Home  |  Archive  |  AJA @ Nature  |  Online Submission  |  News & Events  |  Subscribe  |  APFA  |  Society  |  Links  |  Contact Us  |  中文版

Molecular aspects of mammalian fertilization

Hector Serrano1,  Dolores Garcia-Suarez2

1Dept. Health Sciences, 2Dept. Biology, Universidad Autonma Metropolitana-Iztapalapa, Ave. Michoacan and Purisima, Mexico City, DF 09340, Mexico

Asian J Androl  2001 Dec; 3: 243-249


Keywords:  fertilization; sperm maturation; sperm capacitation; sperm-ovum interactions; membrane fusion
Abstract

Mammalian fertilization is a highly regulated process, much of which are not clearly  understood. Here we present some information in order to elaborate a working hypothesis for this process, beginning with the sperm modifications in the epidydimis up to sperm and egg plasmalemma interaction and fusion. We also discuss the still poorly understood capacitation process, the phenomenon of sperm chemo-attraction that brings the capacitated sperm to interact with the oocyte vestments and certain aspects of the acrosome reaction.

1 Introduction

Fertilization is a process that is the end of one developmental process and the beginning of another process. On the one side, it is the end of the oogenesis and spermatogenesis processes. On the other side, it is the starting point of a new organism. Several aspects of the fertilization process have been described by using different techniques ranging from simple microscopical analysis to knock out genes and transgenic animals. Much of our actual knowledge at the molecular level has been developed in the last two decades but regardless the large amountresearch effort, there are some aspects that are still poorly understood. In this review, we are not trying to introduce all known information of mammalian fertilization that has been covered by other authors[1,2],  but to assemble some described models for different topics of the process from the spermatozoa perspective, in order to make them understandable and to detect the still uncovered areas in each model.

2 Sperm maturation and epididymis

The glycoproteins on the sperm plasma membrane are different in origin. Galactosyltransferase, an enzyme participating in gamete interaction in the rat, is located on the plasma membrane of spermatocytes and the postacrosomal region of tubular spermatozoa and later on the acrosome region of the sperm recovered from the female genital tract[3]. Other proteins are not produced by the spermatocytes but are secreted by the epithelial cells lining the epididymis, like the DE protein in the rat[4-6], or by the male accessory glands or the female oviduct, like the oviduct-specific glycoprotein (OSP) that is important in both fertilization and early embryonic development[7].  The secretory and absorptive activities of the epithelia lining the epididymis are responsible for the surface sculpturing of the mammalian sperm. Gene expression along the epididymis actively participates in this change in shape and characteristics of the mammalian spermatozoa[8].

The relatively diverse origin of the proteins actually lead to the establishment of sperm membrane domains responsible for different sperm capabilities during the fertilization process. Some molecules fixed on the sperm membrane during spermiogenesis can be processed later in the prostate or the female genital tract in such a way that this new conformation can acquire a function that otherwise can not be achieved, as is the case of fibronectin, which is exposed on the human sperm surface only after capacitation has occurred[9]. 

These modifications add another dimension to the process since it is made in the right place at the right time so the molecule becomes fully functional only when needed. In studying these modifications, several updated techniques have been used. For example, change in terminal sugar has been detected by labeled lectins[10,11] or specific antibodies[6,12,13].

3 Capacitation and transport in the female genital tract

When sperm are deposited into the female genital tract, they interact with different compounds. Some of these compounds are in the prostatic secretion. Some others, like mucins, are secreted at various parts of the female genital tract and some of them actually combines with those in the prostatic secretions present in ejaculates, composing the first barrier that the sperm will be gradually released.

In most eutheria, even when several million sperm are deposited in the initial parts of the female tract, only a small part are able to be liberated from the initial barrier. Some species like bull, cattle, pigs, rabbits, mouse and hamsters have anatomically or functionally specialized sperm reservoirs[14-16]. In bulls, recent studies have demonstrated that modification or loss of a sperm membrane peripheral protein acquired from the seminal plasma[14] through  interacting with the fructose residues on the epithelial cell membrane is responsible for sperm liberation from the functional storage at different times[16]. The liberated sperm then continue their journey up to the fertilization site[15].

Once the spermatozoa have been released, they are subjected to different physiological processes. One of these is capacitation, which is initiated almost at the same time the sperm is ejaculated. During the sperm-epithelial cells interaction, there are lipid exchange and proteolytic and carbohydrate modification of the sperm surface. Evidences for most of the processes are derived from the requirements of spermatozoa in order to successfully interact with the oocyte[17].

