,
Nakayama
Y
,
Kihara
A
, et al. .
Rapid trafficking of c-Src, a non-palmitoylated Src-family kinase, between the plasma membrane and late endosomes/lysosomes
.
Exp Cell Res
.
2007
;
313
:
2651
2666
.

342

Kasahara
K
,
Nakayama
Y
,
Sato
I
, et al. .
Role of Src-family kinases in formation and trafficking of macropinosomes
.
J Cell Physiol
.
2007
;
211
:
220
232
.

343

Sandilands
E
,
Frame
MC
.
Endosomal trafficking of Src tyrosine kinase
.
Trends Cell Biol
.
2008
;
18
:
322
329
.

344

Korade-Mirnics
Z
,
Corey
SJ
.
Src kinase-mediated signaling in leukocytes
.
J Leukoc Biol
.
2000
;
68
:
603
613
.

345

Xiao
X
,
Mruk
DD
,
Cheng
FL
,
Cheng
CY
.
c-Src and c-Yes are two unlikely partners of spermatogenesis and their roles in blood-testis barrier dynamics
.
Adv Exp Med Biol
.
2012
;
763
:
295
317
.

346

Soriano
P
,
Montgomery
C
,
Geske
R
,
Bradley
A
.
Targeted disruption of the c-src proto-oncogene leads to osteopetrosis in mice
.
Cell
.
1991
;
64
:
693
702
.

347

Lowell
CA
,
Soriano
P
.
Knockouts of Src-family kinases: stiff bones, wimpy T cells, and bad memories
.
Genes Dev
.
1996
;
10
:
1845
1857
.

348

Mitra
SK
,
Hanson
DA
,
Schlaepfer
DD
.
Focal adhesion kinase: in command and control of cell motility
.
Nat Rev Mol Cell Biol
.
2005
;
6
:
56
68
.

349

Abbi
S
,
Guan
JL
.
Focal adhesion kinase: protein interactions and cellular functions
.
Histol Histopathol
.
2002
;
17
:
1163
1171
.

350

Wozniak
MA
,
Modzelewska
K
,
Kwong
L
,
Keely
PJ
.
Focal adhesion regulation of cell behavior
.
Biochim Biophys Acta
.
2004
;
1692
:
103
119
.

351

Harburger
DS
,
Calderwood
DA
.
Integrin signalling at a glance
.
J Cell Sci
.
2009
;
122
:
159
163
.

352

Huttenlocher
A
,
Horwitz
AR
.
Integrins in cell migration
.
Cold Spring Harb Symp Quant Biol
.
2011
;
3
:
a005074
.

353

Hsia
DA
,
Lim
ST
,
Bernard-Trifilo
JA
, et al. .
Integrin α4β1 promotes focal adhesion kinase-independent cell motility via α4 cytoplasmic domain-specific activation of c-Src
.
Mol Cell Biol
.
2005
;
25
:
9700
9712
.

354

Siu
ER
,
Wong
EW
,
Mruk
DD
,
Sze
KL
,
Porto
CS
,
Cheng
CY
.
An occludin-focal adhesion kinase protein complex at the blood-testis barrier: a study using the cadmium model
.
Endocrinology
.
2009
;
150
:
3336
3344
.

355

Siu
ER
,
Wong
EW
,
Mruk
DD
,
Porto
CS
,
Cheng
CY
.
Focal adhesion kinase is a blood-testis barrier regulator
.
Proc Natl Acad Sci USA
.
2009
;
106
:
9298
9303
.

356

Lie
PP
,
Mruk
DD
,
Mok
KW
,
Su
L
,
Lee
WM
,
Cheng
CY
.
Focal adhesion kinase-Tyr407 and -Tyr397 exhibit antagonistic effects on blood-testis barrier dynamics in the rat
.
Proc Natl Acad Sci USA
.
2012
;
109
:
12562
12567
.

357

Li
SY
,
Mruk
DD
,
Cheng
CY
.
Focal adhesion kinase is a regulator of F-actin dynamics: new insights from studies in the testis
.
Spermatogenesis
.
2013
;
3
:
e25385
.

358

Siu
MK
,
Mruk
DD
,
Lee
WM
,
Cheng
CY
.
Adhering junction dynamics in the testis are regulated by an interplay of β1 integrin and focal adhesion complex-associated proteins
.
Endocrinology
.
2003
;
144
:
2141
2163
.

