Open access

More Than a Simple Lock and Key Mechanism: Unraveling the Intricacies of Sperm-Zona Pellucida Binding

Written By

Kate A. Redgrove, R. John Aitken and Brett Nixon

Submitted: 27 November 2011 Published: 19 September 2012

DOI: 10.5772/50499

From the Edited Volume

Binding Protein

Edited by Kotb Abdelmohsen

Chapter metrics overview

4,035 Chapter Downloads

View Full Metrics

1. Introduction

Mammalian fertilization involves a concerted interplay between the male and female gametes that ultimately results in the creation of new life. However, despite the fundamental importance of gamete interaction, the precise molecular mechanisms that underpin and regulate this complex event remain to be fully elucidated. Such knowledge is crucial in our attempts to resolve the global problems of population control and infertility. The current world population has surpassed 7 billion people, and continues to grow at a rate of approximately 200 000 each day (UN, 2009). Alarmingly, the majority of this population growth is occurring in developing nations, and is driven in part by an unmet need for effective and accessible contraceptive technologies. Indeed, a recent study by the Global Health Council revealed that of the 205 million pregnancies recorded worldwide each year, 60-80 million of these are deemed to be unplanned or unwanted (Guttmacher, 2007). These concerning statistics highlight the inadequacies of our current armory of contraceptives and demonstrate the need for the development of novel methods for fertility control. By virtue of its specificity and its ability to be suppressed in both males and females, sperm interaction with the outer vestments of the oocyte, a structure known as the zona pellucida (ZP), represents an attractive target for the development of novel contraceptives. However, the realization of such technologies is predicated on a thorough understanding of the molecular mechanisms that underpin this intricate binding event.

Such knowledge will also contribute to the development of novel diagnostic and therapeutic strategies for the paradoxical increase in male infertility that is being experienced by Western countries. Indeed, male infertility has become a distressingly common condition affecting at least 1 in 20 men of reproductive age (McLachlan and de Kretser, 2001). In a vast majority (>80%) of infertile patients sufficient numbers of spermatozoa are produced to achieve fertilization, however the functionality of these cells has become compromised, making defective sperm function the largest single defined cause of human infertility (Hull, et al., 1985, Ombelet, et al., 1997). Biologically, a major cause of impaired sperm function is a failure of these cells to recognize the surface of the egg. Defective sperm- zona pellucida interactions is thus a major cause of fertilization failure in vitro and bioassays of sperm- zona pellucida interaction are able to predict male infertility in vivo with great accuracy (Arslan, et al., 2006).

In this review we explore our current understanding of the mechanisms that are responsible for sperm- zona pellucida interactions. Consideration is given to well-established paradigms of receptor-ligand binding with an emphasis on the emerging evidence for models involving the participation of multimeric receptor complexes and the maturation events that promote their assembly.

Advertisement

2. Sperm-zona pellucida interactions

2.1. The mammalian zona pellucida

The zona pellucida (ZP) is a porous extracellular matrix that surrounds the oocyte (Dunbar, et al., 1994, Wassarman and Litscher, 2008). In the most widely accepted models of gamete interaction, the zona pellucida plays a critical role in tethering spermatozoa, and inducing the release of their acrosomal contents (Bleil and Wassarman, 1983). Binding to the zona pellucida is a highly selective and carefully regulated process that serves as an inter-species barrier to fertilization by preventing adherence of non-homologous sperm to eggs (Hardy and Garbers, 1994).

Although all mammalian eggs are enclosed in a zona pellucida matrix, it’s thickness (~1-25μm) and protein content (~1-10ng) varies considerably for eggs derived from different species (Wassarman, 1988). In mice, the zona pellucida comprises three major sulfated glycoproteins designated ZP1 (200kDa), ZP2 (120kDa) and ZP3 (83kDa). Current evidence suggests that these proteins assemble into a non-covalently linked structure comprising ZP2-ZP3 dimers that polymerize into filaments and are cross-linked by ZP1 (Greve and Wassarman, 1985, Wassarman and Mortillo, 1991). In addition to orthologues of the three mouse zona pellucida proteins [hZP1 (100kDa), hZP2 (75kDa) and hZP3 (55kDa)], the human zona pellucida comprises a fourth glycoprotein, hZP4 (65kDa) (Bauskin, et al., 1999, Lefievre, et al., 2004), which is thought to be dysfunctional in the mouse (Lefievre, et al., 2004). The biological significance of the increased complexity in the zona pellucida of humans awaits further investigation. Given that the mouse remains the most widely studied model for understanding sperm- zona pellucida interaction, this species will serve as the focus for the following discussion.

2.2. The biochemistry of sperm-zona pellucida recognition

Sperm- zona pellucida interaction encompasses a complex sequence of events that relies on each gamete having achieved an appropriate level of maturity. Spermatozoa that approach the oocyte have undergone a behavioral and functional reprogramming event within the female reproductive tract, termed capacitation (see section 2.3.1.3), which ultimately endows the cells with the competence for fertilization. Notwithstanding recent evidence to the balance of evidence favors a model for sperm- zona pellucida interaction that involves three distinct stages: the first comprises primary binding of acrosome-intact spermatozoa to the zona pellucida, this is then followed by secondary binding of acrosome-reacted spermatozoa to the zona pellucida, and finally penetration of the acrosome-reacted sperm through the zona pellucida and into the perivitelline space (Florman and Storey, 1982, Inoue and Wolf, 1975, Saling, et al., 1979, Swenson and Dunbar, 1982).

Figure 1.

Putative models of sperm-zona pellucida binding. (I) The glycan model proposes that sperm binding is initiated via O-linked glycans that are attached at residues Ser332 and Ser334 of ZP3. After fertilization, these residues are deglycosylated thereby preventing further sperm adhesion. (ii) The supramolecular structure model is based on the premise that the physical structure of the matrix formed by the three zona pellucida glycoproteins is critical for the binding of sperm. Following fertilization, ZP2 is processed in such a way that it prevents further sperm adhesion. (iii) The hybrid model incorporates aspects of both the glycan model and the supramolecular model and proposes that O-linked glycosylation is a critical determinant of sperm recognition. However, the key O-glycans reside on residues other than Ser332 and Ser334. Furthermore, the modification of ZP2 that accompanies fertilization renders these O-glycans inaccessible to sperm. (iv) In contrast, the domain-specific model proposes that sperm bind with a variety of N-linked glycans attached to ZP3 and/or the peptide backbone of the glycoprotein depending upon its glycosylation status. The concepts proposed in this figure are adapted from those of Visconti and Florman, 2010 and Clark, 2010.

The initial stages of primary binding involve a relatively loose, non-species specific attachment that serves to tether spermatozoa to the surface of the oocyte (Schmell and Gulyas, 1980, Swenson and Dunbar, 1982). This weak binding is rapidly followed (within 10 minutes) by an irreversible tight binding event (Bleil and Wassarman, 1983, Hartmann, et al., 1972) that resists physical manipulation (Hartmann, et al., 1972, Inoue and Wolf, 1975) and is commonly species-specific. In the mouse, this latter event appears to involve binding of the spermatozoon to ZP3. This model emerged from early experiments performed by Bleil and Wassarman using crudely purified native zona pellucida that demonstrated that mouse ZP3 is responsible for acting as both a primary sperm ligand, preferentially binding the plasma membrane overlying the acrosome of acrosome-intact sperm, as well as an inducer of the acrosome reaction (Bleil and Wassarman, 1980a, Bleil and Wassarman, 1980b, Bleil and Wassarman, 1986, Vazquez, et al., 1989, Yanagimachi, 1994b). Purified mouse ZP3 was also shown to competitively inhibit binding of spermatozoa to homologous eggs in vitro (Bleil and Wassarman, 1980a, Endo, et al., 1987, Florman, et al., 1984, Florman and Wassarman, 1985, Leyton and Saling, 1989). The bioactive component of ZP3 responsible for mediation of sperm binding was initially traced to specific O-linked carbohydrate moieties that decorate the protein (Florman and Wassarman, 1985, Litscher, et al., 1995). In support of this model, complete deglycosylation, or selective removal of O-linked oligosaccharides eliminated the ability of ZP3 to interact with spermatozoa (Florman and Wassarman, 1985). In addition, the O-linked oligosaccharides released by these procedures were able to bind directly to spermatozoa and competitively inhibit their ability to adhere to the zona pellucida (Florman and Wassarman, 1985). Furthermore, genetically engineered chimeric mouse oocytes expressing human ZP3, acquire the same O-linked glycans as mouse ZP3 and bind mouse, rather than human, spermatozoa (Rankin et al., 1996; Hoodbhoy and Dean, 2004). Mutagenesis studies of ZP3 suggested that that the key O-linked adhesion glycans for sperm are attached to either Ser332 and/or Ser334 residues (Chen, et al., 1998) located within the C-terminal portion of the ZP3 polypeptide chain.

Notwithstanding such compelling evidence in favor of this classical model it has increasingly been drawn into question by a number of recent observations from genetically manipulated mouse models. For instance, female mice bearing targeted deletions of key glycosyltransferase enzymes responsible for the addition of O-linked glycans produce oocytes that display normal sperm binding characteristics (Ellies, et al., 1998). Furthermore, in a series of elegant experiments, transgenic mice have been produced in which the putative sperm binding residues were mutated (Ser329, 333, 334→Ala, Ser331→Val, Ser332→Gly) to eliminate potential O-linked glycosylation sites at Ser332 and Ser334 (Gahlay et al. 2010). Females from these transgenic lines were shown to retain their fertility both in vitro and in vivo, and their oocytes maintained the ability to bind the same number of sperm as wild type mice, strongly suggesting that neither Ser332 nor Ser334 are critical for sperm- zona pellucida recognition. The latter findings are perhaps best explained by detailed glycoproteomic analyses that have revealed Ser332 and Ser334 are in fact unlikely to be glycosylated in mouse ZP3 (Boja et al. 2003; Chalabi et al. 2006).

These collective findings have led to the proposal of a number of alternative models of sperm- zona pellucida adhesion (Fig. 1), including the: (i) original glycan model that proposes the importance of O-linked glycosylation at Ser332 and Ser334; (ii) a supramolecular structure model in which the sperm binding domain is formed by the complex of the three major zona pellucida glycoproteins and regulated by the cleavage status of ZP2 (Rankin et al. 2003), (iii) a hybrid model that incorporates elements of both former models by proposing that sperm bind to an O-glycan that is conjugated to ZP3 at a site other than Ser332 or Ser334 (Visconti and Florman, 2010) and that sperm access to this glycan is regulated by the proteolytic cleavage state of ZP2; (iv) domain specific model that envisages a dual adhesion system in which sperm protein(s) interact with the glycans and/or the protein backbone of ZP3 depending on its glycosylation state (Clark, 2011) and (v) a novel model in which gamete recognition is able to be resolved into at least two distinct binding events, the first of which involves adherence to oviductal glycoproteins that are peripherally associated with the egg coat prior to engaging with a ZP3-dependent ligand (Lyng and Shur, 2009) The evidence in support of each of these models of gamete interaction has been reviewed in depth previously (Dean 2004; Clark 2010, 2011; Visconti and Florman, 2010). What is clear from these studies is that the initiation of gamete interaction is not mediated by a simple lock and key mechanism involving a single receptor-ligand interaction. Rather it is likely that sperm engage in multiple binding events with a variety of ligands within the zona pellucida matrix. An advantage of this complex adhesion system is that it would enhance the opportunities of sperm to bind to the oocyte and thus maximize the chance of fertilization. It may also account for the myriad of sperm receptors that have been implicated in this process (see below).

2.3. Sperm receptor molecules involved in zona pellucida interaction

2.3.1. Acquisition of the ability to engage in sperm-zona pellucida interactions

Prior to interaction with the egg, the sperm cell must undergo a complex, multifaceted process of functional maturation (Fig. 2). This process begins in the testes where spermatogonial stem cells are dramatically remodeled during spermatogenesis to produce one of the most highly differentiated and specialized cells in the body, the spermatozoon. After their initial morphological differentiation, these cells are released from the germinal epithelium of the testes in a functionally immature state, incapable of movement or any of the complex array of cellular interactions that are required for fertilization (Hermo, et al., 2010a). In all mammalian species, the acquisition of functional competence occurs progressively during the cells descent through the epididymis, a long convoluted tubule that connects the testis to the vas deferens (Fig. 2B). A remarkable feature of epididymal maturation is that this process is driven entirely by extrinsic factors in the complete absence of nuclear gene transcription and significant protein translation within the spermatozoa (Engel, et al., 1973). The surface and intracellular changes associated with epididymal maturation prepare the spermatozoa for their final phase of maturation within the female reproductive tract, whereby they realize their potential to bind to the zona pellucida and ultimately fertilize the egg (Bailey, 2010, Fraser, 2010, Yanagimachi, 1994a).

2.3.1.1. Spermatogenesis

Spermatogenesis describes the process by which spermatozoa develop from undifferentiated germ cells within the seminiferous tubules of the testis. It is characterized by three functional stages: proliferation, meiosis and metamorphosis. During the proliferation phase, spermatogonial germ cells undergo several mitotic divisions in order to renew themselves in addition to producing spermatocytes (Brinster, 2002, de Rooij, 2001, Dym, 1994, Oatley and Brinster, 2006). These cells then undergo two meiotic divisions to form haploid spermatids. The latter then develop into spermatozoa via an extremely complex process of cytodifferentiation and metamorphosis. This includes structural modifications to the shape of their nucleus, compaction of the nuclear chromatin, formation of an acrosomal vesicle and establishment of a flagellum allowing for the subsequent

Figure 2.

Acquisition of spermatozoa’s ability to engage in interaction with the oocyte. (A) During spermatogenesis, primordial germ cells undergo several phases of mitotic and meiotic divisions in order to produce morphologically mature but functionally incompetent spermatozoa. Of particular importance is the process of spermiogensis, whereby spermatids undergo a process of cytodifferentiation that culminates in the production of spermatozoa. In the course of this dramatic transformation an acrosomal vesicle is formed in the anterior region of the sperm head and a flagellum develops posteriorly. The plasma membrane is also remodeled to produce zona pellucida (ZP) and hyaluronic acid (HA) binding sites. (B) Upon leaving the testis, spermatozoa traverse the epididymis and acquire the ability for forward progressive movement and to adhere to the zona pellucida surrounding the oocyte. These changes occur as a result of the reorganization of specific lipids and proteins. (C) However, it is not until the spermatozoa undergo a final phase of maturation termed ‘capacitation’ in the female reproductive tract that they realize the potential for zona pellucida interaction via the induction of hyperactivated motility along with the sequential loss of decapacitation factors (DFs), formation of membrane rafts and activation of key signaling cascades.

development of motility. The latter series of modifications that produce terminally differentiated spermatozoa from spermatids is referred to as spermiogenesis. Of particular importance to fertilization, is the formation of the acrosome during this stage. As seen by light microscopy, acrosomal development begins with the production of small proacrosome granules derived from the Golgi apparatus that lies adjacent to the early spermatid nucleus. These subsequently fuse together to form the acrosome, a large secretory vesicle that overlies the nucleus (Leblond and Clermont, 1952). There is also evidence to suggest that, in addition to the Golgi apparatus, the plasma membrane of the cell and endocytotic trafficking may also play a fundamental role in the formation of the this exocytotic vesicle (Ramalho-Santos, et al., 2001, West and Willison, 1996). Once formed, the acrosome remains associated with the nucleus of the spermatid, and subsequently the spermatozoa for the remainder of its life, and is of critical importance during fertilization due to its ability to aid in the penetration of the zona pellucida surrounding the ovulated oocyte. This function is, in turn, attributed to the hydrolytic enzymes enclosed within the acrosome. Notwithstanding recent evidence to the contrary, it is widely held that the release of these enzymes occurs upon engagement of sperm binding to the zona pellucida and facilitates localized digestion of the zona matrix, thereby facilitating sperm penetration through this barrier and providing access to the oocyte. The acrosomal enzymes are mostly derived from the lysosome, although several are unique to this organelle (Tulsiani, et al., 1998). In general terms, the acrosome can be divided into compartments, the first of which contains soluble proteins such as didpetididyl peptidase II and cystein-rich secretory protein 2 (Hardy, et al., 1991). The second compartment is known as the acrosomal matrix and contains the insoluble fraction of the enzymes including apexin (Kim, et al., 2001, Noland, et al., 1994, Westbrook-Case, et al., 1994), acrosin and acrosin-binding protein (Baba, et al., 1994b), and sp56, which has been previously implicated in the ability of sperm to interact with the zona pellucida (Buffone, et al., 2008a, Buffone, et al., 2008b).

In addition to the formation of the acrosome during spermiogenesis, the sperm develop a cytoplasmic droplet as well as undergoing plasma membrane remodeling events. The cytoplasmic droplet was first described by Retzius in 1909 as being a portion of germ cell cytoplasm that remains attached to the neck region of elongating spermatids. In most species, the cytoplasmic droplet migrates along the midpiece from the neck to annulus and is transiently retained by spermatozoa as they migrate through the epididymis (Cooper and Yeung, 2003), while in others it remains on the spermatozoa in the epididymis and is not shed until the time of ejaculation (Cooper, 2005, Harayama, et al., 1996, Kaplan, et al., 1984, Larsen, et al., 1980). The precise function of this residual cytoplasm remains elusive although its retention beyond ejaculation is associated with poor sperm function. For example, the cytoplasmic droplet on human spermatozoa is associated with poor sperm motility (Zini, et al., 1998), abnormal head and midpiece morphology (Gergely, et al., 1999, Gomez, et al., 1996, Huszar and Vigue, 1993), lower fertilizing capacity (Keating, et al., 1997) and reduced zona pellucida binding (Ergur, et al., 2002, Huszar, et al., 1994, Liu and Baker, 1992). The mechanism by which these abnormal sperm exhibit reduced function is attributed to disturbed membrane remodeling (Huszar, et al., 1997) and higher extents of lipid peroxidation (Aitken, et al., 1994, Huszar, et al., 1994, Ollero, et al., 2000). The latter is most likely due to the high levels of ROS produced by the cytoplasmic droplet itself (Aitken, et al., 1994, Gil-Guzman, et al., 2001, Gomez, et al., 1996, Huszar and Vigue, 1993, Ollero, et al., 2000), combined with the enriched polyunsaturated fatty acids derived from the membrane of the droplet (Huszar and Vigue, 1993, Ollero, et al., 2000). The plasma membrane remodeling event involves the formation of zona pellucida binding sites via protein transport, which is thought to be mediated by the molecular chaperone, HSPA2. In agreement with the observations discussed above, immature human sperm that fail to express HSPA2 display cytoplasmic retention and reduced zona pellucida binding (Huszar, et al., 2000). The sperm also develop the machinery necessary for functional motility during spermiogenesis. As the acrosome grows at one pole of the nuclear surface of round spermatids, paired centrioles migrate to the opposite pole where they initiate the formation of the flagellum. The flagellum consists of a neck piece, a mid piece, a principle piece and an endpiece (Fawcett, 1975, Katz, 1991). The motility apparatus of the flagellum consists of a central axoneme of nine microtubular doublets arranges to form a cylinder around a central pair of single microtubules (Fawcett, 1975).

