IMR Press / FBL / Volume 24 / Issue 4 / DOI: 10.2741/4747
Review
Effects of sperm proteins on fertilization in the female reproductive tract
Show Less
1 State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China
2 Beijing Key Laboratory of Animal Genetic Improvement, China Agricultural University, No. 2 Yuanmingyuan West Road, Beijing, 100193, China
3 Changsha Reproductive Medicine Hospital, Hunan, Changsha, 410205, China
4 Reproductive Medicine Center of People’s Hospital of Zhengzhou University, Zhengzhou 450003, China
5 Reproduction Medical Center, Yantai Yuhuangding Hospital of Qingdao University, Yantai 264000, China
6 Key Laboratory of Fertility Preservation and Maintenance of Ministry of Education, Ningxia Medical University, Yinchuan 750004, China
*Correspondence: dengsl@ioz.ac.cn (Shou-Long Deng)
Front. Biosci. (Landmark Ed) 2019, 24(4), 735–749; https://doi.org/10.2741/4747
Published: 1 March 2019
Abstract

Mammalian fertilization that culminates by fusion of the male and female gametes is intricately regulated within the female reproductive tract. To become competent to fertilize an egg, the mammalian spermatozoa that enter the female reproductive tract must undergo a series of physiological changes, including hyperactivation, and capacitation. For reaching full competency, the acrosome, a specialized membrane-bound organelle that covers the anterior part of the sperm head, must undergo an acrosome reaction. For becoming competent to bind an ovum, and to penetrate the zona pellucida and cumulus, many sperm proteins are released in the course of the acrosome reaction. Ultimately, the acrosome binds to the oolemma and fusion of sperm and egg occurs. In this review, we outline current understanding of the roles and effects of some essential sperm proteins and their functions during fertilization in the female reproductive tract.

Keywords
Fertilization
Sperm proteins
Sperm-oocyte fusion
Acrosome
Review
2. INTRODUCTION

The fusion of spermatozoa and oocytes is a complex process, involving capacitation, hyperactivation, the acrosome reaction (AR), binding of the spermatozoon to the zona pellucida, and activating the oocyte reaction (1). Fusion of spermatozoa with the oocyte plasma membrane occurs via the inner acrosomal membrane (IAM) exposed after the AR. In mammals, gamete fusion takes place through a specialized region of the acrosome known as the equatorial segment (ES) which becomes fusogenic only after the AR has been completed (2). Many studies have been performed on the functions of the acrosome and several molecules have been reported. For example, a set of attachment protein receptors has been reported (3), including PH-20 (4), ZP3R (5), SP-10 (6), IZUMO1 (7), and ADAM2 (which belongs to the ADAM family of proteins) (8). In particular, Equatorin, a complex 38–48 kDa protein in mice, is specifically located to the acrosomal membrane in various species, including humans (913). Our previous study showed that Equatorin plays an important role in acrosomal formation and fertilization (12). Research on sperm proteins participating in sperm–oocyte fusion has been the subject of many investigations to identify their roles and improve the diagnosis and treatment of human infertility. These studies have shown that sperm proteins can exert unique effects on fertilization in the female reproductive tract, which is the focus of this review.

3. SPERM STRUCTURE

Normal, healthy mammalian spermatozoa range in length from about 40 to 250μm (a human sperm cell is 60–70μm long) consisting of a head, neck and principal piece (midpiece and tail) (14). The sperm head contains a highly condensed nucleus with haploid DNA surrounded by nuclear membranes (15, 16). The acrosome is a membrane-bound vesicle located outside the nucleus on the apical part of the sperm head. The acrosomal membrane is further subdivided into two parts: one is the inner acrosomal membrane that lies external to the sperm nuclear membrane; the other is the outer acrosomal membrane that covers most of the acrosome and lies below the sperm plasma membrane. Some important enzymes involved in sperm penetration of oocytes and their vestments are found in the acrosome (12, 17). The sperm midpiece contains mitochondria that provide power in the form of ATP for sperm tail movement (14). All structures of the sperm flagellum function to provide energy for swimming toward the oocyte and completing fertilization in vivo (Figure 1).

Figure 1

Sperm structure. (A) The normal healthy mammalian spermatozoon includes the head, neck, midpiece and tail. The sperm head contains a highly condensed nucleus with its haploid genome surrounded by nuclear membranes. The acrosomal membranes include the inner acrosomal membrane which lies external to the sperm nucleus; the other is the outer acrosomal membrane which lies below the sperm cell plasma membrane. The sperm neck is followed by a short midpiece, containing mitochondria providing the power for sperm tail movement. The schematic diagrams show different parts of the sperm head before (B) and after (C) the acrosome reaction. After the acrosome reaction and zona penetration, the spermatozoa lose the plasma and outer acrosomal membranes of the anterior acrosomal portion, releasing the acrosomal matrix and exposing the inner acrosomal membrane (C). The plasma membrane and the equatorial segment of the posterior acrosome remain intact. AAS, Anterior acrosomal sac; ES, equatorial segment; IAM, inner acrosomal membrane; N, nucleus; NM, nuclear membrane; OAM, outer acrosomal membrane; PA, posterior acrosome; PAR, post acrosomal region; PM, sperm plasma membrane; PS, postacrosomal sheath; PT, perinuclear theca.

