Open access peer-reviewed chapter

Glycosylation on Spermatozoa, a Promise for the Journey to the Oocyte

Written By

Shuangjie Wang, Yadong Li, Aijie Xin, Yang Yang, sheng-ce Tao, Yihua Gu and Huijuan Shi

Submitted: 17 May 2022 Reviewed: 10 July 2022 Published: 24 August 2022

DOI: 10.5772/intechopen.106438

From the Edited Volume

Modifications in Biomacromolecules

Edited by Xianquan Zhan and Atena Jabbari

Chapter metrics overview

91 Chapter Downloads

View Full Metrics

Abstract

Spermatozoa experience a long and tough transit in male and female genital tracts before successful fertilization. Glycosylation helps spermatogenesis, epididymal maturation, passing through cervical mucus, avoiding killing of the female immunologic system, and shaking hands between sperm and egg. Changes in glycosylations along the transit ensure that the right things happen at the right time and place on spermatozoa. Aberrant glycosylations on spermatozoa will negatively affect their fertility. Thus, we developed a lectin array method to examine the glycocalyx of spermatozoa, which will help observe glycosylations occurring on spermatozoa in a normal or abnormal conditions, such as spermatozoa with DEF126 mutation and poor freezability. Intriguingly, binding levels of ABA (Agaricus bisporus agglutinin), a lectin marking the inner layer of the glycocalyx, were changed in these subfertile spermatozoa, which indicates that the integrity of glycocalyx is critical for sperm fertility. In this chapter, we reviewed the impacts of glycosylations on sperm fertility, the lectin array method, and its potential application for sperm function assessment.

Keywords

  • spermatogenesis
  • sperm maturation
  • transit through genital tract
  • glycan profile
  • lectin microarray

1. Introduction

Before mating the oocytes, mammal spermatozoa go on a long adventure, experiencing a series of complicated events along the male and female reproductive tracts [1]. From spermatogonia, spermatozoa develop in the testis, with dramatic changes in morphology and chromatin structure. However, spermatozoa in testis could not fertilize oocytes since their motility and capabilities of recognizing and binding zona pellucida are underdeveloped.

Spermatozoa then enters into the epididymis, several meters long male reproductive tract, and undergo an additional maturation. During the epididymal maturation, also called post-testicular maturation ranges from 1 to 21 days depending on species [2], spermatozoa acquire not only capacities for progressive motility, acrosome reaction, zona pellucida binding and recognition, and spermatozoon-oocyte fusion [3, 4, 5, 6, 7], but also further morphology changes [8, 9, 10, 11], such as chromatin condensation, head size decrease, acrosome reshaping, and droplet migration.

After ejaculation, spermatozoa are transferred into the vagina. Passing through the cervix, uterus, and uterotubal junction, spermatozoa stop at a storage reservoir in the oviduct until ovulation. During ovulation, spermatozoa escape the reservoir, complete the capacitation, migrate to the site of mating, and finally achieve insemination. Despite facilitation guiding to the site of fertilization, a successful spermatozoon faces big challenges in the female genital tract, such as selection, which ensures the fittest one to fertilize, and immune defense, which fights evasion of microbial pathogens [12, 13].

Advertisement

2. Glycosylations on mammal spermatozoa and their physiological functions

Along the journey of spermatozoa, events, such as sperm development, maturation, transit, and fertilization, are mediated by cell–cell interaction, and interaction between spermatozoa and the microenvironment [1]. Providing promises to these events, dramatic modifications on sperm surface occur along the transit of spermatozoa [14]. For instance, high sialylation provides a negative charge surrounding the sperm surface, which results in a repulsive interaction against negatively charged mucins for bypassing cervix mucus [15]; a shell, which protects sperm antigens from immune surveillance in the female genital tract [16]; and an anchor, which helps the epithelial cells hold spermatozoa at the oviductal reservoir [17]. However, during capacitation, lipid component changes resulting in a high membrane fluidity for sperm hyperactivation [18], and desialylation occurs on the sperm surface [19, 20], both of which ensure spermatozoa reach the oocyte and fertilize successfully.

2.1 Glycoconjugates on mammal spermatozoa

A dense glycoconjugate coat with a thickness of 20–60 nm called glycocalyx on animal cells [21], is one of the major surface components on spermatozoa, which participate in such modifications and play critical roles in those interactions along the sperm’s journey to the oocyte [22, 23]. The glycocalyx is synthesized during spermatogenesis in testis and is remodeled through epididymal maturation, the mixing process of sperms and seminal plasma secreted by the accessory glands during ejaculation and transit through the female genital tract [21]. Two of three types of glycoconjugates are identified in sperm glycocalyx: glycoproteins and glycolipids [24], which are classified corresponding to types of glycan-bearing carriers, proteins, or lipids. Besides those glycoconjugates inserted or anchored in the plasma membrane, others are non-covalently attached to the membrane via inter-molecule or hydrophobic interaction [21].

2.1.1 Glycoproteins

Glycosylation is a ubiquitous posttranslational modification on proteins, more than half of human proteins are deduced as glycoproteins according to their amino acid sequences [25]. By exploiting their covalently linked glycans, membrane or surface glycoproteins play roles in intercellular signaling, cell–cell interaction, and cell adhesion [26]. With nucleoside diphosphate or monophosphate glycans as precursors, glycosyltransferases create a glycosidic bond between glycan and peptide backbone. Three types of covalent linkages exist naturally between glycans and polypeptide backbone: N-glycosylation, O-glycosylation, and GPI glycosylphosphatidylinositol [27].

As the most common posttranslational modification in the endoplasmic reticulum (ER) [28], N-linked glycosylation attaches glycans to the amide nitrogen of asparagine or arginine residues within the consensus amino acid sequences or called “sequons,” Asn-X-Thr/Ser, which direct protein folding. In a manner of protein-, cell-, or species-specificity [29], remodeling in Golgi apparatus endows N-glycan structural and functional varieties, which ensure functional complexity of cell surface proteins, such as cell–cell, cell-matrix interaction, or immune responses. N-glycosylation is a prominent posttranslational modification of the protein, about 50% of the proteins have N-glycans [30, 31]. N-glycans are also major carbohydrates on the sperm glycocalyx, which are composed of three types of N-glycans, high mannose, hybrid N-glycans, and complex N-glycans [32]. High mannose N-glycans are essential for Sertoli-germ cell attachment during spermatogenesis, which is a critical cell–cell adhesion for sperm development in testis [33].

Initially in the ER then Golgi apparatus, O-glycosylation covalently links glycans to the oxygen atom at the hydroxyl group of serine, threonine, tyrosine, hydroxylysine, or hydroxyproline residue on the peptide backbone [27]. O-glycosylation has no sequon and core structure, and O-glycan moieties range from monosaccharides to sulfonated polysaccharides. β-defensin 126 (DEFB-126) is a well-known O-linked glycoprotein [34], which is acquired through epididymal maturation and is also an important component of sperm glycocalyx. On the surface of DEFB-126 knock-out sperm, the level of O-glycosylation significantly decreased, and the spermatozoa are male infertility [35].

Glycosylphosphatidylinositol (GPI)-anchor is glycolipid, containing a Mana1–6Mana1–4GlcNa1–6PI motif [36]. Performed by ER-situated transamidase in ER, the carboxyl-terminus of proteins is connected to the ethanolamine moiety of GPI-anchor via an amide bond. Located on the extracellular side of the cytoplasmic membrane, the protein is bound to the cell membrane through GPI-anchoring. Structures and functions of GPI-anchored protein are widely diverse and play important roles in many biological processes. During sperm migration in the epididymis, various GPI-anchored proteins are obtained on the surface under the mediation of epididymosomes in the epididymal fluid. A well-studied GPI-anchored protein is sperm adhesion molecule 1(SPAM1/PH-20). The SPAM1 secreted by the epididymis exists in both epididymal fluid and epididymal exosomes; SPAM1 in the latter can be attached to the epididymal sperm surface by GPI anchoring [37].

2.1.2 Glycolipids

In addition, to proteins, lipid moieties are another type of glycan carrier. Glycosphingolipids are major glycolipids in the cell membrane of animals, which are primarily synthesized in the endoplasmic reticulum, and whose carbohydrate moieties are further modified in the Golgi apparatus [38]. Glycosphingolipids contain one or more carbohydrate residues linked to a specific hydrophobic lipid moiety, sphingoids, or ceramides (N-acylated sphingoid), through a glycosidic bond [39]. According to their glycan structures, glycosphingolipids are classified into the following, namely, ganglio-, isoganglio-, lacto-, neolacto-, lactoganglio-, globo-, isoglobo-, muco-, gala-, neogala-, mollu-, arthro-, schisto- and spirometo-series.

Glycolipids participate in cellular functions in animals, and their biosynthesis and distribution change corresponding to different physiological functions and stages [40]. Glycolipids are essential for many processes of male reproduction [41]. Via manipulated deficiency of galactosyl transferase or sulfotransferase, mouse models with targeted deletion of seminolipid, a sulfoglycolipid, show suspension of spermatogenesis at the stage of meiosis [42]. And, a mouse model with complex ganglioside deficiency resulting from targeted deletion of N-acetylgalactosaminyl transferase is reported to be male infertility [43].

2.2 Physiological functions of sperm glycoconjugates

2.2.1 Spermatogenesis

Occurring in seminiferous tubules of testis, spermatogenesis is a highly regulated series of sperm development, involving in the proliferation of spermatogonia, differentiation, and maturation into spermatozoa. Sertoli-germ cell interaction is vital for spermatogenesis [44], which supports, transports, and protects germ cells. Mouse model studies targeted deletion of N-acetylglucosaminyltransferase-II (GnT-II) and α-mannosidase IIx (MX) suggested that a specific sperm N-glycan, GlcNAc-terminated triantennary, and fucosylated N-glycan, mediates the adhesion of germ cells to Sertoli cells via protein-carbohydrate interaction [45, 46].

