You are on page 1of 11

review REVIEW

Spermatogenesis 1:2, 105-115; April/May/June 2011; © 2011 Landes Bioscience

Regulation of blood-testis barrier dynamics


by desmosome, gap junction, hemidesmosome
and polarity proteins
An unexpected turn of events
C. Yan Cheng,1,* Elissa W.P. Wong,1 Pearl P.Y. Lie,1 Michelle W.M. Li,1 Dolores D. Mruk,1 Helen H.N. Yan,1 Ka-Wai Mok,1
Jayakanthan Mannu,2 Premendu P. Mathur,2 Wing-yee Lui,3 Will M. Lee,3 Michele Bonanomi4 and Bruno Silvestrini4
1
The Mary M. Wohlford Laboratory for Male Contraceptive Research; Center for Biomedical Research; The Population Council; New York, NY USA; 2Center for Bioinformatics;
School of Life Sciences; Pondicherry University; Pondicherry, India; 3School of Biological Sciences; The University of Hong Kong; Hong Kong, China;
4
S.B.M. Srl; Via Domenico Tardini; Rome, Italy

Key words: testis, spermatogenesis, seminiferous epithelial cycle, blood-testis barrier, Sertoli cell,
estrogens, bisphenol A, environmental toxicants, adjudin

The blood-testis barrier (BTB) is a unique ultrastructure in the mammalian testis. Unlike other blood-tissue barriers, such
as the blood-brain barrier and the blood-ocular (or blood-retina) barrier which formed by tight junctions (TJ) between
endothelial cells of the microvessels, the BTB is constituted by coexisting TJ, basal ectoplasmic specialization (basal
ES), desmosomes and gap junctions between adjacent Sertoli cells near the basement membrane of the seminiferous
tubule. The BTB also divides the seminiferous epithelium into the apical (or adluminal) and basal compartments so that
meiosis I and II and post-meiotic germ cell development can all take place in a specialized microenvironment in the
apical compartment behind the BTB. While the unusual anatomical features of the BTB have been known for decades,
the physiological function of the coexisting junctions, in particular the desmosome and gap junction, that constitute the
BTB was unknown until recently. Based on recently published findings, we critically evaluate the role of the desmosome
and gap junction that serve as a signaling platform to coordinate the “opening” and “closing” of the TJ-permeability
barrier conferred by TJ and basal ES during the seminiferous epithelial cycle of spermatogenesis. This is made possible by
polarity proteins working in concert with nonreceptor protein tyrosine kinases, such as focal adhesion kinase (FAK) and
c-Src, at the site to regulate endosome-mediated protein trafficking events (e.g., endocytosis, transcytosis, recycling or
protein degradation). These events not only serve to destabilize the existing “old” BTB above preleptotene spermatocytes
in transit in “clones” at the BTB, but also contribute to the assembly of “new” BTB below the transiting spermatocytes.
Furthermore, hemidesmosomes at the Sertoli cell-basement membrane interface also contribute to the BTB restructuring
events at stage VIII of the epithelial cycle. Additionally, the findings that a gap junction at the BTB provides a possible
route for the passage of toxicants [e.g., bisphenol A (BPA)] and potential male contraceptives (e.g., adjudin) across the BTB
also illustrate that these coexisting junctions, while helpful to maintain the immunological barrier integrity during the
transit of spermatocytes, can be the “gateway” to making the BTB so vulnerable to toxicants and/or chemicals, causing
male reproductive dysfunction.

Introduction Sertoli cells which cease to proliferate by ~15–17 days of age post-
partum in rats.4 Thus the Sertoli cell population remains rela-
In the mammalian testis, the blood-testis barrier (BTB) forms tively constant in adult mammals with ~30 million Sertoli cells
during puberty in humans (at ~12–14 years of age) and a func- per testis in adult rats at 150 and 240 days of age post-partum,5
tional BTB is established by ~17–21 day postpartum in rats,1,2 and each Sertoli cell is known to support ~30–50 developing
coinciding with the differentiation of Type B spermatogo- germ cells in the seminiferous epithelium.6,7 However, recent
nia to prepare spermatocytes for meiosis I and meiosis II.3 studies have shown that Sertoli cells are capable of dividing
This timing also coincides with terminal differentiation of even in adult animals including rats8 and mice9 under experi-
mental and even under physiological conditions in humans.9,10
The BTB anatomically divides the seminiferous epithelium into
*Correspondence to: C. Yan Cheng; Email: Y-Cheng@popcbr.rockefeller.edu the basal and apical (or adluminal) compartments (see Fig. 1).
Submitted: 03/14/10; Revised: 04/04/11; Accepted: 04/05/11 This ultrastructure is created by specialized junctions between
DOI: 10.4161/spmg.1.2.15745
adjacent Sertoli cells near the basement membrane, and it is

www.landesbioscience.com Spermatogenesis 105


Figure 1. A schematic illustration of the blood-testis barrier (BTB) in the seminiferous epithelium of adult mammalian testis. This is a schematic
representation of the seminiferous epithelium in the adult rat testis of a stage VII tubule. Anatomically, the BTB divides the seminiferous epithelium
(composed of only Sertoli and germ cells, lying on top of the basement membrane, which is a modified form of extracellular matrix63,64) into the basal
and apical (adluminal) compartments. The BTB is constituted by coexisting TJ, basal ES, desmosome and gap junction. The most obvious feature of
the BTB is the tightly packed actin filament bundles which are lying parallel to the plasma membrane and are sandwiched in between cisternae of
endoplasmic reticulum and the opposing plasma membranes of the two adjacent Sertoli cells. This tripartite structure is known as the basal ES. This
feature is also found at the Sertoli cell-spermatid (step 8–19 spermatids in rats) interface known as the apical ES in the apical compartment, which is
the only anchoring device once it appears during spermiogenesis. At the BTB, TJ and gap junction coexist with basal ES, whereas desmosome is an
intermediate filament-based anchoring junction type at the BTB, illustrating that the actin- and the intermediate filament-based cytoskeletons are
working in concert to regulate BTB dynamics during spermatogenesis. The unusually organized network of actin filament associated with the plasma
membrane at the BTB and apical ES are maintained via the intricate interactions of: (1) several actin regulatory proteins (e.g., Eps8, an actin bundling
and capping protein) and Arp2/3 protein complex (an actin nucleation protein causing the branching of actin filaments, thereby disrupting the actin
filament bundles at the ES, replacing them with branched actin network) and (2) the properly organized cell adhesion protein complexes at the TJ
(e.g., occludin-ZO-1) and basal ES (e.g., N-cadherin-β-catenin), which are maintained by the signaling action of the protein complexes at the desmo-
some (e.g., desmoglein-2/desmocollin-2/c-Src) and the gap junction (e.g., connexin43-plakophilin-2-c-Src) as well as the polarity proteins (e.g., PAR3,
PAR6, 14-3-3, Cdc42).