4. ZONA PELLUCIDA

The zona pellucida (ZP) is a specialized extracellular matrix that contains three major glycoproteins, designated ZP1, ZP2 and ZP3 (18). Early studies demonstrated that ZP3 is the only glycoprotein that potently enables murine sperm–ZP binding and allows spermatozoa to attach to the ZP. Sperm–ZP binding is a critical step of sperm–oocyte fusion and follows the AR (1820). Many studies have shown that degradation of the sperm plasma membrane leads to the loss of ZP3 receptors; a glycoprotein receptor to ZP2 is exposed and replaces the function of that for ZP3 (21). Thus, ZP2 serves as a secondary receptor for spermatozoa during the fertilization process and maintains binding of acrosome-reacted spermatozoa to oocytes. However, these glycoproteins appear to be indispensable elements for the function of oocytes. In mice, knockout of the coding genes for ZP2 or ZP3 prevent the development of a normal ZP, resulting in infertility (22, 23). A clinical study also showed that the ZP3 mutation, c.400G>A (p. Ala134Thr), prevents the assembly of the ZP and leads to oocyte degeneration (24). In addition, development of a normal ZP is prevented because of defective ZP1 and leads to the sequestration of ZP3 into the ooplasm (25). The sperm flagellum continues to provide power to help it penetrate through the ZP and effect fertilization. After spermatozoa penetrate the ZP, they will pass into the perivitelline space. Penetration of the human spermatozoon passing through the ZP takes less than 30 seconds under in vitro conditions (26).

5. STAGE OF FERTILIZATION

Successive stages of mammalian fertilization occur as follows. First, sperm–oocyte recognition initiates via ZP3 and ZP3 binding molecules on the sperm surface. Proteins on the sperm membrane bind to ZP3 (or ZP2) on the outer surface of the ZP. Second, ZP binding and the AR occurs. Sperm–oocyte binding triggers the AR, in which the sperm plasma membrane forms point fusions with the outer acrosomal membrane, causing exocytosis of the acrosomal contents. It is generally thought that that the AR begins after the spermatozoon binds to the ZP. However, oocytes of transgenic mice (ZP2Mut and ZP3Mut) could also be fertilized, demonstrating that the AR occurred at the time of sperm passage through the ZP or before reaching it (27). Third, acrosomal enzymes are released and the spermatozoon penetrates the ZP. Acrosomal enzymes assist in dissolving a hole in the ZP. Accompanied by sperm tail beating, sperm move through the ZP. During this time, cumulus cells also release factors to promote sperm penetration through the oocyte and to enhance sperm motility (28, 29). Fourth, oocyte binding proteins mediate sperm receptor recognition. Egg-binding proteins such as JUNO on the oocyte surface bind to molecules on the sperm cell membrane. Fifth, oocyte–sperm membrane fusion occurs and the sperm head and centriole enter the oocyte together with the midpiece and principal piece in most species. Finally, the male and female pronuclei migrate toward each other with the aid of microtubules and syngamy eventually occurs (16) (Figure 2).

Figure 2

Successive stages of mammalian fertilization. (A) Sperm–oocyte recognition through ZP3 and ZP3 binding molecules. (B) ZP binding triggers the AR, in which the sperm plasma membrane fuses with outer acrosomal membrane and releases acrosomal enzymes. (C) Acrosomal enzymes dissolve a hole in the ZP to help sperm penetrate the oocyte. (D) The oocyte-binding protein (JUNO) on the inner acrosomal membrane binds to a sperm receptor on the oocyte surface. (E) Oocyte–sperm membrane fusion allows the sperm nucleus and centriole to enter the oocyte. (F) The sperm (male) pronucleus and oocyte (female) pronucleus migrate toward each other.

6. ACROSOME REACTION AND ACROSOME FORMATION

The first AR studies were carried out using conventional microscopy in 1952 (30). Studies found that mammalian fertilization requires a series of changes in the AR process, requiring necessary time-dependent physiological changes in spermatozoa in the female reproductive tract, collectively known as capacitation (31, 32). Recently, the terms ‘acrosomal exocytosis (AE)’ and ‘AR’ have been used interchangeably (33, 34). AE is a terminal morphological alteration of the spermatozoon and is a synchronized and tightly regulated progress during mammalian fertilization, including opening of hundreds of fusion pores between outer acrosomal membrane and sperm plasma membrane during sperm penetration of the ZP (30, 31). Moreover, the physiological site of the AE in the mouse has been determined to be the upper isthmus of the oviduct using genetically modified strains that carry DsRed2 in the mitochondria and enhanced green fluorescent protein (EGFP) in the acrosome (35).

The function of the acrosome depends on its characteristic configuration. The common features include three structures: an inner acrosomal membrane (IAM), which is laminated to the sperm nucleus, an outer acrosomal membrane (OAM), which is close to the plasma membrane over the acrosome and an equatorial segment (ES), which forms the posterior acrosomal margin where the IAM and OAM meet (32) (Figure 1). Acrosomal formation during late meiosis in the testis originates from small, proacrosomal vesicles in the Golgi apparatus of pachytene spermatocytes (3638). The haploid spermatids inherit these proacrosomal vesicles containing a variety of proteins assemble and fuse; these proacrosomal vesicles eventually form a head structure that then gradually covers the future anterior segment of the sperm nucleus. The final sperm maturation phase occurs during epididymal transit.

7. SPERM ANTIGEN

As mentioned above, the acrosome is a vital organelle for fertilization, especially for AR and ZP binding. Sperm antigens are released in the course of AR to assist the binding of the spermatozoon to the oocyte. This release relies on a strict time schedule, which is decided by the location of the antigen (39). According to the different receptors involved, we can divide antigens into two main groups: sperm acrosomal antigens involved in binding to the ZP, and other antigens participating in the binding of the sperm and oocyte plasma membrane.