359

Jovic
M
,
Sharma
M
,
Rahajeng
J
,
Caplan
S
.
The early endosome: a busy sorting station for proteins at the crossroads
.
Histol Histopathol
.
2010
;
25
:
99
112
.

360

Platta
HW
,
Stenmark
H
.
Endocytosis and signaling
.
Curr Opin Cell Biol
.
2011
;
23
:
393
403
.

361

Diaz-Rohrer
B
,
Levental
KR
,
Levental
I
.
Rafting through traffic: membrane domains in cellular logistics
.
Biochim Biophys Acta
.
2014
;
1838
:
3003
3013
.

362

Parat
MO
.
The biology of caveolae: achievements and perspectives
.
Int Rev Cell Mol Biol
.
2009
;
273
:
117
162
.

363

Kiss
AL
.
Caveolae and the regulation of endocytosis
. In:
Jasmin
JF
,
Frank
PG
,
Lisanti
MP
, eds.
Caveolins and Caveolae: Roles in Signaling and Disease Mechanisms
.
Austin, TX
:
Landes Bioscience and Springer Science+Business Media
,
2012
.

364

Murata
M
,
Peränen
J
,
Schreiner
R
,
Wieland
F
,
Kurzchalia
TV
,
Simons
K
.
VIP21/caveolin is a cholesterol-binding protein
.
Proc Natl Acad Sci USA
.
1995
;
92
:
10339
10343
.

365

Engqvist-Goldstein
AE
,
Drubin
DG
.
Actin assembly and endocytosis: from yeast to mammals
.
Annu Rev Cell Dev Biol
.
2003
;
19
:
287
332
.

366

Mooren
OL
,
Galletta
BJ
,
Cooper
JA
.
Roles for actin assembly in endocytosis
.
Annu Rev Biochem
.
2012
;
81
:
661
686
.

367

Tehrani
S
,
Tomasevic
N
,
Weed
S
,
Sakowicz
R
,
Cooper
JA
.
Src phosphorylation of cortactin enhances actin assembly
.
Proc Natl Acad Sci USA
.
2007
;
104
:
11933
11938
.

368

Uruno
T
,
Liu
J
,
Zhang
P
, et al. .
Activation of Arp2/3 complex-mediated actin polymerization by cortactin
.
Nat Cell Biol
.
2001
;
3
:
259
266
.

369

Ammer
AG
,
Weed
SA
.
Cortactin branches out: roles in regulating protrusive actin dynamics
.
Cell Motil Cytoskeleton
.
2008
;
65
:
687
707
.

370

MacGrath
SM
,
Koleske
AJ
.
Cortactin in cell migration and cancer at a glance
.
J Cell Sci
.
2012
;
125
:
1621
1626
.

371

Machesky
LM
,
Mullins
RD
,
Higgs
HN
, et al. .
Scar, a WASp-related protein, activates nucleation of actin filaments by the Arp2/3 complex
.
Proc Natl Acad Sci USA
.
1999
;
96
:
3739
3744
.

372

Rohatgi
R
,
Ma
L
,
Miki
H
, et al. .
The interaction between N-WASP and the Arp2/3 complex links Cdc42-dependent signals to actin assembly
.
Cell
.
1999
;
97
:
221
231
.

373

Mullins
RD
,
Heuser
JA
,
Pollard
TD
.
The interaction of Arp2/3 complex with actin: nucleation, high affinity pointed end capping, and formation of branching networks of filaments
.
Proc Natl Acad Sci USA
.
1998
;
95
:
6181
6186
.

374

Stahlhut
M
,
van Deurs
B
.
Identification of filamin as a novel ligand for caveolin-1: evidence for the organization of caveolin-1-associated membrane domains by the actin cytoskeleton
.
Mol Biol Cell
.
2000
;
11
:
325
337
.

375

Sverdlov
M
,
Shinin
V
,
Place
AT
,
Castellon
M
,
Minshall
RD
.
Filamin A regulates caveolae internalization and trafficking in endothelial cells
.
Mol Biol Cell
.
2009
;
20
:
4531
4540
.

