1,873
Views
15
CrossRef citations to date
0
Altmetric
Review

Oolemma Receptors and Oocyte Activation

, &
Pages 365-375 | Received 15 Jul 2009, Accepted 03 Aug 2009, Published online: 16 Apr 2010

Abstract

At fertilization the sperm triggers a series of intracellular calcium oscillations that are pivotal to oocyte activation and development. Although the biological significance of the characteristic intracellular calcium (Ca2+i) oscillations is not fully understood, calcium ions are known to be involved in cortical granule release and in controlling cell cycle progression. Two different hypotheses attempt to explain how sperm initiate (Ca2+i) oscillations in mammalian oocytes. One hypothesis is that spermatozoa interact with a receptor located in the plasma membrane of the oocyte, which results in induction of pathways leading to activation. This receptor is coupled to a GTP-binding protein or to have tyrosine kinase activity and have the ability to induce activation of phospholipase C (PLC). In turn, PLC stimulates the hydrolysis of phosphatidyl inositol (4,5)–bisphosphate (PIP2) to produce diacylglycerol (DAG) and 1,4,5 inositol trisphosphate (IP3), a common Ca2+ releasing compound. Most studies used to develop the mammalian model of oocyte activation have been performed in the mouse. There is a paucity of information from other mammalian models. The predominant mouse model of oocyte activation is that there is a soluble factor (PLC-zeta) delivered to the cytosol after fertilization that induces oocyte activation. However, as data in other mammals is collected, substantial evidence is beginning to support the existence of other more complex oocyte activation pathways in both murine and non-murine systems. Indeed, activation may involve redundant processes, each of which acting alone may be able to induce aspects of oocyte activation. Recent findings demonstrate the involvement of receptors that are known to associate in large, multimeric complexes. This fact leads one to speculate that the process of oocyte activation by the sperm cell is a highly complex and elaborate process that likely involves many more players than perhaps was initially expected.

Abbreviations
PLC:=

phospholipase C

PIP2:=

phosphatidyl inositol (4,5)-bisphosphate

DAG:=

diacylglycerol

IP3:=

1,4,5 inositol trisphosphate

Ca2+:=

intracellular calcium

G-protein:=

GTP-binding protein

SF:=

sperm factor

PLCζ:=

phospholipase C zeta

ICSI:=

intracytoplasmic sperm injections

PAWP:=

postacrosomal sheath WW domain binding protein

ECM:=

extracellular matrix

RGD:=

Arg-Gly-Asp

6-DMAP:=

6-dimethylaminopurine

ADAM1:=

sperm fertilin alpha (also known as A disintegrin and A metalloprotease 1)

ADAM2:=

fertilin beta

ADAM3:=

cyritestin

BOMP:=

bacterial outer membrane

CRISP1:=

cystein-rich secretory protein 1

GPI:=

glycosyl phosphatidylinositol

CTK:=

cytoplasmic tyrosine kinase

FAK:=

focal adhesion kinase

INTRODUCTION

After the sperm cell penetrates the zona pellucida it moves through the perivitelline space and comes into contact with the vitelline membrane. By this time the sperm has undergone the acrosome reaction and thus ligands present on the inner acrosomal membrane are exposed and come into contact with receptors present on the vitelline membrane. These receptors bind the gametes together and likely transduce a signal across the vitelline membrane leading to activational events within the oocyte. Oocyte activation is defined by a series of metabolic changes that occur within the oocyte and include transient release of intracellular calcium [Ca2+i], cortical granule release, resumption of meiosis, and extrusion of the second polar body. There are also receptor interactions that lead to fusion of the sperm and oocyte membranes.

At fertilization, the sperm induces repetitive intracellular calcium transients that are critical to oocyte activation and development in every species that has been studied [Berridge and Galione Citation1988; Kline and Kline Citation1992]. Calcium is a divalent cation that commonly functions as a second messenger, relaying signals downstream so that a cell can respond to various stimuli. The cell has established critical mechanisms, including ATP driven ion channels and ion exchange channels, which precisely maintain intracellular calcium at very low levels. In this way, calcium is either sequestered within the endoplasmic reticulum or in the extracellular space. This low intracellular and high extracellular calcium environment establishes a strong gradient, such that, when an appropriate signal is received, a very rapid influx of calcium occurs. Several enzymes within the cell, including calmodulin and calpain, are designed to respond to this concomitant increase in calcium. These enzymes can, in turn, activate other enzymes thus propagating the signal cascade. The biological significance of the changes in Ca2+i concentration as it relates to oocyte activation is not fully understood, however, calcium ions are known to be involved in cortical granule release which leads to a block to polyspermy and in the control of cell cycle progression [Kline and Kline Citation1992].

Two different hypotheses attempt to explain how sperm initiate Ca2+i oscillations in mammalian oocytes. One hypothesis is that spermatozoa interact with a receptor located in the plasma membrane of the oocyte. This receptor is postulated to be coupled to either a GTP-binding protein (G-protein) or to have tyrosine kinase activity and the ability to activate phospholipase C (PLC) which, in turn, stimulates the production of diacylglycerol (DAG) and 1,4,5 inositol trisphosphate (IP3), a common Ca2+ releasing compound, from phosphatidyl inositol (4,5)-bisphosphate (PIP2).

The second hypothesis proposes that a factor from the sperm (SF) is released into the oocyte and, by interacting with unknown cytosolic targets, generates Ca2+i oscillations. Several lines of evidence support this theory. First, injection of a sperm preparation into hamster and mouse oocytes triggered Ca2+i oscillations similar to those observed during fertilization [Swann Citation1994]. Secondly, in most species, sperm-oocyte fusion appears to precede the initiation of oscillations [Jones et al. Citation1998; Lawrence et al. Citation1997]. Finally, injection of sperm directly, incon junction with intracytoplasmic sperm injection (ICSI), into the cytoplasm of human oocytes results in initiation of Ca2+i oscillations and subsequent embryonic development [Tesarik et al. Citation1994]. Phospholipase C zeta (PLCζ) has been proposed to be the SF involved in activation of murine oocytes and has been shown to initiate Ca2+i oscillations and parthenogenetic development to the blastocyst stage [Saunders et al. Citation2002]. These results suggest that oocyte activation, in mice and humans, may be mediated by a soluble factor independent of a receptor on the plasma membrane of the oocyte.