During capacitation, the sperm undergo several characteristic biochemical and morphological changes. Chlortetracycline staining[18], for example, is able to demonstrate the capacitation status as well as to evaluate the acrosome reaction[19].

Albumin is a characteristic component of in vitro capacitation media that presumably removes the cholesterol from the sperm plasma membrane, thus increasing the membrane fluidity[20]. Therien and co-workers[21] recently proposed that in regard to lipid exchange, bull sperm are subjected to two cholesterol effluxes. One occurs after a short interaction with the seminal plasma where the bovine seminal plasma proteins (BSP) remove a significantly large amount of cholesterol and a small amount of other phospholipids that initially destabilizes (primes) the membrane. These BSP proteins remain attached to the sperm plasma membrane through their interactions with choline phospholipids that prevents further phospholipid movement. In the female genital tract, a second protein, high density lipoprotein from the follicular fluid (FF-HDL), removes the BSP proteins from the plasmalema so the cholesterol and other phospholipids can be removed by low affinity proteins like albumin.

During capacitation, there are changes on the sperm surface related to the modification or releasing of molecules due to the action of modifying or hydrolyzing enzymes in such a way that new antigenic determinants are detected. From the time-based definition of capacitation[22] to its biochemical-physiological aspects[19], our knowledge on capacitation is far from complete.

Calmodulin plays a major role in the capacitation as the process is dependent on extracellular Ca2+. In mouse, calmodulin possibly activates adenylyl cyclase but inhibition of calmodulin does not affect the tyrosine phosphorylation characteristic of capacitation[23]. This controversial result may be explained by the following observation. cAMP is necessary for the activation of certain kinases, but it can effectively act at low concentration that is not affected by calmodulin-cyclase interaction. Calmodulin inhibition also impairs other capacitation indicator. Calmodulin plays an important role in the whiplash bending of the sperm tail known as hyperactivation, which is not cAMP-dependent[23].

In hamster sperm, capacitation-associated changes depend mainly on the bicarbonate-specific transporter associated to the cAMP metabolism. Capacitation is also related to protein phosphorylation by the cAMP-dependent protein kinase (PKA) and some other pathway, however, this kind of activation is not enough to obtain fully capacitated spermatozoa.

Reactive oxygen species (ROS) do regulate oxidation-reduction processes needed for protein phosphorylation. Presence of nitric oxide (NO) affects the most obvious signals of capacitation, i.e., hyperactivated motility and egg-envelope interaction[24]. Recently it has been demonstrated that NO correlates with the protein phosphorylation level in human spermatozoa. It has been proposed that NO and cAMP interaction has a stimulatory effect on specific tyrosine-phosphorylation.

NO could stimulate tyrosine-phosphorylation via a trans-activation of a cGMP-dependent protein kinase or through inhibiting the cAMP degradation by phosphodiesterases[25].

4 Sperm maintaining and attracting substances

In the female genital tract spermatozoa has to be maintained by different energetic sources. Vaginal, tubal and follicular secretions modify not only sperm surface characteristics but also their motility and responsiveness. In vitro assays have tried to emulate some of the different environments the sperm found along its travel to the fertilization site[26].

Sperm movement through the female genital tract has been associated with both physical and chemical entities. Positive rheology has been demonstrated in bull and other sperm[27] and has been the basis for several techniques to assess sperm attracting activity in follicular fluid[28]. These assays have revealed that sperm attracting activity were associated to molecules much different in nature and origin, such as progesterone[29,30] and substance P[31].  Follicular fluid has been shown to contain sperm attracting activity[32-36] correlated with capacitation[37]. Several candidates have been proposed to explain the fact or responsible for this activity, e.g., hyaluronic acid[38], an uncharacterized heat-stable, low molecular weight protein[39] and an 8.6 kilodalton protein whose N-terminal residues resemble those of the apolipoprotein b2[40].

5 Acrosome reaction

Acrosome reaction is a highly specialized event observed in the capacitated spermatozoa.  Although it has been extensively studied and there are several techniques available for its evaluation[18,41-43], the molecular events that trigger the acrosome reaction have not been fully elucidated. Gamete interaction is the induction signal for this exocytic event and it is admitted that the natural acrosome reaction inducer is the zona pellucida. In vitro studies also indicate that factors from the follicular fluid, mainly progesterone(P4), are able to induce the reaction. At least two stimulatory transduction pathways have been implicated.