crucial to confer cell polarity, to limit paracellular transport of cells and developing germ cells.1,12,13 An earlier study has shown
biomolecules, ions, electrolytes and water from the basal to the that when the BTB assembly was delayed by ~4 wk via treatment
apical compartment, and to maintain an immunological barrier of neonatal rats with a toxicant diethylstilbestrol, spermatogo-
to sequester post-meiotic germ cell development from the sys- nial differentiation and the onset of meiosis would be delayed,
temic circulation.1,11 Thus, spermiogenesis and spermiation take and their resumption correlate with the establishment of an
place in a specialized microenvironment at the apical compart- intact and functional BTB.2 A more recent study has shown that
ment of the seminiferous epithelium composed of only Sertoli spermatogenesis failed to reinitiate in rats treated with adjudin

106 Spermatogenesis Volume 1 Issue 2


[1-(2,4-dichorobenzyl)-1H-indazole-3-carbohydrazide] at high Each connexon is composed of a hexamer of gap junction pro-
doses (e.g., 250 mg/kg b.w.) that irreversibly disrupted the BTB teins known as connexins (Cx). At least 20 connexins are found
integrity, even though the population of spermatogonial stem in humans and rodents, such as Cx43, Cx26 and Cx33. The gap
cells/spermatogonia did not reduce significantly.14 Interestingly, junction channels were shown to permit the diffusion of solutes
rats treated with adjudin at a lower dose, such as at 50 mg/kg b.w. of <1–1.5 kDa in molecular mass.31-33 More recent studies have
which also perturbed the BTB integrity transiently and reversibly, shown that substances of large molecular sizes, such as siRNA
managed to re-initiate spermatogonial differentiation and sper- duplexes, polypeptides and cyclic nucleotides can also pass
matogenesis when the BTB functionality was restored.14 These through the gap junction channels with selective permeability.34
findings thus illustrate the significance of the BTB in maintain- Thus, while desmosomes contribute to the cell adhesion func-
ing spermatogenesis. Unlike other blood-tissue barriers, such as tion at the BTB, gap junction communication channels provide
the blood-brain barrier and the blood-ocular barrier which are the means for signaling to coordinate cellular events at the BTB.
constituted by TJ-permeability barrier of the endothelial cells in However, recent findings illustrate that both desmosome and gap
the microvessels in the brain and the eye, respectively,11,15,16 the junction provide an unusual platform for cell signaling at the
BTB is constituted by coexisting tight junction (TJ, also known BTB, which are discussed in the following sections.
as zonula occludens), basal ectoplasmic specialization [basal ES, Role of desmosomes. Recent studies have unequivocally
a testis-specific adherens junction (AJ, also known as zonula demonstrated that desmosomes are performing some unique and
adherens) type], desmosome (also known as macula adherens) significant roles at the BTB. First, it was shown that while the
and gap junction1 (see Fig. 1). More importantly, in addition to knockdown of desmoglein-2 or desmocollin-2 alone in the Sertoli
TJ protein complexes (e.g., occludin-ZO-1, JAM-A-ZO-1, clau- cell epithelium by RNAi using specific siRNA duplexes failed to
din-ZO-1), the BTB is composed of adhesion protein complexes perturb the Sertoli cell TJ-permeability barrier, the knockdown
found in other junction types, including adherens junction (AJ, of either desmoglein-2 alone or desmoglein-2 and desmocollin-2
e.g., N-cadherin/β-catenin; nectin-afadin), desmosome (e.g., combined caused mis-localization of ZO-1, moving from the
desmoglein-2-desmocollin-2) and gap junction (e.g., connexin Sertoli-Sertoli cell surface to cell cytosol.35 More importantly, a
43-plakophilin-2) (see Fig. 1).1,17,18 Yet the roles of desmosome knockdown of both desmoglein-2 and desmocollin-2 by RNAi
and gap junction at the BTB remain unknown for decades since was found to perturb the Sertoli cell TJ-permeability function
their first discoveries in the 1970s.19-21 Nevertheless, recent stud- in addition to a mis-localization of both c-Src (a nonreceptor
ies have shown that they are crucial to provide the necessary protein tyrosine kinase) and coxsackievirus and adenovirus
cross-talk between different junction types at the BTB. Thus, the receptor (CAR, an integral membrane protein and a cell adhe-
“opening/disruption” and “closing/reassembly” of different junc- sion molecule at the BTB,35a,37,58 which likely interacts with the
tion types can be coordinated temporally and spatially, thereby N-cadherin-based adhesion protein complex at the basal ES
facilitating the transit of preleptotene spermatocytes across the via β-catenin as its binding partner37), moving from the cell-
BTB at stage VIII of the seminiferous epithelial cycle while cell interface to cell cytosol.35 Second, the mis-localization of
maintaining the immunological barrier. CAR in the Sertoli cell epithelium induced by the knockdown
of desmoglein-2 and desmocollin-2 was shown to be the result
What are the Roles of the Desmosome of an increase in endocytosis of CAR,35 thereby destabilizing
and Gap Junction at the BTB? the cell adhesion function at the BTB. Third, studies by co-
immunoprecipitation have shown that while desmoglein-2 did
Introduction. A desmosome (also known as macula adherens) is not structurally interact with occludin, ZO-1 or Cx43, it was
an intermediate filament-based cell-cell anchoring junction type tightly associated c-Src; 35 and other studies have shown that
in which the integral membrane proteins (known as the desmo- c-Src interacted with Cx43,36 CAR,37 occludin, N-cadherin
somal cadherins: desmogleins and desmocollins) are anchored and ZO-1.38 Thus, the desmoglein-2/desmocollin-2/c-Src des-
to intermediate filaments (composed of vimentin in adult tes- mosomal protein complex at the BTB may be an important
tes) via the cytolinker desmoplakin.22,23 Desmosomal cadherin- regulatory complex with c-Src being served as the dominant
desmoplakin interactions are further reinforced by the armadillo regulator to confer proper phosphorylation status of proteins at
proteins plakoglobin and plakophilin, forming a distinctive the BTB. The knockdown of desmoglein-2/desmocollin-2 by
electron dense ultrastructure under electron microscopy at the RNAi in the Sertoli cell epithelium led to improper localization
Sertoli-Sertoli cell interface at the BTB.17,24-26 In the testis, besides of c-Src which moved away from the cell-cell interface to cyto-
the BTB, desmosomes can also be found at the Sertoli cell-sper- sol,35 thereby “messing up” the homeostasis of c-Src signaling
matid (limited to step 1–7 spermatids) or Sertoli-spermatocyte/ function, leading to improper phosphorylation status of crucial
spermatogonium interface.19,27 A gap junction, on the other hand, proteins at the Sertoli-Sertoli cell interface (e.g., CAR, ZO-1)
is a cell-cell actin-based communicating junction type.21,28-30 A and affecting their kinetics of endocytosis. All these contribute
functional gap junction communication channel is composed of to a destabilization of the adhesion function at the site, which in
two opposing connexons (the functional units of gap junction turn, affect the TJ-permeability barrier. Collectively, these find-
with each connexon known as a hemichannel) between adjacent ings illustrate that besides conferring adhesion function at the
Sertoli cells at the BTB [or between Sertoli cells and spermatids BTB, desmosome serves as a signaling platform at the BTB via
(step 1–7 spermatids only), spermatocytes or spermatogonia].21,28 its interaction with c-Src.