The most compelling results to date in bovine supporting an SF hypothesis are recent reports by Ross et al. [Citation2008, Citation2009] indicating that the injection of cRNA coding for bovine PLCζ transcript induces activation of bovine oocytes and somatic cell nuclear transfer embryos. The activation response in these two reports is defined as highly efficient development (comparable to IVF controls) to the blastocyst stage and calcium transients that closely resemble those observed at fertilization. However, the main obstacle to establishing this hypothesis as a native mechanism of bovine activation is the inability of any SF isolates to induce complete activation of bovine oocytes [Malcuit et al. Citation2006]. Putative bovine SFs from sperm lysates have been shown to induce activation responses in murine oocytes, but fail to induce similar responses when used in bovine oocytes [Tang et al. Citation2000]. The deficiency in the SF hypothesis is further illustrated by the fact that, contrary to the results observed in human and mouse oocytes following intracytoplasmic sperm injections (ICSI), ICSI-treated bovine oocytes fail to develop [Chung et al. Citation2000; Suttner et al. 2000; Oikawa et al. Citation2005; Abdalla et al. Citation2009; Devito et al. Citation2009]. It is well established that PLC is involved in the activation process of several species including the bovine [Runft et al. Citation2002; Carroll et al. Citation1999, Citation1997; Tokmakov et al. Citation2002]. Therefore, it is not surprising that injection of PLC cRNA, induces efficient activation of the bovine oocyte. The experiment that must be done to confirm that it is only PLCζ that induces activation is to provide other isotypes of PLC to access their potential for activation or reduce egg PLC isoforms to evaluate the resulting impact on activation. If other cRNA isotypes of PLC induce a response similar to that observed following PLCζ injection, this would argue that PLCζ-triggered activation may not be a physiologic response, or at a minimum, not the only event in activation.

In the mouse, reductions in PLCβ1, an egg PLC isoform, results in a concomitant reduction in calcium transient amplitude, but had no impact on duration or frequency following fertilization [Igarashi et al. Citation2007]. In these same experiments, over-expression of PLCβ1 resulted in an altered initial transient following fertilization and subsequent changes in duration and frequency of subsequent transients. Together, these data indicate the potential of a more active role of egg-PLC isoforms in the activation mechanism than previously thought.

A recent report identified a novel alkaline extractable protein named PAWP (postacrosomal sheath WW domain binding protein) that is located within the post acrosomal sheath of the bovine sperm head. Microinjection of PAWP induces pronuclear formation in bovine, porcine, and rhesus monkey oocytes [Wu et al. Citation2007]. This data indicates that protein factors in addition to PLC are able to induce activation events, either directly as a SF or through a membrane associated event. A need for more than one factor could explain the failure of bovine SF isolates and ICSI protocols to effectively induce a complete activation response and development. Possibly, PLC activity must be preceded by a membrane-associated event.

The debate between these two hypotheses has certainly not ended. There is data to indicate that a SF can induce activation and limited development in some species. However, activation in the bovine appears to be refractory to initiation by putative SF. All reports to date indicate SF fails to induce par thenogenetic development in bovine oocytes and injection of bovine sperm into bovine oocytes fail to induce subsequent development [Malcuit et al. Citation2006]. It is understood that the sperm cell, when it undergoes the acrosome reaction (a fusion of cell membranes that release factors allowing the sperm to penetrate the zona pellucida, and exposes ligands for binding with the oocyte vitelline membrane) experiences many events which are similar to oocyte activation, including transient release of Ca2+i [Kirkman-Brown et al. Citation2004, Citation2002, Citation2000; Marquez and Suarez Citation2004]. Additionally, there is significant data in the bovine as well as other species, indicating that the sperm induces activation of the oocyte through a receptor-mediated interaction. Current models for oocyte activation in both echinoderm and Xenopus oocytes do not appear to include PLCζ, but seem to include Src family kinases and PLC isoforms that are endogenous to the oocyte [Stith et al. Citation2005; Townley et al. Citation2005]. The following is a review of possible mediators of oocyte activation particularly pertaining to the bovine model.

OOCYTE RECEPTORS

Literature indicates that there are two primary putative receptor families, integrins and tetraspannins, which are thought to have some interaction with sperm ligands.

Integrins

Integrin molecules are ubiquitously synthesized cell surface adhesion receptors that form a family of transmembrane glycoproteins with heterodimeric structure [Hynes Citation1992]. Mammalian systems include genes for 18 different alpha and 8 different beta subunits that combine in various permutations to form 24 known integrin receptors. The specific subunit pair that composes the integrin receptor is a major determinant of ligand specificity. Integrin receptors function in cellular processes such as attachment, migration, and cell differentiation. Integrins are known to be involved in the onset and continuation of several disease states, including tumor metastasis, viral infections, and osteoporosis. Because of their role in prominent diseases such as cancer and osteoporosis they are a well-understood family of cell adhesion receptors. Integrins facilitate attachment of the cell to the extracellular matrix (ECM) impacting much of the cellular architecture and function, and act as two-way signaling molecules [Sjaastad and Nelson Citation1997]. Integrins have a short cytoplasmic domain that links to cytoskeletal elements, and when adhesion occurs will induce ‘outside in’ signaling mechanisms, which can feedback ‘inside out’ signaling to regulate integrin function, cytoskeletal assembly, cell behavior, and protein synthesis [Hynes Citation1992].

In addition to integrin subunits conferring specificity, the presence of specific integrin recognition sequences in the ligand determines which integrins will bind the ligand. Fibronectin, collagen, vitronectin, as well as pathogens like viruses and certain snake venom proteins influence cells by binding and signaling through integrins. Although there is diversity in the type of ligand that binds cells through integrins, there is a common element to the ligand motif. Most integrins bind to an element that contains an aspartic acid residue (RGD, ECD, LDV, KGD, RTD, and KQAGD) and they recognize the RGD sequence, which appears in ECM proteins and cell surface molecules [Ruoslahti and Pierschbacher Citation1987], and has been implicated in fertilization [Campbell et al. Citation2000; Sessions et al. Citation2006].

An important feature of integrin receptor binding is the requirement of divalent cations. Media commonly used for in vitro fertilization protocols require Ca2+ and Mg2+ for proper fertilization to occur. Campbell et al. [Citation2000] also demonstrated that the synthetic RGD-containing sequence was unable to generate release of intracellular calcium (a hallmark of fertilization) when used in the absence of divalent cations.

Integrins have been shown to be involved in the process of fertilization [Almeida et al. Citation1995; Bronson and Fusi Citation1990a, Citation1990b]. In 1990, Bronson and Fusi showed that addition of RGD-containing peptides in a heterologous system (human sperm and zona-free hamster oocytes) or a homologous system (hamster sperm and zona-free hamster oocytes) resulted in the complete inhibition of fertilization [Bronson and Fusi Citation1990a, Citation1990b]. Integrins were then found on the plasma membrane of unfertilized murine oocytes and, using a combination of antibody inhibition, peptide inhibition, and somatic cell transfection experiments, the integrin α6β1 complex was shown to serve as a sperm receptor [Almeida et al. Citation1995]. Pig oocytes also consistently express αVβ1 integrin on the vitelline membrane, but not other integrin molecules [Linfor and Berger Citation2000]. These results provide circumstantial evidence that a αVβ1 pig oocyte integrin might interact with a ligand on the sperm plasma membrane during fertilization. Bovine oocytes have also been shown to express integrins on their vitelline membrane. The αV, α6, α4, α2, β1, and β3 integrin subunits were all found on the bovine oocyte vitelline membrane [Pate et al. Citation2007]. Prior results demonstrated that a short synthetic RGD-containing peptide could induce activation and parthenogenetic development in bovine oocytes [Campbell et al. Citation2000]. Because RGD is a known integrin binding ligand, this provides clear evidence in support of integrin-mediated bovine oocyte activation.