Let's start with the P4-induced acrosome reaction. After the sperm has traveled up to the isthmus, the interaction that induces sperm membrane changes modifies the structure of previously unavailable determinants to interact. Progesterone interaction with sperm is different from that usually shown in other P4-responsive cells. By using fluorescence-labeled P4 probes, several groups have found a specific receptor located on the plasmalemma of different sperm species from mouse to pigs and bulls[44].

Membrane progesterone receptor is not a G-protein coupled receptor. P4-receptor interaction increases the intracellular Ca2+ concentration as well as the long lasting increase in Na+ concentration due to a flunarizine-sensitive channel[45].

Progesterone stimulation has been revealed in mouse sperm by an intracellular Ca2+ increase. There is controversial evidence about the type of Ca2+ channel involved. Some authors suggest that the rise in Ca2+ concentration is not due to the action of the T-type Ca2+ channel but another one also activated by cGMP[46], whereas others suggest that there are other channels acting during the zona pellucida-induced acrosome reaction[45,47]. The P4 receptor acts first via a cAMP-dependent protein kinase (PKA) and then by an A-kinase anchored protein (AKAP).

This system mediates the cytoplasmic transportation of the PKA active fragment that in other cells interacts with some other cytoplasmic compartments or even with specific molecules such as those involved in Ca2+ regulation: calmodulin, calreticulin and calbindin[48].

If this model also can be applied to the spermatozoa, it is reasonable to postulate that after progesterone interaction with the membrane receptor, the activation of the adenylyl cyclase increases cAMP concentration. This second messenger then permits the activation of the PKA-AKAP system in such a way that the regulatory moiety of the PKA can interact with both calmodulin and some type of Ca2+channel, thus increasing the intracellular Ca2+ concentration. This increase then promotes a change in the cytoskeletal organization in such a way that, the zone comprising the outer acrosomal membrane and the plasma membrane covering the acrosome will fuse at several points and release the acrosome content.

Membrane fusion is also a time-dependent event that at the early stages only small membrane regions are involved and later more will be included[1].Leyton and Saling[49], after partial proteolysis of mouse zona pellucida demonstrated that some small peptides could interact with capacitated, but acrosome intact mouse spermatozoa. These peptides do not induce the acrosome reaction until the zona receptors are aggregated by a bivalent antibody. Carrera and co-workers[50] indicate that incubation of capacitated sperm with zona pellucida promotes a trans-phosphorylation process after aggregation of a tyrosine-kinase type receptor. These phosphorylated receptors interact with kinase-type proteins that could explain the increase in phosphorylated proteins found during the acrosome reaction[19,51].

The pivotal role of Ca2+ in the acrosome reaction has been demonstrated by a wide variety of techniques, as the Ca2+ specific ionosphere and the concentration-sensitive fluorescent probes. To explain the source of Ca2+, some authors have demonstrated that the most important source is the surrounding medium. Acrosome reaction can be observed in sperm incubated in a Ca2+ containing medium, but not in Ca2+ free medium. Polyamines, particularly spermidine, are required for a successful Ca2+ rise[52]. If the Ca2+ source is intracellular, the main source should be the mitochondria. In spermatozoa, Ca2+ regulating proteins other than calmodulin have not been demonstrated.

Calmodulin is a major regulatory protein. At present, there is no evidence of an active reticulum channel that could explain the increase in intracellular calcium concentration. This indirect evidence suggests that the main calcium source is the surrounding medium which is necessary for the activation of a granulate cyclase, leading to cGMP increase and cGMP-sensitive protein kinase (PKG) activation[53].

6 Sperm-egg interaction, zona pellucida penetration and membrane fusion

Recently, several reports using lectins or specific antibodies have characterized some molecules involved in gamete interaction leading to gamete fusion. In the mice, the O-linked carbohydrates and the mouse ZP3 protein core[54], whereas in the pigs, the tri- and tetra- antennary neutral N-linked oligosaccharides[55], a lactosaminoglycan moiety located as discrete layers through the zona pellucida, and three glycoproteins[56], are responsible for sperm-zona interaction. Although the specific sugar residue involved has not been characterized[57], several candidates have been proposed, e.g., nonreducing -linked oligosaccharides[58], -N-acetylglucosamine, mannose[59,60] and fucose[61,62].