7.1. Sperm antigen involving in binding of sperm and ZP
7.1.1. Zona pellucida 3 receptor (ZP3R), sperm fertilization protein-6 (S-56)

Zona pellucida 3 receptor (ZP3R), initially named SP-56, was first identified as a 56 kDa mouse sperm surface protein with a specific receptor for ZP3; however, there is no report about human ZP3R to date. ZP3R was found to be located on the intact sperm surface and was ascertained to be a binding site for ZP3 in acrosome-intact spermatozoa using photoaffinity cross-linking (40). With progression of the AR, proteases can dissolve ZP3R, and then it is released in areas surrounding the anterior acrosome (41). However, some studies found that ZP3R cannot be identified in live sperm surfaces before capacitation. This suggests that ZP3R is an intra-acrosomal component (42). In addition, as recombinant ZP3R can interact with the ZP of unfertilized oocytes but not with that of 2-cell embryos, it suggests that the changes taking place in the ZP during fertilization might influence the binding of ZP3R (5). Although ZP3R is an acrosomal matrix protein, it might have a vital role in sperm–ZP adhesion during fertilization (41). By targeting deletion of the Zp3r gene, the results showed that males and females homozygous for the affected gene exhibited no differences in litter sizes compared to wild-type and heterozygous animals. This study proves that either ZP3R is not involved in sperm-zona pellucida binding or this process might be functionally redundant, involving multiple proteins for gamete interactions (43).

7.1.2. Sperm protein-10 (SP-10)

Sperm protein-10 (SP-10) was first detected in human spermatozoa (44), and is produced via acrosomal vesicle protein 1 (ACRV1), and was later identified to be present in all tested mammalian species, including mice and baboons. SP-10 is specifically expressed in the testes and exclusively restricted to round and elongated spermatids (45). SP-10 can be first detected in the acrosomal vesicle of Golgi phase spermatids, and subsequently appears on the acrosomal vesicle. The involvement of SP-10 in the binding of sperm to the oolemma is supported by research showing that it is located at the ES (46). Furthermore, the coding gene of SP-10 is viewed as an excellent model to research the transcription of male germ-cell-specific genes (47). Using an anti-SP-10 antibody is used to identify the characteristic morphological features of each stage of acrosomal formation; therefore, it has made staging of the spermatogenic cycle relatively easy and clear (6). In some studies, recombinant SP-10 is used as an immunogen that can be made into man contraceptive vaccine in mouse models (48).

7.1.3. Fertilization antigen-1 (FA-1)

Fertilization antigen-1 (FA-1), a glycoprotein found as dimers (51 ± 2 kDa) and monomers (23 kDa), appears at the end of spermatogenesis. FA-1 exists on the surface of human, mouse, and rabbit spermatozoa. Because of the high conservation of its structure, FA-1 antigen might have similar functions across different species. According to research in human fertilization, as FA-1 is released from the acrosome, it can specifically recognize ZP3. On the sperm surface, once FA-1 recognizes the ZP of mouse oocyte as a complementary receptor molecule, it will affect binding of sperm to the ZP, rather than sperm motility. Antibodies to FA-1 antigen could inhabit sperm–ZP binding and shorten sperm capacitation and AR timing by blocking protein phosphorylation at some amino acids, such as tyrosine, serine and threonine residues (49). Female mice vaccinated with recombinant FA-1 antigen showed long-term, reversible contraceptive effects (50, 51).

7.1.4. Posterior head-20 (PH-20)

PH-20, also known as sperm adhesion molecule1 (SPAM1), is a sperm hyaluronidase, identified in the testes of humans, mice, and bulls (4, 52). PH-20 antigen is located in the plasma membrane over the sperm head and on the IAM. During the AR, PH-20 is found in the IAM and in mosaic vesicles originating from the binding of the sperm plasma membrane and the OAM (53). in addition, after the AR, PH-20 is identified in the IAM of the anterior acrosome and plasma membrane of the ES. Some have used Pichia pastoris to produce a recombinant human PH-20 protein cloned from a specific core DNA fragment of the gene encoding human PH-20 (54). PH-20 has been viewed as multifunctional protein: thus, it is not only a hyaluronidase that is a receptor for hyaluronic acid (HA)-induced cell signaling, but also acts as receptor for the ZP during the AR (55). PH-20, as a sperm hyaluronidase, was regarded as an essential enzyme that can help sperm in penetrating the cumulus surrounding the oocyte when fertilizing. However, in mice, there is some evidence suggesting that PH-20 is not important for this process because the double knockout PH-20 mice remain fertile (56). Accordingly, using human kidney 293 cell lines expressing recombinant pig SPAM1, some have shown that PH-20 can break down the oocyte-cumulus complex during in vitro fertilization (57). Additionally, expression of PH-20 in pig seminal plasma showed significant correlation with high farrowing rates after artificial insemination (58).

7.2. Antigens participated in binding of sperm and plasm membrane
7.2.1. ADAM1b/ADAM2 (Fertilin)

A sperm surface antigen involved in fusion of the spermatozoon and oocyte was identified as fertilin (also known as PH-30), which can be divided into ADAM1b (fertilin-α) and ADAM2 (fertilin-β). ADAM1b and ADAM2 constitute a heterodimer via noncovalent binding (59). ADAM1b and ADAM2 belong to the ADAM family (short for a disintegrin and metalloproteinase). Fertilins were first identified in the epididymis of mice and are located in the plasma membrane over the sperm head. In addition, ADAM2 gene knockout mice show functional defects of spermatozoa illustrating that ADAM2 is essential during sperm migration to the oocyte in the female reproductive tract in vivo (60). In mice and monkeys, ADAM2 has been identified as a 100 kDa precursor in the testis, it is processed to a mature form (47 kDa in monkeys, 45 kDa in mice) during sperm maturation (61, 62). However, although ADAM2, in human testis is found as 100 kDa precursors and is found in human spermatogenic cells, western blot analysis did not detect ADAM2 in human spermatozoa (8). In the boar epididymis. migration of fertilin occurs during passage of spermatozoa though the distal corpus, and is complete when spermatozoa pass the proximal cauda (63).