www.landesbioscience.com Spermatogenesis 107


Role of gap junctions. It is known that gap junctions that lie by the differential localization of polarity complexes: the PAR-
behind the junctional complexes (composed of TJ, AJ and desmo- and the Crumbs-based complexes are localized predominantly to
somes) in multiple epithelia provide the necessary cross-talk and TJ and the Scribble-based protein complex displays basolateral
signaling function for the coordination of cellular events to main- localization. Since each of these protein complexes recruit their
tain the homeostasis of an epithelium.30,33,39 However, the func- own binding partners (including adaptors, kinases, phospha-
tion of gap junctions at the BTB remains largely unknown until tases and scaffolding proteins), the mutual exclusion between
recently because they are found to be intermingled with TJ, basal the Scribble-based complex and the TJ localized polarity protein
ES and desmosome instead of being separate entities as in other complexes thus confers apico-basal polarity and asymmetrical
epithelia (see Fig. 1). It was found that in Sertoli cell epithelium distribution of proteins and organelles within an epithelial cell.
with an established functional TJ-barrier and having ultrastruc- These polarity proteins have at least one homolog found in mam-
tures that mimicked the Sertoli cell BTB in vivo, a knockdown mals.41,44 Indeed, recent studies have shown that many of these
of Cx43 or its adaptor protein plakophilin-2 (also an adaptor for proteins are expressed by Sertoli and/or germ cells,45 regulating
desmosome, illustrating a tight structural association between cell polarity in the seminiferous epithelium. For instance, Sertoli
actin-based gap junction and intermediate filament-based desmo- cell nuclei are restricted to the basal compartment near the base-
some at the BTB in the mammalian testis) using specific siRNA ment membrane, and the heads of developing spermatids are all
duplexes failed to perturbed the Sertoli cell TJ-barrier function.36 pointing toward the basement membrane, illustrating cell polar-
However, a simultaneous knockdown of Cx43 and plakophilin-2 ity in the seminiferous epithelium.44
by RNAi led to a disruption of the Sertoli cell TJ-permeability Besides conferring Sertoli cell and spermatid polarity in the
barrier, possibly caused by the mis-localization of occludin and testis, polarity proteins (e.g., PAR3, PAR6) were recently shown
ZO-1, with these proteins moving from the Sertoli-Sertoli cell to be involved in cell adhesion at the Sertoli cell-elongating
interface to cell cytosol, thereby destabilizing cell adhesion at spermatid interface (known as apical ES) and Sertoli-Sertoli
the TJ and leading to barrier disruption.36 More important, even cell interface at the BTB.45 For instance, a knockdown of either
though the knockdown of Cx43 alone did not perturb the Sertoli PAR3 or PAR6 led to a redistribution of JAM-A and α-catenin at
cell TJ-barrier function,36 subsequent studies using the [Ca 2+]- the Sertoli-Sertoli cell interface, moving from cell surface to cell
switch model and the bisphenol A (BPA) model showed that Cx43 cytosol, thereby destabilizing cell adhesion at the TJ-barrier.45
knockdown hindered these cells from “re-assembling” a disrupted These findings also implicate polarity proteins may be involved
TJ-barrier.40 These findings thus illustrate that while Cx43 may in endocytic vesicle-mediated protein trafficking events, such
not be critical for the assembly of a “new” TJ-barrier, it is essential as endocytosis, transcytosis, recycling and endosome-mediated
for the maintenance of an established TJ-barrier, , such as at the protein degradation. In fact, subsequent studies have shown that
BTB in adult mammalian testes, via its effects on TJ reassembly polarity proteins (e.g., 14-3-3 also known as PAR5 and Cdc42
during spermatogenesis when the BTB is transiently disrupted also called cell division control protein 42 and a member of the
during the epithelial cycle (e.g., at stage VIII to facilitate the Rho GTPases are integrated components of the PAR-based polar-
transit of preleptotene spermatocytes), as manifested by a loss of ity protein complex) are involved in protein endocytosis at the
intracellular [Ca 2+] or following the exposure of Sertoli cell epi- Sertoli cell BTB, since a knockdown of 14-3-3 by RNAi led to a
thelium to an environmental toxicant BPA.40 Similar to a number significant increase in the kinetics of endocytosis of JAM-A and
of BTB protein complexes (e.g., desmoglein-2-based complex),35 N-cadherin.46 Additionally, overexpression of a Cdc42 dominant
the Cx43-based gap junction protein complex also contains c-Src negative mutant in Sertoli cells abolished the TGFβ3-induced
as an integrated component,36 illustrating that c-Src is likely to acceleration in occludin and CAR endocytosis in the Sertoli cell
modify the phosphorylation status of connexins at the gap junc- epithelium, thereby blocking the TGFβ3-induced disruption of
tion, thereby regulating the kinetics of their endocytic vesicle- the Sertoli cell TJ-barrier.47 These findings thus illustrate that
mediated protein trafficking events. polarity proteins are crucial for the endocytic vesicle-mediated
protein trafficking events at the Sertoli BTB. Taking these data
What are the Roles of Polarity Proteins collectively, it is likely that the predominant localization of polar-
and Hemidesmosome on BTB Dynamics? ity proteins at the BTB (e.g., PAR6 14-3-3) and their stage-
specific expression during the seminiferous epithelial cycle are
Polarity proteins. Earlier studies in Drosophila melanogaster and crucial for the desmosome- and gap junction-mediated effects on
Caenorhabditis elegans have shown that cellular asymmetry and protein distribution, which require changes in protein endocyto-
apico-basal polarity are established and maintained by polarity sis, transcytosis and/or recycling. The net result of these changes
proteins.41-44 There are three modules of polarity protein com- thus regulates cell adhesion at the apical ES, and basal ES at the
plexes known as (1) the PAR (partitioning defective) protein BTB.
complex [e.g., PAR3/PAR6/aPKC (atypical protein kinase C, Hemidesmosome. Hemidesmosome is an intermediate fil-
a nonreceptor Ser/Thr protein kinase)], (2) the Crumbs pro- ament-based cell-matrix anchoring junction type found at the
tein complex [e.g., Crumbs-3/PALS1 (protein associated with Sertoli cell-basement membrane interface in the mammalian
(LIN-7)-1)/PATJ (PALS1-associated tight-junction protein)] and testis.1 Its composition and function in the testis remain largely
(3) the Scribble complex [e.g., Scribble/LGL1/2 (Lethal giant lar- unknown, except that β1-integrin and α2-laminin chain appear
vae 1/2)/DLG1 (Discs large 1)]. Apico-basal polarity is conferred to be the putative components of the hemidesmosome.48 Most