In contrast to the experiments mentioned above, a number of publications have indicated that there is no role for integrins, especially α6β1 in fertilization [Miller et al. Citation2000; Barraud-Lange et al. 2007]. Knockout mouse models of α6 and β1, as well as antibody inhibition experiments in which αV and β3 subunits were blocked demonstrated no inhibition of binding or fusion between gametes. It was thus concluded that no integrin subunits known to be on the mouse oocyte or known to bind A Disintegrin and A Mettalloprotease domain (ADAMs) is involved in fertilization [He et al. Citation2003].

Tetraspannins

Integrins bind their ligands relatively loosely compared to other receptors, but are present in much higher concentrations on the surface of cells. In order to compensate for the relatively loose binding, these integrin receptors will cluster together at the site of attachment to increase the strength of binding to the ligands. The complexes of integrin and associated molecules that form these clusters are thought to be organized by members of the tetraspannin family, called a tetraspannin web [Ellerman et al. Citation2003]. Tetraspannins are large proteins with 4 hydrophobic regions and therefore span the plasma membrane four times, with 2 extracellular and 3 cytoplasmic domains. The family contains about 30 members in mammals, several of which have been identified in the mouse oocyte (CD9, CD81, CD82, CD151, SAS, Tspan-3, and Tspan-5). CD9, a tetraspannin signaling molecule known to associate with β1 integrins [Chen et al. Citation1999], has been shown to be involved in the process of sperm-oocyte fusion. Oocytes from female CD9–/–mice were unable to fuse with sperm, and hence were infertile [Miller et al. Citation2000; Miyado et al. Citation2000]. In a subsequent study, sperm fusion to CD9-/-mouse oocytes was restored following expression of human CD9 and mouse CD81 in these animals [Kaji et al. Citation2002]. CD9 has been identified and localized in pig oocytes and it has been determined that both binding and fusion of sperm occur in anti-CD9 antibody treated oocytes [Li et al. Citation2004]. It has also been demonstrated that avidity binding between fertilin beta (ADAM 2) and α6β1 requires cooperation between α6β1 and CD9 [Chen et al. Citation1999]. Though tetraspannins were not originally thought to have a role as receptors, it has recently been demonstrated that a member of the immunoglobulin (IgSF)/CEA subfamily that binds to CD9 called PSG17, is involved in the process of sperm-oocyte fusion [Ellerman et al. Citation2003]. The oocyte receptor complex appears to be far more complex than originally thought and is yet to be completely understood.

Sperm Ligands

There are several families of proteins that are involved in sperm-oocyte interactions leading to oocyte activation. Both fertilin α and β, and snake venom disintegrins, are members of a growing family of proteins known as ADAMs. To date there are 15 ADAM family members described and sequenced at the cDNA level in the guinea pig, monkey, mouse, rabbit, rat, and human [Wolfsberg et al. Citation1995a, Citation1995b]. Fertilin α and β, formerly known as PH-30α and PH-30β, are now referred to as ADAMs 1 and 2 [Huang Citation1998; Wolfsberg et al. Citation1995a, Citation1995b]. Every member of this family has a proteolytic, an adhesion, a fusion, an EGF-like, and signaling domains [Wolfsberg et al. Citation1995a, Citation1995b]. The specific identity of a disintegrin molecule, or other RGD containing protein on the bovine sperm inner acrosomal membrane is still to be determined. Although RGD-containing bovine inner acrosomal membrane proteins that interact with oocyte membrane receptors have been identified, their specific biological significance has not yet been elucidated [Pate et al. Citation2008]. Identification of sperm ligands and intracellular signaling molecules would potentially increase the efficiency of in vitro embryo production by nuclear transfer and could be used as a target for inhibiting fertilization in mammalian populations where this is desirable.

Disintegrins

Disintegrins are peptides of about 70 amino acids that contain many cysteine residues that are involved in intramolecular disulphide bonding. They also contain an Arg-Gly-Asp (RGD) tripeptide sequence that acts as the recognition site of adhesion proteins like integrins [Huang Citation1998]. Disintegrins were first discovered as a component of some snake venoms that operate by inhibiting platelet aggregation by binding to integrin receptors on platelets and thereby blocking binding to fibrinogen. The discription of the effect of snake venom disintegrins was a significant development in the study of integrin-ligand interactions [Huang Citation1998; 38]. Furthermore, these antagonists of ligand-integrin binding are approximately 500–1,000 fold more potent inhibitors of cell adhesion than synthetic linear RGD peptides derived from fibronectin and fibrinogen (known integrin ligands that contain the RGD sequence [Hynes Citation1992]). This immense increase in binding affinity is indicative of a functionally optimal 3-dimensional conformation of the RGD-containing protein [Huang Citation1998], and is important in the function of disintegrins in bovine sperm-oocyte interactions.

Although the RGD domain plays a pivotal role in determining the functional characteristics of the protein, the amino acid composition in and around the RGD tripeptide appears to have an impact on the biological activity and functional properties of RGD-containing peptides. Naturally occurring snake venom disintegrins (RGD-containing) appear to be able to impact fertilization. RGD-containing snake venom disintegrins were used in conjunction with an in vitro fertilization protocol and were shown to competitively inhibit fertilization, while control venoms without the RGD tripeptide have no effect [White et al. Citation2006]. Parthenogenetic development results point to several interesting possibilities about the action of RGD-containing peptides and the mechanism and/or factors involved in oocyte activation [Campbell et al. Citation2000]. Despite the primary role of the RGD domain in determining functional characteristics of the ligand protein, it is becoming clear that secondary contact sites, separate and distinct from the RGD tripeptide, contribute to the binding and functional properties of RGD-containing peptides [Bronson and Fusi Citation1990a; Iwao and Fujimura Citation1996; Lu et al. Citation1994]. Even though the linear RGD-containing peptide used in this work was able to block fertilization and induce intracellular Ca2+ transients, a cyclic RGD peptide or modifying the amino acid environment proximal to the tripeptide RGD may enhance the biological activity of this peptide and may negate the observed requirement for 6-DMAP in the parthenogenetic activation protocol [Campbell et al. Citation2000].