Although N-glycans play a role in spermatogenesis has been widely admitted [3233], proteins containing, such as, glycans remain little known. Leukocyte differentiation antigen (basigin, BSG, also known as CD147) is an essential protein during the process of spermatogenesis, and embryo implantation [47, 48]. which is expressed in various stages of spermatogenic cells (spermatogonia, spermatocytes, and spermatids), Sertoli cells, Leydig cells, and epididymis [49]. Mature spermatozoa are absent in the testis of a mouse model with targeted deletion of BSG. Mass spectrometry and lectin studies reported that N-glycans containing fucosylated high-mannose moieties with terminated N-acetylglucosamines were identified in BSG [49, 50, 51], which is reasonably believed that BSG may participate in Sertoli-germ cell adhesion.

As sulfated glycoconjugates, seminolipids are predominant glycolipids on the plasma membrane of mammalian male germ cells [52]. With a very high cell specificity, seminolipids are synthesized in primary spermatocytes and exist in the subsequent male germ cell types [53]. Seminolipid-deficient mice illustrate male infertility and suspension of spermatogenesis before first meiotic division [42], which might result from failure of uptake of lactate for spermatocytes [54]. In vitro studies indicated that Sertoli membrane protein could bind seminolipids, which suggests that seminolipids may take part in Sertoli-germ cell adhesion during spermatogenesis [55].

Gangliosides are sialic acid terminated glycolipids. Complex ganglioside deficient mice showed male infertility, which might result from failure of testosterone transportation in Leydig cells [43]. Due to their reported functions on carbohydrate-dependent cell adhesion [56], gangliosides are also supposed to play roles in Sertoli-spermatocyte interaction [55].

2.2.2 Epididymal maturation

Through the maturation along the epididymal tract, spermatozoa acquire not only fertility, including progressive movement, but further modifications of morphology (especially, migration of the cytoplasmic droplet), molecular components and cell surface.

During the transit from caput down to cauda epididymis, a significant change in sperm surface is an increase in surface sialylation [56, 57]. Sialic acid is a strongly negatively charged acidic oligosaccharide, sialylation occurs on the terminus of N- or O-glycans. Due to the sialylation on the surface, spermatozoa are covered with a negative charge [58], which could facilitate sperm transit along the genital tracts, shield sperm antigens, and protect spermatozoa from an immune response in the female genital tract [24].

Integrating the sialylated glycoproteins or glycopeptides secreted from epididymal epithelium to the cell surface is a major means to sialylate the sperm surface. Several epididymal sialylated proteins have been reported to be associated with the spermatozoa during epididymal maturation, including proteins D and E, basigin, CD59, fertilin, HE2, HE4 [59], and HE5/CD52 [59, 60].

Secreted from the epididymal epithelium into the epididymal lumen, recent studies showed its significance in the sialylation of sperm surface [61]. CD52 is N-glycosylated mainly in a form of three-antennary oligosaccharides with a terminus of sialylation, which results in a negative charge on CD52. CD52 protects sperm from complement-dependent immune responses, facilitates the process of sperm glycocalyx remodeling, and prevent sperms from agglutination with each other [6162]. CD52 is a GPI-anchored protein [61], which is transported and integrated to the sperm surface via epididymosome [63].

Acquisition of membrane lipids from epididymal epithelium also exploits epididymosome [64], relative quantities of cholesterol and sphingomyelin increase during the epididymal maturation. However, other strategies are also applied for the remodeling of sperm surface components.

Originated in the epididymal epithelium and secreted into the lumen, DEFB-126 is also a highly sialylated O-linked glycoprotein, which tightly associates with the surface of human and macaque spermatozoa. It is believed that DEFB-126 is a major component of sperm glycocalyx, and a predominant contributor to the negative charge of sperm surface [34, 35, 65].

The majority of glycosyltransferase activities are found within the epididymal lumen [66]. High activities of both fucosyltransferase and sialyltransferase exist in caput epididymis and decrease gradually at cauda epididymis. It is suggested that further glycosylation may occur on the sperm surface.

2.2.3 Transit along the female genital tract

After ejaculation, spermatozoa will encounter not only selection pressure, which knocks out spermatozoa with poor motility or morphology via retrograde flow in the female genital tract [67], but immune pressures, which eliminates “foreigners,” such as microbial pathogens, and even spermatozoa.

Like CD52 mentioned above, CD55, also a GPI-anchored protein, is a type of complement-regulating protein on the plasma membrane of human sperm against immune attack [68]. CD59 is an inhibitory molecule of the membrane attack complex of the complement system, which is also localized in the sperm plasma membrane [69]. These three GPI-anchored proteins with N-glycans protect spermatozoa from immunological attack in the female genital tract [70].

DEFB-126 is an O-linked glycoprotein. Like CD52, CD55, and CD59, DEFB-126 is integrated to sperm surface during epididymal maturation, but in a different way [65]. High sialylation on DEFB-126 not only protects spermatozoa from phagocytosis in the female genital tract, but helps penetrate through cervix mucus. It is supposed that tri- and tetra-antennary glycans with Lewis-X and Lewis-Y sequences on these glycoproteins acquired in epididymis may help escape immune detection [71], and suppress antigen-immune responses [72].

Besides pressures, the female genital tract also conducts facilitation and regulations, which help spermatozoa do the right thing at the right time, right place. Glycodelin is a secretary protein with N-glycans, which is composed of four glycoforms, namely, glycodelin-A (amniotic fluid) [73, 74], glycodelin-F (follicular fluid) [75], glycodelin-C (cumulus matrix) [76], and glycodelin-S (seminal plasma) [77], according to the tissues and fluids where the isoforms are identified. By fucosyltransferase-5 on sperm surface, glycodelin isoforms bind to spermatozoa with a spatiotemporal specificity [78], and perform diverse functions [79], with their different glycan moieties [80].

After ejaculation, spermatozoa bind glycodelin-S on the whole head [75], an abundant protein in seminal plasma secreted from the seminal vesicle, which inhibits albumin-induced cholesterol efflux and prevents sperm capacitation. After cervix penetration, glycodelin-S on spermatozoa is displaced by glycodelin-A, which is secreted from maternal endometrial epithelial cells into uterine and amniotic fluid [80], and covers the sperm acrosomal region. Glycodelin-A inhibits sperm capacitation and protects spermatozoa from immune attack. In the fallopian tube, glycodelin-F binds to spermatozoa in the acrosomal region and inhibits the progesterone-induced acrosome reaction. GdC in the cumulus matrix displaces sperm-bound glycodelin-A and -F during sperm cumulus penetration, covers spermatozoa in the equatorial region, and promotes the zona binding capacity of the spermatozoa.

Glycodelin-A carries high mannose, hybrid, and complex-type structure [73]. Whereas, glycans on glycodelin-S are much different, which are highly fucosylated, and contain a complex-type structure of bi-antennary glycans with Lewis-X and Lewis-Y antennae. Glycodelin-A, -F, -C have a similar topology of carbohydrate moiety, with different degrees of sialylation. Glycodelin-A and glycodelin-F are diversely sialylated, which are immunosuppressive, while non-sialylated glycodelin-S works on inhibition of capacitation, and low-sialylated glycodelin-C as successful singleton insemination [79].

In the female genital tract, spermatozoa also take part in the remodeling of the sperm surface. Ovulation triggers the release of sperm at the oviductal reservoir and sperm capacitation. During their last stage of the journey to the oocyte, spermatozoa carry out a series of desialylation on the sperm surface, including releasing high-sialylated DEFB-126 from spermatozoa to free the latter in the oviductal reservoir [17], the release of two neuraminidases (NEU1 and NEU3) to remove sialic acids on sperm surface [81], and release of sialylated GPI-anchored glycoprotein via raft redistribution due to cholesterol efflux, such as CD52 [82]. Desialylation and raft redistribution joined by glycolipids, such as ganglioside GM1and seminolipids, help present functional surface proteins for cumulus penetration, such as SPAM1 [83], for sperm-zone pullucide recognition and binding, such as dicarbonyl/L-xylose Reductase (P34H) [84], and cysteine-rich secretory protein-1 (CRISP1) [85].

Advertisement

3. Lectin microarray, a “sugar decipher” for sperm surface glycoconjugates

Glycosylation ubiquitously exists in cellular biology, however, it is still a poorly understood posttranslational modification, due to its complexities. One is the vast diversity of glycan structure [86]. Due to carbohydrate structural and linkage features, it is established that all possible branched or linear oligosaccharide isomers yield 1.05 × 1012 structures.

The other is the complexity of glycan biosynthesis, due to the complicated biosynthesis machinery and no preexisting template molecule. Possibly, 1–2% of the genome produces glycan biosynthesis-related proteins, such as glycosyltransferases, glycosidases, and transporters. Carbohydrate biosynthesis does not follow “the central dogma,” [87] but several factors instead [88], such as levels, activities, and trafficking of glycosyltransferases, availabilities of carriers and nucleotide sugar substrates, different physiological functions and statuses, and a variety of outer stimuli [89], such as nutrition [89], oxidative stress [90], and so on.

In other words, extreme complexity enables glycans as “the third group of bio-informative molecules,” [86] which integrate panoramic information on cell biology [91, 92], including genomes encoding proteins and biosynthesis machinery of lipids or glycans, and interactions between cell and environmental factors. Glycan profiles on cell surfaces help distinguish individuals and physiological statuses. For an instance, α-linked N-acetylgalactosamine (GalNac) and β-linked GalNac on Sda/GM2 is exploited to identify spermatogonial subpopulations in cattle, pigs, and horses [93].

3.1 The lectins and other glycan-binding proteins

Lectins are a group of glycan-binding proteins that selectively bind different glycans with specific structures, which are first described back in the late of the nineteenth century [94]. As more and more plant lectin-glycan pairings have been identified [95], a sophisticated, reliable, and convenient tool set emerged (Summarized in Table 1). Although lectins could not acquire detailed structural information, they are capable to tell the differences between glycan profiles on different cell surfaces [96].