108 Spermatogenesis Volume 1 Issue 2


Figure 2. A molecular modeling study to assess the docked complex of
estradiol-17β with the Cx43-based gap junction communication chan-
nel. (A) Topographic view of the Cx43-gap junction protein complex
with estradiol-17β. (B) Side view of the Cx43-gap junction protein
complex with estradiol-17β. (C) Ball and stick model for the interaction
residues and secondary structure representation for protein between
estradiol-17β and Cx43. The SOSUI WWW server65 was used to predict
transmembrane helices for connexin 43 (Cx43)-based gap junction (GJ)
communicating channel. The characteristic feature of the Cx43-GJ chan-
nel is the spanning of transmembrane helices in the cell membrane that
creates the “pore” which allows passage of small molecules (see “A” and
“B”). The crystal structure of human Cx26 at 3.5 Å resolution in humans
is known,66 and this information was used for the molecular modeling of
Cx43. Initially, the amino acid sequence of Cx43 [UniProtKB ID: P08050]
was retrieved from UniProtKB database (www.uniprot.org).67 The crystal
structure of Cx26 was retrieved from Protein Data Bank (PDB ID: 2ZW3).68
The MODELLER 9v7 program69 was used for sequence alignment and
model building.70 The sequence and structural alignment of Cx43 to
human Cx26 was carried out using ‘align2d’ program in MODELLER 9v7.
This alignment study illustrated 45% sequence identity in the trans-
membrane region between the two proteins, which suggested that the
most important pore region is conserved between Cx26 and Cx43. The
produced alignment was then used to generate hemichannel struc-
ture with ‘model-multichain’ program in MODELLER 9v7. The quality of
the structure was assessed by submitting the modeled structure into
PROCHECK.71 The loop building and energy minimization were carried
out with Swiss-PdbViewer. The energy computations were done by GRO-
MOS96 implemented in Swiss-PdbViewer. The modeled three dimen-
sional structure of Cx43 was prepared using Schrödinger Suite 2009 Pro-
tein Preparation Wizard. All the hydrogens were added to the backbone
of the structure and bond orders was assigned appropriately. Exhaustive
sampling method was used for the optimization of the hydrogen bond-
ing network, in which the orientation of hydroxyl groups, amide groups
of Asn and Gln, and imidazole ring of His residues were optimized. The
RMSD (root mean square deviation) of atom displacement for terminat-
ing the minimization was set to be 0.30 Å. The minimization was carried
out with OPLS2005 force field. The ligand structure of estradiol-17β (CID:
5757), bisphenol A (CID: 6623) (Fig. 3) and adjudin (CID: 9819086) (Fig.
4) were retrieved from NCBI-PubChem database. LigPrep (Version 2.3,
Schrödinger, LLC, New York, NY 2009) was used to generate 3D structure
with correct chiralities for each ligand used in this study. Grid files repre-
sent the volume of the receptor that can be searched for ligand docking.
Grid box was set on the centriod of all the residues in the Cx43 and no
constraints were selected. The Standard Precision (SP) mode of Glide
(Version 5.5, Schrödinger, LLC, New York, NY 2009) was used for the flex-
ible ligand docking. The best binding conformation was selected based
on the one having the lowest docking energy from the generated dock-
ing solutions. Molecular docking of estradiol-17β into Cx43 generated 32
solutions, in which the best binding conformation having the docking
score of -5.40 kcal/mol. The size of binding site surface area of Cx43 was
864.562 Å, and the size of ligand surface was 271.946 Å. These docking
studies revealed that side chain oxygen atom of Glu42 (E42, Chain A)
makes a hydrogen bond formation with estradiol-17β in the bond length
of 1.790 Å. In addition, Gly2 (G2, Chain B), Asp3 (D3, Chain B), Trp4 (W4,
Chain A and B), Ser5 (S5, Chain A), Ile31 (I31, Chain A), Ile34 (I34, Chain
A), Leu35 (L35, Chain A), Leu37 (L37, Chain A), Gly38 (G38, Chain A) and
Val41 (V41, Chain A) were involved in non-bonded interactions such as
van der Waals forces. It is important to note that estradiol-17β binds with
amino acid residues of N-terminal helix (NTH) and Trans membrane helix
1 (TMH1) of chain A and B alone and it leaves empty space in the pore
region. Thus, it is possible that additional units of estradiol-17β may also
bind with remaining chains to alter the Cx43 gating mechanism.

www.landesbioscience.com Spermatogenesis 109


Figure 3. A molecular modeling study to assess the docked complex of
bisphenol A (BPA) with Cx43-based gap junction communication chan-
nel. (A) Topographic view of the Cx43-gap junction protein complex
with BPA. (B) Side view of the Cx43-gap junction protein complex with
BPA. (C) Ball and stick model for the interaction residues and secondary
structure representation for protein between BPA and Cx43. Molecular
docking of bisphenol A into Cx43 shows formation of strong interac-
tions by making two hydrogen bonds in the complex. The side chain
oxygen atom of Asp3 (D3, Chain A) and backbone nitrogen atoms of
Gly2 (G2, Chain A) were involved in hydrogen bond formation at the
bond distance of 1.587 Å and 1.748 Å, respectively. In addition to these
residues, Gly2 (G2, Chain B and F), Asp3 (D3, Chain B–F), Trp4 (W4,
Chain A and F), Ser5 (S5, Chain F), Ala6 (A6, Chain B, E and F) and Leu7
(L7, Chain B and C) were involved in non-bonded interactions, such as
van der Waals forces. Asp3 (D3) and Trp4 (W4) are the major residues
involved in the interaction in this docked complex. Glide docking score
of the complex was -4.07 kcal/mol. The binding site surface and ligand
surface area of the docked complex were 1494.841 Å and 253.687 Å,
respectively. It is noted that the residues of N-terminal helix (NTH) in
Cx43 alone was involved in the interactions with BPA. The compound
BPA appears to occupy largest binding site surface area in the Cx43
and can block the channel based on the molecular modeling, which is
consistent with recent findings using a functional GJ communication
assay.40

affect TJ-barrier function, illustrating a physiological link exists


between junctions at the BTB and hemidesmosome.

Does the Gap Junction that Provide the Necessary


Cross-Talk between Different Junction Types
at the BTB also Make the BTB More Vulnerable
to Environmental Toxicants?