In combination, these fundamental changes in structure and biochemisty result in terminally differentiated, highly polarized and morphologically mature spermatozoa. However, despite this level of specialization the spermatozoa that leave the testis are functionally incompetent, as yet unable to move forward progressively, nor interact with the zona pellucida and fertilize the oocyte. They must first traverse the epididymis, a highly convoluted tubule adjacent to the testis, during which time they undergo further biochemical and biophysical changes.

2.3.1.2. Epididymal maturation

Upon leaving the testes, the first region of the epididymis that immature sperm encounter is that of the caput (head). Within this region, the sperm are concentrated by a mechanism of resorption that rapidly removes almost all the testicular fluid/proteins that enter the epididymis. As they leave this environment and enter the corpus (body) epididymis, sperm begin to acquire their motility and fertilizing ability. These attributes continue to develop as the sperm move through the corpus, and reach an optimum level as they reach the cauda (tail) region where they are stored in a quiescent state prior to ejaculation (Fig. 2B) (Cornwall, 2009, Gatti, et al., 2004). Ground breaking research performed in the 1960’s and 1970’s provided the first evidence that the epididymis played an active role for the epididymis in sperm development (Bedford, 1963, Bedford, 1965, Bedford, 1967, Bedford, 1968, Orgebin-Crist, 1967 a, Orgebin-Crist, 1967b, Orgebin-Crist, 1968, Orgebin-Crist, 1969). Most importantly, it was discovered that if sperm were held in the testis via ligation of the epididymal duct, they were unable to develop the ability to fertilize an ovum, and as such their maturation is not an intrinsic property (Cooper and Orgebin-Crist, 1975, Cooper and Orgebin-Crist, 1977).

Consistent with this notion, sperm maturation within the epididymis is not under genomic control, since the cells enter the ductal system in a transcriptionally inactive state with limited biosynthetic capacity (Eddy, 2002). Any subsequent molecular changes must therefore be driven by the dynamic intraluminal milieu in which they are bathed as they transit the length of the epididymal tubule (Cooper, 1986). This epididymal microenvironment is characterized by dramatic sequential changes in its composition, a reflection of segment-specific gene expression (Dube, et al., 2007, Jelinsky, et al., 2007, Jervis and Robaire, 2001, Johnston, et al., 2007) and protein secretion (Dacheux, et al., 2006, Dacheux, et al., 2009, Guyonnet, et al., 2011, Nixon, et al., 2002, Syntin, et al., 1996). The unique physiological compartments established by this activity are thought to have evolved to not only to support the maturation of spermatozoa, but to also to provide protection for the vulnerable cells during their transport and prolonged storage.

It is well established that as sperm descend through the epididymis they acquire the potential for forward motility (reviewed (Amann, et al., 1993, Cooper, 1993, Moore and Akhondi, 1996, Soler, et al., 1994). This progressive motion not only allows the sperm to negotiate the female reproductive tract, but has also been suggested to play a role in penetration of the oocytes outer protective barriers, including the cumulus oophorous and the zone pellucida. To date, the mechanisms underlying the acquisition of forward motility by cauda epididymal sperm have not been completely elucidated. However, a number of potential contributing factors have been identified. On a biochemical level, proteins from caput epididymal sperm contain a greater number of sulfhydryl groups than disulfide bonds. Importantly, the oxidation of these sulfhydryl groups during epididymal transit is correlated with stabilization of flagella, as well as the promotion of protein tyrosine phosphorylation on specific sperm proteins involved in key signaling pathways (Calvin and Bedford, 1971, Cornwall, et al., 1988, Seligman, et al., 2004). Additionally, there is also recent evidence to suggest that sperm isolated from the caput epididymis possess the ability to become motile, but that this activity is suppressed through the action of the cannaboid receptor CNR1, which upon engagement with its ligand, t ennocanaboid 2-arachidonoylglcerol, suppresses the capacity for motility (Cobellis, et al., 2010). Furthermore, changes in the luminal environment, as well as specific post-translational modification to sperm proteins have been shown to affect the motility status of these cells during their transit through the epididymis. In relation to the former, acidification of the luminal contents of the epididymis work to maintain sperm in an immotile state. This is finely regulated by epididymal clear cells which are capable of sensing a rise in luminal pH or bicarbonate concentrations via the sperm specific adenylyl cyclase (SACY)-dependent rise in cyclic-adenosinemonophosphate (cAMP) (Pastor-Soler, et al., 2003, Shum, et al., 2009). In terms of post-translational modifications, proteomic analyses of sperm proteins within the epididymis have identified a number of potential targets affected by changes in expression, disulfide bond status, proteolysis and alterations such as phosphorylation (Baker, et al., 2005). Finally, glycolysis plays an essential role as an energy pathway to fuel forward progressive movement in mouse spermatozoa. This is evidenced by the observation that male mice with genetic ablations of the sperm-specific forms of key glycolytic enzymes (glyceraldehydes 3-phosphate dehydrogenase S or phosphoglycerate kinase 2) are infertile or have very low fertility (Danshina, et al., 2010, Miki, et al., 2004). In part this can be explained by significantly decreased levels of ATP production (4 to 10-times lower than wildtype sperm) resulting in poor, or sluggish motility. Furthermore, the spermatogenic cell-specific type 7 hexokinase that is present in mouse spermatozoa undergoes cleavage of dilsulfide bonds during epididymal transit, resulting in increased hexokinase activity which, in turn, has been causally associated with the initiation of sperm motility (Nakamura, et al., 2008). This indicates that specific structural changes to proteins during epididymal maturation have functional consequences, improving sperm competence for motility, and subsequently their ability to engage in fertilization.

In addition to the maturation of the motility apparatus, the acquisition of zona pellucida binding is also temporally associated with the exposure of spermatozoa to two distinct subsets of macromolecular structures in the epididymal lumen: the first being amorphous chaperone-laden ‘dense bodies’ (Asquith, et al., 2005) and the second being membrane bound prostasome-like particles known as epididymosomes (Saez, et al., 2003). It has been suggested that these epididymal granules facilitate the transfer of proteins to the sperm surface during their transit of the organ (Asquith, et al., 2005, Saez, et al., 2003, Yano, et al., 2010). This is in keeping with the demonstration that biotinylated proteins are able to be transferred between epididymosomes and the sperm surface (Saez, et al., 2003). At present it remains to be determined how this transfer is mediated and the number of cargo proteins that are delivered to the maturing spermatozoa in this manner. Nevertheless, a number of proteins have been shown to be acquired by the sperm during epididymal transit. A non-exhaustive list of these proteins include HE5/CD52 (Kirchhoff and Hale, 1996), members of the ADAM family (Girouard, et al., 2011, Oh, et al., 2009), SPAM1 (Zhang and Martin-Deleon, 2003) and other hyaluronidases (Frenette and Sullivan, 2001, Legare, et al., 1999), macrophage migration inhibitory factor (MIF) (Eickhoff, et al., 2001, Frenette, et al., 2003, Girouard, et al., 2011) as well as a number of enzymes including aldose reductase and sorbitol dehydrogenase (Frenette, et al., 2004, Frenette, et al., 2006, Kobayashi, et al., 2002, Thimon, et al., 2008). Collectively these proteins are believed to participate in the modification of the sperm biochemistry and surface architecture conferring the potential to engage in oocyte interactions.

2.3.1.3. Capacitation

Although spermatozoa acquire the potential to fertilize an egg within the epididymis, the expression of this functional competence is suppressed until their release from this environment at the moment of ejaculation. Indeed they must first spend a period of time within the female reproductive tract (Austin, 1952, Chang, 1951) during which they undergo the final phase of post-testicular maturation, a process known as capacitation. Capacitation is characterized by a series of biochemical and biophysical alterations to the cell including changes in intracellular pH, remodeling of the cell surface architecture, changes in motility patterns and initiation of complex signal transduction pathways. These events have been correlated with a dramatic global up-regulation of tyrosine phosphorylation across a number of key proteins. The ensuing activation of these target proteins has, in turn, been causally linked to the initiation of hyperactivated motility, ability to recognize and adhere to the zona pellucida, and the ability to undergo acrosomal exocytosis (Nixon, et al., 2007). For the purpose of this review, focus will be placed on the molecular mechanisms that culminate in the ability of the sperm to interact with the zona matrix. Furthermore, as this is a cell-surface mediated event, discussion will be centered on the capacitation-associated pathways that mediate sperm surface remodeling.

One of the more widely accepted sequences for mammalian capacitation begins with the loss of surface-inhibitory factors, known as decapacitation factors. These factors mostly originate in the epididymis and accessory organs, and their removal from non-capacitated spermatozoa results in a rapid increase in their fertilizing ability (Fraser, 1984). Furthermore, as capacitation is a reversible process, addition of these decapacitation factors into a population of capacitating spermatozoa potently suppress their ability to recognize and fertilize an oocyte (Fraser, et al., 1990). A number of candidates with potential decapacitation activity have been identified including: DF glycoprotein (Fraser, 1998), phosphatidylethanolamine binding protein 1 (PEB1) (Gibbons, et al., 2005, Nixon, et al., 2006), sperm antigen 36, CRISP1 and plasma membrane fatty acid binding protein (Nixon, et al., 2006) and NYD-SP27 (Bi, et al., 2009). Following the release of these decapacitation factors, spermatozoa experience a dramatic efflux of cholesterol from the plasma membrane (Davis, 1981). This efflux appears to be driven by active sequestration upon exposure of the spermatozoa to an environment rich in appropriate cholesterol sinks (Davis, et al., 1979, Langlais, et al., 1988, Visconti, et al., 1999), and accounts for a striking increase in membrane fluidity. Bovine serum albumin is commonly used within in vitro capacitating media as a cholesterol acceptor, although analogous acceptor(s) are believed to be present within the female reproductive tract. Indeed, studies of human follicular fluid have identified the presence high concentrations of albumin and other cholesterol sinks (Langlais, et al., 1988). Cholesterol efflux from the plasma membrane has also been correlated with an influx of bicarbonate ions (HCO3-) into the cell (Boatman and Robbins, 1991, Chen, et al., 2000, Garty and Salomon, 1987, Okamura, et al., 1985). In addition to its key role in initiation of critical signal transduction cascades, HCO3- has itself been shown to have a more direct role in sperm surface remodeling via stimulation of phospholipid scramblase activity (Gadella and Harrison, 2000, Gadella and Harrison, 2002). The ensuing random translocation of phospholipids between the outer and inner leaflets of the bilayer serves to disrupt the characteristic membrane asymmetry, (Flesch, et al., 2001a). This redistribution of phospholipids has been suggested to prime the sperm plasma membrane for cholesterol efflux, thus rendering the cell more ‘fusogenic’ and responsive to zona pellucida glycoproteins (Harrison and Gadella, 2005).

A further consequence of capacitation-associated cholesterol efflux is the formation of membrane rafts and/or the polarized coalescence of these microdomains and their protein cargo into the anterior region of the sperm head, the precise location that mediate zona pellucida binding (Fig. 3). Membrane rafts are generally defined as small, heterogeneous domains that serve to compartmentalize cellular processes (Pike, 2006), and regulate the distribution of membrane proteins, the activation of receptors and initiation of signaling cascades (Brown and London, 1998, Brown and London, 2000, Simons and Ikonen, 1997, Simons and Toomre, 2000). Membrane rafts are highly stable structures due to the inflexible steroid backbone of cholesterol (Martinez-Seara, et al., 2008) and are therefore extremely resistant to solubilization by a number of non-ionic detergents (Schuck, et al., 2003). As such they are often referred to as detergent-resistant membranes (DRMs). However despite their stability, rafts remain highly dynamic entities and have been observed to display considerable lateral movement in various cell types as a response to physical stimuli or cellular activation events (Simons and Vaz, 2004). In sperm, membrane rafts have been identified by the presence of several somatic cell raft markers including GM1 gangliosides, flotillin and proteins that have raft affinity due to the presence of glycosylphophatidylinositol (GPI) anchors, including CD59 and SPAM1 (Nixon, et al., 2009, Sleight, et al., 2005, van Gestel, et al., 2005). Notably, the spatial distribution of membrane rafts within the sperm membrane is dramatically influenced by the capacitation status of the cells. Indeed, the uniform localization of rafts characteristically observed in non-capacitated spermatozoa is replaced by a pattern of confinement within the peri-acrosomal region of the sperm head following the induction of capacitation (Boerke, et al., 2008, Nixon, et al., 2009, Shadan, et al., 2004). This particularly interesting finding raises the possibility that membrane rafts are of significance in coordinating the functional competence of spermatozoa (Bou Khalil, et al., 2006). In keeping with this notion, recent studies have shown isolated DRMs are capable of binding to the zona pellucida of homologous oocytes with a high degree of affinity and specificity (Bou Khalil, et al., 2006, Nixon, et al., 2009, Nixon, et al., 2011) and that these membrane fractions contain a number of key molecules that have been previously implicated in sperm-zona pellucida interactions (Bou Khalil, et al., 2006, Nixon, et al., 2009, Nixon, et al., 2011, Sleight, et al., 2005). Taken together, such findings encourage speculation that sperm membrane rafts may serve as platforms that act to spatially constrain key zona pellucida recognition molecules and deliver them to their site of action on the anterior region of the sperm head during capacitation (Nixon, et al., 2009, Nixon, et al., 2011). Consistent with this notion, elegant real time tracking studies have demonstrated that cholesterol efflux initiates diffusion (and possibly formation) of novel membrane raft-like structures containing zona-binding molecules over the acrosome of live spermatozoa. Furthermore, following head-to-head agglutination spermatozoa show contact-induced coalescence of GM1 gangliosides suggestive of a specific mechanosensitive response that concentrates important molecules to the appropriate site on the sperm surface to mediate zona binding (Jones, et al., 2010).

In addition to stimulating the loss of cholesterol from the plasma membrane, and promoting aggregation of membrane rafts, the elevation of intracellular HCO3- also activates a unique form of soluble adenylyl cyclase (SACY), which synthesizes cAMP from adenine triphosphate (ATP) (Aitken, et al., 1998, White and Aitken, 1989). Calcium has also been shown to coordinate with bicarbonate to stimulate SACY, although the precise mechanism that underpins this interaction remains to be elucidated (Carlson, et al., 2007, Litvin, et al., 2003). The importance of SACY has been demonstrated by the fact that sperm from Sacy-null male mice display limited motility (Esposito, et al., 2004). Furthermore, inhibition of SACY activity in wildtype mice results in the obstruction of capacitation-associated tyrosine phosphorylation and in vitro fertilization (Hess, et al., 2005). In addition to SACY, intracellular levels of cAMP are also regulated by cAMP phosphodiesterases (PDEs) that degrade cAMP to 5’-AMP (Fig. 3). The initial production of cAMP activates protein kinase A (PKA) through association with the regulatory subunits of the enzyme, promoting dissociation and activation of the catalytic subunits that in turn catalyze the phosphorylation of serine/threonine residues (Urner and Sakkas, 2003). Activation of PKA

Figure 3.

Model of mammalian sperm capacitation. Cholesterol efflux during the early phases of capacitation increases plasma membrane fluidity, facilitating the entry of bicarbonate (HCO3-) and calcium ions (Ca2+) into the sperm cytosol through specific membrane channels. Cholesterol is preferentially lost from non-membrane raft portions of the plasma membrane, and appears to promote a polarized redistribution of membrane rafts to the anterior region of the sperm head. This event may serve to reposition key zona pellucida receptor molecules and enable their surface presentation and / or assembly into functional zona pellucida receptor complexes in this region of the sperm head. There is compelling evidence that such dramatic membrane remodeling events may be augmented by the action of molecular chaperones that are themselves activated during capacitation. This activation appears to be underpinned by a complex signaling cascade involving cross-talk between several pathways. In the most well characterized of these, a sperm specific form of soluble adenylyl cyclase (SACY) is activated by increases in intracellular bicarbonate, calcium and pH, leading to the production of the second messenger cyclic AMP (cAMP). cAMP, in turn, initiates the activation of protein kinase A (PKA), which then simultaneously inhibits the activity of protein tyrosine phosphatases (PTP) and activates protein tyrosine kinases (PTK). This dual regulation results in a global increase in protein tyrosine phosphorylation across a myriad of proteins, including a subset of molecular chaperones, and culminates in the functional activation of the cell. Calcium regulated adenylyl cyclases, phosphodiesterases (PDE), tyrosine kinases and tyrosine phosphatases have also been implicated in various aspects of capacitation associated cell signaling in the spermatozoa of a number of mammalian species.

also results in the induction of tyrosine phosphorylation across a number of substrates, most likely through activation of an intermediary protein tyrosine kinase (PTK) and/or inhibition of protein tyrosine phosphatases (PTP), or both. Of the potential candidates, inhibitory studies have implicated the promiscuous SRC kinase-family of PTKs in driving the increase in phosphotyrosine content (Baker, et al., 2006), especially in human spermatozoa (Lawson, et al., 2008, Mitchell, et al., 2008). However, more recent work has demonstrated that the suppression of capacitation-associated parameters induced by SRC kinase inhibitors is able to be overcome by incubation of sperm in the presence of Ser/Thr phosphatase inhibitors. In addition, sperm from Src-null mice contained similar levels of capacitation-associated tyrosine phosphorylation as wild-type sperm. These data indicate that SRC is not directly involved in capacitation-associated changes in tyrosine phosphorylation in mouse spermatozoa. They also provide evidence that capacitation is regulated by two parallel pathways, one requiring activation of PKA and another involving inactivation of Ser/Thr phosphatases, such as PP2A (Krapf, et al., 2010). Other potential candidates include c-ras which has been identified in human sperm (Naz, et al., 1992a), as well as c-abl which has been studied in both mouse (Baker, et al., 2009) and human models (Naz, 1998). It is important to note, that while the above canonical pathway is the primary pathway thought to induce capacitation, there is evidence to suggest that there is significant cross-talk with other signaling pathways. For instance, it has been demonstrated that a subset of the targets for capacitation associated protein tyrosine phosphorylation are activated by the extracellular signal-regulated kinase (ERK) module of the mitogen-activated protein kinase (MAPK) pathway. Interestingly, inhibition of several elements of this pathway results in suppression of sperm surface phosphotyrosine expression and a concomitant reduction in sperm-zona pellucida interactions (Nixon, et al., 2010).