LectinsCarbohydrate specificitySources
AAAFucoseAnguilla Anguilla
AALFucα1–2,3,4Aurentia
ABAGalβ1–3GalNAcAgaricus bisporus
ACAGalβ1–3GalNAcAmaranthus caudatus
AMAManα-Arum maculatum
APAMannoseAllium porum
APPGalNAcα-, GalNAcβ-Aegopodium podagraria
ASAManα1–3Allium sativum
BDAGalNAcα-, GalNAcβ-Bryonia dioica
BBCGalNAcα-, GalNAcβ-Phaseolus vulgaris sp.
BPLGalβ1–3GalNAcBauhinia Purpurea
CAGalβ1–4GlcNAc, GalNAcβ1–4GlcNAcColchicum autumnale
CAAComplex glycans (GlcNAcβ1–2Manα1–3(GlcNAcβ1–2Manα1–6)Manβ1–4GlcNAcβ1–4GlcNAcβ-)Caragana arborescens
CALSEPAMannose, Glucose, Glcα1–4GlcCalystegia sepiem
CCANeu5AcCancer antennarius
Con AManα-, Gluα-Canavalia ensiformis
CPAMannoseCicer arietinum
CSAGalNAcα-Cytisus sessilifolius
DBAGalNAcα1–3GalNAc, GalNAcα1–3GalDolichos biflorus
DSLNeu5Ac-Gal/GalNAcDatura Stramonium
ECAGalβ1–4GlcNAcErythrina cristagalli
EEAGalNAcβ-Euonymus europaeus
GHAGalα-, GalNAcα-Glechoma hederacea
GNAManα-Galanthus nivalis
GS-IGalα-, GalNAcα-Griffonia simplicifolia
GS-IA4GalNAcα-Griffonia simplicifolia
GS-IB4Galα-Griffonia simplicifolia
GS-IIGlcNAcα-, GlcNAcβ-Griffonia simplicifolia
HAAGlcNAcα-, GalNAcα-Helix aspersa
HHAManα-Hippeastrum hybrid
HMAGalNAcα-, Fucα-, Neu5AcHomarus americanus
HPAGalNAcα-Helix pomatia
IAAGalNAcIberis amara
IRAGalNAcα-, GalNAcβ-lris hybrid
JacalinGalα-, Galβ-, GalNAcα-O linkArtocarpus integrifolia
LAAGlcNAcβ-, GlcNAcβ1–4GlcNAcLaburnum alpinum
LALFucoseLaburnum anagyroides
LBAGalNAcα-, complex glycans (GalNAcα1–3(Fucα1–2)Gal)Phaseolus lunatus
LcAMannoseLens culinaris
LcHComplex glycans (Man/GlcNAc core with Fucα1–6)Lens culinaris
LcH AManα-, Glcα-, GlcNAcLens culinaris
LcH BManα-, Glcα-, GlcNAcLens culinaris
LEL (TL)GlcNAcLycopersicon esculentum
LFANeu5AcLimax flavus
LPANeu5AcLimulus polyphemus
LTLFucα1–2,3,4Lotus tetragonolobus
MAANeu5Acα2–3GalMaackia amurensis
MAL-IGalβ1–4GlcNAcMaackia Amurensis
MAL-IINeu5Acα2–3Maackia Amurensis
MNA-GGalα-, Galβ-Morniga G
MNA-MManα-Morniga M
MOAGalα1–3Marasmium oreades
MPAGalα-, GalNAcα-Maclura pomifera
NPAMannoseNarcissus pseudo-narcissus
PSAL-Fucα1,6GlcNAcPisum sativum
PHA-EComplex glycans (Galβ1–4GlcNAcβ1–2(Galβ1–4GlcNAcβ1–6) Man)Phaseolus vulgaris
PHA-LComplex glycans (Galβ1–4GlcNAcβ1–2Man)Phaseolus vulgaris
PMAManα1–3Polygonatum mulitiflorum
PNAGalβ1,3GalNAcArachis hypogaea
PSLComplex glycans (Neu5Acα2–6Galβ1–4 GlcNAc, Neu5Acα2–6Galβ1–4Glc)Polyporus squamosus
PTA GalGalactosePsophocarpus tetragonolobus
PTA GalNAcGalNAcPsophocarpus tetragonolobus
PWM (PWA)GlcNAcβ1–4GlcNAcPhytolacca americana
RCA-IGalβ-Ricinus communis
RCA-IIGalβ-, GalNAcβ-, Galβ1–4Glc (Lactose)Ricinus communis
SBAGalNAcα/β-Soybean
SHAGalNAcSalvia horminum
SJAGalNAcβ-Sophora japonica
SNANeu5Acα2–6Gal/GalNAcSambucus nigra
SNA-INeu5Acα2–6Galβ1–4GlcNAc, Neu5Acα2–6 Galβ1–4GlcSambucus nigra
SNA-IIGalβ-, GalNAcβ-Sambucus nigra
SSAGalNAc-O linkSalvia sclarea
STL (PL)GlcNAc, Neu5AcSolanum Tuberosum
Succinyl-Con AMannoseC. ensiformis
TKAGalβ-, Galβ1–4Glc (Lactose)Trichosanthes kirilowii
TLα-GalNAc, β-GalNAc, GalNAc, Galatose, FucoseTulipa sp.
UDAGalNAcβ-Urtica dioica
UEA-IFucα-, Fucα1–2Galβ1–4GlcNAcUlex europaeus
UEA-IIGlcNAcβ-Ulex europaeus
VAAGalβ-Viscum album
VFAManα-Vicia fava
VGAGalβ1–3GalNAcVicia graminea
VRAGalα-,Vigna radiata
VVAGalNAcα-, GalNAcα1–3GalVicia villosa
VVA ManMannoseVicia villosa
WFAGalNAcα/β-Wisteria floribunda
WGAGlcNAcβ-Triticum vulgaris

Table 1.

Incomplete list of commercial plant lectins.

The lectin carbohydrate specificities were summarized from: 1, the Consortium for Functional Glycomics (http://functionalglycomics.org); 2, Product information from EY or Vector Laboratories, Inc.

Animal lectins are endogenous glycan-binding proteins, which are involved in a variety of biological processes, and could provide more biological function clues for glycans (Summarized in Table 2). Intriguingly, summarized in Table 3, a group of sperm-egg receptors on spermatozoa are reported to have the capability of glycan-binding, including lectin-like proteins, glycosyltransferases, and glycosidases [132, 133]. We identified a chitobiase, lysosomal di-N-acetylchitobiase (CTBS), on mouse spermatozoa [Data unpublished]. CTBS is expressed on spermatozoa with species specificity and shows the binding ability of Lewis-X, which could inhibit mouse sperm-egg binding [134].

Lectin typesSub-typesCommon structuresMembersCarbohydrate specificityReference
R-TypeThe mannose receptor familyType I trans-
membrane glycoproteins containing a single fibronectin type II domain, multiple C-type lectin domains (CTLDs), and an amino-terminal cysteine-rich domain.
The mannose receptorC-type domain:
acetylglucosamine, mannose, and fucose;
C-type domain:
sulfated glycans containing 3-O-sulfated galactose, 3-O-sulfated Lex, and 3-O-sulfated Lea;
[97]
The phospholipase A2 receptorPLA2 neurotoxins[98]
DEC-205Phosphorothioated cytosine–guanosine (CpG) oligonucleotides[99]
Endo180N-acetyl-glucosamine, mannose, and fucose[100]
UDP-GalNAc:
polypeptide α-N-acetylgalactosaminyl-transferases
Type II transmembrane proteins containing an amino-terminal catalytic domain and a carboxy-terminal R-type domainppGalNAcT1–20UDP-GalNAc[101]
L-TypeProtein quality control and sorting relatedType I membrane protein with a lumenal portion containing a Ca++-binding domain a proline-rich long hairpin loop called the P domain, and a L-type lectin domain.Calnexin (CNX) and calreticulin (CRT, missing the cytoplasmic and transmembrane regions)Misfolded or aggregated glycoproteins[102, 103]
Dilysine/diphenylalanine KKFF retention/retrieval motif in cytoplasmic carboxyl terminusERGIC-53 and related proteins ERGL, VIP36, and VIPLOligomannose-type glycans[104]
OthersPentameric proteins containing L-type lectin foldsPentraxins and related proteinsPhosphocholine residues on polysaccharides and on phospholipids;
carbohydrate derivatives on bacterial polysaccharides
[105]
Containing laminin G domain-like (LG) modulesLamininsHeparin. sulfatide, and α-dystroglycan (α-DG)[106]
P-TypeMannose 6
-phosphate receptor
Type I trans-membrane glycoproteins containing large extracytoplasmic domains, transmembrane regions, and carboxy-terminal cytoplasmic domains.Cation-dependent mannose 6-phosphate receptorN-glycans containing mannose 6-phosphate[107, 108]
Cation-independent mannose 6-phosphate receptor
I-TypeSialic acid-binding immunoglobulin-like lectins (Siglecs)Type-1 membrane proteins containing a Sia-binding domain (a V-set domain at amino terminus), and varying numbers of C2-set Ig domains that act as spacers, between Sia-binding site and plasma membraneSubgroup1: Siglec-1(Sn), Siglec-2 (CD22), Siglec-4 (MAG), and Siglec-15; (Based on sequence similarity)Sialic acids[109, 110, 111, 112]
Subgroup2: CD33 related Siglecs. (Siglec-3, -5 to -11, -14 and -16, based on sequence similarity)
OthersImmunoglobulin superfamily (IgSF) members other than Siglecs containing a V-set domain.Paired immunoglobulin-like type-2 receptors (PILRs)Mucin-like O-glycosylated membrane proteins[113, 114]
Platelet endothelial cell adhesion molecule (PECAM)-1α2–6-linked sialic acids[115, 116]
Neural cell adhesion molecule (NCAM) and basigin (CD147)Oligomannose-type glycans[117]
Intercellular adhesion molecule (ICAM)-1Hyaluronan[118]
C-TypeThe Ashwell-Morell receptorTransmembrane heteroligomeric glycoprotein complex composed of ASGPR1 (HL-1) and ASGPR2 (HL-2) subunitsExposed β-linked galactose residues on asialoglycoproteins[119, 120]
CollectinsTransmembrane proteins containing an N-terminal triple-helical collagenous region, an α-helical coiled-coil trimerizing neck region, and a C-terminal C-type lectin domain.Proteins containing 185-Glu-Pro-AsnMannose-like sugars[121]
Proteins containing 185-Gln-Pro-AspGalactose-like sugars
Myeloid C-type lectinsTransmembrane proteins containing immunoreceptor tyrosine-based activation motif (ITAM) motifs in their cytoplasmic portion and extracellualr C-type lectin domainsDectin-1 cluster, including dectin-1, CLEC-1, CLEC-2, LOX-1, CLEC12b, and CLEC9aVariate of glycans. Dectin-1 bings β-glucans, which are polymers of a backbone of β1–3-linked glucose and side chains of β1–6-linked glucose[122, 123]
Dectin-2 cluster including dectin-2, blood DC antigen 2 (BDCA-2), DC immunoactivating receptor (DCAR), DC immunoreceptor (DCIR), CTL superfamily 8 (CLECSF8), and MINCLE (CLEC4E)Variate of glycans. Dectin-2 bings α-mannans, which are polymers of α1–6-linked mannose with α1–2-linked mannose side chains
SelectinsType-1 transmembrane glycoproteins, containing an N-terminal C-type lectin domain followed by a consensus epidermal growth factor (EGF)-like domain, a series of consensus repeats, and a transmembrane domain with a short intracellular tailP-selectin on plateletsP-ctin glycoprotein ligand-1 (PSGL-1),
mucins containing highly clustered O-glycans bearing SLex antigens and sulfate esters,
SLex/a rich sulfated mucins
[124, 125, 126, 127, 128]
E-selectin on endothelial cellsPSGL-1,
other glycoproteins that express the SLex antigen on either N- or O-glycans,
long-chain glycosphingolipids expressing the SLex antigen
L-selectin on leukocytesPSGL-1,
ligands containing sulfated glycans, such as 6-sulfo-SLex on both core-2 O-glycans and on extended core-1 O-glycans
Other proteins with C-type lectin domainsA number of proteins with CTLDs have been identified in the pancreas and kidneyUnknown
GalectinsProto-typical GalectinsContaining a carbohydrate-recognition domain (CRD)Galectin-1, -2, -7, -10, -11, -13, -14, and -15Glycans with terminal β-Gal residues[129, 130]
Chimera-type GalectinsContaining a single CRD and an amino terminus with high ratio of proline, glycine, and tyrosine residuesGalectin-3Glycans with repeating [-3Galβ1–4GlcNAcβ-]n or poly-N-acetyllactosamine sequences
Tandem-repeat GalectinsContaining two CRDs connected by a peptide linkerGalectin-4, -8, -9, and -12α2–3-sialylated glycans, blood group A determinant on a LacNAc core
Glycosaminoglycan (GAG)-binding proteinsDo not have common foldsVarious GAG-binding proteinsGlycosaminoglycan (GAG)[131]