7.2.2. IZUMO1

IZUMO1 is one of the immunoglobulin superfamily (IgSF) of proteins and was first detected as a sperm antigen against the OBF13 antibody, which can affect sperm–oocyte fusion in the mouse reproductive tract. In intact spermatozoa, IZUM1 is localized the acrosome and is not detectable on the sperm surface. At the middle stage of the AR, IZUMO1 is released to the sperm surroundings to promote fertilization. In IZUMO–/– male mice, although their spermatozoa can undergo the AR and penetrate the ZP, they are infertile because of a failure in sperm–oocyte membrane fusion (64). JUNO is the oocyte receptor of IZUMO1, and both of them are viewed as vital proteins for sperm–oocyte fusion (65). Moreover, human IZUMO1 can interact with hamster JUNO, illustrating that binding of IZUMO1 and JUNO has been conserved across several species (66). Studies have shown that IZUMO1 undergoes specific phosphorylation changes in the sperm tail during epididymal transit (67), and this region might play an essential role in fertilization. However, another study utilized mice whose sites of IZUMO1 phosphorylation were truncated via the CRISPR/Cas9 system and found that the fertility of the mutated mice was unaffected, indicating that phosphorylation of IZUMO1 appears to be unimportant for fertilization (68).

8. EQUATORIN

The equatorin gene, also called MN9 or AFAF in the human genome, encodes this acrosomal membrane protein. Equatorin localization is limited to the IAM and OAM from round spermatids to mature sperm stage and is most likely involved in acrosomal biogenesis. Equatorin is preserved at the ES after the AR and reduced the in vitro fertilization rate in vitro (10, 69). Another in vivo study indicated that mMN9 (an anti-equatorin monoclonal antibody) significantly inhibited the mouse fertilization rate. We found no obvious defects in the acrosome in Eqtn–/– male mice sperm (12), indicating that this protein is not essential for acrosomal biogenesis and that the loss of this gene does not affect acrosomal formation. Our observations revealed that the ES can be identified during acrosome biogenesis as a discrete domain in the acrosomal vesicle as early as the Golgi phase of acrosome biogenesis (70). Equatorin is not likely to be involved in fusion in this region because the OAM and plasma membranes do not fuse in the ES during the AR.

The AR is a universal requisite for sperm–oocyte membrane fusion occurring via the IAM exposed after the AR. Equatorin contained in the posterior acrosome is detectable only after spontaneous or induced ARs following fixation and permeabilization, but not in intact spermatozoa (2). Many studies have shown that equatorin is required for successful sperm–oocytes fusion in vitro and in vivo. We showed that Eqtn–/– male mice were subfertile and their spermatozoa show lower fertilization rates in vivo and in vitro (12) (Figure 3). Equatorin antibodies can significantly inhibit sperm penetration of the oocytes and reduce the in vitro fertilization rate. The mechanism involves calcium-triggered AE by increasing calcium efflux from the acrosome (69) (Figure 4-5). Another study indicated that equatorin interacts with the soluble N-ethyl-maleimide-sensitive factor attachment proteins receptors (SNARE) complex and enables completion of membrane fusion during AE (71). We also demonstrated that the equatorin protein might indirectly interact with SNAP25, a component of the SNARE complex, in a transfected cell line, although its loss did not affect the expression of the SNARE family members (Figure 6).

Figure 3

The in vivo fertilization rate of Eqtn-knockout male mice was decreased significantly. Control and Eqtn–/– males were crossed with wild-type females, and embryos were collected from the oviducts at 1.5. days post copulation (dpc). Approximately 90% of the collected oocytes were fertilized and had developed to the 2-cell stage in the control group (A, C: black arrows in A). However, in the Eqtn–/– group, most oocytes remained at the 1-cell stage (B, C: black arrowheads in B), and fewer than 30% of oocytes were fertilized and had developed to the 2-cell stage (B, C: black arrow in B). This difference was significant. *p < 0.0.1 (12).

Figure 4

Acrosome formation-associated factor (Afaf) is required for Ca2+-triggered acrosomal exocytosis (AE) by acting upstream of acrosomal Ca2+ efflux. (A) Schematic diagram demonstrating the treatment protocol of spermatozoa. (B) A: In the control group without stimulation, Afaf was present in the acrosome. B: In the presence of 10 mM Ca2+ stimulation, only a few spermatozoa showed Afaf staining. *p < 0.01 (69).

Figure 5

In the presence of anti-Afaf antibody and 10 μM Ca2+ (anti-Afaf -Ca2+), the AE index was inhibited dramatically. Da: In the presence of NP followed by anti-Afaf and then Ca2+ and hv (NP-anti-Afaf-Ca2+-hv), AE was inhibited dramatically. Db: In the presence of NP followed by Ca2+ and then anti-Afaf and hv (NP-Ca2+-anti-Afaf-hv), no inhibition of AE was observed. Ab: Antibodies, NP: NP-EGTA-AM (photolabile Ca2+ chelator), hv: flash photolysis of the NP (69).

Figure 6

Schematic representation of the pathways involved in biogenesis of the acrosome. Solid arrows indicate transport steps that have been well documented and verified, whereas dashed arrows indicate hypothetical transport steps. Numbers denote the extra-Golgi transport steps that possibly contribute to formation of the acrosome. Path 1, the direct transport path from the endoplasmic reticulum (ER) to the acrosome. Path 2, the transport path from the plasma membrane through the SE directly to the acrosome (the hypothetical trafficking path of Afaf). Path 3, transport path from LE to the acrosome. Path 4, the transport route from lysosome to acrosome. Abbreviations: SE, sorting endosome; RE, recycling endosome; LE, late endosome; L, lysosome; TGN, trans-Golgi network (13).