Several proteins located on the sperm membrane implicated in gamete interaction have been reported. Galactosyltransferase(GTf) is a 60 Kd protein that uses the UDP galactose to link the sperm to the carbohydrate moiety of the ZP3 glycoprotein[63]. Another candidate to mediate the initial sperm-zona binding is the zonadhesin, a 16.4Kd protein synthesized exclusively in the testis and located at the apical region of the sperm head[64]. Northern blot analysis shows that there is a restricted homology between zonadhesins from mice, pig and human. The pig zonadhesin interacts with the respective zona protein through two cystein residues found in the D0-D1 and D2-D4 domains. There are several molecules like sulfogalactosylglycerolipids[65] or proteins that could interact with the egg envelope but most of them are not yet fully characterized[66,67] and their function in fertilization is not known.

Both galactosyltransferase and sperm zonadhesin are implicated in acrosome-intact spermatozoa interaction with the zona pellucida's primary sperm receptor.

However, acrosome-reacted spermatozoa can also interact with another sperm receptor glycoprotein located in the zona pellucida. The secondary sperm receptor in the zona pellucida is the ZP2 glycoprotein. It has been proposed that the protein moiety as well as the carbohydrates participate in the interaction. According to the model proposed by Hedrick and co-workers[68] and supported by experimental evidences[69,70], acrosin embedded in the inner acrosomal membrane interacts with the carbohydrates of ZP2 by a lectin domain exposed by this acrosin type,whereas non-membrane bound acrosin actively digests the ZP1 matrix. An equivalent model has been proposed for the guinea pig but in this case is a lectin-like site independent from the hyaluronidase site of the sperm surface PH-20 protein, that is involved in secondary sperm-egg interaction[71]. An analog protein to PH20 has been found in bovine sperm[13].

It has been reported that GnRH increases human sperm binding to homologous zona pellucida. This increase depends on extracellular Ca2+ transported through a L-type Ca2+ channel of the sperm plasma membrane[72]. Once the sperm has traveled through the zona pellucida by a mechanical and protease-assisted activity, the acrosome-reacted sperm enters the perivitellyne space. Interaction between the equatorial segment of the sperm head and the egg plasma membrane compels the tangential orientation of the sperm and thus can interact with the egg plasmalema by the only plasma membrane covered region in the equatorial segment[1].

Gamete membrane fusion has been associated to the interaction of several proteins. It has been proposed that gamete fusion is mediated by the interaction of anintegrin component in the egg plasma membrane with the heterodimeric fertilin in the sperm equatorial segment membrane[73,74].  When fertilin  was disrupted in mice, fertility is highly affected[75]. This is not the case with human sperm when fertilin gene is not functional[76].

Several integrins types have been detected by immunofluorescence in guinea pig[77], mouse[73], pig[78], and human eggs. When mouse eggs were pretreated with monoclonal or polyclonal antibodies against the 61 integrin and then exposed to sperm, less spermatozoa fuses[73].

According to this model[79,80], the binding domain of ADAM2 in the sperm plasma membrane binds to the egg integrin through the disintegrin domain. This interaction induces a tridimensional change in the fusion domain (in the other subunit of ADAM2) that permits their interaction with the hydrophobic surface of the egg plasmalemma. An hemifusion intermediate is formed when several fusogenic ADAM domains interact. This aggregation acts as a fusion pore allowing the entry of the sperm nucleus.

This model has been extended and is now highly indicative that ADAM2 or an analogue is not the only substance responsible of plasmalema fusion. Cohen and co-workers[5] have shown that at least in mice, the DE protein, an epididymal protein, helps the ADAM2- mediate sperm-egg fusion process since when present in co-incubation media, DE impairs the fusion process. Also, Chen and co-workers[81] have shown evidence that the ADAM-integrin interaction requires the cooperation of the CD9 protein expressed in the uterine-oviduct junction in order to have a high affinity interaction and assist the sperm passage to the oviduct.

7  Concluding remarks

In this mini review we have compiled relevant information that may help understand the structure and physiology of the sperm surface that are needed for a proper interaction with the oocyte. As in all reviews, there are still a lot of questions that are still unanswered or the experimental evidences are far from being conclusive. We believe, however, that it is necessary to have an starting point and to use it as a working hypothesis in such a way we can obtain more information that could make it closer to the real fertilization phenomena.