Irrespective of the mechanisms, capacitation-associated tyrosine phosphorylation has been causally related to the induction of hyperactivated motility, increasing the ability of sperm to bind to the zona pellucida, priming of the cells for acrosomal exocytosis and ultimately enhancing their capacity to fertilize an oocyte (Leclerc, et al., 1997, Sakkas, et al., 2003, Urner and Sakkas, 2003, Visconti, et al., 1995b). The diversity of functions regulated by phosphorylation is consistent with the demonstration that this process occurs in a specific sequence within different compartments of the sperm cell, and is altered again upon binding to the zona pellucida (Sakkas, et al., 2003). In mouse spermatozoa, overt capacitation-associated increases in protein tyrosine phosphorylation have been documented in the flagellum, with principal piece phosphorylation preceding that of the midpiece. Several targets have been identified including aldolase A, NADH dehydrogenase, acrosin binding protein (sp32), proteasome subunit alpha type 6B, and voltage-dependent anion channel 2 among others (Arcelay, et al., 2008). In human spermatozoa however, this increase appears to be restricted to the principal piece, with evidence that both A-kinase anchor protein (AKAP) 3 and AKAP4 are targets (Ficarro, et al., 2003, Sakkas, et al., 2003). The tyrosine phosphorylation of proteins in the sperm flagellum has been causally related to the induction of hyperactivated motility (Mahony and Gwathmey, 1999, Nassar, et al., 1999, Si and Okuno, 1999), a vigorous pattern of motility that is required for spermatozoa to penetrate through the cumulus cell layer and the zona pellucida in order to reach the inner membrane of the oocyte. In addition, to the increased phosphorylation, hyperactivation requires the alkalinization of the sperm and is also calcium-dependent. The calcium required for the induction of hyperactivation can be mobilized into sperm from the external milieu by plasma membrane channel, and can also be released from intracellular stores, including the redundant nuclear envelope located at the base of the sperm flagellum, or the acrosome (Costello, et al., 2009, Herrick, et al., 2005, Ho and Suarez, 2003). Of particular importance in importing calcium into sperm are the CATSPER (cation channel, sperm associated) family of calcium channel proteins, which are sensitive to intracellular alkalinization, and thus are critical for capacitation (Kirichok, et al., 2006, Lobley, et al., 2003, Qi, et al., 2007, Quill, et al., 2001, Ren, et al., 2001). Male mice null for each of the four individual Catsper genes have been shown to be infertile as they are incapable of the hyperactivated motility required for zona pellucida penetration (Carlson, et al., 2005, Jin, et al., 2007, Qi, et al., 2007, Quill, et al., 2001, Ren, et al., 2001).

In addition to the more widely studied phosphorylation of flagellum proteins, capacitation-associated increases in tyrosine phosphorylation have also been reported in an alternate set of proteins located in the sperm head (Asquith, et al., 2004, Flesch, et al., 2001b, Tesarik, et al., 1993, Urner, et al., 2001). Although these proteins represent only a minor proportion of the total pool of phosphorylation substrates in mouse spermatozoa, their importance has been highlighted by the observation that they are expressed on the surface of live, capacitated spermatozoa in a position compatible with a role in mediation of sperm-zona pellucida interactions (Asquith, et al., 2004, Piehler, et al., 2006). Furthermore, these phosphoproteins are present on virtually all sperm that are competent to adhere to the zona pellucida opposed to less than one quarter of sperm in the free swimming population. Although such findings invite speculation that a subset of proteins targeted for phosphotyrosine residues may directly participate sperm-zona pellucida adhesion, this conclusion is at odds with the fact that pre-incubation of sperm with anti-phosphotyrosine antibodies has no discernible effect on their subsequent fertilizing ability (Asquith, et al., 2004). Rather it has been suggested that, following their activation via phosphorylation, these proteins play an indirect role by mediating sperm surface remodeling to render cells competent to engage in zona pellucida adhesion (Fig. 3). In agreement with this proposal, a subset of phosphorylated proteins have been identified in the mouse as the molecular chaperone proteins heat shock protein (HSP) 60 (HSPD1) and endoplasmin (HSP90B1) (Asquith, et al., 2004). Such proteins have well-characterized roles in the folding and trafficking proteins, the assembly of multi-protein structures, and the translocation of proteins across membranes (Nixon, et al., 2005) In addition to mice, a similar cohort of molecular chaperone proteins have also been detected on the surface of sperm from other species including bull (Kamaruddin, et al., 2004), boar (Spinaci, et al., 2005) and human (Miller, et al., 1992, Naaby-Hansen and Herr, 2010), although their phosphorylation status in these species is less clear.

Maturational PhaseChanges contributing to acquisition of zona pellucida binding abilityReferences
SpermatogenesisPrimordial germ cells undergo multiple stages of mitotic and meiotic divisions, followed by a process of cytodifferentiation which results in a highly polarized cell
In early spermatids the Golgi apparatus is transformed into the acrosome
The flagellum is formed to provide sperm with the ability for forward progressive movement
Expression of the molecular chaperone in elongating spermatids is correlated with plasma membrane remodeling that results in the formation of zona pellucida and hyaluronic acid binding sites. These HA binding sites are thought to be responsible for the sperm to penetrate the cumulus cell layer surrounding the oocyte
(Berruti and Paiardi, 2011, Hermo, et al., 2010b, Hermo, et al., 2010c, Huszar, et al., 2007)
Epididymal TransitLipid architecture is remodeled in preparation for the formation of membrane rafts during capacitation
Protein architecture is altered. Existing proteins are unmasked or undergo post-translational modifications, or alternatively novel proteins are integrated into the plasma membrane via epididymosomes and intraluminal fluid
Motility machinery is matured in preparation for acquisition of motility
Upon reaching the cauda epididymis spermatozoa are capable of a sinusoidal movement pattern characterized by a symmetrical tail motion at high frequency and low amplitude
Increase in ability to recognize and interact with zona pellucida
(Cooper, 1986, Cooper and Orgebin-Crist, 1975, Dacheux and Paquignon, 1980, Jones, 1998, Jones, et al., 2007)
CapacitationLoss of specific decapacitation factors (DFs) allows freshly ejaculated spermatozoa to commence capacitation
Cholesterol efflux from the plasma membrane increases membrane fluidity promoting lateral movement of integral proteins, as well as the formation of membrane rafts
Influx of HCO3- activates key signaling cascades whereby SACY stimulates cAMP and in turn PKA. This results in increased tyrosine phosphorylation of specific sperm proteins
In the tail, AKAPs become activated via this phosphorylation and induce a hyperactivated form of motility which allows the sperm to navigate through the oviduct to the site of ovulation.
Key zona pellucida recognition molecules aggregate to the apical region of the sperm head, using membrane rafts as a platform to mediate zona pellucida interaction
(Fraser, 1984, Jones, et al., 2010, Nixon, et al., 2009, Nixon, et al., 2011, Sleight, et al., 2005, Suarez, 2008, Visconti, et al., 1995a)

Table 1.

Summary of specific biochemical- and biophysical-changes that occur during mammalian sperm maturation.

Although the precise role that these surface expressed chaperones play in preparing the sperm for their interaction with the oocyte remains to be established, one possibility is that they promote the presentation and/or assembly of oocyte receptor complex(es) on the sperm surface (Asquith, et al., 2004) (Fig. 3). This notion is supported by the observation that a subset of chaperones have been shown to be the subject of dynamic redistribution during capacitation, leading to their exposure on the anterior region of the sperm head (Asquith, et al., 2005, Dun, et al., 2011). Despite this relocation, a direct role for the chaperones in the mediation of sperm-zona pellucida interactions has been discounted on the basis that anti-chaperone antibodies consistently fail to compromise sperm-zona pellucida adhesion (Asquith, et al., 2005, Dun, et al., 2011, Walsh, et al., 2008). The chaperones do however form stable interactions with a number of putative zona pellucida adhesion molecules which, as discussed below (see Section 2.3.2), appears to indicate that they play an indirect role in gamete interaction. Whether a similar role extends to molecular chaperones in the spermatozoa of other species, such as our own, remains somewhat more controversial. A study by Mitchell et al (2007) failed to localize any of the prominent chaperones to the sperm surface, nor secure evidence for the capacitation-associated phosphorylation of these chaperone proteins (Mitchell, et al., 2007). However, a more recent study by Naaby-Hansen and Herr (2009) demonstrated the expression of seven members from four different chaperone families on the surface of human spermatozoa. They also demonstrated that inhibition of several isoforms of HSPA2 results in decreased fertilization rates in vitro (Naaby-Hansen and Herr, 2010). These studies are supported by earlier work which suggests that the absence of HSPA2 is correlated with decreased ability of sperm to bind to the zona pellucida (Huszar, et al., 2007).

2.3.2. Zona pellucida receptor candidates

Consistent with the apparent complexity of the zona pellucida ligands to which spermatozoa bind, a plethora of candidates have been proposed to act as primary receptors capable of interacting with the carbohydrate moieties and or protein present within the zona pellucida matrix. In most species the list is constantly being refined as new candidates emerge and others are disproven through, for example, the production of knockout models bearing targeted deletions of the putative receptors. Consistent with the notion that primary sperm- zona pellucida interaction involves engagement with specific carbohydrate structures on ZP3, a number of the identified sperm receptors possess lectin-like affinity for specific sugar residues (McLeskey, et al., 1998, Topfer-Petersen, 1999, Wassarman, 1992). In the mouse, the most widely studied model, these receptors include, but are not limited to: β-1,4-galatosyltransferase (GalT1) (Lopez, et al., 1985, Nixon, et al., 2001, Shur and Bennett, 1979, Shur and Hall, 1982a), ZP3R (or sp56) (Bookbinder, et al., 1995, Cheng, et al., 1994, Cohen and Wassarman, 2001), α-D-mannosidase (Cornwall, et al., 1991) and zonadhesin (Gao and Garbers, 1998, Tardif and Cormier, 2011, Topfer-Petersen, et al., 1998) (see Table 1). However, despite the wealth of knowledge accumulated about each of these putative zona pellucida receptors it is now apparent that none are uniquely capable of directing sperm- zona pellucida adhesion. For example, the targeted disruption of GalT1 in knockout mice fails to result in infertility (Lu, et al., 1997). Although sperm from GalT1 null mice bind poorly to ZP3 and fail to undergo a zona-induced acrosome reaction, they retain the ability to bind to the ovulated egg coat in vitro (Lu and Shur, 1997). In a similar vein, a number of zona pellucida binding molecules have been identified in human spermatozoa, including sperm autoantigenic protein 17 (SPA17) (Grizzi, et al., 2003), fucosyltransferase 5 (FUT5) (Chiu, et al., 2003a, Chiu, et al., 2004), and mannose binding receptor (Rosano, et al., 2007). However, further analyses of these receptor molecules have compromised their status as being the single molecule responsible for zona pellucida interaction (see Table 2). In fact prevailing evidence now strongly suggests that no individual receptor is exclusively responsible for regulating gamete interaction. Underscoring the amazing complexity of this interaction, it has instead been proposed to rely on the coordinated action of several zona receptor molecules, which may be assembled into a functional multimeric complex.

Candidate (synonyms)SpeciesEvidenceReferences
Angiotensin-converting enzyme (ACE)Mouse
Rat
Horse
Human
Testis-specific form is found within developing spermatids and mature sperm
ACE KO mice are infertile due to defective transport in the oviducts as well as decreased zona pellucida binding
Play significant role in re-distribution of ADAM3 to the sperm surface
(Esther, et al., 1996, Foresta, et al., 1991, Kohn, et al., 1995, Langford, et al., 1993, Sibony, et al., 1993)
A disintegrin and metalloproteinase (ADAMs)Mouse
Rat
Pig
Human
Family of transmembrane proteins that have varying roles in maturation of spermatozoa
ADAM3 has important role in zona pellucida binding
ADAM2 KO mice show strong suppression of zona pellucida binding and difficulty in moving through female reproductive tract, due to absence of ADAM3 in these mice
ADAM1a KO mice are fertile, but show decreased levels of ADAM3 on the sperm surface
ADAM1b KO mice are fertile
(Kim, et al., 2004, Kim, et al., 2006a, Kim, et al., 2006b, Nishimura, et al., 2004, Nishimura, et al., 2007, Yamaguchi, et al., 2009)
α-D-mannosidase (MAN2B2)Mouse
Rat
Hamster
Human
Integral plasma membrane protein that may facilitate sperm-zona pellucida binding by adhering to mannose-containing zona pellucida oligosaccharides
Pre-incubation of sperm with either D-mannose or anti-MAN2B2 antibody elicits a dose-dependent inhibition of zona pellucida binding
(Cornwall, et al., 1991, Pereira, et al., 1998, Tulsiani, et al., 1993, Tulsiani, et al., 1989, Yoshida-Komiya, et al., 1999),
Arylsulfatase A (AS-A; ARSA)Mouse
Human
Boar
Acquired onto the sperm surface during epididymal transit
Addition of exogenous ARSA, or anti-ARSA antibodies inhibit zona pellucida binding in a dose-dependent manner
ARSA-null males are fertile but fertility decreases with age
(Carmona, et al., 2002, Hess, et al., 1996, Tantibhedhyangkul, et al., 2002, Weerachatyanukul, et al., 2003)
Calmegin (CLGN)/Calnexin/Calspernin (CALR3)MouseCLGN- and CALR3-deficient mice are infertile due to defective sperm migration from uterus into the oviduct, as well as defective zona pellucidabinding
CLGN is required for ADAM1a/ADAM2 dimerization
CALR3 is required for ADAM3 maturation
(Ikawa, et al., 2001, Ikawa, et al., 2011, Yamagata, et al., 2002)
GalT1 (β-1,4-galactosyltransferase; GAlTase; GALT; B4GALT1)Mouse
Rat
Human
Guinea Pig
Rabbit
Bull
Boar
Stallion
Transmembrane protein located on the sperm head overlying the intact acrosome
Transgenic mice overexpressing GalTase are hypersensitive to ZP3 and undergo precocious acrosome reactions
Sperm from mice bearing targeted deletions in GalTase are unable to bind ZP3 or undergo ZP3-dependent acrosomal exocytosis
GalTase-null sperm retain ability to bind to zona pellucida
(Lopez, et al., 1985, Lopez and Shur, 1987, Shi, et al., 2004, Shur and Hall, 1982a, Shur and Hall, 1982b)
Fertization antigen 1 (FA1)Mouse
Human
Bull
Localized to the postacrosomal region of sperm head
Anti-FA-1 antibodies have been implicated in immune infertility in humans
No recorded knockout
(Coonrod, et al., 1994, Menge, et al., 1999, Naz, et al., 1992b, Naz, et al., 1984, Naz and Zhu, 1998)
Fucosyltransferase 5 (FUT5)HumanLocalized to the acrosomal region of the sperm head
Pre-treatment of sperm with antibodies directed against FUT5 inhibits zona pellucida binding
(Chiu, et al., 2003b, Chiu, et al., 2004)
Milk fat globule-EGF factor 8 (MFGE8; p47; SED1)Mouse
Boar
Protein is applied to the sperm acrosome during epididymal transit
Binds specifically to the zona pellucida of unfertilized, but not fertilized eggs
Recombinant MFGE8 and anti-MFGE8 antibodies competitively inhibits zona pellucida binding
MFGE8 null males are subfertile and their sperm are unable to bind to the zona pellucida in vitro
(Ensslin, et al., 1995, Ensslin and Shur, 2003)
Proacrosin (acrosin)Mouse
Boar
Localizes to acrosome and inner acrosomal membrane
Mediates secondary zona pellucida binding via interaction with ZP2
Binding to zona pellucida is non-enzymatic and thought to involve recognition of polysulfate groups on zona pellucida glycoproteins
Acrosin null males are fertile but displaycompromised zona pellucida penetration
(Baba, et al., 1994a, Baba, et al., 1994b, Howes, et al., 2001, Howes and Jones, 2002, Moreno, et al., 1998, Urch and Patel, 1991)
Sperm adhesion molecule 1 (SPAM1; PH-20)All mammalsWidely conserved sperm surface protein
Localized to plasma membrane over anterior region of sperm head
Possesses hyaluronidase activity that aids in the digestion of cumulus cells
Relocalizes to inner acrosomal membrane following acrosome reaction; potentially participates in secondary zona pellucida binding
SPAM1 null males are fertile although their sperm areless efficient in cumulus cell dispersal
(Baba, et al., 2002, Hunnicutt, et al., 1996a, Hunnicutt, et al., 1996b, Lin, et al., 1994, Morales, et al., 2004, Myles and Primakoff, 1997)
Sperm autoantigenic protein 17 (SPA17; SP17)Mouse
Rabbit
Human
Primates
Highly conserved protein localized to the acrosome and fibrous sheath
Has been implicated in regulation of sperm maturation, capacitation, acrosomal exocytosis and zona pellucida binding
Shown to bind to specific mannose components of the zona pellucida
(Chiriva-Internati, et al., 2009, Grizzi, et al., 2003, Yamasaki, et al., 1995)
Spermadhesins (AWN; AQN-1; AQN-3)Boar
Stallion
Bull
Are major components of seminal plasma
May be involved in several sequential steps of fertilization through multifuncational ability to bind to carbohydrates, sulfated glycosaminoglycans, phospholipids and protease inhibitors
(Petrunkina, et al., 2000, Sinowatz, et al., 1995, Topfer-Petersen, et al., 1998)
Sulfogalactosylglycerolipid (SGG)Mouse
Rat
Human
Boar
SGG is a major sperm sulfoglycolipid that putatively facilitates uptake of sulfolipid-immobilizing protein-1 (SLIP1) and ARSA
Following capacitation, SGG is predominantly found in membrane rafts, microdomains that possess zona pellucida affinity
Pre-incubation of sperm with monovalent anti-SGG Fab fragments significantly inhibits zona pellucida binding
(Bou Khalil, et al., 2006, Kornblatt, 1979, Tanphaichitr, et al., 1990, Tanphaichitr, et al., 1993, Weerachatyanukul, et al., 2001, White, et al., 2000)
Zonadhesin (ZAN)Mouse
Hamster
Rabbit
Boar
Bull
Horse
Primates
Localizes to the apical region of the sperm head following spermatogenesis and epididymal maturation
Features a mosaic protein architecture with several domains that potentially enable the protein to participate in multiple cell adhesion processes including zona pellucida binding
Appears to confer species specificity to sperm-zona pellucida adhesion in that sperm from Zan -/- males are able to bind promiscuously to the zona pellucida of non-homologous species
(Bi, et al., 2003, Gasper and Swanson, 2006, Hardy and Garbers, 1994, Hardy and Garbers, 1995, Herlyn and Zischler, 2008, Hickox, et al., 2001, Olson, et al., 2004, Tardif, et al., 2010)
ZP3R (sp56)MouseLocalized to the surface of the sperm head
Pre-incubation of sperm with anti-ZP3R antibodies blocks zona pellucida binding
Pre-treatment of sperm with recombinant ZP3R inhibits fertilization in vivo
EM localizes ZP3R within acrosomal matrix, but the protein appears to undergo a capacitation-associated relocation to the surface of the anterior region of the sperm head
ZP3r -/- males are fertile and their spermatozoa retain their ability to bind zonae of unfertilized eggs and undergo acrosomal exocytosis
(Hardy, et al., 2004, Muro, et al., 2012, Wassarman, 2009)

Table 2.