Table 2.

Major human carbohydrate-binding proteins.

Classification from Varki A, Cummings RD, Esko JD, et al., editors. Essentials of Glycobiology [Internet]. 3rd edition. Cold Spring Harbor (NY): Cold Spring Harbor Laboratory Press; 2015–2017. Available from: https://www.ncbi.nlm.nih.gov/books/NBK310274/

SpeciesSperm receptorReference
Mouseβ-1,4-galactosyltransferase (β-1,4-GalT)[132]
α-D-mannosidase[132]
sulphoglycolipid immobilizing protein (SLIP1)[132]
56 kDa galactose binding protein (sp56)[132]
α-fucosidase[133]
fructosyltransferase[133]
di-N-acetylchitobiase (CTBS)Data unpublished
Ratα-D-mannosidase[132]
galactose receptor[132]
Humanmannose-binding protein[132]
α-D-mannosidase[132]
β-1,4-galactosyltransferase (β-1,4-GalT)[132]
galactose lectin[132]
selectin-like molecule[132]
PH-20[133]
α-fucosidase[133]
fucosyltransferase-5 (FUT5)[78]
dicarbonyl/L-xylulose reductase (DCXR)[133]

Table 3.

Sperm surface glycan-binding receptors.

Microbial nontoxic B subunits and developed antibodies against specific carbohydrate structures are also glycan-binding proteins, which are exploited to probe the specific glycans [135, 136]. Cholera toxin subunit B (CTxB) is the nontoxic subunit of CTx, which specially bind to the oligosaccharide moiety of a raft-associated glycosphingolipid, ganglioside GM1, and mediates entry of microbe into the host cells [137]. In male fertility, GM1 localization on human spermatozoa detected by CTxB is used as a diagnostic tool for evaluating sperm response to stimuli for capacitation [138].

3.2 Lectin microarray methods

Originated from protein chips, the technique of lectin microarray is established in 2005 for high throughput analysis of glycans [139]. Compared with mass spectrometry-based approaches, analysis of lectin microarray is simple, convenient, and low time-consuming [140]. Due to its non-quantitation and incomplete determination of glycan structures, lectin microarray is more appropriate for analyzing differences between glycan profiles.

We first established the methodology of lectin microarray for the glycan profiling of sperm surface [141]. The major process is summarized in Figure 1. Briefly, the process includes the following steps:

  1. Pre-treatment of lectin microarray and propidium iodide (PI)-labeling of fixed spermatozoa

  2. Incubation of spermatozoa with the microarray

  3. Rinsing the microarray to remove the excess or unbound spermatozoa

  4. Air-drying the microarray, and record the signals on the chip

  5. Data analysis

  6. Verification of the results by using flow cytometry with fluorescence-labeled lectins.

Figure 1.

Scheme of lectin microarray analysis on spermatozoa.

3.3 Sugar codes on spermatozoa with lectin microarray

By using a chip with 91 plant lectins following a previous study [142], we compared surface glycan profiles of five mammalian spermatozoa, human, boar, bull, goat, and rabbit [143]. Roughly, 50 lectins were observed to bind to spermatozoa of 5 all mammalian species, which illustrates a diversity of glycan structures on spermatozoa. Although lectin binding profiles showed a similarity among the five species, a subtle difference was discovered. SBA, HPA, and VVL (binding to GalNac) and GSL I (recognizing galactose) showed strong binding to spermatozoa of boar, bull, goat, and rabbit, while MAL II, PHA-L, PHA-E + L, PHA-E and SNA bound to human spermatozoa strongly. The results suggested that complex glycan structures (recognized by MAL II, PHA-L, PHA-E + L, and PHA-E) and terminal sialic acids (recognized by SNA) are richer in human spermatozoa.

DEFB-126 is an O-linked glycoprotein, which is a major component of human sperm glycocalyx. It is postulated that DEF-126 carries terminal sialylation that is critical for sperm penetration through cervix mucus. We compared glycan profiles between human DEFB-126 mutant and wild-type spermatozoa with lectin microarray analysis [143]. Intriguingly, compared with wild-type spermatozoa, binding of ABA and Jacalin (both are O-linked glycosylation specific lectins) decreased dramatically, while binding of SNA (a lectin recognizing sialic acids deleted) did not change on DEFB-126 mutant spermatozoa. It is suggested that deficiency of DEFB-126 will cause a decrease of O-linked glycans, while the level of sialylation around spermatozoa does not change, which indicates that a more complicated mechanism may happen for subfertility resulting from DEFB-126 mutation.

We also studied the impacts of cryopreservation on sperm fertility [144]. Decreased MAA (a sialic acid-specific lectin) and increased ABA (O-linked glycosylation specific lectin, generally recognizing the inner layer of glycocalyx) were discovered. Which suggested that cryopreservation may destroy the outermost layer of sialylation on sperm glycocalyx. Furthermore, we found a weak ABA binding to cryopreservation tolerant human spermatozoa, which indicates the importance of the integrity of sperm glycocalyx on cryopreservation tolerance [145].

Due to the significance of glycan structural specificity and biological function, we utilized 60 human lectins and lectin-like proteins for microarray analysis of human spermatozoa [146]. Strong bindings were observed for 5 lectins, galectin-1, 7, 8, GalNAc-T6, and ERGIC-53 (LMAN1). Among them, galectin-8 has definite sperm physiology, which could significantly enhance the acrosome reaction.

Advertisement

4. Conclusion

In modern society, concerns are rising on male fertility. About 10–15% of couples worldwide suffer from infertility, of which male factors account for 50% [147]. Although spermatogenesis is a complicated process, involving in more than 2300 genes that are regulated tempo-spatially to develop spermatogonia to spermatozoa [148], only 30% of male infertility are associated with genetic abnormalities [149150]. Furthermore, about 50% of infertility are idiopathic [151, 152], which is a long-term unsolved andrological question, resulting from many possible factors, such as environmental factors, lifestyle, and so on [153]. Thus, a comprehensive andrological assessment is required for a male infertility work-up, including a detailed history analysis, physical examination, well-established semen analysis, endocrine assessment, DNA and epigenetic deficiencies, and so on [154].

The specific molecular mechanism of sperm development, maturation, capacitation, and fertilization could provide clues to solve male reproductive problems. As discussed above, glycan profiling covers panoramic information about genetic background, previous and current environmental impacts, and biological functions of spermatozoa [92]. Due to the universality of glycosylation modifications along the journey of spermatozoa, it is of great significance to study the physiological function of glycosylation on spermatozoa, which could reveal the targets for diagnosis and treatment of male infertility. As a post-genome technique platform, lectin microarray is capable to decode the “sugar code,” the glycoconjugate characteristics of cells [139, 155], and its research on spermatozoa can provide new ideas for diagnosis and management of male reproductive system diseases and infertility.

Advertisement

Acknowledgments

We would like to express our special thanks to our all family members and friends who supported us a lot during this harsh lockdown time. We are really thankful for them.

Advertisement

Conflict of interest

The authors declare no conflict of interest.

Advertisement

Author contributions

SJW, YDL, AJX, and YY wrote the manuscript; SCT conceived the part of lectin microarray; YHG and HJS are joint corresponding authors.