The ability of the RGD-containing peptide to initiate intracellular Ca2+ transients but not full activation as evidenced by parthenogenetic development may suggest the need for two factors to produce “complete” oocyte activation. Based on the totality of the results in the bovine, a reasonable conclusion is that intracellular calcium transients induced by the ligand-receptor pair works in conjunction with a soluble factor produced by and released from the sperm into the oocyte cytoplasm upon fusion with the oocyte, or working with existing PLC isoforms found within the oocyte. This soluble factor might mimic the effects of 6-DMAP, which inhibits protein phosphorylation, and suggest that two signaling pathways are required for “complete” activation [Moses et al. Citation1995; Moses and Masui Citation1995]. It seems highly probable that multiple sites or receptors may be required for the “complete” activation of the bovine oocyte. It is also possible that this requirement for 6-DMAP could be negated by using a more appropriate 3-D structure than the short synthetic linear peptide used by Campbell et al. [Citation2000].

There is a strong correlation between biological activity and the surrounding 3–D structure of the disintegrin [White et al. Citation2006]. Bovine oocytes were responsive to naturally occurring RGD-containing snake venom (kistrin, echistatin, and elegantin) as evidenced by their ability to induce parthenogenetic development to the blastocyst stage regardless of the presence or absence of 6-dimethylaminopurine (6-DMAP). In contrast, the non-RGD-containing venom (erabutoxin b) failed to induce parthenogenetic development at all concentrations evaluated.

Substitutions within the RGD tripeptide sequence, as well as in regions flanking the RGD sequence, affect the ability of RGD to parthenogenetically activate bovine oocytes [Sessions et al. Citation2006]. It is apparent that the amino acids in and around the RGD motif have an effect on the functional properties of the sequence as it relates to fertilization. The peptide resulting in the strongest inhibition of fertilization, PRGDMPPDD, contained two consecutive negatively charged side chains near its negatively charged c-terminus of the peptide, indicating the negatively charged carboxy terminus might allow for a more stable conformation resulting in a better association with the integrin receptor [Sessions et al. Citation2006]. Also, the overall acidic nature of the peptide may have resulted in increased biological activity and better conformation that enabled more efficient binding to the integrin receptor. Sessions et al. [Citation2006] also indicated that synthetic peptides with weakly hydrophobic residues in close proximity to each other and dispersed throughout the peptide resulted in better inhibition of sperm binding. Though evidence exists to demonstrate the importance of surrounding regions in integrin interactions, to date no 3-D disintegrin structure has been shown to be clearly optimal for binding and activation of bovine oocytes.

We recently reported [Pate et al. Citation2008] a list of sperm proteins that interact with receptors on the oocyte plasma membrane. Among those of greatest interest are a bacterial outer membrane (BOMP)-like protein and RIKEN cDNA hypothetical protein. BOMP proteins such as activin mediate the invasion of non-phagocytic eukaryotic cells through beta 1 integrins and utilize focal adhesion kinase and Src-family kinase pathways [Alrutz and Isberg Citation1998]. The RIKEN protein is postulated to have an RGD sequence. Both of these, could mediate integrin binding and signaling events during bovine oocyte activation. Work to determine the specific biological activity of these and other candidate proteins is ongoing.

Fertilin

One of the mammalian sperm proteins thought to be involved in adhesion and fusion of gametes is fertilin. Fertilin is a heterodimeric membrane protein composed of an α and β subunit [Blobel et al. Citation1992]. The fertilin ligand has been linked to sperm-oocyte binding and fusion. Sperm from mice lacking fertilin β are deficient in their ability to adhere to and fuse with oocytes [Frayne and Hall Citation1999; Nishimura et al. Citation2001]. Fertilin β on murine sperm is also known to bind the α6β1 integrin, and requires the CD9 as a co-receptor [Chen et al. Citation1999].

The involvement of sperm fertilin alpha (also known as A Disintegrin and A Metalloprotease 1, ADAM1), fertilin beta (ADAM2), cyritestin (ADAM3), and CRISP 1 (Cystein-Rich Secretory Protein 1) in sperm-oocyte adhesion are demonstrated by studies in which monoclonal antibodies, short peptides, and native or recombinant proteins were used [Frayne and Hall Citation1999; Nishimura et al. Citation2001]. It was demonstrated that fertilin beta-/- and cyritestin-/- mice had a marked inability to adhere to the vitelline membrane. In a few cases where sperm were able to adhere to the vitelline membrane, there was no noticeable reduction in fusion between the gametes [Nishimura et al. Citation2001]. This implicates both fertilin beta and cyritestin as mediators of gamete binding, but not fusion. Because of the disintegrin domain contained within the sequence of these sperm proteins, it has been hypothesized that integrin receptors on the oocyte vitelline membrane are likely to be involved in the process of sperm-oocyte binding, though it is not known whether there is any role in fusion.

Izumo

Recently, a novel protein from the immunoglobulin superfamily was discovered to have a role in fusion of mouse sperm and oocytes. This protein, named Izumo, is a sperm-specific, 56.4–kDa antigen that can be blocked with antibodies to prevent fusion. It was also demonstrated, using Izumo-/- mice, that fusion of gametes did not occur [Inoue et al. Citation2005; Sleight et al. Citation2005]. Although little is presently known about this specific antigen, it has been found in human sperm as well, and may prove to be a critical player in gamete interactions as more information is gathered.

Cyritestin

Several functional studies have provided evidence that fertilin α, fertilin β, and cyritestin participate in the sperm-oocyte interaction [Yuan et al. Citation1997]. Antibodies to these proteins bind to sperm and inhibit fertilization in IVF assays. Recombinant forms of fertilin α, fertilin β, and cyritestin bind to the mouse oocytes plasma membrane, and inhibit sperm-oocyte binding resulting in a reduction of fertilization. Cyritestin (ADAM3) is an ADAM that was first identified in the mouse and monkey [Wolfsberg et al. Citation1995a, Citation1995b]. Studies indicate that the disintegrin domain of fertilin functions in sperm-egg adhesion leading to fusion. These data indicate that a second ADAM family member, cyritestin, may function with fertilin in sperm-egg plasma membrane adhesion and fusion. [Yuan et al. Citation1997].

GPI-Anchored Proteins

A glycosyl phosphatidylinositol (GPI)-anchored protein may be required for fertilization. Studies have shown that removal of GPI-anchored proteins with phosphatidylinositol-specific phospholipase C (PI–PLC) results in the extensive inhibition of sperm-oocyte adhesion and fusion [Coonrod et al. Citation1999a, Citation1999b]. The GPI-anchored proteins appear to have some interaction with the bovine oocyte plasma membrane during fertilization [Pate et al. Citation2008]. Angiotensin-converting enzyme has been demonstrated to have GPI GPI-anchored protein releasing activity and is necessary for proper sperm penetration of the zona pellucida in mice and is localized to the acrosomal region of mouse sperm [Saha et al. Citation2000]. A role for this protein in bovine fertilization is being determined.

The specific identity of a disintegrin, ADAM, or other RGD containing protein on the sperm inner acrosomal membrane is still to be determined. The identification of sperm membrane proteins that bind to the vitelline membrane protein should lead to an understanding of intracellular signals, processes, and pathways that are involved in bovine oocyte activation. Identification of sperm ligands and intracellular signaling molecules will potentially increase the efficiency of in vitro embryo production by nuclear transfer, increase efficiency of ICSI, or help in the reduction of species in which overpopulation is a concern. Intracellular signaling pathways will help to clarify and confirm previously published data [Campbell et al. Citation2000; Sessions et al. Citation2006] that has demonstrated receptor-mediated oocyte activation in the bovine model.