Putative sperm- zona pellucida receptor candidates

2.4. Toward an integrated model of sperm- zona pellucida interaction

2.4.1. Multimeric protein complexes in zona pellucida binding

Despite decades of research, the specific molecular mechanisms that drive the initial interaction between the male and female gametes remain elusive. As stated previously, a myriad of diverse candidate molecules have been proposed as putative mediators of sperm binding to the zona matrix (Table 1). Regardless of this, prevailing evidence now indicates that none are uniquely responsible for directing or maintaining this interaction (Nixon, et al., 2007). Indeed, the classical model of a simple lock and key mechanism that prevailed in this field of research for several decades has been largely disproven. The fact that spermatozoa contain a multiplicity of zona pellucida receptor candidates allows for a level of functional redundancy commensurate with the overall importance of this fundamental cellular interaction. It also accounts for the succession of both low affinity and high affinity interactions (Thaler and Cardullo, 1996, Thaler and Cardullo, 2002) that characterize gamete interaction. Although the biochemical basis of this multifaceted adhesion process remains obscure, it is unlikely that it could be regulated by the activity of a single receptor. Furthermore, mammalian spermatozoa undergo considerable changes in their already complex surface architecture during epididymal transit and the capacitation process in the female reproductive tract. Prior to these events, the cells are unable to recognize or bind to the zona pellucida. A simple lock and key mechanism involving a constitutively expressed surface receptor does not account for the need to undergo such radical alterations prior to obtaining affinity for the zonae.. Collectively, these data have led to an alternative hypothesis that sperm maturation leads to the surface expression and/or assembly of multimeric complex(es) compromising a multitude of zona pellucida receptors.

The concept that multimeric protein complexes are capable of regulating cell-cell interactions draws on an extensive body of literature. It is well known for instance that the human genome codes for in excess of 500 000 different proteins, of which an estimated 80% function as part of multimeric protein complexes, as opposed to individual proteins (Berggard, et al., 2007). In addition, there are many documented examples of cell-cell adhesion events that require the formation of multimeric protein complexes. As a case in point, β-catenin is well-known to form a complex with several other adhesion proteins, such as cadherin, at sites of cell-cell contact. Interestingly, the formation of these complexes is tightly regulated by phosphorylation and dephosphorylation of the N-terminus of β-catenin (Maher, et al., 2009). Tight junctions have also been shown to rely heavily on the formation of specific protein complexes, comprising transmembrane and membrane-associated proteins (Shen, et al., 2008). Studies with migrating cells, and other cell types that interact in fluid, dynamic environments similar to that in which gametes bind, have illustrated that they most likely rely on the sequential receptor-ligand interactions that are coordinated through the formation of protein adhesion complexes (Sackstein, 2005). In a situation analogous to that recorded in spermatozoa, recent work in cancer cell biology has described the importance of molecular chaperone complexes in increasing the migration and invasiveness of specific cancer types. Breast cancer in particular relies heavily on the action of HSP90α in order to invade other cell types. In this case, HSP90α is excreted by the cancer cell in order to act as a mediator between a complex of co-chaperones outside the cell, including HSP70, HSP40, Hop (HSP70/HSP90 organizing protein) and p23, subsequently activating MMP-2 (matrix metalloproteinase 2) (Eustace, et al., 2004, McCready, et al., 2010, Sims, et al., 2011). MMP-2 then acts to degrade proteins in the extracellular matrix of target cells, thus increasing the invasive ability of the malignant cancer cells (Folgueras, et al., 2004, Jezierska and Motyl, 2009).

The concept of a multimeric zona pellucida receptor complex in spermatozoa was originally proposed by Asquith et al in mouse spermatozoa (Asquith, et al., 2004). This work demonstrated the preferential tyrosine phosphorylation of a specific subset of molecular chaperones during capacitation. A finding that generated considerable interest was that this modification, coincided with the translocation of the chaperones the surface of the sperm head in the precise region that mediates zona pellucida binding. However, the failure of either anti-phosphotyrosine or anti-chaperone antibodies to compromise sperm- zona pellucida interactions led to the proposal that these chaperones may have an indirect role in zona pellucida interaction by virtue of their ability to coordinate the assembly of a zona pellucida receptor complex during capacitation. A key observation in support of this model is that the chaperones, along with numerous putative zona pellucida receptors, partition into lipid microdomains or DRMs (discussed in Section 2.3.1.3). It is proposed that these microdomains may serve as platforms to recruit chaperone clients proteins and/or enhance productive interactions between these two classes of proteins (Nixon, et al., 2009). Indeed, independent evidence indicates that chaperones do form stable protein complexes within membrane rafts during the capacitation of mouse spermatozoa. For instance, Han et al (2011) have recently revealed that the molecular chaperones, HSPA5 and calnexin, associate with a number of client proteins to form a stable supramolecular complex on the surface of mouse spermatozoa. These client proteins include ADAM7 (a disintegrin and metalloprotease 7), a protease that is transferred to the sperm surface via epididymosomes as the cells transit through the epididymis (Oh et al. 2009) and belongs to a family of proteases that have been implicated in sperm migration in the female reproductive tract and adhesion to the zona pellucida (Muro and Okabe 2011) (Cho, et al., 1998, Shamsadin, et al., 1999, Yanagimachi, 2009). Interesting the HSPA5/calnexin/ADAM7 complex resides within DRMs (membrane rafts) and its assembly is promoted by sperm capacitation (Han et al. 2011). In addition, recent work performed by Dun et al (2011) demonstrated the presence of a number of high molecular weight protein complexes expressed on the surface of capacitated mouse sperm utilizing the technique of Blue Native PAGE (Dun, et al., 2011). Of particular interest was the identification of the chaperonin-containing TCP-1 complex (CCT/TRiC) and its ability to form a stable complex with zona pellucida binding protein 2 (ZPBP2). In addition to independent evidence that ZPBP2 participates in zona pellucida binding (Lin, et al., 2007) the CCT/TRiC / ZPBP2 complex was also shown to display affinity for homologous zonae. Importantly, a complex of similar size and compromising the same combination of the CCT/TRiC / ZPBP2 complex was also recently identified via application of the same methodology in human spermatozoa and again shown to participate in zona pellucida interaction in this species (Redgrove, et al., 2011). The conservation of this complex implies that it may be involved in mediation of non-species specific initial interactions, which are relatively weak and forgiving of species barriers. The same may also be true of the 20S proteasome complex that has been shown to display a high level of conservation among the spermatozoa of different species. For instance, the proteasome complex has been described in the spermatozoa of pig, mouse and human and, in each of these species, it has been implicated in zonae interactions (Morales, et al., 2003, Pasten, et al., 2005, Yi, et al., 2010, Zimmerman, et al., 2011). Although this is a constitutively expressed complex, there is evidence that certain subunits of the complex may be subjected to post-translational modifications, including tyrosine phosphorylation, during capacitation (Redgrove et al., 2011). In this context it is noteworthy that the tyrosine phosphorylation of similar proteasome subunits has been shown to influence the substrate specificity of the complex in other cell types (Bose et al., 1999; Castano et al., 1996; Mason et al., 1996; Wehren et al., 1996). Taken together, these findings raise the possibility the proteasome complex may be activated during sperm maturation in preparation for its functional role(s) in sperm–oocyte interactions. These roles appear to extend beyond that of zona pellucida recognition (Zimmerman, et al., 2011) to include regulation of the acrosome reaction in addition to penetration of the zona matrix (Kong, et al., 2009, Sutovsky, et al., 2004), (Morales, et al., 2003).

Interestingly, the indirect role of molecular chaperones in sperm- zona pellucida interactions appears to extend beyond the capacitation-associated remodeling of the sperm surface. Indeed, chaperones such as calmegin, calspernin, calnexin, and HSPA2 have been implicated in additional remodeling events during spermatogenesis and epididymal maturation. With respect to calspernin and calmegin, it has been shown that mice lacking these genes are incapable of binding to the zona pellucida, a defect that is attributable to the role these chaperones play in the maturation of ADAM3 (a protein required for fertilization), as well as the dimerization of an ADAM1 / ADAM2 heterodimer (Ikawa, et al., 2011). In contrast, calnexin has a primary role in retaining unfolded or unassembled N-linked glycoproteins in the ER (Sitia and Braakman, 2003). Importantly however, calnexin has also been shown to be present on the surface of mouse spermatozoa where it partitions into membrane rafts (Nixon, et al., 2009, Stein, et al., 2006). In addition to these lectin-like chaperones, testis-specific HSPA2 has been shown to be essential in several stages of spermatogenesis (Govin, et al., 2006) and, in the human, it has a prominent role in plasma membrane remodeling through the formation of zona pellucida and hyaluronic acid binding sites (Huszar, et al., 2007, Huszar, et al., 2006).

Advertisement

3. Summary

For decades, researchers have strived to find the key molecule on the sperm surface that is responsible for directing its binding to the zona pellucida in a cell and specifies specific manner. However, this premise of a simple lock and key mechanism has been increasingly drawn into question since it fails to account for the myriad of potential receptor molecules that have been identified over the intervening years and the fact that sperm- zona pellucida binding can be resolved into a number of sequential recognition events of varying affinity. Instead, owing largely to the application of elegant genetic manipulation strategies, it is now apparent that the interaction between the two gametes relies on an intricate interplay between a multitude of receptors and their complementary ligands, none of which are uniquely responsible. Such a level of functional redundancy is commensurate with the overall importance that this interaction holds in the initiation of a new life.

An important question that arises from this work is how the activity of such a diverse array of receptors is coordinated to ensure they are presented in the correct sequence to enable productive interactions with the zonae. One possibility is that the zona pellucida binding proteins are organized into functional receptor complexes that are assembled on the anterior region of the sperm head during the different phases of sperm maturation. Such a model may account for the need for the dramatic membrane remodeling events that accompany epididymal maturation and capacitation. Until recently a major challenge to this model has been the lack of direct evidence that sperm harbor multimeric protein complexes on their surface. However, through the application of a variety of novel techniques, independent laboratories have now verified that sperm do express high molecular weight protein complexes on their surface, a subset of which possess affinity for homologous zonae. Furthermore, there is compelling evidence that the assembly and / or surface presentation of these complexes is regulated by the capacitation status of the cells (Dun, et al., 2011, Han, et al., 2010, Morales, et al., 2003, Redgrove, et al., 2011, Sutovsky, et al., 2004).

The conservation of complexes such as the 20S proteasome and CCT/TRiC implies that they are not involved in high-affinity species specific binding to homologous zonae. Rather they may mediate the initial loose tethering of sperm to the zona pellucida and / or downstream events in the fertilization cascade. It is therefore considered likely that the higher affinity, species-specific zona pellucida interactions that follow are executed by additional protein complexes that have been shown to reside in human and mouse spermatozoa (Dun, et al., 2011, Redgrove, et al., 2011) but have yet to be characterised. The proteomic profiling and functional characterization of these additional multiprotein complexes therefore promises to shed new light on the intricacies of sperm-egg interactions.