Advertisement

Abbreviations

BSG

Leukocyte differentiation antigen basigin also known as CD147

CRISP1

Cysteine-rich secretory protein 1

CTBS

Lysosomal di-N-acetylchitobiase

CTx

Cholera toxin

CTxB

Cholera toxin subunit B

DEFB126

β-defensin 126

ER

Endoplasmic reticulum

GalNac

N-acetylgalactosamine

GalNAc-T6

Polypeptide N-acetylgalactosaminyltransferase 6

GM2

Gangioside GM2

GPI

Glycosylphosphatidylinositol

GnT-II

N-acetylglucosaminyltransferase-II

LMAN1

Lectin mannose-binding 1, endoplasmic reticulum-Golgi intermediate compartment protein 53

MX

α-mannosidase IIx

NEU

Neuraminidases

P34H

Dicarbonyl/L-xylose Reductase

PI

Propidium Iodide

Sda

Sid blood group antigen

SPAM1/PH-20

Sperm adhesion molecule 1

References

  1. 1. Yanagimachi R. Mammalian fertilization. The Physiology of Reproduction. 1994;1:189-317. DOI: 1571135649640364928
  2. 2. Rowley MJ, Teshima F, Heller CG. Duration of transit of spermatozoa through the human male ductular system. Fertility and Sterility. 1970;21:390-396. DOI: S0015028216375021
  3. 3. Lakoski KA, Carron CP, Cabot CL, et al. Epididymal maturation and the acrosome reaction in mouse sperm: Response to zona pellucida develops coincident with modification of M42 antigen. Biology of Reproduction. 1988;38:221-233. DOI: 10.1095/biolreprod38.1.221
  4. 4. Yeung CH, Perez-Sanchez F, Soler C, et al. Maturation of human spermatozoa (from selected epididymides of prostatic carcinoma patients) with respect to their morphology and ability to undergo the acrosome reaction. Human Reproduction Update. 1997;3:205-213. DOI: 10.1093/humupd/3.3.205
  5. 5. Moore HDM, Hartman TD, Pryor JP. Development of the oocyte penetrating capacity of spermatozoa in the human epididymis. International Journal of Andrology. 1983;6:310-318. DOI: 10.1111/j.1365-2605.1983.tb00545.x
  6. 6. Harayama H, Kusunoki H, Kato S. Capacity of rete testicular and cauda epididymal boar spermatozoa to undergo the acrosome reaction and subsequent fusion with egg plasma membrane. Molecular Reproduction and Development. 1993;35:62-68. DOI: 10.1002/mrd.1080350111
  7. 7. Avella MA, Xiong B, Dean J. The molecular basis of gamete recognition in mice and humans. Molecular Human Reproduction. 2013;19:279-289. DOI: 10.1093/molehr/gat004
  8. 8. Haidl G, Badura B, Schill WB. Function of human epididymal spermatozoa. Journal of Andrology. 1994;15(Suppl):23S-27S. DOI: 10.1002/j.1939-4640.1994.tb01699.x
  9. 9. Cooper TG, Yeung CH. Acquisition of volume regulatory response of sperm upon maturation in the epididymis and the role of the cytoplasmic droplet. Microscopy Research and Technique. 2003;61:28-38. DOI: 10.1002/jemt.10314
  10. 10. Toshimori K. Biology of spermatozoa maturation: An overview with an introduction to this issue. Microscopy Research and Technique. 2003;61:1-6. DOI: 10.1002/jemt.10311
  11. 11. Cooper TG. The epididymis, cytoplasmic droplets and male fertility. Asian Journal of Andrology. 2011;13:130-138. DOI: 10.1038/aja.2010.97
  12. 12. Suarez SS. Mammalian sperm interactions with the female reproductive tract. Cell and Tissue Research. 2016;363(1):185-194. DOI: 10.1007/s00441-015-2244-2
  13. 13. Miller DJ. Review: The epic journey of sperm through the female reproductive tract. Animal. 2018;12(s1):s110-s120. DOI: 10.1017/S1751731118000526
  14. 14. Rickard JP, Pool KR, Druart X, et al. The fate of spermatozoa in the female reproductive tract: A comparative review. Theriogenology. 2019;137:104-112. DOI: 10.1016/j.theriogenology.2019.05.044
  15. 15. Tollner TL, Yudin AI, Treece CA, et al. Macaque sperm coating protein DEFB126 facilitates sperm penetration of cervical mucus. Human Reproduction. 2008;23(11):2523-2534. DOI: 10.1093/humrep/den276
  16. 16. Czuppon AB. Biochemical characterization of a human spermatozoal sialoglycoprotein with respect to antigenicity masking by its sialic acid moieties. Biochemistry International. 1984;8:9-18 PMID: 6206868
  17. 17. Tollner TL, Yudin AI, Tarantal AF, et al. Beta-defensin 126 on the surface of macaque sperm mediates attachment of sperm to oviductal epithelia. Biology of Reproduction. 2008;78:400-412. DOI: 10.1095/biolreprod.107.064071
  18. 18. Leahy T, Gadella BM. New insights into the regulation of cholesterol efflux from the sperm membrane. Asian Journal of Andrology. 2015;17(4):561-567. DOI: 10.4103/1008-682X.153309
  19. 19. Iqbal N, Hunter AG. Comparison of various bovine sperm capacitation systems for their ability to alter the net negative surface charge of spermatozoa. Journal of Dairy Science. 1995;78:84-90. DOI: 10.3168/jds.S0022-0302(95)76619-X
  20. 20. Iqbal N, Hunter AG. Comparison of bovine sperm capacitation systems for ability of sperm to penetrate zona-free hamster oocytes and bovine oocytes matured in vitro. Journal of Dairy Science. 1995;78:77-83. DOI: 10.3168/jds.S0022-0302(95)76618-8
  21. 21. Schröter S, Osterhoff C, McArdle W, Ivell R. The glycocalyx of the sperm surface. Human Reproduction Update. 1999;5(4):302-313. DOI: 10.1093/humupd/5.4.302
  22. 22. Diekman A. Glycoconjugates in sperm function and gamete interactions: How much sugar does it take to sweet-talk the egg? CMLS. Cellular and Molecular Life Sciences. 2003;60:298-308. DOI: 10.1007/s000180300025
  23. 23. Cooper TG. Interactions between epididymal secretions and spermatozoa. Journal of Reproduction and Fertility. Supplement. 1998;53:119-136 PMID: 10645272
  24. 24. Tecle E, Gagneux P. Sugar-coated sperm: Unraveling the functions of the mammalian sperm glycocalyx. Molecular Reproduction and Development. 2015;82:635-650. DOI: 10.1002/mrd.22500
  25. 25. Apweiler R, Hermjakob H, Sharon N. On the frequency of protein glycosylation, as deduced from analysis of the SWISS-PROT database. Biochimica et Biophysica Acta. 1999;1473:4-8. DOI: 10.1016/s0304-4165(99)00165-8
  26. 26. Colley KJ, Varki A, Kinoshita T. Cellular organization of glycosylation. In: Ajit V, Richard DC, editors. Essentials of Glycobiology. 3rd ed. Cold Spring Harbor: Cold Springs Harbor Laboratory Press; 2015. pp. 41-49. DOI: 10.1101/glycobiology.3e.004
  27. 27. Spiro RG. Protein glycosylation: Nature, distribution, enzymatic formation, and disease implications of glycopeptide bonds. Glycobiology. 2002;12(4):43R-56R. DOI: 10.1093/glycob/12.4.43r
  28. 28. Yan A, Lennarz WJ. Unraveling the mechanism of protein N-glycosylation. The Journal of Biological Chemistry. 2005;280(5):3121-3124. DOI: 10.1074/jbc.R400036200
  29. 29. Aebi M. N-linked protein glycosylation in the ER. Biochimica et Biophysica Acta. 2013;1833:2430-2437. DOI: 10.1016/j.bbamcr.2013.04.001
  30. 30. Varki A, Lowe J. Biological roles of glycans. In: Ajit V, Richard DC, editors. Essentials of Glycobiology. 2nd ed. Vol. 2009. New York: Cold Spring Harbor Laboratory Press; 2009. pp. 75-88 PMID: 20301233
  31. 31. Dennis JW, Nabi IR, Demetriou M. Metabolism, cell surface organization, and disease. Cell. 2009;139:1229-1241. DOI: 10.1016/j.cell.2009.12.008
  32. 32. Pang PC, Tissot B, Drobnis EZ, et al. Expression of bisecting type and Lewis-x/Lewis-y terminated N-glycans on human sperm. The Journal of Biological Chemistry. 2007;282:36593-36602. DOI: 10.1074/jbc.M705134200
  33. 33. Huang HH, Stanley P. A testis-specific regulator of complex and hybrid N-glycan synthesis. The Journal of Cell Biology. 2010;190:893-910. DOI: 10.1083/jcb.201004102
  34. 34. Yudin AI, Treece CA, Tollner TL, et al. The carbohydrate structure of DEFB126, the major component of the cynomolgus macaque sperm plasma membrane glycocalyx. The Journal of Membrane Biology. 2005;207:119-129. DOI: 10.1007/s00232-005-0806-z
  35. 35. Tollner TL, Venners SA, Hollox EJ, et al. A common mutation in the defensin DEFB126 causes impaired sperm function and subfertility. Science Translational Medicine. 2011;3:92ra65. DOI: 10.1126/scitranslmed.3002289
  36. 36. Nosjean O, Briolay A, Roux B. Mammalian GPI proteins: Sorting, membrane residence and functions. Biochimica et Biophysica Acta. 1997;1331:153-186. DOI: 10.1016/s0304-4157(97)00005-1
  37. 37. Griffiths GS, Galileo DS, Reese K, Martin-Deleon PA. Investigating the role of murine epididymosomes and uterosomes in GPI-linked protein transfer to sperm using SPAM1 as a model. Molecular Reproduction and Development. 2008;75(11):1627-1636. DOI: 10.1002/mrd.20907
  38. 38. Maccioni HJ. Glycosylation of glycolipids in the Golgi complex. Journal of Neurochemistry. 2007;103(Suppl 1):81-90. DOI: 10.1111/j.1471-4159.2007.04717.x