References

[1] Yanagimachi R. Mammalian fertilization. In: Knobil E, Neil JD, editors. The Physiology of Reproduction, 2nd ed. New York: Raven Press; 1994. p 189-317.
[2] Wassarman PA, Albertini DF. The mammalian ovum. In: Knobil E, Neil JD, editors. The Physiology of Reproduction 2nd ed. New York: Raven Press; 1994: p 79-122.
[3] Shur BD, Neely CA. Plasma membrane association, purification, and partial characterization of mouse sperm beta 1,4-galactosyltransferase. J Biol Chem 1988;263: 17706-14. 
[4] Cameo MS, Blaquier JA. Androgen-controlled specific proteins in rat epididymis. J Endocrinol 1976; 69: 47-55.
[5] Cohen DJ, Ellerma DA, Cuasnic PS. Mammalian sperm-egg fusion: evidence that epididymal protein DE plays a role in gamete fusion. Biol Reprod 2000; 63: 462-8.
[6] Cohen DJ, Munuce MJ, Cuasnic PS. Mammalian sperm-egg fusion: the development of rat oolemma fusibility during oogenesis involves the appearance of binding sites for sperm protein DE. Biol Reprod 1996; 55: 200-6.
[7] Kouba AJ, Abeydeera LR, Alvarez IA, Day BN, Buhi WC. Effects of porcine oviduct-specific glycoprotein on fertilization, polyspermy, and embryonic development in vitro. Biol Reprod 2000; 63: 242-50.
[8] Jervis KM, Robaire B. Dynamic changes in gene expression along the  rat epididymis. Biol Reprod 2001; 65: 696-703.
[9] Fusi FM, Bronson RA. Sperm surface fibronectin. Expression following capacitation. J Androl 1992; 13: 28-35.
[10] Fierro R, Foliguet B, Grignon G, Daniel M, Bene MC, Faure GC, et al. Lectin-binding sites on human sperm during acrosome reaction: modifications judged by electron microscopy/flow cytometry. Arch Androl 1996;  36: 187-96.
[11] Srivastava A, Olson GE. Glycoprotein changes in the rat sperm plasma membrane during maturation in the epididymis. Mol Reprod Develop 1991;  29: 357-64.
[12] Primakoff P, Myles DG. A map of guinea pig sperm membrane protein  constructed with monoclonal antibodies. Dev Biol 1983;  98: 417-28.
[13] Lalancette C, Dorval V, Leblanc V, Leclerc P. Characterization of an 80-kilodalton bull sperm protein identified as PH-20. Biol Reprod 2001;  65: 628-36.
[14] Ignotz GG, Lo MC, Perez CL, Gwathmey TM, Suarez SS. Characterization of a fucose-binding protein from bull sperm and seminal plasma that may be responsible for formation of the oviductal sperm reservoir. Biol Reprod 2001;  64:1806-11.
[15] Revah I, Gadella BM, Flesch FM, Colenbrander B, Suarez SS. Physiological state of bull sperm affects fucose- and mannose-binding properties. Biol Reprod 2000;  62: 1010-5.
[16] Suarez S, Redfern K, Raynor P, Martin F, Phillips DM. Attachment of boar sperm to mucosal explants of oviduct in vitro: possible role formation of a sperm reservoir. Biol Reprod 1991;  44: 998-1004.
[17] Wassarman PM. Mammalian fertilization: molecular aspects of gamete adhesion, exocytosis, and fusion. Cell 1999;  96. 175-83.
[18] Ward CR, Storey BT. Determination of the time course of capacitation in mouse using a chlortetracycline fluorescence assay. Dev Biol 1984;  171: 554-63.
[19] Visconti PE, Bailey JL, Moore GD, Pan D, Olds-Clarke P, Kopf GS. Capacitation in mouse spermatozoa I. Correlation between capacitation state and protein tyrosine phoshorylation. Development 1995;  121: 1129-37.
[20] Go KJ, Wolf DP. Albumin-mediated changes in sperm sterol content during capacitation. Biol Reprod 1985;  32: 145-53.
[21] Therien I, Bousquet D, Manjunath P.  Effect of seminal phospholipid-binding
 proteins and follicular fluid on bovine sperm capacitation. Biol Reprod 2001;  65: 41-51.
[22] Chang MC. The meaning of sperm capacitation. J Androl 1984;  5: 45-50.