376

Nakamura
F
,
Stossel
TP
,
Hartwig
JH
.
The filamins: organizers of cell structure and function
.
Cell Adh Migr
.
2011
;
160
169
.

377

Zhou
AX
,
Hartwig
JH
,
Akyürek
LM
.
Filamins in cell signaling, transcription and organ development
.
Trends Cell Biol
.
2010
;
20
:
113
123
.

378

Li
S
,
Seitz
R
,
Lisanti
MP
.
Phosphorylation of caveolin by src tyrosine kinases. The α-isoform of caveolin is selectively phosphorylated by v-Src in vivo
.
J Biol Chem
.
1996
;
271
:
3863
3868
.

379

Lee
H
,
Park
DS
,
Wang
XB
,
Scherer
PE
,
Schwartz
PE
,
Lisanti
MP
.
Src-induced phosphorylation of caveolin-2 on tyrosine 19. Phospho-caveolin-2 (Tyr(P)19) is localized near focal adhesions, remains associated with lipid rafts/caveolae, but no longer forms a high molecular mass hetero-oligomer with caveolin-1
.
J Biol Chem
.
2002
;
277
:
34556
34567
.

380

Goode
BL
,
Drubin
DG
,
Barnes
G
.
Functional cooperation between the microtubule and actin cytoskeletons
.
Curr Opin Cell Biol
.
2000
;
12
:
63
71
.

381

Mitchison
T
,
Kirschner
M
.
Dynamic instability of microtubule growth
.
Nature
.
1984
;
312
:
237
242
.

382

Wade
RH
.
On and around microtubules: an overview
.
Mol Biotechnol
.
2009
;
43
:
177
191
.

383

Wade
RH
.
Microtubules: an overview
.
Methods Mol Med
.
2007
;
137
:
1
16
.

384

Desai
A
,
Mitchison
TJ
.
Microtubule polymerization dynamics
.
Annu Rev Cell Dev Biol
.
1997
;
13
:
83
117
.

385

Apodaca
G
.
Endocytic traffic in polarized epithelial cells: role of the actin and microtubule cytoskeleton
.
Traffic
.
2001
;
2
:
149
159
.

386

Anitei
M
,
Hoflack
B
.
Bridging membrane and cytoskeleton dynamics in the secretory and endocytic pathways
.
Nat Cell Biol
.
2012
;
14
:
11
19
.

387

Murray
JW
,
Wolkoff
AW
.
Roles of the cytoskeleton and motor proteins in endocytic sorting
.
Adv Drug Deliv Rev
.
2003
;
55
:
1385
1403
.

388

de Forges
H
,
Bouissou
A
,
Perez
F
.
Interplay between microtubule dynamics and intracellular organization
.
Int J Biochem Cell Biol
.
2012
;
44
:
266
274
.

389

Shen
L
,
Turner
JR
.
Actin depolymerization disrupts tight junctions via caveolae-mediated endocytosis
.
Mol Biol Cell
.
2005
;
16
:
3919
3936
.

390

Bruewer
M
,
Utech
M
,
Ivanov
AI
,
Hopkins
AM
,
Parkos
CA
,
Nusrat
A
.
Interferon-γ induces internalization of epithelial tight junction proteins via a macropinocytosis-like process
.
FASEB J
.
2005
;
19
:
923
933
.

391

Fletcher
SJ
,
Rappoport
JZ
.
Tight junction regulation through vesicle trafficking: bringing cells together
.
Biochem Soc Trans
.
2014
;
42
:
195
200
.

392

Fletcher
SJ
,
Rappoport
JZ
.
The role of vesicle trafficking in epithelial cell motility
.
Biochem Soc Trans
.
2009
;
37
:
1072
1076
.

393

Findley
MK
,
Koval
M
.
Regulation and roles for claudin-family tight junction proteins
.
IUBMB Life
.
2009
;
61
:
431
437
.

394

Hagiwara
M
,
Shirai
Y
,
Nomura
R
, et al. .
Caveolin-1 activates Rab5 and enhances endocytosis through direct interaction
.
Biochem Biophys Res Commun
.
2009
;
378
:
73
78
.

395

Coyne
CB
,
Shen
L
,
Turner
JR
,
Bergelson
JM
.
Coxsackievirus entry across epithelial tight junctions requires occludin and the small GTPases Rab34 and Rab5
.
Cell Host Microbe
.
2007
;
2
:
181
192
.