  39. 39. Yu RK, Tsai YT, Ariga T, Yanagisawa M. Structures, biosynthesis, and functions of gangliosides—An overview. Journal of Oleo Science. 2011;60(10):537-544. DOI: 10.5650/jos.60.537
  40. 40. Iwamori M. A new turning point in glycosphingolipid research. Human Cell. 2005;18(3):117-133. DOI: 10.1111/j.1749-0774.2005.tb00002.x
  41. 41. Iwamori M, Adachi S, Lin B, et al. Spermatogenesis-associated changes of fucosylated glycolipids in murine testis. Human Cell. 2020;33(1):23-28. DOI: 10.1007/s13577-019-00304-x
  42. 42. Honke K, Hirahara Y, Dupree J, Suzuki K, Popko B, Fukushima K, et al. Paranodal junction formation and spermatogenesis require sulfoglycolipids. Proceedings of the National Academy of Sciences of the United States of America. 2002;99(7):4227-4232. DOI: 10.1073/pnas.032068299
  43. 43. Takamiya K, Yamamoto A, Furukawa K, et al. Complex gangliosides are essential in spermatogenesis of mice: Possible roles in the transport of testosterone. Proceedings of the National Academy of Sciences of the United States of America. 1998;95(21):12147-12152. DOI: 10.1073/pnas.95.21.12147
  44. 44. Mruk DD, Cheng CY. Sertoli-Sertoli and Sertoli-germ cell interactions and their significance in germ cell movement in the seminiferous epithelium during spermatogenesis. Endocrine Reviews. 2004;25:747-806. DOI: 10.1210/er.2003-0022
  45. 45. Wang Y, Tan J, Sutton-Smith M, et al. Modeling human congenital disorder of glycosylation type IIa in the mouse: Conservation of asparagine-linked glycan-dependent functions in mammalian physiology and insights into disease pathogenesis. Glycobiology. 2001;11:1051-1070. DOI: 10.1093/glycob/11.12.1051
  46. 46. Akama TO, Nakagawa H, Sugihara K, et al. Germ cell survival through carbohydrate-mediated interaction with Sertoli cells. Science. 2002;295:124-127. DOI: 10.1126/science.1065570
  47. 47. Igakura T, Kadomatsu K, Kaname T, et al. A null mutation in basigin, an immunoglobulin superfamily member, indicates its important roles in peri-implantation development and spermatogenesis. Developmental Biology. 1998;194:152-165. DOI: 10.1006/dbio.1997.8819
  48. 48. Chen L, Bi J, Nakai M, et al. Expression of basigin in reproductive tissues of estrogen receptor-α or -β null mice. Reproduction. 2010;139:1057-1066. DOI: 10.1530/REP-10-0069
  49. 49. Bi J, Li Y, Sun F, et al. Basigin null mutant male mice are sterile and exhibit impaired interactions between germ cells and Sertoli cells. Developmental Biology. 2013;380:145-156. DOI: 10.1016/j.ydbio.2013.05.023
  50. 50. Tang W, Chang SB, Hemler ME. Links between CD147 function, glycosylation, and caveolin-1. Molecular Biology of the Cell. 2004;15:4043-4050. DOI: 10.1091/mbc.e04-05-0402
  51. 51. Loong JH, Wong TL, Tong M, Sharma R, Zhou L, Ng KY, et al. Glucose deprivation-induced aberrant FUT1-mediated fucosylation drives cancer stemness in hepatocellular carcinoma. The Journal of Clinical Investigation. 2021;131(11):e143377. DOI: 10.1172/JCI143377
  52. 52. Vos JP, Lopescardozo M, Gadella BM. Metabolic and functional-aspects of sulfogalactolipids. Biochimica et Biophysica Acta (BBA)—Lipids and Lipid Metabolism. 1994;1211:125-149. DOI: 10.1016/0005-2760(94)90262-3
  53. 53. Tanphaichitr N, Bou Khalil M, Weerachatyanukul W, et al, Physiological and biophysical properties of male germ cell sulfogalactosylglycerolipid, in: De Vriese S (Ed.), Lipid Metabolism and Male Fertility, AOCS Press, Champaign, IL, 2003, pp. 125-148. DOI: 10.1201/9781439822234.ch11
  54. 54. Honke K. Biosynthesis and biological function of sulfoglycolipids. Proceedings of the Japan Academy. Series B, Physical and Biological Sciences. 2013;89(4):129-138. DOI: 10.2183/pjab.89.129
  55. 55. Tanphaichitr N et al. Properties, metabolism and roles of sulfogalactosylglycerolipid in male reproduction. Progress in Lipid Research. 2018;72:18-41. DOI: 10.1016/j.plipres.2018.08.002
  56. 56. Holt WV. Surface-bound sialic acid on ram and bull spermatozoa: Deposition during epididymal transit and stability during washing. Biology of Reproduction. 1980;23:847-857. DOI: 10.1095/biolreprod23.4.847
  57. 57. Yanagimachi R, Noda Y, Fujimoto M, et al. The distribution of negative surface charges on mammalian spermatozoa. The American Journal of Anatomy. 1972;135:497-519. DOI: 10.1002/aja.1001350405
  58. 58. Pang PC, Chiu PCN, Lee CL, et al. Human sperm binding is mediated by the sialyl-Lewisx oligosaccharide on the zona pellucida. Science. 2011;333:1761-1764. DOI: 10.1126/science.1207438
  59. 59. Mark AB. Proteomics of post-translational modifications of mammalian spermatozoa. Cell and Tissue Research. 2016;363(1):279-287. DOI: 10.1007/s00441-015-2249-x
  60. 60. Kirchhoff C, Schröter S. New insights into the origin, structure and role of CD52: A major component of the mammalian sperm glycocalyx. Cells, Tissues, Organs. 2001;168:93-104. DOI: 10.1159/000016810
  61. 61. Koyama K, Ito K, Hasegawa A. Role of male reproductive tract CD52 (mrt-CD52) in reproduction. Society of Reproduction and Fertility Supplement. 2007;63:103-110 PMID: 17566265
  62. 62. Diekman AB, Norton EJ, Klotz KL, et al. N-linked glycan of a sperm CD52 glycoform associated with human infertility. The FASEB Journal. 1999;13:1303-1313. DOI: 10.1096/fasebj.13.11.1303
  63. 63. Sullivan R, Frenette G, Girouard J. Epididymosomes are involved in the acquisition of new sperm proteins during epididymal transit. Asian Journal of Andrology. 2007;9(4):483-491. DOI: 10.1111/j.1745-7262.2007.00281.x
  64. 64. Rejraji H, Sion B, Prensier G, et al. Lipid remodeling of murine epididymosomes and spermatozoa during epididymal maturation. Biology of Reproduction. 2006;74(6):1104-1113. DOI: 10.1095/biolreprod.105.049304
  65. 65. Tollner TL, Bevins CL, Cherr GN. Multifunctional glycoprotein DEFB126—A curious story of defensin-clad spermatozoa. Nature Reviews. Urology. 2012;9(7):365-375. DOI: 10.1038/nrurol.2012.109
  66. 66. Tulsiani D, Skudlarek MD, Holland MK, et al. Glycosylation of rat sperm plasma membrane during epididymal maturation. Biology of Reproduction. 1993;48:417-428. DOI: 10.1095/biolreprod48.2.417
  67. 67. Kolle S. Transport, distribution and elimination of mammalian sperm following natural mating and insemination. Reproduction in Domestic Animals. 2015;50:2-6. DOI: 10.1111/rda.12576
  68. 68. Cummerson JA, Flanagan BF, Spiller DG, et al. The complement regulatory proteins CD55 (decay accelerating factor) and CD59 are expressed on the inner acrosomal membrane of human spermatozoa as well as CD46 (membrane cofactor protein). Immunology. 2006;118:333-342. DOI: 10.1111/j.1365-2567.2006.02374.x
  69. 69. Rooney IA, Atkinson JP, Krul ES, et al. Physiologic relevance of the membrane attack complex inhibitory protein CD59 in human seminal plasma: CD59 is present on extracellular organelles (prostasomes), binds cell membranes, and inhibits complement-mediated lysis. The Journal of Experimental Medicine. 1993;177:1409-1420. DOI: 10.1084/jem.177.5.1409
  70. 70. Kirchhoff C, Hale G. Cell-to-cell transfer of glycosylphosphatidylinositol-anchored membrane proteins during sperm maturation. Molecular Human Reproduction. 1996;2:177-184. DOI: 10.1093/molehr/2.3.177
  71. 71. El Ouagari K, Teissié J, Benoist H. Glycophorin a protects K562 cells from natural killer cell attack. Role of oligosaccharides. Journal of Biological Chemistry. 1995;270:26970-26975. DOI: 10.1074/jbc.270.45.26970
  72. 72. Bergman M, Del Prete G, van Kooyk Y, et al. Helicobacter pylori phase variation, immune modulation and gastric autoimmunity. Nature Reviews. Microbiology. 2006;4:151-159. DOI: 10.1038/nrmicro1344
  73. 73. Dell A, Morris HR, Easton RL, et al. Structural analysis of the oligosaccharides derived from glycodelin, a human glycoprotein with potent immunosuppressive and contraceptive activities. The Journal of Biological Chemistry. 1995;270:24116-24126. DOI: 10.1074/jbc.270.41.24116
  74. 74. Koistinen H, Easton RL, Chiu PC, et al. Differences in glycosylation and sperm-egg binding inhibition of pregnancy-related glycodelin. Biology of Reproduction. 2003;69:1545-1551. DOI: 10.1095/biolreprod.103.017830
  75. 75. Chiu PC, Chung MK, Koistinen R, et al. Cumulus oophorus-associated glycodelin-C displaces sperm-bound glycodelin-A and -F and stimulates spermatozoa-zona pellucida binding. The Journal of Biological Chemistry. 2007;282:5378-5388. DOI: 10.1074/jbc.M607482200
  76. 76. Tse JY, Chiu PC, Lee KF, et al. The synthesis and fate of glycodelin in human ovary during folliculogenesis. Molecular Human Reproduction. 2002;8:142-148. DOI: 10.1093/molehr/8.2.142