9. PERSPECTIVE

AR is triggered by sperm–ZP binding, which is a necessary prerequisite for successful fertilization. It is an interaction that allows the sperm head’s OAM and sperm plasma membrane to fuse, which enables the exposure of acrosomal contents through the formation of mosaic vesicles and is a critical step in fertilization. During the AR, sperm proteins are released. These can recognize the ZP, and help spermatozoa penetrate the ZP and cumulus. ZP3R acts a vital role in sperm–ZP adhesion, and PH-20 is not merely a receptor for ZP components but helps the spermatozoa pass through the cumulus as a special enzyme. When spermatozoa arrive at the oolemma, other sperm proteins play a role in binding. ADAM2 and IZUMO1 are essential in ensuring that sperm can migrate to the oocyte. However, IZUMO1 is unimportant for fertilization. Equatorin localization is limited to the ES and the IAM and OAM. It is not essential for acrosome biogenesis and knockout of its encoding gene does not affect acrosomal formation but is required for successful sperm–oocyte fusion in vitro and in vivo. These findings suggest that equatorin is essential for sperm–oocyte fusion, so it might be a candidate target for human contraception.

10. ACKNOWLEDGEMENT

Tie-Cheng Sun and Jia-Hao Wang contributed equally to this work. This work was supported by the National Natural Science Foundation of China (grant no. 31501953 to Shou-Long Deng; 31471352 to Yi-Xun Liu); Clinical Capability Construction Project for Liaoning Provincial Hospitals (grant no. LNCCC-C09-2015, LNCCC-D50-2015) to Yi-Xun Liu and Academician Workstation Support (Shenyang, Changsha and Shandong) to Yi-Xun Liu.