Intracellular Signaling Pathways

When integrins bind to form cell-matrix or cell-cell interactions they cluster together. As the integrins cluster on the plasma membrane to initiate a signal, a direct consequence of this process is the recruitment of a cytoplasmic tyrosine kinase (CTK) referred to as focal adhesion kinase (FAK). The association of integrins to various different intracellular elements results in the recruitment and clustering of FAK enzymes following binding to their ligands; making FAK an important component of the integrin-regulated signaling mechanism. These two proteins may have a role in communication across cell-matrix and cell-cell junctions. FAK is known to be involved in the regulation of N-cadherin-based cell-cell adhesion [Schaller Citation2004; Yano et al. Citation2004]. FAK molecules cross-phosphorylate each other on tyrosine residues which act as attachment sites for various CTKs from the Src family; the Src family kinases are responsible for phosphorylating other tyrosines on FAK as well as other proteins that have been recruited to the focal adhesion, thereby activating them. FAK is considered to be a regulator of focal adhesions and once activated, initiate many intracellular signaling pathways [Parsons et al. Citation2000].

It is hypothesized that integrins mediate sperm-oocyte interactions and thus a variety of CTKs, including FAK and the Src family, are implicated for a possible role in oocyte activation. Tyrosine kinase involvement in oocyte activation pathways has also been detected in mouse oocytes [Mori et al. Citation1992], pig oocytes [Kim et al. Citation1999], and Xenopus eggs [Abassi et al. Citation2000; Abassi and Foltz Citation1994; Moore and Kinsey Citation1995]. Although the majority of results to date indicate that one or more tyrosine kinases are involved, it is not yet unclear which specific kinase is involved, and their complete role in mammalian fertilization remains to be elucidated.

The largest family of cell-surface receptors in eukaryotes is G-protein-linked receptors. When extracellular signaling molecules bind receptors, the receptors undergo a conformational change that activates G-proteins. G-proteins are trimers composed of α, β, and γ subunits. There are several known isoforms of alpha subunits, which are used to classify the various G-protein signaling trimers. Activation of a G-protein follows induction of the α subunit to exchange a bound GDP molecule for a GTP molecule resulting in a production of an α subunit and a βγ subunit. Each type of α subunit and each βγ subunit can act as a signaling molecule, targeting specific enzymes. Gs α can activate Ca2+ channels, while Go βγ can inactivate Ca2+ channels. Several subunits can also activate phospholipase isoforms. In 1994 it was reported that injection of guanosine 5′–0–(2-thiodiphosphate) (GDPβ[S]), a G-protein antagonist, into fertilized rabbit oocytes resulted in inhibition of intracellular Ca2+ oscillations [Fissore and Robl Citation1994]. GDPβ[S] is a non-hydrolizable GDP analog that competitively inhibits G-protein activation by GTP. It was hypothesized that G-proteins were possibly involved in the production of IP3 [Fissore and Robl Citation1994]. Acetylcho-line, known to interact with plasma membrane-coupled G-protein receptors [Williams et al. Citation1992], and injection of GTPγ[S], an activator of G-proteins [Swann and Whitaker Citation1990], elicits Ca2+i oscillations. An exogenously added rat M1 muscarinic receptor mediated porcine oocyte activation by a G protein coupled signal transduction pathway and led to oocyte activation [Kim et al. Citation1998]. The Gβγ subunit is responsible for the modulation of IP3 binding to IP3 receptors (IP3R) and stabilizes IP3Rs in a channel conformation [Zeng et al. Citation2003]. Zeng et al. [Citation2003] suggested Gβγ as an alternative to IP3 in activating IP3R. Our preliminary data also indicate that G-proteins are functional in bovine oocyte development, as oocytes microinjected with the GDPβ[S] inhibitor do not cleave as often as control groups. More specific inhibitors of G-protein sub-units can be used to determine which subunits are specifically involved in fertilization pathways.

We recently published data that implicated tyrosine kinases, particularly Src-family kinases and focal adhesion kinase (FAK) in bovine oocyte activation. Of particular note is the involvement of FAK because of its relationship to integrin signaling pathways. FAK levels were reported to be relatively high in mature bovine oocytes compared to 2, 4, or 8-cell embryos. FAK levels then rise sharply during morula and blastocyst stages as would be expected as cell-cell interactions become necessary for differentiation of blastomeres. The fact that FAK levels are high in the mature oocyte could be an indication that they are necessary for fertilization. The requirement for FAK in bovine oocyte activation was demonstrated by using a dominant competitive inhibitor (FRNK) of FAK to block release of intracellular calcium (; [Pate et al. 2009]). Pretreatment of bovine oocytes with FRNK at a final concentration of 12 μM (estimated IC50 = 120 pM) completely blocked calcium transients following fertilization by sperm. Further evidence of the involvement of FAK in the activation mechanism was observed following pretreatment of bovine oocytes with specific antibody directed against FAK which also inhibited calcium transients in fertilized oocytes to unfertilized control levels [Pate et al. 2009]. The apparent involvement of FAK, as well as other tyrosine kinases, specifically from the Src family, is consistent with the integrin-mediated oocyte activation hypothesis.

Table 1.  Involvement of FAK: Focal Adhesion Kinase; PLC: phospholipase C; and SRC: proto-oncogenic tyrosine kinase in Induction of Activation-Associated Calcium Transients After Fertilization by Sperm. FRNK: Focal Adhesion-Related Non-Kinase; PP1: 1 –(1,1 Dimethylethyl)–1–(4–methylphenyl)-1 H-pyrazolo[3, 4–d]-pyrimidin-4-amine.

CONCLUSIONS

A significant amount of data has been generated to elucidate the specific molecules that are involved in interactions between gametes of many species. Most of this data was collected using mouse, Xenopus, and ascidian models. Many of the findings have been contradictory, and there is certainly no clear-cut demonstration that specific molecules are or are not involved in sperm-oocyte binding, fusion, activation, or development. Recent findings demonstrate the involvement of receptors that are known to associate in large, multimeric complexes. This fact partially explains why it has been so difficult to specifically identify molecules and their particular role. It also leads one to speculate that the process of oocyte activation by the sperm cell is a highly complex and elaborate process that involves many more players than perhaps was initially expected. Investigation of the bovine sperm proteome should reveal putative binding molecules and the biological activity of these ligands could be identified, shedding light on this critical mechanism.

Declaration of Interest: The authors report no conflicts of interest. The authors alone are responsible for the content and writing of the paper.