References

  1. 1. AitkenJ.KrauszC.BuckinghamD.1994Relationships between biochemical markers for residual sperm cytoplasm, reactive oxygen species generation, and the presence of leukocytes and precursor germ cells in human sperm suspensions.Mol Reprod Dev 39(3): 2682790104-0452X
  2. 2. AitkenR. J.HarkissD.KnoxW.PatersonM.IrvineD. S.1998A novel signal transduction cascade in capacitating human spermatozoa characterised by a redox-regulated, cAMP-mediated induction of tyrosine phosphorylation.J Cell Sci 111 ( Pt 5)(6456560021-9533
  3. 3. AmannR. P.HammerstedtR. H.VeeramachaneniD. N.1993The epididymis and sperm maturation: a perspective.Reprod Fertil Dev 5(4): 3613811031-3613
  4. 4. ArcelayE.SalicioniA. M.WertheimerE.ViscontiP. E.2008Identification of proteins undergoing tyrosine phosphorylation during mouse sperm capacitation.Int J Dev Biol 52(5-6): 4634720214-6282
  5. 5. ArslanM.MorshediM.ArslanE. O.TaylorS.KanikA.DuranH. E.OehningerS.2006Predictive value of the hemizona assay for pregnancy outcome in patients undergoing controlled ovarian hyperstimulation with intrauterine inseminationFertil Steril 85(6): 169717071556-5653
  6. 6. AsquithK. L.BaleatoR. M.Mc LaughlinE. A.NixonB.AitkenR. J.2004Tyrosine phosphorylation activates surface chaperones facilitating sperm-zona recognition.J Cell Sci 117(Pt 16): 364536570021-9533
  7. 7. AsquithK. L.HarmanA. J.Mc LaughlinE. A.NixonB.AitkenR. J.2005Localization and significance of molecular chaperones, heat shock protein 1, and tumor rejection antigen gp96 in the male reproductive tract and during capacitation and acrosome reaction.Biol Reprod 72(2): 3283370006-3363
  8. 8. AustinC. R.1952The capacitation of the mammalian sperm.Nature3260028-0836
  9. 9. BabaD.KashiwabaraS.HondaA.YamagataK.WuQ.IkawaM.OkabeM.BabaT.2002Mouse sperm lacking cell surface hyaluronidase PH-20 can pass through the layer of cumulus cells and fertilize the egg.J Biol Chem 277(33): 30310303140021-9258
  10. 10. BabaT.AzumaS.KashiwabaraS.ToyodaY.1994aSperm from mice carrying a targeted mutation of the acrosin gene can penetrate the oocyte zona pellucida and effect fertilization.J Biol Chem 269(50): 31845318490021-9258
  11. 11. BabaT.NiidaY.MichikawaY.KashiwabaraS.KodairaK.TakenakaM.KohnoN.GertonG. L.AraiY.1994bAn acrosomal protein, sp32, in mammalian sperm is a binding protein specific for two proacrosins and an acrosin intermediate.J Biol Chem 269(13): 10133101400021-9258
  12. 12. BaileyJ. L.2010Factors regulating sperm capacitationSyst Biol Reprod Med 56(5): 3343481939-6376
  13. 13. BakerM. A.HetheringtonL.AitkenR. J.2006Identification of SRC as a key PKA-stimulated tyrosine kinase involved in the capacitation-associated hyperactivation of murine spermatozoaJ Cell Sci 119(Pt 15): 318231920021-9533
  14. 14. BakerM. A.HetheringtonL.CurryB.AitkenR. J.2009Phosphorylation and consequent stimulation of the tyrosine kinase c-Abl by PKA in mouse spermatozoa; its implications during capacitationDev Biol 333(1): 57660109-5564X
  15. 15. BakerM. A.WitherdinR.HetheringtonL.Cunningham-SmithK.AitkenR. J.2005Identification of post-translational modifications that occur during sperm maturation using difference in two-dimensional gel electrophoresis.Proteomics100310121615-9853
  16. 16. BauskinA. R.FrankenD. R.EberspaecherU.DonnerP.1999Characterization of human zona pellucida glycoproteins.Mol Hum Reprod 5(6): 5345401360-9947
  17. 17. BedfordJ. M.1963Morphological changes in rabbit spermatozoa during passage through the epididymis.J Reprod Fertil 5(1691770022-4251
  18. 18. BedfordJ. M.1965Changes in fine structure of the rabbit sperm head during passage through the epididymis.J Anat 99(Pt 4): 8919060021-8782
  19. 19. BedfordJ. M.1967Effects of duct ligation on the fertilizing ability of spermatozoa from different regions of the rabbit epididymis.J Exp Zool 166(2): 2712810002-2104X
  20. 20. BedfordJ. M.1968Ultrastructural changes in the sperm head during fertilization in the rabbit.Am J Anat 123(2): 3293580002-9106
  21. 21. BerggardT.LinseS.JamesP.2007Methods for the detection and analysis of protein-protein interactions.Proteomics283328421615-9853
  22. 22. BerrutiG.PaiardiC.2011Acrosome biogenesis: Revisiting old questions to yield new insightsSpermatogenesis95982156-5562
  23. 23. BiM.HickoxJ. R.WinfreyV. P.OlsonG. E.HardyD. M.2003Processing, localization and binding activity of zonadhesin suggest a function in sperm adhesion to the zona pellucida during exocytosis of the acrosome." Biochem J 375(Pt 2): 4774881470-8728
  24. 24. BiY.XuW. M.WongH. Y.ZhuH.ZhouZ. M.ChanH. C.ShaJ. H.2009NYD-SP27, a novel intrinsic decapacitation factor insperm." Asian J Androl 11(2): 2292390100-8682X
  25. 25. BleilJ. D.WassarmanP. M.1980aMammalian sperm-egg interaction: identification of a glycoprotein in mouse egg zonae pellucidae possessing receptor activity for sperm.Cell8738820092-8674
  26. 26. BleilJ. D.WassarmanP. M.1980bStructure and function of the zona pellucida: identification and characterization of the proteins of the mouse oocyte’s zona pellucida.Dev Biol 76(1): 1852020012-1606
  27. 27. BleilJ. D.WassarmanP. M.1983Sperm-egg interactions in the mouse: sequence of events and induction of the acrosome reaction by a zona pellucida glycoprotein.Dev Biol 95(2): 3173240012-1606
  28. 28. BleilJ. D.WassarmanP. M.1986Autoradiographic visualization of the mouse egg’s sperm receptor bound to spermJ Cell Biol 102(4): 136313710021-9525
  29. 29. BoatmanD. E.RobbinsR. S.1991Bicarbonate: carbon-dioxide regulation of sperm capacitation, hyperactivated motility, and acrosome reactions.Biol Reprod 44(5): 8068130006-3363
  30. 30. BoerkeA.TsaiP. S.Garcia-GilN.BrewisI. A.GadellaB. M.2008Capacitation-dependent reorganization of microdomains in the apical sperm head plasma membrane: functional relationship with zona binding and the zona-induced acrosome reactionTheriogenology118811960009-3691X
  31. 31. BojaE. S.HoodbhoyT.FalesH. M.DeanJ.2003Structural characterization of native mouse zona pellucida proteins using mass spectrometry.J Biol Chem 278(36): 34189342020021-9258
  32. 32. BookbinderL. H.ChengA.BleilJ. D.1995Tissue- and species-specific expression of sp56, a mouse sperm fertilization protein.Science 269(5220): 86890036-8075
  33. 33. BoseS.MasonG. G.RivettA. J.1999Phosphorylation of proteasomes in mammalian cells.Mol Biol Rep 26(1-2): 11140301-4851
  34. 34. BouKhalil. M.ChakrabandhuK.XuH.WeerachatyanukulW.BuhrM.BergerT.CarmonaE.VuongN.KumarathasanP.WongP. T.CarrierD.TanphaichitrN.2006Sperm capacitation induces an increase in lipid rafts having zona pellucida binding ability and containing sulfogalactosylglycerolipid.Dev Biol 290(1): 2202350012-1606
  35. 35. BrinsterR. L.2002Germline stem cell transplantation and transgenesis.Science 296(5576): 217421761095-9203
  36. 36. BrownD. A.LondonE.1998Functions of lipid rafts in biological membranes.Annu Rev Cell Dev Biol 14(1111361081-0706
  37. 37. BrownD. A.LondonE.2000Structure and function of sphingolipid- and cholesterol-rich membrane rafts.J Biol Chem 275(23): 17221172240021-9258
  38. 38. BuffoneM. G.FosterJ. A.GertonG. L.2008aThe role of the acrosomal matrix in fertilization.Int J Dev Biol 52(5-6): 5115220214-6282
  39. 39. BuffoneM. G.ZhuangT.OrdT. S.HuiL.MossS. B.GertonG. L.2008bRecombinant mouse sperm ZP3-binding protein (ZP3R/sp56) forms a high order oligomer that binds eggs and inhibits mouse fertilization in vitroJ Biol Chem 283(18): 12438124450021-9258
  40. 40. CalvinH. I.BedfordJ. M.1971Formation of disulphide bonds in the nucleus and accessory structures of mammalian spermatozoa during maturation in the epididymis.J Reprod Fertil Suppl 13(pp. Suppl 1365750449-3087
  41. 41. CarlsonA. E.HilleB.BabcockD. F.2007External Ca2+ acts upstream of adenylyl cyclase SACY in the bicarbonate signaled activation of sperm motility.Dev Biol 312(1): 1831920109-5564X
  42. 42. CarlsonA. E.QuillT. A.WestenbroekR. E.SchuhS. M.HilleB.BabcockD. F.2005Identical phenotypes of CatSper1 and CatSper2 null sperm.J Biol Chem 280(37): 32238322440021-9258
  43. 43. CarmonaE.WeerachatyanukulW.XuH.FluhartyA.AnupriwanA.ShoushtarianA.ChakrabandhuK.TanphaichitrN.2002Binding of arylsulfatase A to mouse sperm inhibits gamete interaction and induces the acrosome reaction.Biol Reprod 66(6): 182018270006-3363
  44. 44. CastanoJ. G.MahilloE.AriztiP.ArribasJ.1996Phosphorylation of C8 and C9 subunits of the multicatalytic proteinase by casein kinase II and identification of the C8 phosphorylation sites by direct mutagenesis.Biochemistry378237890006-2960
  45. 45. ChalabiS.PanicoM.Sutton-SmithM.HaslamS. M.PatankarM. S.LattanzioF. A.MorrisH. R.ClarkG. F.DellA.2006Differential O-glycosylation of a conserved domain expressed in murine and human ZP3.Biochemistry6376470006-2960
  46. 46. ChangM. C.1951Fertilizing capacity of spermatozoa deposited into the fallopian tubes.Nature6976980028-0836
  47. 47. ChenJ.LitscherE. S.WassarmanP. M.1998Inactivation of the mouse sperm receptor, mZP3, by site-directed mutagenesis of individual serine residues located at the combining site for spermProc Natl Acad Sci U S A 95(11): 619361970027-8424
  48. 48. ChenY.CannM. J.LitvinT. N.IourgenkoV.SinclairM. L.LevinL. R.BuckJ.2000Soluble adenylyl cyclase as an evolutionarily conserved bicarbonate sensor.Science 289(5479): 6256280036-8075
  49. 49. ChengA.Le T.PalaciosM.BookbinderL. H.WassarmanP. M.SuzukiF.BleilJ. D.1994Sperm-egg recognition in the mouse: characterization of sp56, a sperm protein having specific affinity for ZP3J Cell Biol 125(4): 8678780021-9525
  50. 50. Chiriva-InternatiM.GaglianoN.DonettiE.CostaF.GrizziF.FranceschiniB.AlbaniE.Levi-SettiP. E.GioiaM.JenkinsM.CobosE.KastW. M.2009Sperm protein 17 is expressed in the sperm fibrous sheathJ Transl Med 7(611479-5876
  51. 51. ChiuP. C.KoistinenR.KoistinenH.SeppalaM.LeeK. F.YeungW. S.2003aBinding of zona binding inhibitory factor-1 (ZIF-1) from human follicular fluid on spermatozoa.J Biol Chem 278(15): 13570135770021-9258
  52. 52. ChiuP. C.KoistinenR.KoistinenH.SeppalaM.LeeK. F.YeungW. S.2003bZona-binding inhibitory factor-1 from human follicular fluid is an isoform of glycodelin.Biol Reprod 69(1): 3653720006-3363
  53. 53. ChiuP. C.TsangH. Y.KoistinenR.KoistinenH.SeppalaM.LeeK. F.YeungW. S.2004The contribution of D-mannose, L-fucose, N-acetylglucosamine, and selectin residues on the binding of glycodelin isoforms to human spermatozoa.Biol Reprod 70(6): 171017190006-3363
  54. 54. ChoC.BunchD. O.FaureJ. E.GouldingE. H.EddyE. M.PrimakoffP.MylesD. G.1998Fertilization defects in sperm from mice lacking fertilin beta.Science 281(5384): 185718590036-8075
  55. 55. ClarkG. F.2010The mammalian zona pellucida: a matrix that mediates both gamete binding and immune recognition?Syst Biol Reprod Med 56(5): 3493641939-6376
  56. 56. ClarkG. F.2011aThe molecular basis of mouse sperm-zona pellucida binding: a still unresolved issue in developmental biology."Reproduction3773811741-7899
  57. 57. ClarkG. F.2011bMolecular models for mouse sperm-oocyte bindingGlycobiology351460-2423
  58. 58. CobellisG.RicciG.CacciolaG.OrlandoP.PetrosinoS.CascioM. G.BisognoT.De PetrocellisL.ChioccarelliT.AltucciL.FasanoS.MeccarielloR.PierantoniR.LedentC.Di MarzoV.2010A gradient of 2-arachidonoylglycerol regulates mouse epididymal sperm cell start-upBiol Reprod 82(2): 4514581529-7268
  59. 59. CohenN.WassarmanP. M.2001Association of egg zona pellucida glycoprotein mZP3 with sperm protein sp56 during fertilization in mice.Int J Dev Biol 45(3): 5695760214-6282
  60. 60. CoonrodS. A.WesthusinM. E.NazR. K.1994Monoclonal antibody to human fertilization antigen-1 (FA-1) inhibits bovine fertilization in vitro: application in immunocontraception.Biol Reprod 51(1): 14230006-3363
  61. 61. CooperT. G.1993The human epididymis--is it necessary?Int J Androl 16(4): 2453000105-6263
  62. 62. CooperT. G.2005Cytoplasmic droplets: the good, the bad or just confusing?Hum Reprod 20(1): 9110268-1161
  63. 63. CooperT. G.Orgebin-CristM. C.1975The effect of epididymal and testicular fluids on the fertilising capacity of testicular and epididymal spermatozoa.Andrologia85930303-4569
  64. 64. CooperT. G.Orgebin-CristM. C.1977Effect of aging on the fertilizing capacity of testicular spermatozoa from the rabbit.Biol Reprod 16(2): 2582660006-3363
  65. 65. CooperT. G.YeungC. H.2003Acquisition of volume regulatory response of sperm upon maturation in the epididymis and the role of the cytoplasmic droplet.Microsc Res Tech 61(1): 28380105-9910X
  66. 66. CornwallG. A.2009New insights into epididymal biology and functionHum Reprod Update 15(2): 2132271460-2369
  67. 67. CornwallG. A.TulsianiD. R.Orgebin-CristM. C.1991Inhibition of the mouse sperm surface alpha-D-mannosidase inhibits sperm-egg binding in vitro.Biol Reprod 44(5): 9139210006-3363
  68. 68. CornwallG. A.VindivichD.TillmanS.ChangT. S.1988The effect of sulfhydryl oxidation on the morphology of immature hamster epididymal spermatozoa induced to acquire motility in vitro.Biol Reprod 39(1): 1411550006-3363
  69. 69. CostelloS.MichelangeliF.NashK.LefievreL.MorrisJ.Machado-OliveiraG.BarrattC.Kirkman-BrownJ.PublicoverS.2009Ca2+-stores in sperm: their identities and functions." Reproduction 138(3): 4254371741-7899
  70. 70. DacheuxJ. L.BelghaziM.LansonY.DacheuxF.2006Human epididymal secretome and proteomeMol Cell Endocrinol 250(1-2): 36420303-7207
  71. 71. DacheuxJ. L.BelleanneeC.JonesR.LabasV.BelghaziM.GuyonnetB.DruartX.GattiJ. L.DacheuxF.2009Mammalian epididymal proteomeMol Cell Endocrinol 306(1-2): 45501872-8057
  72. 72. DacheuxJ. L.PaquignonM.1980Relations between the fertilizing ability, motility and metabolism of epididymal spermatozoa.Reprod Nutr Dev 20(4A): 108510990181-1916
  73. 73. DanshinaP. V.GeyerC. B.DaiQ.GouldingE. H.WillisW. D.KittoG. B.Mc CarreyJ. R.EddyE. M.O’BrienD. A.2010Phosphoglycerate kinase 2 (PGK2) is essential for sperm function and male fertility in miceBiol Reprod 82(1): 1361451529-7268
  74. 74. DavisB. K.1981Timing of fertilization in mammals: sperm cholesterol/phospholipid ratio as a determinant of the capacitation intervalProc Natl Acad Sci U S A 78(12): 756075640027-8424
  75. 75. DavisB. K.ByrneR.HungundB.1979Studies on the mechanism of capacitation. II. Evidence for lipid transfer between plasma membrane of rat sperm and serum albumin during capacitation in vitro.Biochim Biophys Acta 558(3): 2572660006-3002
  76. 76. de RooijD. G.2001Proliferation and differentiation of spermatogonial stem cells.Reproduction 121(3): 3473541470-1626
  77. 77. DeanJ.2004Reassessing the molecular biology of sperm-egg recognition with mouse genetics.Bioessays 26(1): 29380265-9247
  78. 78. DubeE.ChanP. T.HermoL.CyrD. G.2007Gene expression profiling and its relevance to the blood-epididymal barrier in the human epididymisBiol Reprod 76(6): 103410440006-3363
  79. 79. DunM. D.SmithN. D.BakerM. A.LinM.AitkenR. J.NixonB.2011The chaperonin containing TCP1 complex (CCT/TRiC) is involved in mediating sperm-oocyte interactionJ Biol Chem pp. 0108-33511083351X
  80. 80. DunbarB. S.AveryS.LeeV.PrasadS.SchwahnD.SchwoebelE.SkinnerS.WilkinsB.1994The mammalian zona pellucida: its biochemistry, immunochemistry, molecular biology, and developmental expression.Reprod Fertil Dev 6(3): 3313471031-3613
  81. 81. DymM.1994Spermatogonial stem cells of the testis.Proc Natl Acad Sci U S A 91(24): 11287112890027-8424
  82. 82. EddyE. M.2002Male germ cell gene expression.Recent Prog Horm Res 57(1031280079-9963
  83. 83. EickhoffR.WilhelmB.RennebergH.WennemuthG.BacherM.LinderD.BucalaR.SeitzJ.MeinhardtA.2001Purification and characterization of macrophage migration inhibitory factor as a secretory protein from rat epididymis: evidences for alternative release and transfer to spermatozoa.Mol Med 7(1): 27351076-1551
  84. 84. ElliesL. G.TsuboiS.PetryniakB.LoweJ. B.FukudaM.MarthJ. D.1998Core 2 oligosaccharide biosynthesis distinguishes between selectin ligands essential for leukocyte homing and inflammation.Immunity8818901074-7613
  85. 85. EndoY.MatteiP.KopfG. S.SchultzR. M.1987Effects of a phorbol ester on mouse eggs: dissociation of sperm receptor activity from acrosome reaction-inducing activity of the mouse zona pellucida protein, ZP3.Dev Biol 123(2): 5745770012-1606
  86. 86. EngelJ. C.BernardE. A.WassermannG. F.1973Protein synthesis by isolated spermatozoa from cauda and caput epididymis of rat.Acta Physiol Lat Am 23(5): 3583620001-6764
  87. 87. EnsslinM.CalveteJ. J.TholeH. H.SierraltaW. D.AdermannK.SanzL.Topfer-PetersenE.1995Identification by affinity chromatography of boar sperm membrane-associated proteins bound to immobilized porcine zona pellucida. Mapping of the phosphorylethanolamine-binding region of spermadhesin AWN.Biol Chem Hoppe Seyler 376(12): 7337380177-3593