As briefly discussed above regarding the role of desmosome and


gap junction at the BTB, these two junction types at the BTB
likely provide the signaling function and cross-talk between
different junction types, maintaining the immunological bar-
rier during the epithelial cycle. Furthermore, molecular mod-
eling studies have shown that toxicants (e.g., BPA), estrogens
(e.g., estradiol-17β) and male contraceptives (e.g., adjudin) that
can modulate Sertoli cell BTB function can interact with the
gap junction communicating channel with putative docking
domains (see Figs. 2–4). These interactions can possibly lead
to the blockage of Cx43-based gap junction-based communica-
tion, as well as the penetration of the BTB by utilizing the gap
junction communication junction as a “gateway” and gaining
entry into the apical compartment (Figs. 2–4). Indeed, BPA
was recently shown to perturb gap junction communication
based on a functional dye-transfer assay.40 Based on these find-
importantly, a knockdown of β1-integrin in Sertoli cell epithe- ings and those discussed above,1,18,49 we herein propose a hypo-
lium (without any residual elongating/elongated spermatids and thetical model (see Fig. 5) summarizing the molecular basis
thus no apical ES was present) not only perturbed the hemides- of BTB restructuring during spermatogenesis. In brief, during
mosome function, but also led to a disruption of the Sertoli cell stage VIII of the epithelial cycle, “new” TJ-fibrils are being
TJ-barrier function. This effect resulted from the induction of established behind the preleptotene spermatocytes in transit at
occludin endocytosis and its redistribution from cell surface to the BTB via testosterone-induced de novo protein synthesis,50-53
cell cytosol, thereby disrupting cell adhesion function at the as well as testosterone- or cytokine-mediated transcytosis and
BTB.48 It remains to be determined the involvement of polarity recycling of integral membrane protein complexes from the
proteins in this event. These findings thus demonstrate that a “old” BTB site above the transiting spermatocytes.54-56 Once
disruption of β1-integrin function at the hemidesmosome would the “new” BTB is established, the “old” BTB site undergoes

110 Spermatogenesis Volume 1 Issue 2


Figure 4. A molecular modeling study to assess the docked complex of
adjudin with Cx43-based gap junction communication channel.
(A) Topographic view of the Cx43-gap junction protein complex with
adjudin. (B) Side view of the Cx43-gap junction protein complex with
adjudin. (C) Ball and stick model for the interaction residues and sec-
ondary structure representation for protein between adjudin and Cx43.
Docking of adjudin into Cx43 shows strong interactions in the pore lin-
ing residues of N-terminal helix (NTH). The amino acid residues such as
Gly2 (G2, Chain B) and Asp3 (D3, Chain A and B) were involved in multi-
ple hydrogen bond formation with adjudin (C). The backbone nitrogen
atom of Gly2 (G2, Chain B) and side chain nitrogen atom of Asp3 (D3,
Chain B) were shown to have hydrogen bond interaction with adjudin
in the bond distances of 2.305 Å and 1.940 Å, respectively. Whereas the
side chain oxygen of Asp3 (D3, Chain A) and backbone nitrogen atom
of Asp3 (D3, Chain A) were also involved in hydrogen bond formation
in the bond distance of 1.703 Å and 2.227 Å, respectively. In addition
to hydrogen bonds, Gly2 (G2, Chain A, C, D and F), Asp3 (D3, Chain C),
Trp3 (W3, Chain A and B), Ser5 (S5, Chain A) and Ala6 (A6, Chain A) were
involved in non-bonded interaction to further stabilize the interaction.
The docking score of this binding complex was -4.47 kcal/mol. The size
of surface area for binding site and ligand were 978.172 Å and 290.038
Å, respectively. This docking result has shown that adjudin binds with
pore lining N-terminal helix residues of chain (A–D) and (F) except chain
(E). Hence, it has the possibility that adjudin can block the gating of
this channel protein. From these docking studies, we have observed
that large size of both binding site surface in BPA and ligand surface in
adjudin makes very strong binding into N-terminal helix of Cx43. On the
other hand, docking of estradiol-17β reveals very little docking energy
when compared to docking of other two compounds, which indicates
estradiol-17β can make very stable binding into Cx43-based GJ com-
munication channel. It can also be noted that both NTH and TMH1
amino residues are involved in the binding pocket for estradiol-17β
interactions. Our docking results disclose that each of the compounds
is forming at least one hydrogen bond with the binding site residues in
the Cx43-based communication channel. As a whole we concluded that
all three compounds can regulate the function of Cx43 by binding with
pore lining residues of this protein.

complete degeneration induced by cytokines (e.g., TNFα,


TGFβ3, IL-1α).54-57 Furthermore, TJ, basal ES, desmosome and
gap junction proteins expressed by germ cells (e.g., preleptotene
spermatocytes) during their transit at the BTB can also form
inter-locking interactions with the corresponding proteins in
Sertoli cells, even “loosely” associated as recently proposed.58
This thus avoids the formation of a “disrupted” or an “open”
configuration at the BTB to elicit an unwanted immunologi-
cal response in the host immune system. In short, these events,
as discussed in above sections and recently published,35,36,40 are
regulated and coordinated by gap junctions and desmosomes
at the BTB, so that different junction types can be “disrupted”
and “re-established” orderly and perhaps in sequence, illustrat-
ing the physiological significance of gap junctions and desmo-
somes at the BTB.
Recent studies using a bisphenol A (BPA) model (note: BPA
is an estrogenic environmental toxicant that interacts with estro- this BPA-induced Sertoli cell TJ-barrier disruption is mediated
gen receptor in mammalian cells59-61) have shown that the BPA- by a blockade of the gap junction function due to the physical
induced disruption of the Sertoli cell TJ-barrier, both in vitro interaction of BPA with the Cx43-based gap junction channel.
and in vivo in immature rats,62 is likely mediated by a disrup- Using molecular modeling approach, it was found that similar
tion of the gap junction communication channel function as to estradiol-17β (Fig. 2), BPA (Fig. 3) was capable of interacting
demonstrated by a functional dye-transfer assay.40 These find- with Cx43-based gap junction channel via a putative “docking”
ings also provide the first functional proof regarding the role of domain in the Cx43-based connexon. These findings thus pro-
gap junctions at the BTB. They also prompted us to examine if vide a molecular basis that BPA-induced blockage of gap junction

www.landesbioscience.com Spermatogenesis 111


Figure 5. For figure legend, see page 113.

channel-mediated dye transfer40 is likely to be the result of BPA A recent study has shown that rats treated with adjudin at
“docking” in the functional channel, disrupting the necessary 50 mg/kg b.w. (by gavage) displayed a transient BTB disruption
cross-talk maintained by the Cx43-based gap junctions at the (wherein BTB was disrupted by ~6–12 wk, but it was fully recov-
BTB. The molecular docking findings (Figs. 2 and 3) also illus- ered by 20 wk post adjudin treatment), but when adjudin was
trate that BPA can possibly traverse the BTB to enter the apical used at a higher dose at 250 mg/kg b.w. (by gavage), BTB was
compartment via the gap junction channels. disrupted by 2 wk but did not recover up to 30 wk at the end of