[23] Si Y, Olds-Clarke P. Evidence for the involvement of Calmodulin in mouse sperm capacitation. Biol Reprod 2000;  62: 1231-9.
[24] Roselli M, Keller PJ, Dubey RK. Role of nitric oxide in the biology, physiology and pathophysiology of reproduction. Hum Reprod Update 1998;  4: 3-24.
[25] Herrero MB, de Lamirande E, Gagnon C. Nitric oxide regulates human sperm capacitation and proteintyrosine phosphorylation in vitro. Biol Reprod 1999; 61: 575-81.
[26] Hong Cy, Chao HF, Lee SL, Wei YH. Modification of human sperm function by human follicular fluid. A review. Int J Androl 1993;  16: 93-6.
[27] Roberts  AM. Motility of spermatozoa in fluid streams. Nature 1970;  228: 375-6.
[28] Eisenbach M. Sperm chemotaxis. Rev Reprod 1999a;  4: 56-66.
[29] VillanuevaDaz C, Arias-Martnez J, Bermejo-Martnez L, Vadillo-Ortega F. Progesterone induces human sperm chemotaxis. Fertil Steril 1995;  64: 1113-8.
[30] Navarro MC, Valencia J, Vzquez C, Czar E, Villanueva C. Crude mare follicular fluid exerts chemotactic effects on stallion spermatozoa. Reprod Dom  Anim 1998;  33: 321-4.
[31] Sliwa L. Substance P and beta-endorphin act as possible chemoattractants of mouse sperm. Arch Androl 2001;  46: 135-40.
[32] Nichol R, Hunter RH, de Lamaride E, Gagnon C, Cooke GM. Motility of spermatozoa in hydrosalpingeal and follicular fluid of pigs. J Reprod Fertil 1997;  110. 79-86.
[33] Fabbri R, Porcu E, Lenzi A, Gandini L, Marsella T, Flamifni C.  Follicular fluid and human granulosa cell cultures: influence on sperm kinetic parameters, hyperactivation and acrosome reaction. Fertil Steril 1998;  69: 112-7.
[34] Oliveira RG, Tomasi L, Rovasio RA, Giojalas LC. Increased velocity and induction of chemotactic response in mouse spermatozoa by follicular and oviductal fluids. J Reprod Fertil 1999;  115: 23-7.
[35] Yao Y, Ho P, Yeung WS. Effects of human follicular fluid on the capacitation and motility of human spermatozoa. Fertil Steril 2000;  73: 680-6.
[36] Tacconis P, Revelli  A, Massobrio M, Batista La Sala G, Tesarik J. Chemotactic responsiveness of human spermatozoa to follicular fluid is enhanced by capacitation but is impaired in dyspermic semen. J Assist Reprod Genet 2001;  18. 36-44. 
[37] Cohen-Dayag A, Tur-Kaspa I, Dor J, Mashiach S, Eisenbach M. Sperm capacitation in humans is transient and correlates with chemotactic responsiveness to follicular factors. Proc Nat Acad Sci USA 1995;  92:11039-43.
[38] Sliwa L. Hyaluronic acid and chemoattractant substances from follicular fluid: in vitro effect of human sperm migration. Arch Androl 1999;  43: 73-6.
[39] Eisenbach M. Mammalian sperm chemotaxis and its association with capacitation. Dev Genet 1999b;  25: 87-94.
[40] Serrano H, Canchola E, GarcaSurez MD. Sperm-attracting activity in follicular fluid associated to an 8.6-kDa protein. Biochem Biophys Res Commun 2001; 283: 782-4.
[41] Aitken RJ, Brindle JP. Analysis of the ability of three probes targeting the outer acrosomal membrane or acrosomal contents to detect the acrosome reactionin human spermatozoa. Hum Reprod 1993;  8: 1663-9.
[42] Serrano H, Diaz-Esparza L, Garcia-Suarez MD. Pig sperm membrane integrity evaluated by lectin labeling. Arch Androl 2001;  47: 59-65.
[43] Tesarik J, Mendoza C, Carreras A. Fast acrosome reaction measure: A highly sensitive method for evaluating stimulus-induced acrosome reaction. Fertil Steril 1993;  59: 424-30.
[44] Rommerts FFG. Cell surface actions of steroids: A complementary mechanism for regulation of spermatogenesis? In: Nieschlag E, Habenicht UF, editors. Spermatogenesis, Fertilization, Contraception. Heidelberg: Springer-Verlag; 1992: p 1-19.