396

Matsuda
M
,
Kubo
A
,
Furuse
M
,
Tsukita
S
.
A peculiar internalization of claudins, tight junction-specific adhesion molecules, during the intercellular movement of epithelial cells
.
J Cell Sci
.
2004
;
117
:
1247
1257
.

397

Fong
JT
,
Kells
RM
,
Falk
MM
.
Two tyrosine-based sorting signals in the Cx43 C-terminus cooperate to mediate gap junction endocytosis
.
Mol Biol Cell
.
2013
;
24
:
2834
2848
.

398

Piehl
M
,
Lehmann
C
,
Gumpert
A
,
Denizot
JP
,
Segretain
D
,
Falk
MM
.
Internalization of large double-membrane intercellular vesicles by a clathrin-dependent endocytic process
.
Mol Biol Cell
.
2007
;
18
:
337
347
.

399

Falk
MM
,
Fong
JT
,
Kells
RM
,
O'Laughlin
MC
,
Kowal
TJ
,
Thévenin
AF
.
Degradation of endocytosed gap junctions by autophagosomal and endo-/lysosomal pathways: a perspective
.
J Membr Biol
.
2012
;
245
:
465
476
.

400

Leithe
E
,
Sirnes
S
,
Fykerud
T
,
Kjenseth
A
,
Rivedal
E
.
Endocytosis and post-endocytic sorting of connexins
.
Biochim Biophys Acta
.
2012
;
1818
:
1870
1879
.

401

Yan
HH
,
Mruk
DD
,
Lee
WM
,
Cheng
CY
.
Blood-testis barrier dynamics are regulated by testosterone and cytokines via their differential effects on the kinetics of protein endocytosis and recycling in Sertoli cells
.
FASEB J
.
2008
;
22
:
1945
1959
.

402

Su
L
,
Mruk
DD
,
Lee
WM
,
Cheng
CY
.
Differential effects of testosterone and TGF-β3 on endocytic vesicle-mediated protein trafficking events at the blood-testis barrier
.
Exp Cell Res
.
2010
;
316
:
2945
2960
.

403

Tuma
P
,
Hubbard
AL
.
Transcytosis: crossing cellular barriers
.
Physiol Rev
.
2003
;
83
:
871
932
.

404

Deborde
S
,
Perret
E
,
Gravotta
D
, et al. .
Clathrin is a key regulator of basolateral polarity
.
Nature
.
2008
;
452
:
719
723
.

405

Beardsley
A
,
Fang
K
,
Mertz
H
,
Castranova
V
,
Friend
S
,
Liu
J
.
Loss of caveolin-1 polarity impedes endothelial cell polarization and directional movement
.
J Biol Chem
.
2005
;
280
:
3541
3547
.

406

Grande-García
A
,
del Pozo
MA
.
Caveolin-1 in cell polarization and directional migration
.
Eur J Cell Biol
.
2008
;
87
:
641
647
.

407

Du
M
,
Young
JN
,
De Asis
M
, et al. .
A novel subcellular machine contributes to basal junction remodeling in the seminiferous epithelium
.
Biol Reprod
.
2013
;
88
:
1
17
.

408

Pelletier
RM
.
Cyclic modulation of Sertoli cell junctional complexes in a seasonal breeder: the mink (Mustela vison)
.
Am J Anat
.
1988
;
183
:
68
102
.

409

Vogl
AW
,
Young
JS
,
Du
M
.
New insights into roles of tubulobulbar complexes in sperm release and turnover of blood-testis barrier
.
Int Rev Cell Mol Biol
.
2013
;
303
:
319
355
.

410

Russell
LD
.
Observations on the inter-relationships of Sertoli cells at the level of the blood-testis barrier: evidence for formation and resorption of Sertoli-Sertoli tubulobulbar complexes during the spermatogenic cycle of the rat
.
Am J Anat
.
1979
;
155
:
259
279
.

411

Young
JS
,
Guttman
JA
,
Vaid
KS
,
Vogl
AW
.
Tubulobulbar complexes are intercellular podosome-like structures that internalize intact intercellular junctions during epithelial remodeling events in the rat testis
.
Biol Reprod
.
2009
;
80
:
162
174
.