  88. 88. EnsslinM. A.ShurB. D.2003Identification of mouse sperm SED1, a bimotif EGF repeat and discoidin-domain protein involved in sperm-egg binding.Cell4054170092-8674
  89. 89. ErgurA. R.DokrasA.GiraldoJ. L.HabanaA.KovanciE.HuszarG.2002Sperm maturity and treatment choice of in vitro fertilization (IVF) or intracytoplasmic sperm injection: diminished sperm HspA2 chaperone levels predict IVF failure.Fertil Steril 77(5): 9109180015-0282
  90. 90. EspositoG.JaiswalB. S.XieF.Krajnc-FrankenM. A.RobbenT. J.StrikA. M.KuilC.PhilipsenR. L.van DuinM.ContiM.GossenJ. A.2004Mice deficient for soluble adenylyl cyclase are infertile because of a severe sperm-motility defectProc Natl Acad Sci U S A 101(9): 299329980027-8424
  91. 91. EstherC. R.Jr HowardT. E.MarinoE. M.GoddardJ. M.CapecchiM. R.BernsteinK. E.1996Mice lacking angiotensin-converting enzyme have low blood pressure, renal pathology, and reduced male fertility.Lab Invest 74(5): 9539650023-6837
  92. 92. EustaceB. K.SakuraiT.StewartJ. K.YimlamaiD.UngerC.ZehetmeierC.LainB.TorellaC.HenningS. W.BesteG.ScrogginsB. T.NeckersL.IlagL. L.JayD. G.2004Functional proteomic screens reveal an essential extracellular role for hsp90 alpha in cancer cell invasiveness.Nat Cell Biol 6(6): 5075141465-7392
  93. 93. FawcettD. W.1975Gametogenesis in the male: prospects for its control.Symp Soc Dev Biol 33): 2553
  94. 94. FicarroS.ChertihinO.WestbrookV. A.WhiteF.JayesF.KalabP.MartoJ. A.ShabanowitzJ.HerrJ. C.HuntD. F.ViscontiP. E.2003Phosphoproteome analysis of capacitated human sperm. Evidence of tyrosine phosphorylation of a kinase-anchoring protein 3 and valosin-containing protein/p97 during capacitation.J Biol Chem 278(13): 11579115890021-9258
  95. 95. FleschF. M.BrouwersJ. F.NievelsteinP. F.VerkleijA. J.van GoldeL. M.ColenbranderB.GadellaB. M.2001aBicarbonate stimulated phospholipid scrambling induces cholesterol redistribution and enables cholesterol depletion in the sperm plasma membrane.J Cell Sci 114(Pt 19): 354335550021-9533
  96. 96. FleschF. M.WijnandE.van de LestC. H.ColenbranderB.van GoldeL. M.GadellaB. M.2001bCapacitation dependent activation of tyrosine phosphorylation generates two sperm head plasma membrane proteins with high primary binding affinity for the zona pellucida.Mol Reprod Dev 60(1): 1071150104-0452X
  97. 97. FlormanH. M.BechtolK. B.WassarmanP. M.1984Enzymatic dissection of the functions of the mouse egg’s receptor for sperm.Dev Biol 106(1): 2432550012-1606
  98. 98. FlormanH. M.StoreyB. T.1982Mouse gamete interactions: the zona pellucida is the site of the acrosome reaction leading to fertilization in vitro.Dev Biol 91(1): 1211300012-1606
  99. 99. FlormanH. M.WassarmanP. M.1985O-linked oligosaccharides of mouse egg ZP3 account for its sperm receptor activity.Cell3133240092-8674
  100. 100. FolguerasA. R.PendasA. M.SanchezL. M.Lopez-OtinC.2004Matrix metalloproteinases in cancer: from new functions to improved inhibition strategies.Int J Dev Biol 48(5-6): 4114240214-6282
  101. 101. ForestaC.MioniR.RossatoM.VarottoA.ZorziM.1991Evidence for the involvement of sperm angiotensin converting enzyme in fertilization.Int J Androl 14(5): 3333390105-6263
  102. 102. FraserL. R.1984Mouse sperm capacitation in vitro involves loss of a surface-associated inhibitory component.J Reprod Fertil 72(2): 3733840022-4251
  103. 103. FraserL. R.1998Interactions between a decapacitation factor and mouse spermatozoa appear to involve fucose residues and a GPI-anchored receptor.Mol Reprod Dev 51(2): 1932020104-0452X
  104. 104. FraserL. R.2010The "switching on" of mammalian spermatozoa: molecular events involved in promotion and regulation of capacitation." Mol Reprod Dev 77(3): 1972081098-2795
  105. 105. FraserL. R.HarrisonR. A.HerodJ. E.1990Characterization of a decapacitation factor associated with epididymal mouse spermatozoa.J Reprod Fertil 89(1): 1351480022-4251
  106. 106. FrenetteG.LessardC.MadoreE.FortierM. A.SullivanR.2003Aldose reductase and macrophage migration inhibitory factor are associated with epididymosomes and spermatozoa in the bovine epididymis.Biol Reprod 69(5): 158615920006-3363
  107. 107. FrenetteG.LessardC.SullivanR.2004Polyol pathway along the bovine epididymis.Mol Reprod Dev 69(4): 4484560104-0452X
  108. 108. FrenetteG.SullivanR.2001Prostasome-like particles are involved in the transfer of P25b from the bovine epididymal fluid to the sperm surface.Mol Reprod Dev 59(1): 1151210104-0452X
  109. 109. FrenetteG.ThabetM.SullivanR.2006Polyol pathway in human epididymis and semen.J Androl 27(2): 2332390196-3635
  110. 110. GadellaB. M.HarrisonR. A.2000The capacitating agent bicarbonate induces protein kinase A-dependent changes in phospholipid transbilayer behavior in the sperm plasma membrane.Development 127(11): 240724200950-1991
  111. 111. GadellaB. M.HarrisonR. A.2002Capacitation induces cyclic adenosine 3’,5’-monophosphate-dependent, but apoptosis-unrelated, exposure of aminophospholipids at the apical head plasma membrane of boar sperm cells.Biol Reprod 67(1): 3403500006-3363
  112. 112. GahlayG.GauthierL.BaibakovB.EpifanoO.DeanJ.2010Gamete recognition in mice depends on the cleavage status of an egg’s zona pellucida proteinScience2162191095-9203
  113. 113. GaoZ.GarbersD. L.1998Species diversity in the structure of zonadhesin, a sperm-specific membrane protein containing multiple cell adhesion molecule-like domains.J Biol Chem 273(6): 341534210021-9258
  114. 114. GartyN. B.SalomonY.1987Stimulation of partially purified adenylate cyclase from bull sperm by bicarbonate.FEBS Lett 218(1): 1481520014-5793
  115. 115. GasperJ.SwansonW. J.2006Molecular population genetics of the gene encoding the human fertilization protein zonadhesin reveals rapid adaptive evolutionAm J Hum Genet 79(5): 8208300002-9297
  116. 116. GattiJ. L.CastellaS.DacheuxF.EcroydH.MetayerS.ThimonV.DacheuxJ. L.2004Post-testicular sperm environment and fertility.Anim Reprod Sci 82-83(3213390378-4320
  117. 117. GergelyA.KovanciE.SenturkL.CosmiE.VigueL.HuszarG.1999Morphometric assessment of mature and diminished-maturity human spermatozoa: sperm regions that reflect differences in maturity.Hum Reprod 14(8): 200720140268-1161
  118. 118. GibbonsR.Adeoya-OsiguwaS. A.FraserL. R.2005A mouse sperm decapacitation factor receptor is phosphatidylethanolamine-binding protein 1.Reproduction 130(4): 4975081470-1626
  119. 119. Gil-GuzmanE.OlleroM.LopezM. C.SharmaR. K.AlvarezJ. G.ThomasA. J.Jr AgarwalA.2001Differential production of reactive oxygen species by subsets of human spermatozoa at different stages of maturationHum Reprod 16(9): 192219300268-1161
  120. 120. GirouardJ.FrenetteG.SullivanR.2011Comparative proteome and lipid profiles of bovine epididymosomes collected in the intraluminal compartment of the caput and cauda epididymidis.Int J Androl 34(5 Pt 2): e475e4861365-2605
  121. 121. GomezE.BuckinghamD. W.BrindleJ.LanzafameF.IrvineD. S.AitkenR. J.1996Development of an image analysis system to monitor the retention of residual cytoplasm by human spermatozoa: correlation with biochemical markers of the cytoplasmic space, oxidative stress, and sperm function.J Androl 17(3): 2762870196-3635
  122. 122. GovinJ.CaronC.EscoffierE.FerroM.KuhnL.RousseauxS.EddyE. M.GarinJ.KhochbinS.2006Post-meiotic shifts in HSPA2/HSP70.2 chaperone activity during mouse spermatogenesis.J Biol Chem 281(49): 37888378920021-9258
  123. 123. GreveJ. M.WassarmanP. M.1985Mouse egg extracellular coat is a matrix of interconnected filaments possessing a structural repeat.J Mol Biol 181(2): 2532640022-2836
  124. 124. GrizziF.Chiriva-InternatiM.FranceschiniB.HermonatP. L.SodaG.LimS. H.DioguardiN.2003Immunolocalization of sperm protein 17 in human testis and ejaculated spermatozoa.J Histochem Cytochem 51(9): 12451248Print)0022-1554 (Linking).
  125. 125. GuyonnetB.DacheuxF.DacheuxJ. L.GattiJ. L.2011The epididymal transcriptome and proteome provide some insights into new epididymal regulationsJ Androl 32(6): 6516641939-4640
  126. 126. HanC.ParkI.LeeB.JinS.ChoiH.KwonJ. T.KwonY. I.doKimH.ParkZ. Y.ChoC.2010Identification of heat shock protein 5, calnexin and integral membrane protein 2B as Adam7-interacting membrane proteins in mouse sperm.J Cell Physiol 226(5): 118611951097-4652
  127. 127. HarayamaH.ShibukawaT.MiyakeM.KannanY.KatoS.1996Fructose stimulates shedding of cytoplasmic droplets from epididymal boar spermatozoa.Reprod Fertil Dev 8(7): 103910431031-3613
  128. 128. HardyC. M.ClydesdaleG.MobbsK. J.2004Development of mouse-specific contraceptive vaccines: infertility in mice immunized with peptide and polyepitope antigensReproduction 128(4): 3954071470-1626
  129. 129. HardyD. M.GarbersD. L.1994Species-specific binding of sperm proteins to the extracellular matrix (zona pellucida) of the egg.J Biol Chem 269(29): 19000190040021-9258
  130. 130. HardyD. M.GarbersD. L.1995A sperm membrane protein that binds in a species-specific manner to the egg extracellular matrix is homologous to von Willebrand factor.J Biol Chem 270(44): 26025260280021-9258
  131. 131. HardyD. M.OdaM. N.FriendD. S.HuangT. T.Jr 1991A mechanism for differential release of acrosomal enzymes during the acrosome reaction.Biochem J 275 ( Pt 3)(7597660264-6021
  132. 132. HarrisonR. A.GadellaB. M.2005Bicarbonate-induced membrane processing in sperm capacitation.Theriogenology3423510009-3691X
  133. 133. HartmannJ. F.GwatkinR. B.HutchisonC. F.1972Early contact interactions between mammalian gametes in vitro: evidence that the vitellus influences adherence between sperm and zona pellucidaProc Natl Acad Sci U S A 69(10): 276727690027-8424
  134. 134. HerlynH.ZischlerH.2008The molecular evolution of sperm zonadhesin.Int J Dev Biol 52(5-6): 7817900214-6282
  135. 135. HermoL.PelletierR. M.CyrD. G.SmithC. E.2010aSurfing the wave, cycle, life history, and genes/proteins expressed by testicular germ cells. Part 1: background to spermatogenesis, spermatogonia, and spermatocytes.Microsc Res Tech 73(4): 2412781097-0029
  136. 136. HermoL.PelletierR. M.CyrD. G.SmithC. E.2010bSurfing the wave, cycle, life history, and genes/proteins expressed by testicular germ cells. Part 2: changes in spermatid organelles associated with development of spermatozoa." Microsc Res Tech 73(4): 2793191097-0029
  137. 137. HermoL.PelletierR. M.CyrD. G.SmithC. E.2010cSurfing the wave, cycle, life history, and genes/proteins expressed by testicular germ cells. Part 5: intercellular junctions and contacts between germs cells and Sertoli cells and their regulatory interactions, testicular cholesterol, and genes/proteins associated with more than one germ cell generation." Microsc Res Tech 73(4): 4094941097-0029
  138. 138. HerrickS. B.SchweissingerD. L.KimS. W.BayanK. R.MannS.CardulloR. A.2005The acrosomal vesicle of mouse sperm is a calcium store.J Cell Physiol 202(3): 6636710021-9541
  139. 139. HessB.SaftigP.HartmannD.CoenenR.Lullmann-RauchR.GoebelH. H.EversM.vonFigura. K.D’HoogeR.NagelsG.De DeynP.PetersC.GieselmannV.1996Phenotype of arylsulfatase A-deficient mice: relationship to human metachromatic leukodystrophyProc Natl Acad Sci U S A 93(25): 14821148260027-8424
  140. 140. HessK. C.JonesB. H.MarquezB.ChenY.OrdT. S.KamenetskyM.MiyamotoC.ZippinJ. H.KopfG. S.SuarezS. S.LevinL. R.WilliamsC. J.BuckJ.MossS. B.2005The "soluble" adenylyl cyclase in sperm mediates multiple signaling events required for fertilization." Dev Cell 9(2): 2492591534-5807
  141. 141. HickoxJ. R.BiM.HardyD. M.2001Heterogeneous processing and zona pellucida binding activity of pig zonadhesin.J Biol Chem 276(44): 41502415090021-9258
  142. 142. HoH. C.SuarezS. S.2003Characterization of the intracellular calcium store at the base of the sperm flagellum that regulates hyperactivated motility.Biol Reprod 68(5): 159015960006-3363
  143. 143. HoodbhoyT.DeanJ.2004Insights into the molecular basis of sperm-egg recognition in mammals.Reproduction 127(4): 4174221470-1626
  144. 144. HowesE.PascallJ. C.EngelW.JonesR.2001Interactions between mouse ZP2 glycoprotein and proacrosin; a mechanism for secondary binding of sperm to the zona pellucida during fertilization.J Cell Sci 114(Pt 22): 412741360021-9533
  145. 145. HowesL.JonesR.2002Interactions between zona pellucida glycoproteins and sperm proacrosin/acrosin during fertilization.J Reprod Immunol 53(1-2): 1811920165-0378
  146. 146. HullM. G.GlazenerC. M.KellyN. J.ConwayD. I.FosterP. A.HintonR. A.CoulsonC.LambertP. A.WattE. M.DesaiK. M.1985Population study of causes, treatment, and outcome of infertility." Br Med J (Clin Res Ed) 291(6510): 169316970267-0623
  147. 147. HunnicuttG. R.MahanK.LathropW. F.RamaraoC. S.MylesD. G.PrimakoffP.1996aStructural relationship of sperm soluble hyaluronidase to the sperm membrane protein PH-20.Biol Reprod 54(6): 134313490006-3363
  148. 148. HunnicuttG. R.PrimakoffP.MylesD. G.1996bSperm surface protein PH-20 is bifunctional: one activity is a hyaluronidase and a second, distinct activity is required in secondary sperm-zona binding.Biol Reprod 55(1): 80860006-3363
  149. 149. HuszarG.JakabA.SakkasD.OzenciC. C.CayliS.DelpianoE.OzkavukcuS.2007Fertility testing and ICSI sperm selection by hyaluronic acid binding: clinical and genetic aspects.Reprod Biomed Online 14(5): 6506631472-6483
  150. 150. HuszarG.OzkavukcuS.JakabA.Celik-OzenciC.SatiG. L.CayliS.2006Hyaluronic acid binding ability of human sperm reflects cellular maturity and fertilizing potential: selection of sperm for intracytoplasmic sperm injection.Curr Opin Obstet Gynecol 18(3): 2602670104-0872X
  151. 151. HuszarG.SbraciaM.VigueL.MillerD. J.ShurB. D.1997Sperm plasma membrane remodeling during spermiogenetic maturation in men: relationship among plasma membrane beta 1,4-galactosyltransferase, cytoplasmic creatine phosphokinase, and creatine phosphokinase isoform ratios.Biol Reprod 56(4): 102010240006-3363
  152. 152. HuszarG.StoneK.DixD.VigueL.2000Putative creatine kinase M-isoform in human sperm is identifiedas the 70-kilodalton heat shock protein HspA2.Biol Reprod 63(3): 9259320006-3363
  153. 153. HuszarG.VigueL.1993Incomplete development of human spermatozoa is associated with increased creatine phosphokinase concentration and abnormal head morphology.Mol Reprod Dev 34(3): 2922980104-0452X
  154. 154. HuszarG.VigueL.OehningerS.1994Creatine kinase immunocytochemistry of human sperm-hemizona complexes: selective binding of sperm with mature creatine kinase-staining pattern.Fertil Steril 61(1): 1361420015-0282
  155. 155. IkawaM.NakanishiT.YamadaS.WadaI.KominamiK.TanakaH.NozakiM.NishimuneY.OkabeM.2001Calmegin is required for fertilin alpha/beta heterodimerization and sperm fertility." Dev Biol 240(1): 2542610012-1606
  156. 156. IkawaM.TokuhiroK.YamaguchiR.BenhamA. M.TamuraT.WadaI.SatouhY.InoueN.OkabeM.2011Calsperin is a testis-specific chaperone required for sperm fertilityJ Biol Chem 286(7): 563956460108-3351X
  157. 157. InoueM.WolfD. P.1975Fertilization-associated changes in the murine zona pellucida: a time sequence study.Biol Reprod 13(5): 5465510006-3363
  158. 158. JelinskyS. A.TurnerT. T.BangH. J.FingerJ. N.SolarzM. K.WilsonE.BrownE. L.KopfG. S.JohnstonD. S.2007The rat epididymal transcriptome: comparison of segmental gene expression in the rat and mouse epididymidesBiol Reprod 76(4): 5615700006-3363
  159. 159. JervisK. M.RobaireB.2001Dynamic changes in gene expression along the rat epididymis.Biol Reprod 65(3): 6967030006-3363
  160. 160. JezierskaA.MotylT.2009Matrix metalloproteinase-2 involvement in breast cancer progression: a mini-reviewMed Sci Monit 15(2): RA32RA401643-3750
  161. 161. JinJ.JinN.ZhengH.RoS.TafollaD.SandersK. M.YanW.2007Catsper3 and Catsper4 are essential for sperm hyperactivated motility and male fertility in the mouseBiol Reprod 77(1): 37440006-3363
  162. 162. JohnstonD. S.TurnerT. T.FingerJ. N.OwtscharukT. L.KopfG. S.JelinskyS. A.2007Identification of epididymis-specific transcripts in the mouse and rat by transcriptional profilingAsian J Androl 9(4): 5225270100-8682X
  163. 163. JonesR.1998Plasma membrane structure and remodelling during sperm maturation in the epididymis.J Reprod Fertil Suppl 53(73840449-3087
  164. 164. JonesR.HowesE.DunneP. D.JamesP.BruckbauerA.KlenermanD.2010Tracking diffusion of GM1 gangliosides and zona pellucida binding molecules in sperm plasma membranes following cholesterol effluxDev Biol 339(2): 3984060109-5564X
  165. 165. JonesR.JamesP. S.HowesL.BruckbauerA.KlenermanD.2007Supramolecular organization of the sperm plasma membrane during maturation and capacitationAsian J Androl 9(4): 4384440100-8682X
  166. 166. KamaruddinM.KroetschT.BasrurP. K.HansenP. J.KingW. A.2004Immunolocalization of heat shock protein 70 in bovine spermatozoaAndrologia3273340303-4569