References
[1]
M Okabe The cell biology of mammalian fertilization Development 2013 140 22 4471 9 DOI:10.1242/dev.090613 PMid:24194470
[2]
G Manandhar K Toshimori Exposure of sperm head equatorin after acrosome reaction and its fate after fertilization in mice Biol Reprod 2001 65 5 1425 36 DOI:10.1095/biolreprod65.5.1425 PMid:11673259
[3]
J Ramalho-Santos RD Moreno P Sutovsky AW Chan L Hewitson GM Wessel CR Simerly G Schatten SNAREs in mammalian sperm: possible implications for fertilization Dev Biol 2000 223 1 54 69 DOI:10.1006/dbio.2000.9745 PMid:10864460
[4]
K Sabeur GN Cherr AI Yudin P Primakoff MW Li JW Overstreet The PH-20 protein in human spermatozoa J Androl 1997 18 2 151 8
[5]
MG Buffone T Zhuang TS Ord L Hui SB Moss GL Gerton Recombinant mouse sperm ZP3-binding protein (ZP3R/sp56) forms a high order oligomer that binds eggs and inhibits mouse fertilization in vitro J Biol Chem 2008 283 18 12438 45 DOI:10.1074/jbc.M706421200 PMid:18316377 PMCid: PMC2431013
[6]
HP Osuru JE Monroe AP Chebolu J Akamune P Pramoonjago SA Ranpura PP Reddi The acrosomal protein SP-10 (Acrv1) is an ideal marker for staging of the cycle of seminiferous epithelium in the mouse Mol Reprod Dev 2014 81 10 896 907 DOI:10.1002/mrd.22358 PMid:25158006 PMCid: PMC4198580
[7]
N Inoue M Ikawa M Okabe The mechanism of sperm-egg interaction and the involvement of IZUMO1 in fusion Asian J Androl 2011 13 1 81 7 DOI:10.1038/aja.2010.70 PMid:21057513 PMCid: PMC3739395
[8]
H Choi S Jin JT Kwon J Kim J Jeong J Kim S Jeon ZY Park KJ Jung K Park C Cho Characterization of mammalian ADAM2 and its absence from human sperm PLoS One 2016 11 6 e0158321 DOI:10.1371/journal.pone.0158321 PMid:27341348 PMCid: PMC4920383
[9]
K Toshimori DK Saxena I Tanii K Yoshinaga An MN9 antigenic molecule, equatorin, is required for successful sperm-oocyte fusion in mice Biol Reprod 1998 59 1 22 9 DOI:10.1095/biolreprod59.1.22 PMid:9674989
[10]
K Yoshinaga DK Saxena T Oh-oka I Tanii K Toshimori Inhibition of mouse fertilization in vivo by intra-oviductal injection of an anti-equatorin monoclonal antibody Reproduction 2001 122 4 649 55 DOI:10.1530/rep.0.1220649 PMid:11570972
[11]
K Yamatoya K Yoshida C Ito M Maekawa M Yanagida K Takamori H Ogawa Y Araki K Miyado Y Toyama K Toshimori Equatorin: identification and characterization of the epitope of the MN9 antibody in the mouse Biol Reprod 2009 81 5 889 97 DOI:10.1095/biolreprod.109.077438 PMid:19605790
[12]
J Hao M Chen S Ji X Wang Y Wang X Huang L Yang Y Wang X Cui L Lv Y Liu F Gao Equatorin is not essential for acrosome biogenesis but is required for the acrosome reaction Biochem Biophys Res Commun 2014 444 4 537 42 DOI:10.1016/j.bbrc.2014.01.080 PMid:24480441
[13]
YC Li XQ Hu KY Zhang J Guo ZY Hu SX Tao LJ Xiao QZ Wang CS Han YX Liu Afaf, a novel vesicle membrane protein, is related to acrosome formation in murine testis FEBS Lett 2006 580 17 4266 73 DOI:10.1016/j.febslet.2006.06.010 PMid:16831425
[14]
WC Ford Comments on the release of the 5th edition of the WHO Laboratory Manual for the Examination and Processing of Human Semen Asian J Androl 2010 12 1 59 63 DOI:10.1038/aja.2008.57 PMid:20111082 PMCid: PMC3739684
[15]
M Smit JC Romijn MF Wildhagen RF Weber GR Dohle Sperm chromatin structure is associated with the quality of spermatogenesis in infertile patients Fertil Steril 2010 94 5 1748 52 DOI:10.1016/j.fertnstert.2009.10.030 PMid:20004379
[16]
PM Wassarman Early events in mammalian fertilization Annu Rev Cell Biol 1987 3 109 42 DOI:10.1146/annurev.cb.03.110187. 000545 PMid:3318876
[17]
Y Kanemori Y Koga M Sudo W Kang S Kashiwabara M Ikawa H Hasuwa K Nagashima Y Ishikawa N Ogonuki A Ogura T Baba Biogenesis of sperm acrosome is regulated by pre-mRNA alternative splicing of Acrbp in the mouse Proc Natl Acad Sci USA 2016 113 26 E3696 705 DOI:10.1073/pnas.1522333113 PMid:27303034 PMCid: PMC4932935
[18]
JD Bleil PM Wassarman Structure and function of the zona pellucida: identification and characterization of the proteins of the mouse oocyte's zona pellucida Dev Biol 1980 76 1 185 202 DOI:10.1016/0012-1606(80)90371-1
[19]
SX Tao J Guo XS Zhang YC Li ZY Hu CS Han YX Liu Germ cell apoptosis induced by experimental cryptorchidism is mediated by multiple molecular pathways in Cynomolgus Macaque Front Biosci 2006 11 1077 89 DOI:10.2741/1864 PMid:16146798
[20]
YQ Shi QZ Wang SY Liao Y Zhang YX Liu CS Han In vitro propagation of spermatogonial stem cells from KM mice Front Biosci 2006 11 2614 22 DOI:10.2741/1995 PMid:16720338
[21]
JD Bleil JM Greve PM Wassarman Identification of a secondary sperm receptor in the mouse egg zona pellucida: role in maintenance of binding of acrosome-reacted sperm to eggs Dev Biol 1988 128 2 376 85 DOI:10.1016/0012-1606(88)90299-0
[22]
TL Rankin M O'Brien E Lee K Wigglesworth J Eppig J Dean Defective zonae pellucidae in Zp2-null mice disrupt folliculogenesis, fertility and development Development 2001 128 7 1119 26
[23]
C Liu ES Litscher S Mortillo Y Sakai RA Kinloch CL Stewart PM Wassarman Targeted disruption of the mZP3 gene results in production of eggs lacking a zona pellucida and infertility in female mice Proc Natl Acad Sci USA 1996 93 11 5431 6 DOI:10.1073/pnas.93.11.5431 PMid:8643592
[24]
T Chen Y Bian X Liu S Zhao K Wu L Yan M Li Z Yang H Liu H Zhao Z Chen A Recurrent Missense Mutation in ZP3 Causes Empty Follicle Syndrome and Female Infertility Am J Hum Genet 2017 101 3 459 465 DOI:10.1016/j.ajhg.2017.08.001 PMid:28886344 PMCid: PMC5590947
[25]