  77. 77. Morris HR, Dell A, Easton RL, et al. Gender-specific glycosylation of human glycodelin affects its contraceptive activity. The Journal of Biological Chemistry. 1996;271:32159-32167. DOI: 10.1074/jbc.271.50.32159
  78. 78. Chiu PC, Chung MK, Koistinen R, et al. Glycodelin-A interacts with fucosyltransferase on human sperm plasma membrane to inhibit spermatozoa-zona pellucida binding. Journal of Cell Science. 2007;120(Pt 1):33-44. DOI: 10.1242/jcs.03258
  79. 79. Uchida H, Maruyama T, Nishikawa-Uchida S, et al. Glycodelin in reproduction. Reproductive Medicine & Biology. 2013;12(3):79-84. DOI: 10.1007/s12522-013-0144-2
  80. 80. Dell A, Morris HR, Easton RL, et al. Secretory endometrium synthesizes placental protein 14. Endocrinology. 1986;118:1782-1786. DOI: 10.1210/endo-118-5-1782
  81. 81. Ma F, Wu D, Deng L, et al. Sialidases on mammalian sperm mediate deciduous sialylation during capacitation. The Journal of Biological Chemistry. 2012;287:38073-38079. DOI: 10.1074/jbc.M112.380584
  82. 82. Watanabe H, Takeda R, Hirota K, et al. Lipid raft dynamics linked to sperm competency for fertilization in mice. Genes to Cells. 2017;22(5):493-500. DOI: 10.1111/gtc.12491
  83. 83. Griffiths GS, Miller KA, Galileo DS, et al. Murine SPAM1 is secreted by the estrous uterus and oviduct in a form that can bind to sperm during capacitation: Acquisition enhances hyaluronic acid-binding ability and cumulus dispersal efficiency. Reproduction. 2008;135:293-301. DOI: 10.1530/REP-07-0340
  84. 84. Boué F, Bérubé B, De Lamirande E, et al. Human sperm-zona pellucida interaction is inhibited by an antiserum against a hamster sperm protein. Biology of Reproduction. 1994;51:577-587. DOI: 10.1095/biolreprod51.4.577
  85. 85. Da Ros VG, Weigel Muñoz M, Battistone MA, et al. From the epididymis to the egg: Participation of CRISP proteins in mammalian fertilization. Asian Journal of Andrology. 2015;17:711-715. DOI: 10.4103/1008-682X.155769
  86. 86. Laine RA. A calculation of all possible oligosaccharide isomers both branched and linear yields 1.05 x 10(12) structures for a reducing hexasaccharide: The isomer barrier to development of single-method saccharide sequencing or synthesis systems. Glycobiology. 1994;4:759-767. DOI: 10.1093/glycob/4.6.759
  87. 87. Broussard AC, Boyce M, Bement W. Life is sweet: The cell biology of glycoconjugates. Molecular Biology of the Cell. 2019;30(5):525-529. DOI: 10.1091/mbc.E18-04-0247
  88. 88. Almeida A, Kolarich D. The promise of protein glycosylation for personalised medicine. Biochimica et Biophysica Acta. 2016;1860(8):1583-1895. DOI: 10.1016/j.bbagen.2016.03.012
  89. 89. Boyer SW, Johnsen C, Morava E. Nutrition interventions in congenital disorders of glycosylation. Trends in Molecular Medicine. 2022;S1471-4914(22):00081-00088. DOI: 10.1016/j.molmed.2022.04.003
  90. 90. Tredan O, Galmarini CM, Patel K, et al. Drug resistance and the solid tumor microenvironment. Journal of the National Cancer Institute. 2007;99:1441-1454. DOI: 10.1093/jnci/djm135
  91. 91. Knezević A, Polasek O, Gornik O, et al. Variability, heritability and environmental determinants of human plasma N-glycome. Journal of Proteome Research. 2009;8(2):694-701. DOI: 10.1021/pr800737u
  92. 92. Lauc G. Precision medicine that transcends genomics: Glycans as integrators of genes and environment. Biochimica et Biophysica Acta. 2016;1860(8):1571-1573. DOI: 10.1016/j.bbagen.2016.05.001
  93. 93. Klisch K, Contreras DA, Sun X, et al. The Sda/GM2-glycan is a carbohydrate marker of porcine primordial germ cells and of a subpopulation of spermatogonia in cattle, pigs, horses and llama. Reproduction. 2011;142(5):667-674. DOI: 10.1530/REP-11-0007
  94. 94. Sharon N, Lis H. History of lectins: From hemagglutinins to biological recognition molecules. Glycobiology. 2004;14(11):53R-62R. DOI: 10.1093/glycob/cwh122
  95. 95. Sharon N, Lis H. Lectins: Cell-agglutinating and sugar-specific proteins. Science. 1972;177(4053):949-959. DOI: 10.1126/science.177.4053.949
  96. 96. Gabius HJ, Kayser K. Introduction to glycopathology: The concept, the tools and the perspectives. Diagnostic Pathology. 2014;20(9):4. DOI: 10.1186/1746-1596-9-4
  97. 97. Wileman TE, Lennartz MR, Stahl PD. Identification of the macrophage mannose receptor as a 175-kDa membrane protein. Proceedings of the National Academy of Sciences of the United States of America. 1986;83:2501-2505. DOI: 10.1073/pnas.83.8.2501
  98. 98. Lambeau G, Ancian P, Barhanin J, et al. Cloning and expression of a membrane receptor for secretory phospholipases A2. The Journal of Biological Chemistry. 1994;269:1575-1578 PMID: 8294398
  99. 99. Jiang W, Swiggard WJ, Heufler C, et al. The receptor DEC-205 expressed by dendritic cells and thymic epithelial cells is involved in antigen processing. Nature. 1995;375:151-155. DOI: 10.1038/375151a0
  100. 100. East L, Rushton S, Taylor ME, et al. Characterization of sugar binding by the mannose receptor family member, Endo180. The Journal of Biological Chemistry. 2002;277:50469-50475. DOI: 10.1074/jbc.M208985200
  101. 101. Bennett EP, Mandel U, Clausen H, et al. Control of mucin-type O-glycosylation: A classification of the polypeptide GalNAc-transferase gene family. Glycobiology. 2012;22(6):736-756. DOI: 10.1093/glycob/cwr182
  102. 102. Kuribara T, Usui R, Totani K. Glycan structure-based perspectives on the entry and release of glycoproteins in the calnexin/calreticulin cycle. Carbohydrate Research. 2021;502:108273. DOI: 10.1016/j.carres.2021.108273
  103. 103. Caramelo JJ, Parodi AJ. A sweet code for glycoprotein folding. FEBS Letters. 2015;589(22):3379-3387. DOI: 10.1016/j.febslet.2015.07.021
  104. 104. Satoh T, Suzuki K, Yamaguchi T, et al. Structural basis for disparate sugar-binding specificities in the homologous cargo receptors ERGIC-53 and VIP36. PLoS One. 2014;9:e87963. DOI: 10.1371/journal.pone.0087963
  105. 105. Agrawal A, Singh PP, Bottazzi B, et al. Pattern recognition by pentraxins. Advances in Experimental Medicine and Biology. 2009;653:98-116. DOI: 10.1007/978-1-4419-0901-5_7
  106. 106. Grady RM, Grange RW, Lau KS, et al. Role for alpha-dystrobrevin in the pathogenesis of dystrophin-dependent muscular dystrophies. Nature Cell Biology. 1999;1(4):215-220. DOI: 10.1038/12034
  107. 107. Kaplan A, Achord DT, Sly WS. Phosphohexosyl components of a lysosomal enzyme are recognized by pinocytosis receptors on human fibroblasts. Proceedings of the National Academy of Sciences. 1977;74:2026-2030. DOI: 10.1073/pnas.74.5.2026
  108. 108. Hasilik A, Klein U, Waheed A, et al. Phosphorylated oligosaccharides in lysosomal enzymes: Identification of α-N-acetylglucosamine(1) phospho(6)mannose diester groups. Proceedings of the National Academy of Sciences. 1980;77:7074-7078. DOI: 10.1073/pnas.77.12.7074
  109. 109. Varki A, Schnaar RL, Crocker PR. I-type lectins. In: Varki A, Cummings RD, Esko JD, Stanley P, Hart GW, Aebi M, et al., editors. Essentials of Glycobiology. 3rd ed. Cold Spring Harbor: Cold Springs Harbor Laboratory Press; 2015. pp. 453-467. DOI: 10.1101/glycobiology.3e.035
  110. 110. Macauley MS, Crocker PR, Paulson JC. Siglec-mediated regulation o immune cell function in disease. Nature Reviews. Immunology. 2014;14:10. DOI: 10.1038/nri3737
  111. 111. Duan S, Paulson JC. Siglecs as immune cell checkpoints in disease. Annual Review of Immunology. 2020;38:365-395. DOI: 10.1146/annurev-immunol-102419-035900
  112. 112. Büll C, Heise T, Adema GJ, et al. Sialic acid mimetics to target the sialic acid-Siglec axis. Trends in Biochemical Sciences. 2016;41:6. DOI: 10.1016/j.tibs.2016.03.007
  113. 113. Shiratori I, Ogasawara K, Saito T, et al. Activation of natural killer cells and dendritic cells upon recognition of a novel CD99-like ligand by paired immunoglobulin-like type 2 receptor. The Journal of Experimental Medicine. 2004;199:525-533. DOI: 10.1084/jem.20031885
  114. 114. Mousseau DD, Banville D, L'Abbe D, et al. PILRα, a novel immunoreceptor tyrosine-based inhibitory motif-bearing protein, recruits SHP-1 upon tyrosine phosphorylation and is paired with the truncated counterpart PILRβ. The Journal of Biological Chemistry. 2000;275:4467-4474. DOI: 10.1074/jbc.275.6.4467
  115. 115. Kirschbaum NE, Gumina RJ, Newman PJ. Organization of the gene for human platelet/endothelial cell adhesion molecule-1 shows alternatively spliced isoforms and a functionally complex cytoplasmic domain. Blood. 1994;84:4028-4037 PMID: 7994021