[45] Patrat C, Serres C, Juannet P. Induction of a sodium ion influx by progesterone in human spermatozoa. Biol Reprod 2000;  62: 1380-6.
[46] Kobori H, Miyazaki S, Kuwabara Y. Characterization of intracellular Ca2+ increase in response to progesterone and cyclic nucleotides in mouse spermatozoa. Biol Reprod 2000;  63: 113-20.
[47] Florman HM, Arnoult C, Kazam IG, Li C, O'Toole CMB. A perspective of the control of mammalian fertilization by egg-activated ion channels in sperm. A tale of two channels. Biol Reprod 1998;  59: 12-6.
[48] Jordan JD, Landau EM, Iyengar R. Signal networks: The origins of cellular multitasking. Cell 2000;  103: 193-200.
[49] Leyton L, Saling PA. Evidence that aggregation of mouse sperm receptors by ZP3 triggers the acrosome reaction. J Cell Biol 1989;  108: 2163-8.
[50] Carrera A, Moos J, Ning  XP, Gerton GL, Tesarik J, Kopf GS, Moss SB. Regulation of protein tyrosine phosphorylation in human sperm by a Calcium/Calmodulind-ependent mechanism: identification of a kinase anchor protein as major substrates for tyrosine phosphorylation. Dev Biol 1996;  180: 284-96.
[51] Visconti PE, Moore GD, Bailey JL, Pan D, Leclerc P, Connors S, et al. Capacitation in mouse spermatozoa. II. capacitation and protein tyrosine phosphorylation are regulated by a cAMP-dependent pathway. Development 1995;  121: 1139-50.
[52] Meizel S, Turner KO. Effects of polyamine biosynthesis inhibitors on the progesterone-initiated increase in intracellular free Ca2+ and acrosome reaction in human sperm. Mol Reprod Dev 1993;  34: 457-65.
[53] Revelli A, Costamagna C, Moffa F, Aldieri E, Ochetti S, Bosia A, et al. Signalling pathway of nitric oxide\|induced acrosome reaction in human spermatozoa. Biol Reprod 2001;  64: 1708-12.
[54] Florman AM, Wassarman PA. O-linked oligosaccharides of mouse egg ZP3 accounts for its sperm receptor activity. Cell 1985;  41: 313-24.
[55] Nakano M, Yonezawa N, Hatanaka Y, Noguchi S. Structure and function of the N-linked carbohydrate chains of pig zona pellucida glycoproteins. J Reprod Fertil 1996;  50: 25-34.
[56] Dunbar BS, Timmons TM, Skinner SM, Prasad SV. Molecular analysis of a carbohydrate antigen involved in the structure and function of zona pellucida glycoproteins. Biol Reprod 2001;  65: 951-60.
[57] Yurewicz EC, Pack BA, Sacco AG. Isolation, composition and biological activity of sugar chains of porcine oocyte zona pellucida 55K glycoproteins. Mol Reprod Develop 1991;  30: 126-34.
[58] Litscher ES, Juntunen K, Seppo A, Pentil L, Niemal R, Renkonen O, Wassarman PM. Oligosaccharide constructs with defined structures that inhibit binding of mouse sperm to unfertilized eggs in vitro. Biochemistry 1995;  34: 4662-9.
[59] Cormwall GA, Tulsiani DRP, Orgebin-Crist MC.  Inhibition of the mouse sperm surface -D-mannosidase inhibits sperm-egg binding in vitro. Biol Reprod 1991;  44: 913-21.
[60] Miller DI, Macek MB, Shur BD. Complementarity between sperm surface  1, 4- galactosyltransferase and egg-coat ZP3 mediates sperm-egg binding in vitro. Nature 1992;  357: 589-93.
[61] Huang TTF, Yanagimachi R. Fucoidin inhibits attachment of guinea pig spermatozoa to the zona pellucida through binding to the inner acrosomal membrane and equatorial domains. Exp Cell Res 1984;  153: 363-73.
[62] Apter FM, Baltz JM, Millette CF. A possible role for cell surface fucosyl transferase (FT) activity during sperm zona- pellucida binding in the mouse. J Cell Biol 1988;  107: 175a.
[63] Hamilton DW. UDP-galactose: N-acetylglucosamine galactosyltransferase in rat testis and epidydimis. Biol Reprod 1980;  23: 377-85.