REFERENCES

  • Abassi, Y. A., Carroll, D. J., Giusti, A. F., Belton, R. J., Jr and Foltz, K. R. (2000) Evidence that Src-type tyrosine kinase activity is necessary for initiation of calcium release at fertilization in sea urchin eggs. Dev Biol 218(2):206–219.
  • Abassi, Y. A. and Foltz, K. R. (1994) Tyrosine phosphorylation of the egg receptor for sperm at fertilization. Dev Biol 164(2):430–443.
  • Abdalla, H., Shimoda, M., Hirabayashi, M. and Hochi, S. (2009) A combined treatment of ionomycin with ethanol improves blastocyst development of bovine oocytes harvested from stored ovaries and microinjected with spermatozoa. Theriogenology. May 20. [Epub ahead of print].
  • Almeida, E. A., Huovila, A. P., Sutherland, A. E., Stephens, L. E., Calarco, P. G., Shaw, L. M., Mercurio, A. M., Sonnenberg, A., Primakoff, P., Myles, D. G., et al. (1995) Mouse egg integrin alpha 6 beta 1 functions as a sperm receptor. Cell 81(7):1095–1104.
  • Alrutz, M. A. and Isberg, R. R. (1998) Involvement of focal adhesion kinase in invasin-mediated uptake. Proc Natl Acad Sci USA 95(23):13658–13663.
  • Berridge, M. J. and Galione, A. (1988) Cytosolic calcium oscillators. Faseb J 2(15):3074–3082.
  • Blobel, C. P., Wolfsberg, T. G., Turck, C. W., Myles, D. G., Primakoff, P. and White, J. M. (1992) A potential fusion peptide and an integrin ligand domain in a protein active in sperm-egg fusion. Nature 356(6366):248–252.
  • Bronson, R. A. and Fusi, F. (1990a) Evidence that an Arg-Gly-Asp adhesion sequence plays a role in mammalian fertilization. Biol Reprod 43(6):1019–1025.
  • Bronson, R. A. and Fusi, F. (1990b) Sperm-oolemmal interaction: role of the Arg-Gly-Asp (RGD) adhesion peptide. Fertil Steril 54(3):527–529.
  • Campbell, K. D., Reed, W. A. and White, K. L. (2000) Ability of integrins to mediate fertilization, intracellular calcium release, and parthenogenetic development in bovine oocytes. Biol Reprod 62(6):1702–1709.
  • Carroll, D. J., Ramarao, C. S., Mehlmann, L. M., Roche, S., Terasaki, M. and Jaffe, L. A. (1997) Calcium release at fertilization in starfish eggs is mediated by phospholipase Cgamma. J Cell Biol 138:1303–1311.
  • Carroll, D. J., Albay, D. T., Terasaki, M., Jaffe, L. A. and Foltz, K. R. (1999) Identification of PLCγ-dependent and independent events during fertilization of sea urchin eggs. Dev Biol 206:232–247.
  • Chen, M. S., Tung, K. S., Coonrod, S. A., Takahashi, Y., Bigler, D., Chang, A., Yamashita, Y., Kincade, P. W., Herr, J. C. and White, J. M. (1999) Role of the integrin-associated protein CD9 in binding between sperm ADAM 2 and the egg integrin alpha6beta1: implications for murine fertilization. Proc Natl Acad Sci USA 96(21):11830–11835.
  • Chung, J. T., Keefer, C. L. and Downey, B. R. (2000) Activation of bovine oocytes following intracytoplasmic sperm injection (ICSI). Theriogenology Apr 1 53(6):1273–1284.
  • Coonrod, S., Naaby-Hansen, S., Shetty, J. and Herr, J. (1999a) PI-PLC releases a 25–40 kDa protein cluster from the hamster oolemma and affects the sperm penetration assay. Mol Hum Reprod 5(11):1027–1033.
  • Coonrod, S. A., Naaby-Hansen, S., Shetty, J., Shibahara, H., Chen, M., White, J. M. and Herr, J. C. (1999b) Treatment of mouse oocytes with PI-PLC releases 70-kDa (pI 5) and 35- to 45-kDa (pI 5.5) protein clusters from the egg surface and inhibits sperm-oolemma binding and fusion. Dev Biol 207(2):334–349.
  • Devito, L. G., Fernandes, C. B., Blanco, I., Tsuribe, P. M., Landim-Alvarenga, F. C., 2009. Use of a Piezo Drill for Intracytoplasmic Sperm Injection into Cattle Oocytes Activated with Ionomycin Associated with Roscovitine. Reprod Domest Anim. Jan 8. [Epub ahead of print].
  • Ellerman, D. A., Ha, C., Primakoff, P., Myles, D. G. and Dveksler, G. S. (2003) Direct binding of the ligand PSG17 to CD9 requires a CD9site essential for sperm-egg fusion. Mol Biol Cell 14(12):5098–5103.
  • Fissore, R. A. and Robl, J. M. (1994) Mechanism of calcium oscillations in fertilized rabbit eggs. Dev Biol 166(2):634–642.
  • Frayne, J. and Hall, L. (1999) Mammalian sperm-egg recognition: does fertilin beta have a major role to play? Bioessays 21(3):183–187.
  • He, Z. Y., Brakebusch, C., Fassler, R., Kreidberg, J. A., Primakoff, P. and Myles, D. G. (2003) None of the integrins known to be present on the mouse egg or to be ADAM receptors are essential for sperm-egg binding and fusion. Dev Biol 254(2):226–237.
  • Huang, T. F. (1998) What have snakes taught us about integrins? Cell Mol Life Sci 54(6):527–540.
  • Hynes, R. O. (1992) Integrins: versatility, modulation, and signaling in cell adhesion. Cell 69(1):11–25.
  • Igarashi, H., Knott, J. G., Schultz, R. M. and Williams, C. J. (2007) Alterations of PLCβ1 in mouse eggs change calcium oscillatory behavior following fertilization. Dev. Biol. 312(1):321–330.
  • Inoue, N., Ikawa, M., Isotani, A. and Okabe, M. (2005) The immuno-globulin superfamily protein Izumo is required for sperm to fuse with eggs. Nature 434(7030):234–238.
  • Iwao, Y. and Fujimura, T. (1996) Activation of Xenopus eggs by RGD-containing peptides accompanied by intracellular Ca2+ release. Dev Biol 177(2):558–567.
  • Jones, K. T., Soeller, C. and Cannell, M. B. (1998) The passage of Ca2+ and fluorescent markers between the sperm and egg after fusion in the mouse. Development 125(23):4627–635.
  • Kaji, K., Oda, S., Miyazaki, S. and Kudo, A. (2002) Infertility of CD9-deficient mouse eggs is reversed by mouse CD9, human CD9, or mouse CD81; polyadenylated mRNA injection developed for molecular analysis of sperm-egg fusion. Dev Biol 247(2):327–334.
  • Kim, J. H., Do, H. J., Wang, W. H., Machaty, Z., Han, Y. M., Day, B. N. and Prather, R. S. (1999) A protein tyrosine phosphatase inhibitor, sodium orthovanadate, causes parthenogenetic activation of pig oocytes via an increase in protein tyrosine kinase activity. Biol Reprod 61(4):900–905.