112 Spermatogenesis Volume 1 Issue 2


Figure 5 (See opposite page). A schematic illustration of a molecular/biochemical model on the regulation of BTB restructuring during the seminifer-
ous epithelial cycle of spermatogenesis in adult mammalian testes. This model was prepared based on recent findings in studies in the rat testis (see
text). On the left part, an intact and functional BTB is shown in which the BTB is constituted by actin-based TJ-, basal ES- and gap junction-protein
complexes, as well as the intermediate filament-based desmosome. The adhesion function of these protein complexes are maintained by the actin
filament bundles sandwiched in between cisternae of endoplasmic reticulum and the apposing plasma membranes of two adjacent Sertoli cells
provided by the basal ES, and the integrity of the actin filament bundles is maintained by their intriguing interactions with Eps8 and Arp2/3 protein
complex. Intermediate filaments at the desmosome also contribute to the integrity of the BTB, and it is obvious that there are cross-talks between
intermediate filaments and actin-based cytoskeleton to maintain the BTB function. Both protein kinases and polarity proteins are also involved in
maintaining the proper phosphorylation status and the cellular distribution of the adhesion protein complexes at the BTB. During stage VIII of the
epithelial cycle when BTB undergoes restructuring to accommodate the transit of preleptotene spermatocytes (which are interconnected by intercel-
lular bridges as “clones”72-75), “new” TJ-fibrils that create a “new” BTB are being assembled behind the transiting spermatocytes via de novo synthesis
of BTB proteins under the influence of androgen and estrogen. Furthermore, proteins at the “old” BTB site also undergo endocytosis facilitated by
polarity proteins (e.g., 14-3-3, PAR6). These protein trafficking events are perhaps triggered by c-Src- and/or FAK-mediated phosphorylation under the
influence of cytokines (TGFβ3, TNFα, IL-1α) and steroids (e.g., testosterone, estradiol-17β), so that they can be transcytosed and recycled to the “new”
BTB site, while some unwanted endocytosed proteins are targeted to degradation via the endosome- or ubiquitin-mediated pathways. Through this
mechanism, the integrity of the immunological barrier can be maintained because there is no “actual” “opening” or “degeneration” of the BTB at any
time during the transit of preleptotene spermatocytes at the site. Furthermore, TJ, basal ES, desmosome and gap junction integral membrane proteins
are also expressed on the cell surface of spermatocytes, so that they can form interlocking interactions with the corresponding proteins on the Sertoli
cell plasma membrane to maintain “loosely” associated protein complexes during their transit at the BTB.

the experimental period suggesting a permanent BTB damage.14 of spermatogenesis. A loss of the function of these proteins, such
Interestingly, spermatogenesis failed to “re-initiate” in the high- as Cx43, plakophillin-2, desmoglein-2, desmocollin-2, PAR3,
dose treated group while spermatogenesis re-initiates gradually in PAR6, 14-3-3 and Cdc42, even transiently, leads to a disrup-
the low-dose adjudin treated group. Thus, we sought to examine tion of the Sertoli cell TJ-permeability barrier by causing mis-
if adjudin could also interact with the Cx43-based gap junction localization of proteins at the TJ (e.g., occludin, ZO-1) and the
channel via a “docking” domain. As shown in Figure 4, adju- basal ES (e.g., N-cadherin, β-catenin). These effects apparently
din was found to interact with the gap junction channel. These are mediated by an alteration of the endocytic vesicle-mediated
observations are significant since gap junctions at the BTB, protein trafficking events at the site (e.g., endocytosis, recycling),
unlike other blood-tissue barriers, are not sequestered from the which in turn are regulated by the PAR-based polarity proteins
junctional complexes behind the TJ and the AJ and desmosome (e.g., PAR6, 14-3-3, Cdc42). These findings thus prompt us to
plaques. Instead, they are intermingled with other junction types propose a likely molecular regulatory pathway that maintains
and are immediately accessible to toxicants (e.g., BPA) or chemi- the immunological barrier during the transit of preleptotene
cals (e.g., adjudin) when the host, including rodents and humans, spermatocytes at the BTB at stage VIII of the epithelial cycle
is exposed to harmful substances that exert their effects via the as depicted in Figure 5, in which desmosomes, gap junctions
gap junction. This thus renders the BTB (and the testis) more and polarity proteins are working in concert with protein kinases
vulnerable to environmental toxicants than the TJ-permeability (e.g., FAK, c-Src) to establish a “new” BTB behind the tran-
barrier of the microvessels in the interstitium. This apparently siting spermatocytes while an orderly disassembly of the “old”
new concept also opens a possible new window of research to BTB can take place to avoid a disruption or an unwanted “fall
“manage” toxicant-induced male reproductive dysfunction by through” of the immunological barrier.
protecting or sequestering gap junctions and desmosomes from
these toxicants. Acknowledgments
Studies in the authors’ laboratories were supported by grants from
Concluding Remarks and Future Perspectives the National Institutes of Health (R01 HD056034 to C.Y.C.;
R01 HD056034-02-S1 to C.Y.C.; U54 HD029990, Project 5
Herein, we review some unexpected findings based on recent to C.Y.C.; R03 HD061401 to D.D.M.); Hong Kong Research
reports regarding the role of gap junctions and desmosomes that Grants Council (HKU772009 to W.Y.L.; HKU7693/07M
coexist with TJ and basal ES at the BTB, illustrating these junc- to W.M.L.); and the Government of India, Department of
tions provide the necessary cross-talks and serve as the signal- Biotechnology (BT/BI/03/015/2002 to P.P.M.) and Department
ing platform to maintain the homeostasis of different junction of Information Technology (DIT/R&D/BIO/15(9)/2007 to
types in the seminiferous epithelium during the cyclic restruc- P.P.M.).
turing of the BTB throughout the seminiferous epithelial cycle
3. Clermont Y, Perry B. Quantitative study of the cell 6. Weber JE, Russell LD, Wong V, Peterson RN. Three
References
population of the seminiferous tubules in immature dimensional reconstruction of a rat stage V Sertoli cell:
1. Cheng CY, Mruk DD. A local autocrine axis in rats. J Anat 1957; 100:241-67. II. Morphometry of Sertoli-Sertoli and Sertoli-germ
the testes that regulates spermatogenesis. Nature Rev 4. Orth JM. Proliferation of Sertoli cells in fetal and post- cell relationships. Am J Anat 1983; 167:163-79.
Endocrinol 2010; 6:380-95. natal rats: A quantitative autoradiographic study. Anat 7. Wong V, Russell LD. Three-dimensional reconstruc-
2. Toyama Y, Ohkawa M, Oku R, Maekawa M, Yuasa S. Rec 1982; 203:485-92. tion of a rat stage V Sertoli cell: I. Methods, basic
Neonatally administered diethylstilbestrol retards the 5. Berndtson WE, Thompson TL. Changing relationships configuration and dimensions. Am J Anat 1983;
development of the blood-testis barrier in the rat. J between testis size, Sertoli cell number and spermatogen- 167:143-61.
Androl 2001; 22:413-23. esis in Sprague-Dawley rats. J Androl 1990; 11:429-35.