[64] Hardy DM, Garbers DL. A sperm membrane protein that binds in a species-specific manner to the egg extracellular matrix is a homolog to von Willebrand factor. J Biol Chem 1995;  270: 26025-8.
[65] White D, Weerachatyanukul W, Gadella B, Kamolvarin N, Attar M. Role of sulfogalactosylglycerolipid in mouse sperm\|zona pellucida binding. Biol Reprod 2000;  63: 147-55.
[66] Moos J, Peknikov J, Surneva-Nakova TN, Pavlk M. Identification of 17-kDa zona pellucida binding proteins from boar spermatozoa. FEBS Lett 1990;  280: 183-6.
[67] Zayas H, Bonilla E, Ducolomb Y, Casas E, Crdova A, Betancourt M. Affinity of pig oocyte zona pellucida peptides for sperm proteins. Med Sci Res 1995;  23: 831-2.
[68] Hedrick JL, Urch UA, Hardy DM. Structure-function properties of the sperm enzyme acrosin. In: Witaker JR, Sonnett PE, editors. Biocatalysisi in Agricultural Biotechnology. Washington:  American Chemical Society;  1989. p 212-9.
[69] Liu DY, Baker HWG. Inhibition of acrosin activity with a trypsin inhibitor blocks human sperm penetration of the zona pellucida. Biol Reprod 1993;  48: 340-8.
[70] Crosby JA, Barros C. Effect of recombinant boar -acrosin on sperm binding to intact zona pellucida during in vitro fertilization. Biol Reprod 1999;  61: 1535-40.
[71] Hunnicut GR, Primakoff P, Myles DG. Sperm surace protein PH20 is bifunctional: one activity is a hyaluronidase and a second, distinct activity is required in secondary sperm-zona binding. Biol Reprod 1996: 55: 80-6.
[72] Morales P, Pizarro E, Kong M, Kerr B, Ceric F, Vigil P. Gonadotropin-releasing hormone-stimulated sperm binding to the human zona pellucida is mediated by a calcium influx. Biol Reprod 2000;  63: 635-42.
[73] Almeida EAC, Houvila APJ, Sutherland AE, Stephens LE, Calarco PG, Shaw LM, et al. Mouse egg integrin 61 funcions as a sperm receptor. Cell 1995;  81: 1095-104.
[74] Evans JP, Schultz RM, Kopf GS. Mouse sperm-egg plasma membrane interactions. Analysis of roles of egg integrin and the mouse homologue of PH-30 (fertilin) . J Cell Sci 1995;  108: 3267-78.
[75] Cho C, O'Dell Bunch D, Faure JE, Goulding EH, Eddy EM, Primakoff P, Myles DG. Fertilization defects in sperm from mice lacking fertilin . Science 1998;  281: 1857-9.
[76] Jury JA, Frayne J, Hall L. The human fertilin gene is non-functional: implications for its proposed role in fertilization. Biochem J 1997;  321: 577-81.
[77] Blobel CP, Wolfsberg TG, Turck CW, Myles DG, Primakof P, White JM. A potential fusion peptide and an integrin ligand domain in a protein active in sperm-egg fusion. Nature 1992;  356: 248-52.
[78] Linfor J, Berger T. Potential role of v and 1 integrins as oocyte adhesion molecules during fertilization in pigs. J Reprod Fertil 2000;  120: 65-72.
[79] Bigler D, Chen M, Waters S, White JM. A model for sperm-egg binding and fusion based on ADAMs and integrins. Trend Cell Biol 1997;  7: 220-5.
[80] Schlondorff J, Blobel CP. Metalloprotease-disintegrins: modular proteins capable of promoting cell-cell interactions and triggering signals by protein-ectodomain shedding. J Cell Sci 1999;  112: 3603-17.
[81] Chen MS, Tung KSK, Coonrod SA, Takahashi Y, Bigler D, Chang A, et al. Role of the integrin-associated protein CD9 in binding between sperm ADAM2 and the egg integrin 61: implications for murine fertilization. Proc Nat Acad Sci USA. 1999;  96: 11830-5
.

home


Correspondence to: Dr. Hector Serrano, Dept. Health Sciences, UniversidadAutnoma Metropolitana-Iztapalapa, Ave. Michoacan and Purisima, Mexico City, DF 09340, Mexico.
Tel: +52-5-804 4733,     Fax: +52-5-804 4727
E-mail: hectorserrano@hotmail.com
Received 2001-10-19    Accepted 2001-11-21