  • Kim, S. J., Koh, E. M., Kang, T. M., Kim, Y. C., So, I., Isenberg, G. and Kim, K. W. (1998) Ca2+ influx through carbachol-activated non-selective cation channels in guinea-pig gastric myocytes. J Physiol 513(Pt 3):749–760.
  • Kirkman-Brown, J. C., Barratt, C. L. and Publicover, S. J. (2004) Slow calcium oscillations in human spermatozoa. Biochem J 378(Pt 3):827–832.
  • Kirkman-Brown, J. C., Bray, C., Stewart, P. M., Barratt, C. L. and Publicover, S. J. (2000) Biphasic elevation of [Ca(2 + )]?〈i〉 in individual human spermatozoa exposed to progesterone. Dev Biol 222(2):326–335.
  • Kirkman-Brown, J. C., Punt, E. L., Barratt, C. L. and Publicover, S. J. (2002) Zona pellucida and progesterone-induced Ca2+ signaling and acrosome reaction in human spermatozoa. J Androl 23(3):306–315.
  • Kline, D. and Kline, J. T. (1992) Repetitive calcium transients and the role of calcium in exocytosis and cell cycle activation in the mouse egg. Dev Biol 149(1):80–89.
  • Lawrence, Y., Whitaker, M. and Swann, K. (1997) Sperm-egg fusion is the prelude to the initial Ca2+| increase at fertilization in the mouse. Development 124(1):233–241.
  • Li, Y. H., Hou, Y., Ma, W., Yuan, J. X., Zhang, D., Sun, Q. Y. and Wang, W. H. (2004) Localization of CD9 in pig oocytes and its effects on sperm-egg interaction. Reproduction 127(2):151–157.
  • Linfor, J. and Berger, T. (2000) Potential role of alphav and beta1 integrins as oocyte adhesion molecules during fertilization in pigs. J Reprod Fertil 120(1):65–72.
  • Lu, X., Williams, J. A., Deadman, J. J., Salmon, G. P., Kakkar, V. V., Wilkinson, J. M., Baruch, D., Authi, K. S. and Rahman, S. (1994) Preferential antagonism of the interactions of the integrin alpha IIb beta 3 with immobilized glycoprotein ligands by snake-venom RGD (Arg-Gly-Asp) proteins Evidence supporting a functional role for the amino acid residues flanking the tripeptide RGD in determining the inhibitory properties of snake-venom RGD proteins. Biochem J 304(Pt 3):929–936.
  • Malcuit, C., Maserati, M., Takahashi, Y., Page, R. and Fissore, R. A. (2006) Intracytoplasmic sperm injection in the bovine induces abnormal [Ca2 + ]i responses and oocyte activation. Reprod Fertil Dev 18(1–2):39–51.
  • Marquez, B. and Suarez, S. S. (2004) Different signaling pathways in bovine sperm regulate capacitation and hyperactivation. Biol Reprod 70(6):1626–1633.
  • Miller, B. J., Georges-Labouesse, E., Primakoff, P. and Myles, D. G. (2000) Normal fertilization occurs with eggs lacking the integrin alpha6beta1 and is CD9-dependent. J Cell Biol 149(6):1289–1296.
  • Miyado, K., Yamada, G., Yamada, S., Hasuwa, H., Nakamura, Y., Ryu, F., Suzuki, K., Kosai, K., Inoue, K., Ogura, A., Okabe, M. and Mekada, E. (2000) Requirement of CD9 on the egg plasma membrane for fertilization. Science 287(5451):321–324.
  • Moore, K. L. and Kinsey, W. H. (1995) Effects of protein tyrosine kinase inhibitors on egg activation and fertilization-dependent protein tyrosine kinase activity. Dev Biol 168(1):1–10.
  • Mori, T., Gou, M. W., Yoshida, H., Saito, S. and Mori, E. (1992) Expression of the signal transducing regions of CD4-like and lck genes in murine egg. Biochem Biophys Res Commun 182(2):527–533.
  • Moses, R. M., Kline, D. and Masui, Y (1995) Maintenance of meta-phase in colcemid-treated mouse eggs by distinct calcium- and 6-dimethylaminopurine (6-DMAP)-sensitive mechanisms. Dev Biol 167(1):329–337.
  • Moses, R. M. and Masui, Y (1995) Metaphase arrest in newly matured or microtubule-depleted mouse eggs after calcium stimulation. Zygote 3(1):1–8.
  • Nishimura, H., Cho, C., Branciforte, D. R., Myles, D. G. and Primakoff, P. (2001) Analysis of loss of adhesive function in sperm lacking cyritestin or fertilin beta. Dev Biol 233(1):204–213.
  • Oikawa, T., Takada, N., Kikuchi, T., Numabe, T., Takenaka, M. and Horiuchi, T. (2005) Evaluation of activation treatments for blastocyst production and birth of viable calves following bovine intracytoplasmic sperm injection. Anim Reprod Sci 86(3–4):187–194.
  • Parsons, J. T., Martin, K. H., Slack, J. K., Taylor, J. M. and Weed, S. A. (2000) Focal adhesion kinase: a regulator of focal adhesion dynamics and cell movement. Oncogene 19(49):5606–5613.
  • Pate, B. J., White, K. L., Winger, Q. A., Rickords, L. F., Aston, K. I., Sessions, B. R., Li, G. P., Campbell, K. D., Weimer, B. and Bunch, T. D. (2007) Specific Integrin Subunits In Bovine Oocytes, Including Novel Sequences For Alpha 6 And Beta 3 Subunits. Mole. Reprod. Devel. 74:600–607.
  • Pate, B. J., White, K. L., Chen, D., Aston, K. I., Sessions, B. R., Bunch, T. D. and Weimer, B. C. (2008) A Novel Approach To Identify Bovine Sperm Membrane Proteins That Interact With Receptors On The Vitelline Membrane Of Bovine Oocytes. Mole. Reprod. Devel. (Published online September 20, 2007) 75:641–649.
  • Pate, B. J., White, K. L., DeWald, D. B., Rickords, L. F., Li, G-P., Aston, K. I., Sessions, B. R. and Bunch, T. D. (2010) Involvement of Tyrosine Kinases, Specifically Src Family Kinases, Focal Adhesion Kinase (FAK), and Agonist-Induced PLC in the Activation and Development of Bovine Oocytes. In Press.
  • Ross, J. P., Beyhan, Z., Iager, A. E., Yoon, S. Y., Malcuit, C., Schellander, K., Fissore, R. A. and Cibelli, J. B. (2008) Parthenogenetic activation of bovine oocytes using bovine and murine phospholipase C zeta. BMC Developmental Biology 8:16.
  • Ross, J. P., Rodriguez, R. M., Iager, A. E., Beyhan, Z., Wang, K., Ragina, N. P., Yoon, S. Y., Fissore, R. A. and Cibelli, J. B. (2009) Activation of bovine somatic cell nuclear transfer embryos by PLCZ cRNA injection. Reproduction 137:427–437.
  • Runft, L. L., Jaffe, L. and Mehlmann, L. (2002) Egg activation at fertilization: where it all begins. Dev. Biol. 245:237–254.