www.landesbioscience.com Spermatogenesis 113


8. Chaudhary J, Sadler-Riggleman I, Ague JM, Skinner 28. Li MWM, Mruk DD, Cheng CY. Gap junctions 48. Yan HHN, Mruk DD, Wong EWP, Lee WM, Cheng
MK. The helix-loop-helix inhibitor of differentiation and blood-tissue barriers. In: Cheng CY, Ed. Biology CY. An autocrine axis in the testis that coordinates
(ID) proteins induce post-mitotic terminally differenti- and Regulation of Blood-Tissue Barriers. Austin, TX: spermiation and blood-testis barrier restructuring dur-
ated Sertoli cells to re-enter the cell cycle and prolifer- Landes Bioscience and Springer Science + Business ing spermatogenesis. Proc Natl Acad Sci USA 2008;
ate. Biol Reprod 2005; 72:1205-17. Media LLC 2011; In press. 105:8950-5.
9. Ahmed EA, Barten-van Rijbroek AD, Kal HB, Sadri- 29. Pointis G, Gilleron J, Carette D, Segretain D. 49. Cheng CY, Wong EWP, Yan HHN, Mruk DD.
Ardekani H, Mizrak SC, van Pelt AM, de Rooij DG. Physiological and physiopathological aspects of con- Regulation of spermatogenesis in the microenviron-
Proliferative activity in vitro and DNA repair indicate nexins and communicating gap junctions in spermato- ment of the seminiferous epithelium: New insights and
that adult mouse and human Sertoli cells are not termi- genesis. Philos Trans R Soc Lond B Biol Sci 2010; advances. Mol Cell Endocrinol 2010; 315:49-56.
nally differentiated, quiescent cells. Biol Reprod 2009; 365:1607-20. 50. Wang RS, Decrock E, De Vuyst E, Ponsaerts R,
80:1084-91. 30. Vinken M, Decrock E, De Vuyst E, Ponsaerts R, D’hondt C, Bultynck G, et al. Androgen receptor in
10. Chui K, Trivedi A, Cheng CY, Cherbavaz DB, Dazin D’hondt C, Bultynck G, et al. Connexins: sensors and Sertoli cell is essential for germ cell nursery and junc-
PF, Huynh AL, et al. Characterization and functional- regulators of cell cycling. Biochim Biophys Acta 2011; tion complex formation in mouse testes. Endocrinology
ity of proliferative human Sertoli cells. Cell Transplant 1815:13-25. 2006; 147:5624-33.
2011; In press; PMID: 21054948. 31. Simpson I, Rose B, Loewenstein WR. Size limit of mol- 51. Chung NPY, Cheng CY. Is cadmium chloride-induced
11. Wong CH, Cheng CY. The blood-testis barrier: Its ecules permeaeting the junctional membrane channels. inter-Sertoli tight junction permeability barrier disrup-
biology, regulation and physiological role in spermato- Science 1977; 195:294-6. tion a suitable in vitro model to study the events of
genesis. Curr Topics Dev Biol 2005; 71:263-96. 32. Loewenstein WR. Junctional intercellular commun- junction disassembly during spermatogenesis in the rat
12. O’Donnell L, Nicholls PK, O’Bryan MK, McLachlan ciation: the cell-to-cell membrane channel. Physiol Rev testis? Endocrinology 2001; 142:1878-88.
RI, Stanton PG. Spermiation: the process of sperm 1981; 61:829-913. 52. Meng J, Holdcraft RW, Shima JE, Griswold MD,
release. Spermatogenesis 2011; 1:14-35. 33. Maeda S, Tsukihara T. Structure of the gap junction Braun RE. Androgens regulate the permeability of the
13. Cheng CY, Mruk DD. Regulation of spermiogenesis, channel and its implications for its biological functions. blood-testis barrier. Proc Natl Acad Sci USA 2005;
spermiation and blood-testis barrier dynamics: novel Cell Mol Life Sci 2011; 68:1115-29. 102:16696-70.
insights from studies on Eps8 and Arp3. Biochem J 34. Harris AL. Connexin channel permeability to cytoplas- 53. Kaitu’u-Lino TJ, Sluka P, Foo CF, Stanton PG.
2011; 435:553-62. mic molecules. Prog Biophys Mol Biol 2007; 94:120-43. Claudin-11 expression and localisation is regulated by
14. Mok KW, Mruk DD, Lee WM, Cheng CY. 35. Lie PPY, Cheng CY, Mruk DD. Crosstalk between des- androgens in rat Sertoli cells in vitro. Reproduction
Spermatogonial stem cells alone are not sufficient to moglein-2/desmocollin-2/Src kinase and coxsackie and 2007; 133:1169-79.
re-initiate spermatogenesis in the rat testis follow- adenovirus receptor/ZO-1 protein complexes, regulates 54. Yan HHN, Mruk DD, Lee WM, Cheng CY. Blood-
ing adjudin-induced infertility. Int J Androl 2011; blood-testis barrier dynamics. Int J Biochem Cell Biol testis barrier dynamics are regulated by testosterone and
DOI:10.1111/j.1365-2605.2011.01183.x. 2010; 42:975-86. cytokines via their differential effects on the kinetics
15. Easton AS. Regulation of permeability across the 35a. Mirza M, Petersen C, Nordqvist K, Sollerbrandt K. of protein endocytosis and recycling in Sertoli cells.
blood-brain barrier. In: Cheng CY, Ed. Biology and Coxsackievirus and adenovirus receptor is up-regulated FASEB J 2008; 22:1945-59.
Regulation of Blood-Tissue Barriers. Austin, TX: in migratory germ cells during passage of the blood- 55. Xia W, Wong EWP, Mruk DD, Cheng CY. TGFβ3
Landes Bioscience and Springer Science Business testis barrier. Endocrinology 2007; 148:5459-69. and TNFα perturb blood-testis barrier (BTB) dynam-
Media LLC 2011; In press. 36. Li MWM, Mruk DD, Lee WM, Cheng CY. Connexin ics by accelerating the clathrin-mediated endocytosis
16. Hosoya KI, Tachikawa M. The inner blood-retinal 43 and plakophilin-2 as a protein complex that regu- of integral membrane proteins: A new concept of BTB
barrier: Molecular structure and transport biology. lates blood-testis barrier dynamics. Proc Natl Acad Sci regulation during spermatogenesis. Dev Biol 2009;
In: Cheng CY, Ed. Biology and Regulation of Blood- USA 2009; 106:10213-8. 327:48-61.
Tissue Barriers. Austin, TX: Landes Bioscience and 37. Wang CQF, Mruk DD, Lee WM, Cheng CY. 56. Su L, Mruk DD, Lee WM, Cheng CY. Differential
Springer Science+Business Media LLC 2011; In press. Coxsackie and adenovirus receptor (CAR) is a product effects of testosterone and TGFβ3 on endocytic vesicle-
17. Lie PPY, Cheng CY, Mruk DD. The biology of the of Sertoli and germ cells in rat testes which is localized mediated protein trafficking events at the blood-testis
desmosome-like junction: A versatile anchoring junc- at the Sertoli-Sertoli and Sertoli-germ cell interface. barrier. Exp Cell Res 2010; 316:2945-60.
tion and signal transducer in the seminiferous epithe- Exp Cell Res 2007; 313:1373-92. 57. Lie PPY, Cheng CY, Mruk DD. Interleukin-1α is a
lium. Int Rev Cell Mol Biol 2011; 286:223-69. 38. Lee NPY, Cheng CY. Protein kinases and adherens regulator of the blood-testis barrier. FASEB J 2011;
18. Cheng CY, Mruk DD. An intracellular trafficking junction dynamics in the seminiferous epithelium of 25:1244-53.
pathway in the seminiferous epithelium regulating the rat testis. J Cell Physiol 2005; 202:344-60. 58. Wang CQF, Cheng CY. A seamless trespass: germ cell
spermatogenesis: a biochemical and molecular perspec- 39. Goodenough DA, Paul DL. Gap junctions. Cold migration across the seminiferous epithelium during
tive. Crit Rev Biochem Mol Biol 2009; 44:245-63. Spring Harbor Perspect Biol 2009; 1:2576. spermatogenesis. J Cell Biol 2007; 178:549-56.
19. Russell LD. Desmosome-like junctions between Sertoli 40. Li MWM, Mruk DD, Lee WM, Cheng CY. Connexin 59. Hunt PA, Susiarjo M, Rubio C, Hassold TJ. The
and germ cells in the rat testis. Am J Anat 1977; 43 is critical to maintain the homeostasis of blood-testis bisphenol A experience: A primer for the analysis of
148:301-12. barrier via its effects on tight junction reassembly. Proc environmental effects on mammalian reproduction.
20. Vogl A, Vaid K, Guttman J. The Sertoli cell cytoskel- Natl Acad Sci USA 2010; 107:17998-8003. Biol Reprod 2009; 81:807-13.
eton. In: Cheng CY, Ed. Molecular Mechanisms in 41. Assemat E, Bazellieres E, Pallesi-Pocachard E, Le Bivic 60. Wong EWP, Cheng CY. Impacts of environmental
Spermatogenesis. Austin, TX: Landes Bioscience and A, Massey-Harroche D. Polarity complex proteins. toxicants on male reproductive dysfunction. Trends
Springer Science + Business Media LLC 2008; 186-211. Biochim Biophys Acta 2008; 1778:614-30. Pharmacol Sci 2011; 32:290-9. DOI: 10.1016/j.
21. Enders G. Sertoli-Sertoli and Sertoli-germ cell commu- 42. Iden S, Collard JG. Crosstalk between small GTPases tips.2011.01.001.
nications. In: Russell L, Griswold M, Eds. The Sertoli and polarity proteins in cell polarization. Nature Rev 61. Cheng CY, Wong EWP, Lie PPY, Li MWM, Su L, Siu
Cell. St. Louis, MO; Cache River Press 1990; 447-60. Mol Cell Biol 2008; 9:846-59. ER, et al. Environmental toxicants and male reproduc-
22. Thomason HA, Scothern A, McHarg S, Garrod DR. 43. Goldstein B, Macara IG. The PAR proteins: tive function. Spermatogenesis 2011; 1:2-13.
Desmosomes: adhesive strength and signalling in health Fundamental players in animal cell polarization. Dev 62. Li MWM, Mruk DD, Lee WM, Cheng CY. Disruption
and disease. Biochem J 2010; 429:419-33. Cell 2007; 13:609-22. of the blood-testis barrier integrity by bisphenol A
23. Delva E, Tucker DK, Kowalczyk AP. The desmosome. 44. Wong EWP, Cheng CY. Polarity proteins and cell-cell in vitro: Is this a suitable model for studying blood-
Cold Spring Harb Pespect Biol 2009; 1:2543. interactions in the testis. Int Rev Cell Mol Biol 2009; testis barrier dynamics? Int J Biochem Cell Biol 2009;
24. Green KJ, Getsios S, Troyanovsky S, Godsel LM. 278:309-53. 41:2302-14.
Intercellular junction assembly, dynamics and homeo- 45. Wong EWP, Mruk DD, Lee WM, Cheng CY. Par3/ 63. Dym M. Basement membrane regulation of Sertoli
stasis. Cold Spring Harb Perspect Biol 2010; 2:125. Par6 polarity complex coordinates apical ectoplasmic cells. Endocr Rev 1994; 15:102-15.
25. Mruk DD, Cheng CY. Desmosomes in the testis: specialization and blood-testis barrier restructuring 64. Siu MKY, Cheng CY. Dynamic cross-talk between cells
Moving into an unchartered territory. Spermatogenesis during spermatogenesis. Proc Natl Acad Sci USA 2008; and the extracellular matrix in the testis. BioEssays
2011; 1:47-51. 105:9657-62. 2004; 26:978-92.
26. McCrea PD, Gu D. The catenin family at a glance. J 46. Wong EWP, Sun S, Li MWM, Lee WM, Cheng CY. 65. Hirokawa T, Boon-Chieng S, Mitaku S. SOSUI: clas-
Cell Sci 2010; 123:637-42. 14-3-3 protein regulates cell adhesion in the seminif- sification and secondary structure prediction system for
27. Russell LD, Peterson RN. Sertoli cell junctions: mor- erous epithelium of rat testes. Endocrinology 2009; membrane proteins. Bioinformatics 1998; 14:378-9.
phological and functional correlates. Int Rev Cytol 150:4713-23. 66. Maeda S, Nakagawa S, Suga M, Yamashita E, Oshima
1985; 94:177-211. 47. Wong EWP, Mruk DD, Lee WM, Cheng CY. A, Fujiyoshi Y, Tsukihara T. Structure of the Cx26 gap
Regulation of blood-testis barrier dynamics by TGFβ3 junction channel at 3.5 Å resolution. Nature 2009;
is a Cdc42-dependent protein trafficking event. Proc 458:597-602.
Natl Acad Sci USA 2010; 107:11399-404.