  167. 167. KaplanM.RussellL. D.PetersonR. N.MartanJ.1984Boar sperm cytoplasmic droplets: their ultrastructure, their numbers in the epididymis and at ejaculation and their removal during isolation of sperm plasma membranes.Tissue Cell 16(3): 4554680040-8166
  168. 168. KatzD. F.1991Characteristics of sperm motility.Ann N Y Acad Sci 637(4094230077-8923
  169. 169. KeatingJ.GrundyC. E.FiveyP. S.ElliottM.RobinsonJ.1997Investigation of the association between the presence of cytoplasmic residues on the human sperm midpiece and defective sperm function.J Reprod Fertil 110(1): 71770022-4251
  170. 170. KimE.NishimuraH.IwaseS.YamagataK.KashiwabaraS.BabaT.2004Synthesis, processing, and subcellular localization of mouse ADAM3 during spermatogenesis and epididymal sperm transport." J Reprod Dev 50(5): 5715780916-8818
  171. 171. KimE.YamashitaM.NakanishiT.ParkK. E.KimuraM.KashiwabaraS.BabaT.2006aMouse sperm lacking ADAM1b/ADAM2 fertilin can fuse with the egg plasma membrane and effect fertilization.J Biol Chem 281(9): 563456390021-9258
  172. 172. KimK. S.FosterJ. A.GertonG. L.2001Differential release of guinea pig sperm acrosomal components during exocytosis.Biol Reprod 64(1): 1481560006-3363
  173. 173. KimT.OhJ.WooJ. M.ChoiE.ImS. H.YooY. J.KimD. H.NishimuraH.ChoC.2006bExpression and relationship of male reproductive ADAMs in mouse.Biol Reprod 74(4): 7447500006-3363
  174. 174. KirchhoffC.HaleG.1996Cell-to-cell transfer of glycosylphosphatidylinositol-anchored membrane proteins during sperm maturation.Mol Hum Reprod 2(3): 1771841360-9947
  175. 175. KirichokY.NavarroB.ClaphamD. E.2006Whole-cell patch-clamp measurements of spermatozoa reveal an alkaline-activated Ca2+ channel.Nature7377401476-4687
  176. 176. KobayashiT.KanekoT.IuchiY.MatsukiS.TakahashiM.SasagawaI.NakadaT.FujiiJ.2002Localization and physiological implication of aldose reductase and sorbitol dehydrogenase in reproductive tracts and spermatozoa of male rats.J Androl 23(5): 6746830196-3635
  177. 177. KohnF. M.MiskaW.SchillW. B.1995Release of angiotensin-converting enzyme (ACE) from human spermatozoa during capacitation and acrosome reaction.J Androl 16(3): 2592650196-3635
  178. 178. KongM.DiazE. S.MoralesP.2009Participation of the human sperm proteasome in the capacitation process and its regulation by protein kinase A and tyrosine kinaseBiol Reprod 80(5): 102610350006-3363
  179. 179. KornblattM. J.1979Synthesis and turnover of sulfogalactoglycerolipid, a membrane lipid, during spermatogenesis.Can J Biochem 57(3): 2552580008-4018
  180. 180. KrapfD.ArcelayE.WertheimerE. V.SanjayA.PilderS. H.SalicioniA. M.ViscontiP. E.2010Inhibition of Ser/Thr phosphatases induces capacitation-associated signaling in the presence of Src kinase inhibitorsJ Biol Chem 285(11): 797779850108-3351X
  181. 181. LangfordK. G.ZhouY.RussellL. D.WilcoxJ. N.BernsteinK. E.1993Regulated expression of testis angiotensin-converting enzyme during spermatogenesis in mice.Biol Reprod 48(6): 121012180006-3363
  182. 182. LanglaisJ.KanF. W.GrangerL.RaymondL.BleauG.RobertsK. D.1988Identification of sterol acceptors that stimulate cholesterol efflux from human spermatozoa during in vitro capacitation.Gamete Res 20(2): 1852010148-7280
  183. 183. LarsenR. E.ShopeR. E.Jr LemanA. D.KurtzH. J.1980Semen changes in boars after experimental infection with pseudorabies virus.Am J Vet Res 41(5): 7337390002-9645
  184. 184. LawsonC.GoupilS.LeclercP.2008Increased activity of the human sperm tyrosine kinase SRC by the cAMP-dependent pathway in the presence of calciumBiol Reprod 79(4): 6576660006-3363
  185. 185. LeblondC. P.ClermontY.1952Spermiogenesis of rat, mouse, hamster and guinea pig as revealed by the periodic acid-fuchsin sulfurous acid technique.Am J Anat 90(2): 1672150002-9106
  186. 186. LeclercP.de LamirandeE.GagnonC.1997Regulation of protein-tyrosine phosphorylation and human sperm capacitation by reactive oxygen derivatives.Free Radic Biol Med 22(4): 6436560891-5849
  187. 187. LefievreL.ConnerS. J.SalpekarA.OlufowobiO.AshtonP.PavlovicB.LentonW.AfnanM.BrewisI. A.MonkM.HughesD. C.BarrattC. L.2004Four zona pellucida glycoproteins are expressed in the human.Hum Reprod 19(7): 158015860268-1161
  188. 188. LegareC.BerubeB.BoueF.LefievreL.MoralesC. R.El -AlfyM.SullivanR.1999Hamster sperm antigen P26h is a phosphatidylinositol-anchored protein.Mol Reprod Dev 52(2): 2252330104-0452X
  189. 189. LeytonL.SalingP.1989Evidence that aggregation of mouse sperm receptors by ZP3 triggers the acrosome reactionJ Cell Biol 108(6): 216321680021-9525
  190. 190. LinY.MahanK.LathropW. F.MylesD. G.PrimakoffP.1994A hyaluronidase activity of the sperm plasma membrane protein PH-20 enables sperm to penetrate the cumulus cell layer surrounding the eggJ Cell Biol 125(5): 115711630021-9525
  191. 191. LinY. N.RoyA.YanW.BurnsK. H.MatzukM. M.2007Loss of zona pellucida binding proteins in the acrosomal matrix disrupts acrosome biogenesis and sperm morphogenesis.Mol Cell Biol 27(19): 679468050270-7306
  192. 192. LitscherE. S.JuntunenK.SeppoA.PenttilaL.NiemelaR.RenkonenO.WassarmanP. M.1995Oligosaccharide constructs with defined structures that inhibit binding of mouse sperm to unfertilized eggs in vitro.Biochemistry466246690006-2960
  193. 193. LitvinT. N.KamenetskyM.ZarifyanA.BuckJ.LevinL. R.2003Kinetic properties of "soluble" adenylyl cyclase. Synergism between calcium and bicarbonate.J Biol Chem 278(18): 15922159260021-9258
  194. 194. LiuD. Y.BakerH. W.1992Morphology of spermatozoa bound to the zona pellucida of human oocytes that failed to fertilize in vitro.J Reprod Fertil 94(1): 71840022-4251
  195. 195. LobleyA.PierronV.ReynoldsL.AllenL.MichalovichD.2003Identification of human and mouse CatSper3 and CatSper4 genes: characterisation of a common interaction domain and evidence for expression in testisReprod Biol Endocrinol 1(531477-7827
  196. 196. LopezL. C.BaynaE. M.LitoffD.ShaperN. L.ShaperJ. H.ShurB. D.1985Receptor function of mouse sperm surface galactosyltransferase during fertilizationJ Cell Biol 101(4): 150115100021-9525
  197. 197. LopezL. C.ShurB. D.1987Redistribution of mouse sperm surface galactosyltransferase after the acrosome reactionJ Cell Biol 105(4): 166316700021-9525
  198. 198. LuQ.HastyP.ShurB. D.1997Targeted mutation in beta1,4-galactosyltransferase leads to pituitary insufficiency and neonatal lethality.Dev Biol 181(2): 2572670012-1606
  199. 199. LuQ.ShurB. D.1997Sperm from beta 1,4-galactosyltransferase-null mice are refractory to ZP3-induced acrosome reactions and penetrate the zona pellucida poorly.Development 124(20): 412141310950-1991
  200. 200. LyngR.ShurB. D.2009Mouse oviduct-specific glycoprotein is an egg-associated ZP3-independent sperm-adhesion ligandJ Cell Sci 122(Pt 21): 389439061477-9137
  201. 201. MaherM. T.FlozakA. S.StockerA. M.ChennA.GottardiC. J.2009Activity of the beta-catenin phosphodestruction complex at cell-cell contacts is enhanced by cadherin-based adhesion.J Cell Biol 186(2): 2192281540-8140
  202. 202. MahonyM. C.GwathmeyT.1999Protein tyrosine phosphorylation during hyperactivated motility of cynomolgus monkey (Macaca fascicularis) spermatozoa.Biol Reprod 60(5): 123912430006-3363
  203. 203. Martinez-SearaH.RogT.Pasenkiewicz-GierulaM.VattulainenI.KarttunenM.ReigadaR.2008Interplay of unsaturated phospholipids and cholesterol in membranes: effect of the double-bond positionBiophys J 95(7): 329533051542-0086
  204. 204. Mc CreadyJ.SimsJ. D.ChanD.JayD. G.2010Secretion of extracellular hsp90alpha via exosomes increases cancer cell motility: a role for plasminogen activation.BMC Cancer2941471-2407
  205. 205. Mc LachlanR. I.de KretserD. M.2001Male infertility: the case for continued research.Med J Aust 174(3): 1161170002-5729X
  206. 206. Mc LeskeyS. B.DowdsC.CarballadaR.WhiteR. R.SalingP. M.1998Molecules involved in mammalian sperm-egg interaction.Int Rev Cytol 177(571130074-7696
  207. 207. MengeA. C.ChristmanG. M.OhlD. A.NazR. K.1999Fertilization antigen-1 removes antisperm autoantibodies from spermatozoa of infertile men and results in increased rates of acrosome reaction.Fertil Steril 71(2): 2562600015-0282
  208. 208. MikiK.QuW.GouldingE. H.WillisW. D.BunchD. O.StraderL. F.PerreaultS. D.EddyE. M.O’BrienD. A.2004Glyceraldehyde 3-phosphate dehydrogenase-S, a sperm-specific glycolytic enzyme, is required for sperm motility and male fertilityProc Natl Acad Sci U S A 101(47): 16501165060027-8424
  209. 209. MillerD.BroughS.al-HarbiO.1992Characterization and cellular distribution of human spermatozoal heat shock proteins.Hum Reprod 7(5): 6376450268-1161
  210. 210. MitchellL. A.NixonB.AitkenR. J.2007Analysis of chaperone proteins associated with human spermatozoa during capacitationMol Hum Reprod 13(9): 6056131360-9947
  211. 211. MitchellL. A.NixonB.BakerM. A.AitkenR. J.2008Investigation of the role of SRC in capacitation-associated tyrosine phosphorylation of human spermatozoaMol Hum Reprod 14(4): 2352431460-2407
  212. 212. MooreH. D.AkhondiM. A.1996In vitro maturation of mammalian spermatozoa.Rev Reprod 1(1): 54601359-6004
  213. 213. MoralesC. R.BadranH.El -AlfyM.MenH.ZhangH.Martin DeLeon. P. A.2004Cytoplasmic localization during testicular biogenesis of the murine mRNA for Spam1 (PH-20), a protein involved in acrosomal exocytosis.Mol Reprod Dev 69(4): 4754820104-0452X
  214. 214. MoralesP.KongM.PizarroE.PastenC.2003Participation of the sperm proteasome in human fertilizationHum Reprod 18(5): 10101017Print)
  215. 215. Linking).
  216. 216. 0967-19947583Moreno, R. D., Sepulveda, M. S., de Ioannes, A. and Barros, C. (1998). "The polysulphate binding domain of human proacrosin/acrosin is involved in both the enzyme activation and spermatozoa-zona pellucida interaction." Zygote 6(1): pp. 75-83, ISSN 0967-1994
  217. 217. MuroY.BuffoneM. G.OkabeM.GertonG. L.2012Function of the acrosomal matrix: zona pellucida 3 receptor (ZP3R/sp56) is not essential for mouse fertilization.Biol Reprod 86(1): 161529-7268
  218. 218. MuroY.OkabeM.2011Mechanisms of fertilization--a view from the study of gene-manipulated mice.J Androl 32(3): 2182251939-4640
  219. 219. MylesD. G.PrimakoffP.1997Why did the sperm cross the cumulus? To get to the oocyte. Functions of the sperm surface proteins PH-20 and fertilin in arriving at, and fusing with, the eggBiol Reprod 56(2): 3203270006-3363
  220. 220. Naaby-HansenS.HerrJ. C.2010Heat shock proteins on the human sperm surfaceJ Reprod Immunol 84(1): 32401872-7603
  221. 221. NakamuraN.Miranda-VizueteA.MikiK.MoriC.EddyE. M.2008Cleavage of disulfide bonds in mouse spermatogenic cell-specific type 1 hexokinase isozyme is associated with increased hexokinase activity and initiation of sperm motilityBiol Reprod 79(3): 5375450006-3363
  222. 222. NassarA.MahonyM.MorshediM.LinM. H.SrisombutC.OehningerS.1999Modulation of sperm tail protein tyrosine phosphorylation by pentoxifylline and its correlation with hyperactivated motility.Fertil Steril 71(5): 9199230015-0282
  223. 223. NazR. K.1998c-Abl proto-oncoprotein is expressed and tyrosine phosphorylated in human sperm cell.Mol Reprod Dev 51(2): 2102170104-0452X
  224. 224. NazR. K.AhmadK.KaplanP.1992aExpression and function of ras proto-oncogene proteins in human sperm cells.J Cell Sci 102 ( Pt 3)(4874940021-9533
  225. 225. NazR. K.BrazilC.OverstreetJ. W.1992bEffects of antibodies to sperm surface fertilization antigen-1 on human sperm-zona pellucida interaction.Fertil Steril 57(6): 130413100015-0282
  226. 226. NazR. K.RosenblumB. B.MengeA. C.1984Characterization of a membrane antigen from rabbit testis and sperm isolated by using monoclonal antibodies and effect of its antiserum on fertility.Proc Natl Acad Sci U S A 81(3): 8578610027-8424
  227. 227. NazR. K.ZhuX.1998Recombinant fertilization antigen-1 causes a contraceptive effect in actively immunized mice.Biol Reprod 59(5): 109511000006-3363
  228. 228. NishimuraH.KimE.NakanishiT.BabaT.2004Possible function of the ADAM1a/ADAM2 Fertilin complex in the appearance of ADAM3 on the sperm surface.J Biol Chem 279(33): 34957349620021-9258
  229. 229. NishimuraH.MylesD. G.PrimakoffP.2007Identification of an ADAM2-ADAM3 complex on the surface of mouse testicular germ cells and cauda epididymal sperm.J Biol Chem 282(24): 17900179070021-9258
  230. 230. NixonB.AitkenR. J.Mc LaughlinE. A.2007New insights into the molecular mechanisms of sperm-egg interactionCell Mol Life Sci 64(14): 18051823X (Print)
  231. 231. 1420682X (Linking).
  232. 232. NixonB.AsquithK. L.JohnAitken. R.2005The role of molecular chaperones in mouse sperm-egg interactions.Mol Cell Endocrinol 240(1-2): 1100303-7207
  233. 233. NixonB.BielanowiczA.AndersonA. L.WalshA.HallT.Mc CloghryA.AitkenR. J.2010Elucidation of the signaling pathways that underpin capacitation-associated surface phosphotyrosine expression in mouse spermatozoaJ Cell Physiol 224(1): 71831097-4652
  234. 234. NixonB.BielanowiczA.Mc LaughlinE. A.TanphaichitrN.EnsslinM. A.AitkenR. J.2009Composition and significance of detergent resistant membranes in mouse spermatozoaJ Cell Physiol 218(1): 1221341097-4652
  235. 235. NixonB.JonesR. C.HansenL. A.HollandM. K.2002Rabbit epididymal secretory proteins. I. Characterization and hormonal regulationBiol Reprod 67(1): 1331390006-3363
  236. 236. NixonB.LuQ.WasslerM. J.FooteC. I.EnsslinM. A.ShurB. D.2001Galactosyltransferase function during mammalian fertilizationCells Tissues Organs46571422-6405
  237. 237. NixonB.MacIntyre. D. A.MitchellL. A.GibbsG. M.O’BryanM.AitkenR. J.2006The identification of mouse sperm-surface-associated proteins and characterization of their ability to act as decapacitation factors.Biol Reprod 74(2): 2752870006-3363
  238. 238. NixonB.MitchellL. A.AndersonA. L.Mc LaughlinE. A.O’BryanM. K.AitkenR. J.2011Proteomic and functional analysis of human sperm detergent resistant membranes.J Cell Physiol 226(10): 265126651097-4652
  239. 239. NolandT. D.FridayB. B.MaulitM. T.GertonG. L.1994The sperm acrosomal matrix contains a novel member of the pentaxin family of calcium-dependent binding proteins.J Biol Chem 269(51): 32607326140021-9258
  240. 240. OatleyJ. M.BrinsterR. L.2006Spermatogonial stem cells." Methods Enzymol 419(2592820076-6879
  241. 241. OhJ. S.HanC.ChoC.2009ADAM7 is associated with epididymosomes and integrated into sperm plasma membraneMol Cells 28(5): 4414460219-1032
  242. 242. OkamuraN.TajimaY.SoejimaA.MasudaH.SugitaY.1985Sodium bicarbonate in seminal plasma stimulates the motility of mammalian spermatozoa through direct activation of adenylate cyclase.J Biol Chem 260(17): 969997050021-9258
  243. 243. OlleroM.PowersR. D.AlvarezJ. G.2000Variation of docosahexaenoic acid content in subsets of human spermatozoa at different stages of maturation: implications for sperm lipoperoxidative damage.Mol Reprod Dev 55(3): 3263340104-0452X
  244. 244. OlsonG. E.WinfreyV. P.BiM.HardyD. M.NagDas. S. K.2004Zonadhesin assembly into the hamster sperm acrosomal matrix occurs by distinct targeting strategies during spermiogenesis and maturation in the epididymis.Biol Reprod 71(4): 112811340006-3363
  245. 245. OmbeletW.WoutersE.BoelsL.CoxA.JanssenM.SpiessensC.VereeckenA.BosmansE.SteenoO.1997Sperm morphology assessment: diagnostic potential and comparative analysis of strict or WHO criteria in a fertile and a subfertile population.Int J Androl 20(6): 3673720105-6263
  246. 246. Orgebin-CristM. C.1967aFertility in does inseminated with epididymal spermatozoaJ Reprod Fertil 14(2): 3463470022-4251
  247. 247. Orgebin-CristM. C.1967bSperm maturation in rabbit epididymis.Nature8168180028-0836
  248. 248. Orgebin-CristM. C.1968Maturation of spermatozoa in the rabbit epididymis: delayed fertilization in does inseminated with epididymal spermatozoa.J Reprod Fertil 16(1): 29330022-4251
  249. 249. Orgebin-CristM. C.1969Studies on the function of the epididymis.Biol Reprod 1(pp. Suppl 1155750006-3363
  250. 250. PastenC.MoralesP.KongM.2005Role of the sperm proteasome during fertilization and gamete interaction in the mouse.Mol Reprod Dev 71(2): 2092190104-0452X
  251. 251. Pastor-SolerN.BeaulieuV.LitvinT. N.DaSilva. N.ChenY.BrownD.BuckJ.LevinL. R.BretonS.2003Bicarbonate-regulated adenylyl cyclase (sAC) is a sensor that regulates pH-dependent V-ATPase recycling.J Biol Chem 278(49): 49523495290021-9258
  252. 252. PereiraB. M.Abou-HailaA.TulsianiD. R.1998Rat sperm surface mannosidase is first expressed on the plasma membrane of testicular germ cells.Biol Reprod 59(6): 128812950006-3363
  253. 253. PetrunkinaA. M.HarrisonR. A.Topfer-PetersenE.2000Only low levels of spermadhesin AWN are detectable on the surface of live ejaculated boar spermatozoaReprod Fertil Dev 12(7-8): 3613711031-3613