  • Ruoslahti, E. and Pierschbacher, M. D. (1987) New perspectives in cell adhesion: RGD and integrins. Science 238(4826):491–497.
  • Saha, L., Garg, S. K., Bhargava, V. K. and Mazumdar, S. (2000) Role of angiotensin-converting enzyme inhibitor, lisinopril, on spermatozoal functions in rats. Methods Find Exp Clin Pharmacol 22(3):159–162.
  • Saunders, C. M., Larman, M. G., Parrington, J., Cox, L. J., Royse, J., Blayney, L. M., Swann, K. and Lai, F. A. (2002) PLC zeta: a sperm-specific trigger of Ca(2 + ) oscillations in eggs and embryo development. Development 129(15):3533–3544.
  • Schaller, M. D. (2004) FAK and paxillin: regulators of N-cadherin adhesion and inhibitors of cell migration? J Cell Biol 166(2):157–159.
  • Sessions, B. R., Aston, K. I., Davis, A. P., Pate, B. J. and White, K. L. (2006) Effects of amino acid substitutions in and around the arginine-glycine-aspartic acid (RGD) sequence on fertilization and partheno-genetic development in mature bovine oocytes. Mol Reprod Dev 73(5):651–657.
  • Sjaastad, M. D. and Nelson, W. J. (1997) Integrin-mediated calcium signaling and regulation of cell adhesion by intracellular calcium. Bioessays 19(1):47–55.
  • Sleight, S. B., Miranda, P. V., Plaskett, N. W., Maier, B., Lysiak, J., Scrable, H., Herr, J. C. and Visconti, P. E. (2005) Isolation and proteomic analysis of mouse sperm detergent-resistant membrane fractions: evidence for dissociation of lipid rafts during capacitation. Biol Reprod 73(4):721–729.
  • Stith, B., Stafford, J., Chang, Y., Kane, T., Bates, R., Nickle, A. and Petcoff, D. (2005) The role of phosphatidic acid in regulation of intracellular calcium during fertilization in xenopus. The American Society for Cell Biology 45th Annual Meeting, San Francisco, CA, USA. Abstract Number 515.
  • Sutner, R., Zakhartchenko, V., Stojkovic, P., Mller, S., Alberio, R., Medjugorac, I., Brem, G., Wolf, E. and Stojkovic, M. (2000) Intra-cytoplasmic sperm injection in bovine: effects of oocyte activation, sperm pretreatment and injection technique. Theriogenology. Oct 1 54(6):935–948.
  • Swann, K. (1994) Ca2 + oscillations and sensitization of Ca2 + release in unfertilized mouse eggs injected with a sperm factor. Cell Calcium 15(4):331–339.
  • Swann, K. and Whitaker, M. J. (1990) Second messengers at fertilization in sea-urchin eggs. J Reprod Fertil Suppl 42:141–153.
  • Tang, T. S., Dong, J. B., Huang, X. Y. and Sun, F. Z. (2000) Ca(2 + ) oscillations induced by a cytosolic sperm protein factor are mediated by a maternal machinery that functions only once in mammalian eggs. Development. Mar 127(5):1141–1150.
  • Tesarik, J., Sousa, M. and Testart, J. (1994) Human oocyte activation after intracytoplasmic sperm injection. Hum Reprod 9(3):511–518.
  • Tokmakov, A. A., Sato, K. I., Iwasaki, T. and Fukami, Y. (2002) Src kinase induces calcium release in Xenopus egg extracts via PLCγ and IP3-dependent mechanism. Cell Calcium 32:11–20.
  • Townley, I. K., Raisch, M., Reade, A. and Foltz, K. (2005) Characterization of Src family kinase signaling in echinoderm egg activation. The American Society for Cell Biology 45th Annual Meeting, San Francisco, CA, USA. Abstract Number 530.
  • White, K., Passipieri, M., Bunch, T., Campbell, K. and Pate, B. (2006) Effects of Arginine-Glycine-Aspartic Acid (RGD) Containing Snake Venom Peptides on Parthenogenetic Development and In Vitro Fertilization of Bovine Oocytes. Mol Reprod Dev 74:88–96.
  • Williams, C. J., Schultz, R. M. and Kopf, G. S. (1992) Role of G proteins in mouse egg activation: stimulatory effects of acetylcholine on the ZP2 to ZP2f conversion and pronuclear formation in eggs expressing a functional m1 muscarinic receptor. Dev Biol 151(1):288–296.
  • Wolfsberg, T. G., Primakoff, P., Myles, D. G. and White, J. M. (1995a) ADAM, a novel family of membrane proteins containing A Disintegrin And Metalloprotease domain: multipotential functions in cell-cell and cell-matrix interactions. J Cell Biol 131(2):275–278.
  • Wolfsberg, T. G., Straight, P. D., Gerena, R. L., Huovila, A. P., Primakoff, P., Myles, D. G. and White, J. M. (1995b) ADAM, a widely distributed and developmentally regulated gene family encoding membrane proteins with a disintegrin and metalloprotease domain. Dev Biol 169(1):378–383.
  • Wu, A. T., Sutovsky, P., Manandhar, G., Xu, W., Katayama, M., Day, B. N., Park, K. W., Yi, Y. J., Xi, Y. W., Prather, R. S. and Oko, R. (2007) PAWP, a sperm-specific WW domain-binding protein, promotes meiotic resumption and pronuclear development during fertilization. J Biol Chem. 2007 Apr 20 282(16):12164–12175.
  • Yano, H., Mazaki, Y., Kurokawa, K., Hanks, S. K., Matsuda, M. and Sabe, H. (2004) Roles played by a subset of integrin signaling molecules in cadherin-based cell-cell adhesion. J Cell Biol 166(2):283–295.
  • Yuan, R., Primakoff, P. and Myles, D. G. (1997) A role for the disintegrin domain of cyritestin, a sperm surface protein belonging to the ADAM family, in mouse sperm-egg plasma membrane adhesion and fusion. J Cell Biol 137(1):105–112.
  • Zeng, W., Mak, D. O., Li, Q., Shin, D. M., Foskett, J. K. and Muallem, S. (2003) A new mode of Ca2+ signaling by G protein-coupled receptors: gating of IP3 receptor Ca2+ release channels by Gbetagamma. Curr Biol 13(10):872–876.

Reprints and Corporate Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

To request a reprint or corporate permissions for this article, please click on the relevant link below:

Academic Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

Obtain permissions instantly via Rightslink by clicking on the button below:

If you are unable to obtain permissions via Rightslink, please complete and submit this Permissions form. For more information, please visit our Permissions help page.