HL Huang C Lv YC Zhao W Li XM He P Li AG Sha X Tian CJ Papasian HW Deng GX Lu HM Xiao Mutant ZP1 in familial infertility N Engl J Med 2014 370 13 1220 6 DOI:10.1056/NEJMoa1308851 PMid:24670168 PMCid: PMC4076492
[26]
M Bungum L Bungum P Humaidan A prospective study, using sibling oocytes, examining the effect of 30 seconds versus 90 minutes gamete co-incubation in IVF Hum Reprod 2006 21 2 518 23 DOI:10.1093/humrep/dei350 PMid:16239314
[27]
M Jin E Fujiwara Y Kakiuchi M Okabe Y Satouh SA Baba K Chiba N Hirohashi Most fertilizing mouse spermatozoa begin their acrosome reaction before contact with the zona pellucida during in vitro fertilization Proc Natl Acad Sci USA 2011 108 12 4892 4896 DOI:10.1073/pnas.1018202108 PMid:21383182 PMCid: PMC3064341
[28]
I Tanii T Aradate K Matsuda A Komiya H Fuse PACAP-mediated sperm–cumulus cell interaction promotes fertilization Reproduction 2011 141 2 163 171 DOI:10.1530/REP-10-0201 PMid:21071464
[29]
M Shimada Y Yanai T Okazaki N Noma I Kawashima T Mori JS Richards Hyaluronan fragments generated by sperm-secreted hyaluronidase stimulate cytokine/chemokine production via the TLR2 and TLR4 pathway in cumulus cells of ovulated COCs, which may enhance fertilization Development 2008 135 11 2001 2011 DOI:10.1242/dev.020461 PMid:18434414
[30]
I Kligman M Glassner BT Storey GS Kopf Zona pellucida-mediated acrosomal exocytosis in mouse spermatozoa: characterization of an intermediate stage prior to the completion of the acrosome reaction Dev Biol 1991 145 2 344 55 DOI:10.1016/0012-1606(91)90133-N
[31]
MG Buffone Since its discovery in 1952, acrosomal exocytosis (also called acrosome reaction) has been a fascinating process Adv Anat Embryol Cell Bio 2016 l 220 v vi
[32]
C Stival C Puga Molina Ldel B Paudel MG Buffone PE Visconti D Krapf Sperm capacitation and acrosome reaction in mammalian sperm Adv Anat Embryol Cell Biol 2016 220 93 106 DOI:10.1007/978-3-319-30567-7_5 PMid:27194351
[33]
HM Florman NL First The regulation of acrosomal exocytosis. I. Sperm capacitation is required for the induction of acrosome reactions by the bovine zona pellucida in vitro Dev Biol 1988 128 2 453 63 DOI:10.1016/0012-1606(88)90307-7
[34]
S Morisawa GN Cherr Acrosome reaction in spermatozoa from hagfish (Agnatha) Eptatretus burgeri and Eptatretus stouti: acrosomal exocytosis and identification of filamentous actin Dev Growth Differ 2002 44 4 337 44 DOI:10.1046/j.1440-169X.2002.00643.x PMid:12175368
[35]
FA La Spina LC Puga Molina A Romarowski AM Vitale TL Falzone D Krapf N Hirohashi MG Buffone Mouse sperm begin to undergo acrosomal exocytosis in the upper isthmus of the oviduct Dev. Biol 2016 411 2 172 182
[36]
OO Anakwe GL Gerton Acrosome biogenesis begins during meiosis: evidence from the synthesis and distribution of an acrosomal glycoprotein, acrogranin, during guinea pig spermatogenesis Biol Reprod 1990 42 2 317 28 DOI:10.1095/biolreprod42.2.317 PMid:1692485
[37]
D Escalier JM Gallo M Albert G Meduri D Bermudez G David J Schrevel Human acrosome biogenesis: immunodetection of proacrosin in primary spermatocytes and of its partitioning pattern during meiosis Development 1991 113 3 779 88
[38]
G Berruti C Paiardi Acrosome biogenesis: Revisiting old questions to yield new insights Spermatogenesis 2011 1 2 95 98 DOI:10.4161/spmg.1.2.16820 PMid:22319656 PMCid: PMC3271650
[39]
C Ito K Toshimori Acrosome markers of human sperm Anat Sci Int 2016 91 2 128 42 DOI:10.1007/s12565-015-0323-9 PMid:26748928
[40]
JD Bleil PM Wassarman Identification of a ZP3-binding protein on acrosome-intact mouse sperm by photoaffinity crosslinking Proc Natl Acad Sci U S A 1990 87 14 5563 7 DOI:10.1073/pnas.87.14.5563 PMid:2371290 PMCid: PMC54365
[41]
PM Wassarman Mammalian fertilization: the strange case of sperm protein 56 Bioessays 2009 31 2 153 8 DOI:10.1002/bies.200800152 PMid:19204987
[42]
JA Foster BB Friday MT Maulit C Blobel VP Winfrey GE Olson KS Kim GL Gerton AM67, a secretory component of the guinea pig sperm acrosomal matrix, is related to mouse sperm protein sp56 and the complement component 4-binding proteins J Biol Chem 1997 272 19 12714 22 DOI:10.1074/jbc.272.19.12714 PMid:9139729
[43]
Y Muro MG Buffone M Okabe GL Gerton Function of the acrosomal matrix: zona pellucida 3 receptor (ZP3R/sp56) is not essential for mouse fertilization Biol Reprod 2012 86 1 1 6 DOI:10.1095/biolreprod.111.095877 PMid:21998167 PMCid: PMC3313668
[44]
JC Herr CJ Flickinger M Homyk K Klotz E John Biochemical and morphological characterization of the intra-acrosomal antigen SP-10 from human sperm Biol Reprod 1990 42 1 181 93 DOI:10.1095/biolreprod42.1.181 PMid:2310816
[45]
HP Osuru P Pramoonjago MM Abhyankar E Swanson LA Roker H Cathro PP Reddi Immunolocalization of TAR DNA-binding protein of 43 kDa (TDP-43) in mouse seminiferous epithelium Mol Reprod Dev 2017 84 8 675 685 DOI:10.1002/mrd.22851 PMid:28600885 PMCid: PMC5577912
[46]
K Hoshi H Sasaki K Yanagida A Sato A Tsuiki Localization of fibronectin on the surface of human spermatozoa and relation to the sperm-egg interaction Fertil Steril 1994 61 3 542 7 DOI:10.1016/S0015-0282(16)56590-X
[47]
AS Lalmansingh CJ Urekar PP Reddi TDP-43 is a transcriptional repressor: the testis-specific mouse acrv1 gene is a TDP-43 target in vivo J Biol Chem 2011 286 13 10970 82 DOI:10.1074/jbc.M110.166587 PMid:21252238 PMCid: PMC3064152
[48]
D Ripoche J Gout RM Pommier R Jaafar CX Zhang L Bartholin P Bertolino Generation of a conditional mouse model to target Acvr1b disruption in adult tissues Genesis 2013 51 2 120 7 DOI:10.1002/dvg.22352 PMid:23109354