  254. 254. PiehlerE.PetrunkinaA. M.Ekhlasi-HundrieserM.Topfer-PetersenE.2006Dynamic quantification of the tyrosine phosphorylation of the sperm surface proteins during capacitation.CytometryA 69(10): 106210701552-4922
  255. 255. PikeL. J.2006Rafts defined: a report on the Keystone Symposium on Lipid Rafts and Cell Function.J Lipid Res 47(7): 159715980022-2275
  256. 256. QiH.MoranM. M.NavarroB.ChongJ. A.KrapivinskyG.KrapivinskyL.KirichokY.RamseyI. S.QuillT. A.ClaphamD. E.2007All four CatSper ion channel proteins are required for male fertility and sperm cell hyperactivated motilityProc Natl Acad Sci U S A 104(4): 121912230027-8424
  257. 257. QuillT. A.RenD.ClaphamD. E.GarbersD. L.2001A voltage-gated ion channel expressed specifically in spermatozoaProc Natl Acad Sci U S A 98(22): 12527125310027-8424
  258. 258. Ramalho-SantosJ.MorenoR. D.WesselG. M.ChanE. K.SchattenG.2001Membrane trafficking machinery components associated with the mammalian acrosome during spermiogenesisExp Cell Res 267(1): 45600014-4827
  259. 259. RankinT. L.ColemanJ. S.EpifanoO.HoodbhoyT.TurnerS. G.CastleP. E.LeeE.Gore-LangtonR.DeanJ.2003Fertility and taxon-specific sperm binding persist after replacement of mouse sperm receptors with human homologs.Dev Cell 5(1): 33431534-5807
  260. 260. RedgroveK. A.AndersonA. L.DunM. D.Mc LaughlinE. A.O’BryanM. K.AitkenR. J.NixonB.2011Involvement of multimeric protein complexes in mediating the capacitation-dependent binding of human spermatozoa to homologous zonae pellucidaeDev Biol 356(2): 4604740109-5564X
  261. 261. RenD.NavarroB.PerezG.JacksonA. C.HsuS.ShiQ.TillyJ. L.ClaphamD. E.2001A sperm ion channel required for sperm motility and male fertility.Nature6036090028-0836
  262. 262. RosanoG.CailleA. M.Gallardo-RiosM.MunuceM. J.2007D-Mannose-binding sites are putative sperm determinants of human oocyte recognition and fertilization.Reprod Biomed Online 15(2): 1821901472-6483
  263. 263. SacksteinR.2005The lymphocyte homing receptors: gatekeepers of the multistep paradigm.Curr Opin Hematol 12(6): 4444501065-6251
  264. 264. SaezF.FrenetteG.SullivanR.2003Epididymosomes and prostasomes: their roles in posttesticular maturation of the sperm cells.J Androl 24(2): 1491540196-3635
  265. 265. SakkasD.Leppens-LuisierG.LucasH.ChardonnensD.CampanaA.FrankenD. R.UrnerF.2003Localization of tyrosine phosphorylated proteins in human sperm and relation to capacitation and zona pellucida binding.Biol Reprod 68(4): 146314690006-3363
  266. 266. SalingP. M.SowinskiJ.StoreyB. T.1979An ultrastructural study of epididymal mouse spermatozoa binding to zonae pellucidae in vitro: sequential relationship to the acrosome reaction.J Exp Zool 209(2): 2292380002-2104X
  267. 267. SchmellE. D.GulyasB. J.1980Mammalian sperm-egg recognition and binding in vitro. I. Specificity of sperm interactions with live and fixed eggs in homologous and heterologous inseminations of hamster, mouse, and guinea pig oocytes.Biol Reprod 23(5): 107510850006-3363
  268. 268. SchuckS.HonshoM.EkroosK.ShevchenkoA.SimonsK.2003Resistance of cell membranes to different detergentsProc Natl Acad Sci U S A 100(10): 579558000027-8424
  269. 269. SeligmanJ.ZipserY.KosowerN. S.2004Tyrosine phosphorylation, thiol status, and protein tyrosine phosphatase in rat epididymal spermatozoa.Biol Reprod 71(3): 100910150006-3363
  270. 270. ShadanS.JamesP. S.HowesE. A.JonesR.2004Cholesterol efflux alters lipid raft stability and distribution during capacitation of boar spermatozoa.Biol Reprod 71(1): 2532650006-3363
  271. 271. ShamsadinR.AdhamI. M.NayerniaK.HeinleinU. A.OberwinklerH.EngelW.1999Male mice deficient for germ-cell cyritestin are infertile.Biol Reprod 61(6): 144514510006-3363
  272. 272. ShenL.WeberC. R.TurnerJ. R.2008The tight junction protein complex undergoes rapid and continuous molecular remodeling at steady stateJ Cell Biol 181(4): 6836951540-8140
  273. 273. ShiS.WilliamsS. A.SeppoA.KurniawanH.ChenW.YeZ.MarthJ. D.StanleyP.2004Inactivation of the Mgat1 gene in oocytes impairs oogenesis, but embryos lacking complex and hybrid N-glycans develop and implantMol Cell Biol 24(22): 992099290270-7306
  274. 274. ShumW. W.DaSilva. N.BrownD.BretonS.2009Regulation of luminal acidification in the male reproductive tract via cell-cell crosstalkJ Exp Biol 212(Pt 11): 175317610022-0949
  275. 275. ShurB. D.BennettD.1979A specific defect in galactosyltransferase regulation on sperm bearing mutant alleles of the T/t locus.Dev Biol 71(2): 2432590012-1606
  276. 276. ShurB. D.HallN. G.1982a). "A role for mouse sperm surface galactosyltransferase in sperm binding to the egg zona pellucida." J Cell Biol 95(2 Pt 1): 5745790021-9525
  277. 277. ShurB. D.HallN. G.1982bSperm surface galactosyltransferase activities during in vitro capacitationJ Cell Biol 95(2 Pt 1): 5675730021-9525
  278. 278. SiY.OkunoM.1999Role of tyrosine phosphorylation of flagellar proteins in hamster sperm hyperactivation.Biol Reprod 61(1): 2402460006-3363
  279. 279. SibonyM.GascJ. M.SoubrierF.Alhenc-GelasF.CorvolP.1993Gene expression and tissue localization of the two isoforms of angiotensin I converting enzyme.HypertensionPt 1): 8278350019-4911X
  280. 280. SimonsK.IkonenE.1997Functional rafts in cell membranes.Nature5695720028-0836
  281. 281. SimonsK.ToomreD.2000Lipid rafts and signal transduction.Nat Rev Mol Cell Biol 1(1): 31391471-0072
  282. 282. SimonsK.VazW. L.2004Model systems, lipid rafts, and cell membranes." Annu Rev Biophys Biomol Struct 33(2692951056-8700
  283. 283. SimsJ. D.Mc CreadyJ.JayD. G.2011Extracellular heat shock protein (Hsp)70 and Hsp90alpha assist in matrix metalloproteinase-2 activation and breast cancer cell migration and invasion." PLoS One 6(4): e188481932-6203
  284. 284. SinowatzF.AmselgruberW.Topfer-PetersenE.CalveteJ. J.SanzL.PlendlJ.1995Immunohistochemical localization of spermadhesin AWN in the porcine male genital tract.Cell Tissue Res 282(1): 1751790030-2766X
  285. 285. SitiaR.BraakmanI.2003Quality control in the endoplasmic reticulum protein factoryNature 426(6968): 8918941476-4687
  286. 286. SleightS. B.MirandaP. V.PlaskettN. W.MaierB.LysiakJ.ScrableH.HerrJ. C.ViscontiP. E.2005Isolation and proteomic analysis of mouse sperm detergent-resistant membrane fractions: evidence for dissociation of lipid rafts during capacitationBiol Reprod 73(4): 7217290006-3363
  287. 287. SolerC.YeungC. H.CooperT. G.1994Development of sperm motility patterns in the murine epididymis.Int J Androl 17(5): 2712780105-6263
  288. 288. SpinaciM.VolpeS.BernardiniC.De AmbrogiM.TamaniniC.SerenE.GaleatiG.2005Immunolocalization of heat shock protein 70 (Hsp 70) in boar spermatozoa and its role during fertilization.Mol Reprod Dev 72(4): 5345410104-0452X
  289. 289. SteinK. K.GoJ. C.LaneW. S.PrimakoffP.MylesD. G.2006Proteomic analysis of sperm regions that mediate sperm-egg interactions.Proteomics353335431615-9853
  290. 290. SuarezS. S.2008Control of hyperactivation in spermHum Reprod Update 14(6): 6476571460-2369
  291. 291. SutovskyP.ManandharG.Mc CauleyT. C.CaamanoJ. N.SutovskyM.ThompsonW. E.DayB. N.2004Proteasomal interference prevents zona pellucida penetration and fertilization in mammals.Biol Reprod 71(5): 162516370006-3363
  292. 292. SwensonC. E.DunbarB. S.1982Specificity of sperm-zona interaction.J Exp Zool 219(1): 971040002-2104X
  293. 293. SyntinP.DacheuxF.DruartX.GattiJ. L.OkamuraN.DacheuxJ. L.1996Characterization and identification of proteins secreted in the various regions of the adult boar epididymis.Biol Reprod 55(5): 9569740006-3363
  294. 294. TanphaichitrN.SmithJ.KatesM.1990Levels of sulfogalactosylglycerolipid in capacitated motile and immotile mouse spermatozoa.Biochem Cell Biol 68(2): 5285350829-8211
  295. 295. TanphaichitrN.SmithJ.MongkolsirikieartS.GradilC.LingwoodC. A.1993Role of a gamete-specific sulfoglycolipid immobilizing protein on mouse sperm-egg bindingDev Biol 156(1): 1641750012-1606
  296. 296. TantibhedhyangkulJ.WeerachatyanukulW.CarmonaE.XuH.AnupriwanA.MichaudD.TanphaichitrN.2002Role of sperm surface arylsulfatase A in mouse sperm-zona pellucida binding.Biol Reprod 67(1): 2122190006-3363
  297. 297. TardifS.CormierN.2011Role of zonadhesin during sperm-egg interaction: a species-specific acrosomal molecule with multiple functionsMol Hum Reprod 17(11): 6616681460-2407
  298. 298. TardifS.WilsonM. D.WagnerR.HuntP.GertsensteinM.NagyA.LobeC.KoopB. F.HardyD. M.2010Zonadhesin is essential for species specificity of sperm adhesion to the egg zona pellucidaJ Biol Chem 285(32): 24863248700108-3351X
  299. 299. TesarikJ.MoosJ.MendozaC.1993Stimulation of protein tyrosine phosphorylation by a progesterone receptor on the cell surface of human sperm.Endocrinology3283350013-7227
  300. 300. ThalerC. D.CardulloR. A.1996The initial molecular interaction between mouse sperm and the zona pellucida is a complex binding event.J Biol Chem 271(38): 23289232970021-9258
  301. 301. ThalerC. D.CardulloR. A.2002Distinct membrane fractions from mouse sperm bind different zona pellucida glycoproteins.Biol Reprod 66(1): 65690006-3363
  302. 302. ThimonV.FrenetteG.SaezF.ThabetM.SullivanR.2008Protein composition of human epididymosomes collected during surgical vasectomy reversal: a proteomic and genomic approachHum Reprod 23(8): 169817071460-2350
  303. 303. Topfer-PetersenE.1999Carbohydrate-based interactions on the route of a spermatozoon to fertilization.Hum Reprod Update 5(4): 3143291355-4786
  304. 304. Topfer-PetersenE.RomeroA.VarelaP. F.Ekhlasi-HundrieserM.DostalovaZ.SanzL.CalveteJ. J.1998Spermadhesins: a new protein family. Facts, hypotheses and perspectives.Andrologia2172240303-4569
  305. 305. TulsianiD. R.Abou-HailaA.LoeserC. R.PereiraB. M.1998The biological and functional significance of the sperm acrosome and acrosomal enzymes in mammalian fertilization.Exp Cell Res 240(2): 1511640014-4827
  306. 306. TulsianiD. R.SkudlarekM. D.NagdasS. K.Orgebin-CristM. C.1993Purification and characterization of rat epididymal-fluid alpha-D-mannosidase: similarities to sperm plasma-membrane alpha-D-mannosidase.Biochem J 290 ( Pt 2)(4274360264-6021
  307. 307. TulsianiD. R.SkudlarekM. D.Orgebin-CristM. C.1989Novel alpha-D-mannosidase of rat sperm plasma membranes: characterization and potential role in sperm-egg interactions.J Cell Biol 109(3): 125712670021-9525
  308. 308. UN2009World population to exceed 9 billion by 2050." pp.
  309. 309. UrchU. A.PatelH.1991The interaction of boar sperm proacrosin with its natural substrate, the zona pellucida, and with polysulfated polysaccharides.Development 111(4): 116511720950-1991
  310. 310. UrnerF.Leppens-LuisierG.SakkasD.2001Protein tyrosine phosphorylation in sperm during gamete interaction in the mouse: the influence of glucose.Biol Reprod 64(5): 135013570006-3363
  311. 311. UrnerF.SakkasD.2003Protein phosphorylation in mammalian spermatozoa.Reproduction 125(1): 17261470-1626
  312. 312. van GestelR. A.BrewisI. A.AshtonP. R.HelmsJ. B.BrouwersJ. F.GadellaB. M.2005Capacitation-dependent concentration of lipid rafts in the apical ridge head area of porcine sperm cellsMol Hum Reprod 11(8): 5835901360-9947
  313. 313. VazquezM. H.PhillipsD. M.WassarmanP. M.1989Interaction of mouse sperm with purified sperm receptors covalently linked to silica beads.J Cell Sci 92 ( Pt 4)(7137220021-9533
  314. 314. ViscontiP. E.BaileyJ. L.MooreG. D.PanD.Olds-ClarkeP.KopfG. S.1995aCapacitation of mouse spermatozoa. I. Correlation between the capacitation state and protein tyrosine phosphorylation.Development 121(4): 112911370950-1991
  315. 315. ViscontiP. E.FlormanH. M.2010Mechanisms of sperm-egg interactions: between sugars and broken bonds.Sci Signal 3(142): pe351937-9145
  316. 316. ViscontiP. E.MooreG. D.BaileyJ. L.LeclercP.ConnorsS. A.PanD.Olds-ClarkeP.KopfG. S.1995bCapacitation of mouse spermatozoa. II. Protein tyrosine phosphorylation and capacitation are regulated by a cAMP-dependent pathway.Development 121(4): 113911500950-1991
  317. 317. ViscontiP. E.NingX.FornesM. W.AlvarezJ. G.SteinP.ConnorsS. A.KopfG. S.1999Cholesterol efflux-mediated signal transduction in mammalian sperm: cholesterol release signals an increase in protein tyrosine phosphorylation during mouse sperm capacitation.Dev Biol 214(2): 4294430012-1606
  318. 318. WalshA.WhelanD.BielanowiczA.SkinnerB.AitkenR. J.O’BryanM. K.NixonB.2008Identification of the molecular chaperone, heat shock protein 1 (chaperonin 10), in the reproductive tract and in capacitating spermatozoa in the male mouseBiol Reprod 78(6): 9839930006-3363
  319. 319. WassarmanP. M.1988Zona pellucida glycoproteins.Annu Rev Biochem 57(4154420066-4154
  320. 320. WassarmanP. M.1992Mouse gamete adhesion molecules.Biol Reprod 46(2): 1861910006-3363
  321. 321. WassarmanP. M.2009Mammalian fertilization: the strange case of sperm protein 56Bioessays1531581521-1878
  322. 322. WassarmanP. M.LitscherE. S.2008Mammalian fertilization: the egg’s multifunctional zona pellucida.Int J Dev Biol 52(5-6): 6656760214-6282
  323. 323. WassarmanP. M.MortilloS.1991Structure of the mouse egg extracellular coat, the zona pellucida.Int Rev Cytol 130(851100074-7696
  324. 324. WeerachatyanukulW.RattanachaiyanontM.CarmonaE.FurimskyA.MaiA.ShoushtarianA.SirichotiyakulS.BallakierH.LeaderA.TanphaichitrN.2001Sulfogalactosylglycerolipid is involved in human gamete interaction.Mol Reprod Dev 60(4): 5695780104-0452X
  325. 325. WeerachatyanukulW.XuH.AnupriwanA.CarmonaE.WadeM.HermoL.daSilva. S. M.RippsteinP.SobhonP.SretarugsaP.TanphaichitrN.2003Acquisition of arylsulfatase A onto the mouse sperm surface during epididymal transit.Biol Reprod 69(4): 118311920006-3363
  326. 326. WehrenA.MeyerH. E.SobekA.KloetzelP. M.DahlmannB.1996Phosphoamino acids in proteasome subunits.Biol Chem 377(7-8): 4975031431-6730
  327. 327. WestA. P.WillisonK. R.1996Brefeldin A and mannose 6-phosphate regulation of acrosomic related vesicular trafficking.Eur J Cell Biol 70(4): 3153210171-9335
  328. 328. Westbrook-CaseV. A.WinfreyV. P.OlsonG. E.1994Characterization of two antigenically related integral membrane proteins of the guinea pig sperm periacrosomal plasma membrane.Mol Reprod Dev 39(3): 3093210104-0452X
  329. 329. WhiteD.WeerachatyanukulW.GadellaB.KamolvarinN.AttarM.TanphaichitrN.2000Role of sperm sulfogalactosylglycerolipid in mouse sperm-zona pellucida binding.Biol Reprod 63(1): 1471550006-3363
  330. 330. WhiteD. R.AitkenR. J.1989Relationship between calcium, cyclic AMP, ATP, and intracellular pH and the capacity of hamster spermatozoa to express hyperactivated motility." Gamete Res 22(2): 1631770148-7280
  331. 331. YamagataK.NakanishiT.IkawaM.YamaguchiR.MossS. B.OkabeM.2002Sperm from the calmegin-deficient mouse have normal abilities for binding and fusion to the egg plasma membraneDev Biol 250(2): 3483570012-1606
  332. 332. YamaguchiR.MuroY.IsotaniA.TokuhiroK.TakumiK.AdhamI.IkawaM.OkabeM.2009Disruption of ADAM3 impairs the migration of sperm into oviduct in mouseBiol Reprod 81(1): 1421460006-3363
  333. 333. YamasakiN.RichardsonR. T.O’RandM. G.1995Expression of the rabbit sperm protein Sp17 in COS cells and interaction of recombinant Sp17 with the rabbit zona pellucida.Mol Reprod Dev 40(1): 48550104-0452X
  334. 334. YanagimachiR.1994aFertility of mammalian spermatozoa: its development and relativity.Zygote 2(4): 3713720967-1994
  335. 335. YanagimachiR.2009Germ cell research: a personal perspectiveBiol Reprod 80(2): 2042180006-3363
  336. 336. YanoR.MatsuyamaT.KanekoT.KurioH.MurayamaE.ToshimoriK.IidaH.2010Bactericidal/Permeability-increasing protein is associated with the acrosome region of rodent epididymal spermatozoa.J Androl 31(2): 2012141939-4640
  337. 337. YiY. J.ManandharG.SutovskyM.ZimmermanS. W.JonakovaV.van LeeuwenF. W.OkoR.ParkC. S.SutovskyP.2010Interference with the 19S proteasomal regulatory complex subunit PSMD4 on the sperm surface inhibits sperm-zona pellucida penetration during porcine fertilization.Cell Tissue Res 341(2): 3253401432-0878
  338. 338. Yoshida-KomiyaH.TulsianiD. R.HirayamaT.ArakiY.1999Mannose-binding molecules of rat spermatozoa and sperm-egg interaction.Zygote 7(4): 3353460967-1994
  339. 339. ZhangH.Martin-DeleonP. A.2003Mouse epididymal Spam1 (pH-20) is released in the luminal fluid with its lipid anchor.J Androl 24(1): 51580196-3635
  340. 340. ZimmermanS. W.ManandharG.YiY. J.GuptaS. K.SutovskyM.OdhiamboJ. F.PowellM. D.MillerD. J.SutovskyP.2011Sperm proteasomes degrade sperm receptor on the egg zona pellucida during mammalian fertilizationPLoS One 6(2): e172561932-6203
  341. 341. ZiniA.O’BryanM. K.IsraelL.SchlegelP. N.1998Human sperm NADH and NADPH diaphorase cytochemistry: correlation with sperm motility.Urology4644680090-4295

Written By

Kate A. Redgrove, R. John Aitken and Brett Nixon

Submitted: 27 November 2011 Published: 19 September 2012