114 Spermatogenesis Volume 1 Issue 2


67. Consortium U. The Universal Protein Resource
(UniProt). Nucleic Acid Res 2009; 37:169-74.
68. Berman HM, et al. The Protein Data Bank. Nucleic
Acid Res 2000; 28:235-42.
69. Fiser A, Sali A. Modeller: generation and refinement
of homology-based protein structure models. Methods
Enzymol 2003; 374:461-91.
70. Eswar N, Webb B, Marti-Renom MA, Madhusudhan
MS, Eramian D, Shen MY, et al. Comparative protein
structure modeling with MODELLER. Curr Protoc
Bioinformatics 2006; 15:1-30.
71. Roman A, Rullmannn JA, MacArthur MW, Kaptein
R, Thornton JM. AQUA and PROCHECK-NMR:
Programs for checking the quality of protein structures
solved by NMR. J Biomol NMR 1996; 8:477-86.
72. Miething A. Local desynchronization of cellular devel-
opment within mammalian male germ cell clones. Ann
Anat 2010; 192:247-50.
73. Hamer G, Roepers-Gajadien HL, Gademan IS, Kal HB,
De Rooij DG. Intercellular bridges and apoptosis in
clones of male germ cells. Int J Androl 2003; 26:348-53.
74. Ren HP, Russell LD. Clonal development of intercon-
nected germ cells in the rat and its relationship to the
segmental and subsegmental organization of spermato-
genesis. Am J Anat 1991; 192:121-8.
75. Russell LD, Malone JP, MacCurdy DS. Effect of the
microtubule disrupting agents, colchicine and vinblas-
tine, on seminiferous tubule structure in the rat. Tissue
Cell 1981; 13:349-67.

www.landesbioscience.com Spermatogenesis 115

You might also like