[49]
BK Li X Wang CX Liu SB Zheng HL Li LP Li AB Xu Influence of reproductive tract obstruction on expression of epididymal proteins and their restoration after patency Asian J Androl 2013 15 1 105 9 DOI:10.1038/aja.2012.64 PMid:22922320 PMCid: PMC3739126
[50]
AS Samuel RK Naz Isolation of human single chain variable fragment antibodies against specific sperm antigens for immunocontraceptive development Hum Reprod 2008 23 6 1324 37 DOI:10.1093/humrep/den088 PMid:18372255 PMCid: PMC2902835
[51]
R Naz A Aleem Effect of immunization with six sperm peptide vaccines on fertility of female mice Soc Reprod Fertil Suppl 2007 63 455 64
[52]
G Morin C Lalancette R Sullivan P Leclerc Identification of the bull sperm p80 protein as a PH-20 ortholog and its modification during the epididymal transit Mol Reprod Dev 2005 71 4 523 34 DOI:10.1002/mrd.20308 PMid:15892045
[53]
JW Overstreet Y Lin AI Yudin SA Meyers P Primakoff DG Myles DF Katz CA Vandevoort Location of the PH-20 protein on acrosome-intact and acrosome-reacted spermatozoa of cynomolgus macaques Biol Reprod 1995 52 1 105 14 DOI:10.1095/biolreprod52.1.105 PMid:7711169
[54]
KJ Chen S Sabrina NS El-Safory GC Lee CK Lee Constitutive expression of recombinant human hyaluronidase PH20 by Pichia pastoris J Biosci Bioeng 2016 122 6 673 678 DOI:10.1016/j.jbiosc.2016.06.007 PMid:27373489
[55]
GN Cherr AI Yudin JW Overstreet The dual functions of GPI-anchored PH-20: hyaluronidase and intracellular signaling Matrix Biol 2001 20 8 515 25 DOI:10.1016/S0945-053X(01)00171-8
[56]
C Zhou W Kang T Baba Functional characterization of double-knockout mouse sperm lacking SPAM1 and ACR or SPAM1 and PRSS21 in fertilization J Reprod Dev 2012 58 3 330 7 DOI:10.1262/jrd.2011-006 PMid:22362218
[57]
S Yoon KT Chang H Cho J Moon JS Kim SH Min DB Koo SR Lee SH Kim KE Park YI Park E Kim Characterization of pig sperm hyaluronidase and improvement of the digestibility of cumulus cell mass by recombinant pSPAM1 hyaluronidase in an in vitro fertilization assay Anim Reprod Sci 2014 150 3-4 107 14
[58]
C Perez-Patino I Parrilla I Barranco M Vergara-Barberan EF Simo-Alfonso JM Herrero-Martinez H Rodriguez-Martinez EA Martinez J Roca New in-depth analytical approach of the porcine seminal plasma proteome reveals potential fertility biomarkers J Proteome Res 2018 17 3 1065 1076 DOI:10.1021/acs.jproteome.7b00728 PMid:29411616
[59]
CP Blobel TG Wolfsberg CW Turck DG Myles P Primakoff JM White A potential fusion peptide and an integrin ligand domain in a protein active in sperm-egg fusion Nature 1992 356 6366 248 52 DOI:10.1038/356248a0 PMid:1552944
[60]
C Cho Testicular and epididymal ADAMs: expression and function during fertilization Nat Rev Urol 2012 9 10 550 60 DOI:10.1038/nrurol.2012.167 PMid:22926424
[61]
C Cho H Ge D Branciforte P Primakoff DG Myles Analysis of mouse fertilin in wild-type and fertilin beta(-/-) sperm: evidence for C-terminal modification, alpha/beta dimerization, and lack of essential role of fertilin alpha in sperm-egg fusion Dev Biol 2000 222 2 289 95 DOI:10.1006/dbio.2000.9703 PMid:10837118
[62]
E Kim JW Lee DC Baek SR Lee MS Kim SH Kim CS Kim ZY Ryoo HS Kang KT Chang Processing and subcellular localization of ADAM2 in the Macaca fascicularis testis and sperm Anim Reprod Sci 2010 117 1-2 155 9
[63]
A Fabrega B Guyonnet JL Dacheux JL Gatti M Puigmule S Bonet E Pinart Expression, immunolocalization and processing of fertilins ADAM-1 and ADAM-2 in the boar (Sus domesticus) spermatozoa during epididymal maturation Reprod Biol Endocrinol 2011 9 96
[64]
N Inoue M Ikawa A Isotani M Okabe The immunoglobulin superfamily protein Izumo is required for sperm to fuse with eggs Nature 2005 434 7030 234 8 DOI:10.1038/naturne03362 PMid:15759005
[65]
E Bianchi B Doe D Goulding GJ Wright Juno is the egg Izumo receptor and is essential for mammalian fertilization Nature 2014 508 7497 483 7 DOI:10.1038/naturne13203 PMid:24739963 PMCid: PMC3998876
[66]
E Bianchi GJ Wright Cross-species fertilization: the hamster egg receptor, Juno, binds the human sperm ligand, Izumo1 Philos Trans R Soc Lond B Biol Sci 2015 370 1661 20140101 DOI:10.1098/rstb.2014.0101 PMid:25533103 PMCid: PMC4275915
[67]
MA Baker L Hetherington A Weinberg N Naumovski T Velkov M Pelzing S Dolman MR Condina RJ Aitken Analysis of phosphopeptide changes as spermatozoa acquire functional competence in the epididymis demonstrates changes in the post-translational modification of Izumo1 J Proteome Res 2012 11 11 5252 64 DOI:10.1021/pr300468m PMid:22954305
[68]
SA Young H Miyata Y Satouh M Muto MR Larsen RJ Aitken MA Baker M Ikawa CRISPR/Cas9-mediated mutation revealed cytoplasmic tail is dispensable for IZUMO1 function and male fertility Reproduction 2016 152 6 665 672 DOI:10.1530/REP-16-0150 PMid:27624483
[69]
XQ Hu SY Ji YC Li CH Fan H Cai JL Yang CP Zhang M Chen ZF Pan ZY Hu F Gao YX Liu Acrosome formation-associated factor is involved in fertilization Fertil Steril 2010 93 5 1482 92 DOI:10.1016/j.fertnstert.2009.01.067 PMid:19285662
[70]
MJ Wolkowicz J Shetty A Westbrook K Klotz F Jayes A Mandal CJ Flickinger JC Herr Equatorial segment protein defines a discrete acrosomal subcompartment persisting throughout acrosomal biogenesis Biol Reprod 2003 69 3 735 45 DOI:10.1095/biolreprod.103.016675 PMid:12773409
[71]
CN Tomes The proteins of exocytosis: lessons from the sperm model Biochem J 2015 465 3 359 70 DOI:10.1042/BJ20141169 PMid:25609177
Share
Back to top