  116. 116. Kitazume S, Imamaki R, Ogawa K, et al. α2,6-sialic acid on platelet endothelial cell adhesion molecule (PECAM) regulates its homophilic interactions and downstream antiapoptotic signaling. The Journal of Biological Chemistry. 2010;285:6515-6521. DOI: 10.1074/jbc.M109.073106
  117. 117. Heller M, von der Ohe M, Kleene R, Mohajeri MH, Schachner M. The immunoglobulin-superfamily molecule basigin is a binding protein for oligomannosidic carbohydrates: An anti-idiotypic approach. Journal of Neurochemistry. 2003;84(3):557-565. DOI: 10.1046/j.1471-4159.2003.01537.x
  118. 118. Taylor KR, Gallo RL. Glycosaminoglycans and their proteoglycans: Host-associated molecular patterns for initiation and modulation of inflammation. The FASEB Journal. 2006;20(1):9-22. DOI: 10.1096/fj.05-4682rev
  119. 119. Hoffmeister KM, Falet H. Platelet clearance by the hepatic Ashwell-Morrell receptor: Mechanisms and biological significance. Thrombosis Research. 2016;141(Suppl 2):S68-S72. DOI: 10.1016/S0049-3848(16)30370-X
  120. 120. Li R, Hoffmeister KM, Falet H. Glycans and the platelet life cycle. Platelets. 2016;27(6):505-511. DOI: 10.3109/09537104.2016.1171304
  121. 121. Murugaiah V, Tsolaki AG, Kishore U. Collectins: Innate immune pattern recognition molecules. Advances in Experimental Medicine and Biology. 2020;1204:75-127. DOI: 10.1007/978-981-15-1580-4_4
  122. 122. Del Fresno C, Cueto FJ, Sancho D. A proposal for nomenclature in myeloid C-type lectin receptors. Frontiers in Immunology. 2019;10:2098. DOI: 10.3389/fimmu.2019.02098
  123. 123. Tang C, Makusheva Y, Sun H, Han W, Iwakura Y. Myeloid C-type lectin receptors in skin/mucoepithelial diseases and tumors. Journal of Leukocyte Biology. 2019;106(4):903-917. DOI: 10.1002/JLB.2RI0119-031R
  124. 124. Cappenberg A, Kardell M, Zarbock A. Selectin-mediated signaling—Shedding light on the regulation of integrin activity in neutrophils. Cell. 2022;11:1310. DOI: 10.3390/cells11081310
  125. 125. Kansas GS. Selectins and their ligands: Current concepts and controversies. Blood. 1996;88:3259-3287 PMID: 8896391
  126. 126. Kahn J, Walcheck B, Migaki GI, et al. Calmodulin regulates L-selectin adhesion molecule expression and function through a protease-dependent mechanism. Cell. 1998;92:809-818. DOI: 10.1016/s0092-8674(00)81408-7
  127. 127. Liu Z, Miner JJ, Yago T, et al. Differential regulation of human and murine P-selectin expression and function in vivo. The Journal of Experimental Medicine. 2010;207:2975-2987. DOI: 10.1084/jem.20101545
  128. 128. McEver RP, Moore KL, Cummings RD. Leukocyte trafficking mediated by selectin-carbohydrate interactions. The Journal of Biological Chemistry. 1995;270:11025-11028. DOI: 10.1074/jbc.270.19.11025
  129. 129. Toscano M, Allo VCM, Cutine AM, et al. Untangling galectin-driven regulatory circuits in autoimmune inflammation. Trends in Molecular Medicine. 2018;24:348-363. DOI: 10.1016/j.molmed.2018.02.008
  130. 130. Di Lella S, Sundblad V, Cerliani JP, et al. When galectins recognize glycans: From biochemistry to physiology and back again. Biochemistry. 2011;50:7842-7857. DOI: 10.1021/bi201121m
  131. 131. Ricard-Blum S, Perez S. Glycosaminoglycan interaction networks and databases. Current Opinion in Structural Biology. 2022;74:102355. DOI: 10.1016/j.sbi.2022.102355
  132. 132. Benoff S. Carbohydrates and fertilization: An overview. Molecular Human Reproduction. 1997;3(7):599-637. DOI: 10.1093/molehr/3.7.599
  133. 133. Clark GF. A role for carbohydrate recognition in mammalian sperm-egg binding. Biochemical and Biophysical Research Communications. 2014;450(3):1195-1203. DOI: 10.1016/j.bbrc.2014.06.051
  134. 134. Kerr CL, Hanna WF, Shaper JH, et al. Lewis X-containing glycans are specific and potent competitive inhibitors of the binding of ZP3 to complementary sites on capacitated, acrosome-intact mouse sperm. Biology of Reproduction. 2004;71(3):770-777. DOI: 10.1095/biolreprod.103.023812
  135. 135. Danielewicz N, Rosato F, Dai W, et al. Microbial carbohydrate-binding toxins—From etiology to biotechnological application. Biotechnology Advances. 2022;59:107951. DOI: 10.1016/j.biotechadv.2022.107951
  136. 136. Comer FI, Vosseller K, Wells L, et al. Characterization of a mouse monoclonal specific antibody for O-linked N-acetylglucosamine. Analytical Biochemistry. 2001;293(2):169-177. DOI: 10.1006/abio.2001.5132
  137. 137. O’Neal CJ, Amaya EI, Jobling MG, et al. Crystal structures of an intrinsically active cholera toxin mutant yield insight into the toxin activation mechanism. Biochemistry. 2004;43:3772-3782. DOI: 10.1021/bi0360152
  138. 138. Selvaraj V, Buttke DE, Asano A, et al. GM1 dynamics as a marker for membrane changes associated with the process of capacitation in murine and bovine spermatozoa. Journal of Andrology. 2007;28(4):588-599. DOI: 10.2164/jandrol.106.002279
  139. 139. Hirabayashi J. Lectin-based glycomics: How and when was the technology born? Methods in Molecular Biology. 2014;1200:225-242. DOI: 10.1007/978-1-4939-1292-6_20
  140. 140. Dang K, Zhang W, Jiang S, et al. Application of lectin microarrays for biomarker discovery. Chemistry Open. 2020;9(3):285-300. DOI: 10.1002/open.201900326
  141. 141. Xin AJ, Cheng L, Diao H, et al. Comprehensive profiling of accessible surface glycans of mammalian sperm using a lectin microarray. Clinical Proteomics. 2014;11(1):10. DOI: 10.1186/1559-0275-11-10
  142. 142. Tao SC, Li Y, Zhou J, et al. Lectin microarrays identify cell-specific and functionally significant cell surface glycan markers. Glycobiology. 2008;18(10):761-769. DOI: 10.1093/glycob/cwn063
  143. 143. Xin A, Cheng L, Diao H, et al. Lectin binding of human sperm associates with DEFB126 mutation and serves as a potential biomarker for subfertility. Scientific Reports. 2016;6:20249. DOI: 10.1038/srep20249
  144. 144. Wu YC, Xin AJ, Lu H, et al. Effects of cryopreservation on human sperm glycocalyx. Journal of Reproduction and Contraception. 2017;37(4):6. DOI: https://medcentral.net/doi/full/10.4103/2096-2924.224914
  145. 145. Xin AJ, Wu YC, Lu H, et al. Comparative analysis of human sperm glycocalyx from different freezability ejaculates by lectin microarray and identification of ABA as sperm freezability biomarker. Clinical Proteomics. 2018;15(1):19. DOI: 10.1186/s12014-018-9195-z
  146. 146. Sun Y, Cheng L, Gu Y, et al. A human lectin microarray for sperm surface glycosylation analysis. Molecular and Cellular Proteomics. 2016;15(9):2839-2851. DOI: 10.1074/mcp.M116.059311
  147. 147. Cui W. Mother or nothing: The agony of infertility. Bulletin of the World Health Organization. 2010;88:881-882. DOI: 10.2471/BLT.10.011210
  148. 148. Carrell DT, Aston KI, Oliva R, Emery BR, De Jonge CJ. The "omics" of human male infertility: Integrating big data in a systems biology approach. Cell and Tissue Research. 2016;363(1):295-312. DOI: 10.1007/s00441-015-2320-7
  149. 149. Krzastek SC, Smith RP, Kovac JR. Future diagnostics in male infertility: Genomics, epigenetics, metabolomics and proteomics. Translational Andrology and Urology. 2020;9:S195-S205. DOI: 10.21037/tau.2019.10.20
  150. 150. Hotaling J, Carrell DT. Clinical genetic testing for male factor infertility: Current applications and future directions. Andrology. 2014;2:339-350. DOI: 10.1111/j.2047-2927.2014.00200.x
  151. 151. Agarwal A, Mulgund A, Hamada A, Chyatte MR. A unique view on male infertility around the globe. Reproductive Biology and Endocrinology. 2015;26(13):37. DOI: 10.1186/s12958-015-0032-1
  152. 152. Agarwal A, Parekh N, Panner Selvam MK, et al. Male oxidative stress infertility (MOSI): Proposed terminology and clinical practice guidelines for management of idiopathic male infertility. The World Journal of Men's Health. 2019;37(3):296-312. DOI: 10.5534/wjmh.190055
  153. 153. Minas A, Fernandes ACC, Maciel Júnior VL, Adami L, Intasqui P, Bertolla RP. Influence of physical activity on male fertility. Andrologia. 2022;54(7):e14433. DOI: 10.1111/and.14433
  154. 154. Esteves SC. Evolution of the World Health Organization semen analysis manual: Where are we? Nature Reviews. Urology. 2022;19(7):439-446. DOI: 10.1038/s41585-022-00593-2
  155. 155. André S, Kaltner H, Manning JC, Murphy PV, et al. Lectins: Getting familiar with translators of the sugar code. Molecules. 2015;20(2):1788-1823. DOI: 10.3390/molecules20021788

Written By

Shuangjie Wang, Yadong Li, Aijie Xin, Yang Yang, sheng-ce Tao, Yihua Gu and Huijuan Shi

Submitted: 17 May 2022 Reviewed: 10 July 2022 Published: 24 August 2022