ABSTRACT

STUDY QUESTION

Are there differences in the proteomic profile of exosomes isolated from seminal plasma of normozoospermic (NSP) and severe asthenozoospermic (SA) men, potentially contributing to sperm features?

SUMMARY ANSWER

A relevant group of proteins known to positively regulate sperm functions were over-represented in seminal exosomes of NSP men, i.e. cysteine-rich secretory protein-1 (CRISP1), while the inhibitory protein glycodelin was enriched in exosomes of SA subjects.

WHAT IS KNOWN ALREADY

Exosomes are secreted along the male reproductive tract and are thought to be involved in spermatozoa maturation and function. Ejaculated spermatozoa are still able to capture exosomes; exosomes of NSP individuals improve sperm motility and prompt capacitation, while exosomes of SA men fail to exert similar features.

STUDY DESIGN, SIZE, DURATION

Semen samples from NSP and SA men, aged 18 to 55 and registered at a single IVF center, were considered for this study project. Subjects were subdivided into three groups: a discovery cohort (five NSP men and six SA patients), a validation cohort (seven NSP and seven SA men) and the ‘glycodelin analysis’ cohort (20 NSP and 37 SA men). Exosomes were purified from semen of every participant.

PARTICIPANTS/MATERIALS, SETTING, METHODS

Exosomes were characterized by nanoparticle tracking analysis, transmission electron microscopy and western blot. Comprehensive proteomics analysis of the exosomal proteome was performed by nanoscale liquid chromatographic tandem mass spectrometry analysis. Funrich software was used to determine statistical enrichment of pathways, networks and Gene Ontology terms of the identified proteins. Validation of differentially expressed proteins was performed through ELISA and western blot analysis.

MAIN RESULTS AND THE ROLE OF CHANCE

The comprehensive proteomic analysis identified a total of 2138 proteins for both groups. There were 89 proteins found to be differentially expressed in exosomes of NSP versus SA subjects, of which 37 were increased in the NSP group and 52 were increased in the SA group. One-third of the exosomes-associated proteins highly expressed in NSP samples were involved in the reproductive process; conversely, the over-expressed proteins in exosomes of SA samples were not functionally specific. Quantitative data were confirmed on seminal exosomes from different cohorts of subjects.

LARGE SCALE DATA

N/A

LIMITATIONS, REASONS FOR CAUTION

Transfer of the proteins from exosomes to spermatozoa has been only partially demonstrated and up-take mechanisms are still poorly defined.

WIDER IMPLICATIONS OF THE FINDINGS

Seminal exosomes carry proteins that are potentially able to either favour or inhibit the reproductive process in humans. A better understanding of these phenomena might pave the way for novel intervention measures in terms of male infertility.

STUDY FUNDING/COMPETING INTEREST(S)

This study was funded by the Italian Ministry of Health through an Institution Seed Grant. None of the authors has any competing interests.

Introduction

The molecular mechanisms involved in sperm maturation are still largely unknown; over the last decades, extracellular vesicles (EVs) secreted along the male reproductive tract have been demonstrated to be involved in the process (Machtinger et al., 2016; Simon et al., 2018; da Silveira et al., 2018). EVs are multifaceted carriers containing a plethora of molecules including proteins, nucleic acids and lipids (Lotvall et al., 2014). The presence of EVs in the seminal fluid was originally described by Ronquist in the 1970s (Ronquist et al., 1977; Ronquist et al., 1978) and such vesicles were called prostasomes, because of their principal identified origin. However, the prostate, epididymis and other compartments of the male reproductive tract have been shown to release EVs that eventually can be retrieved from seminal fluid (Sullivan et al., 2007; Aalberts et al., 2013a; Belleannee et al., 2013). Significant discrepancies can be found in the literature concerning the biological functions of vesicles in human seminal plasma (Machtinger et al., 2016; Simon et al., 2018). Pons-Rejraji et al. (2011), for instance, it was reported that prostasomes participate in avoiding premature sperm capacitation and acrosome reaction. In contrast, other in vitro evidence has shown that prostasomes may contribute to both capacitation and acrosome reaction (Arienti et al., 1999; Bechoua et al., 2011; Park et al., 2011). Our group has recently demonstrated that seminal EVs, mostly characterized as exosomes, could prompt sperm capacitation through an increased induction of tyrosine phosphorylation, therefore inducing the acrosome reaction (Murdica et al., 2019).

Given these potential effects on sperm function, several studies have been carried out to elucidate the content of human seminal EVs. In this context, Utleg et al. (2003) identified 139 proteins obtained from healthy men by using microcapillary high-performance liquid chromatography tandem mass spectrometry (LC-MS). Subsequently, Poliakov et al. (2009) were able to investigate the structure of prostasomes by cryo-electron microscopy and their proteomic analysis led to the identification of 440 proteins. Thereafter, Ronquist et al. (2013) performed proteomic studies comparing prostasomes from four difference species, demonstrating that they are able to produce extracellular ATP and thus to activate G protein-coupled receptors (Neary et al., 1991) or ion channels (Bean, 1992) involved in the process of sperm acrosome exocytosis (Florman et al., 1998) and in the interaction of prostasomes with spermatozoa (Ronquist et al., 1990; Ronquist et al., 2013). Likewise, Dubois et al. (2015) reported a comprehensive liquid chromatography-tandem MS profiling of proteins in lipid rafts derived from seminal exosomes (SEs), identifying more than 370 lipid raft-associated proteins. More recently, Yang et al. (2017) performed a comprehensive proteomics analysis of exosomes derived from the seminal plasma of a pool of six healthy donors, identifying as many as 1474 proteins. Surprisingly, all these previous studies have been performed on seminal EV samples without any evaluation of the sperm parameters but assuming a normal condition. Only 54 proteins were found in common among the major above-mentioned studies (Poliakov et al., 2009; Ronquist et al., 2013; Dubois et al., 2015; Yang et al., 2017). To the best of the current knowledge, only García-Rodríguez et al. (2018) have compared protein expression patterns between prostasomal pools obtained from normozoospermic and dyspermic men; in this context, they observed that 7 and 36 proteins were either significantly over- or under-expressed in prostasomes from dyspermic men, respectively, as compared with those of normozoospermic individuals.

Despite the unquestionable scientific support of all these latter studies, little is known about the difference in terms of protein content of SEs from normozoospermic versus severely asthenozoospermic men. Therefore, our study was aimed to: (i) determine potential discrepancies of the proteomic content of SEs between normozoospermic and severe asthenozoospermic white-Caucasian men and (ii) define the role of SEs in different reproductive processes in human beings.

Materials and Methods

Patients enrolment and ethics statement

Men, aged 18–45 years, undergoing semen analysis at a single IVF center (for any reason) were considered for the specific purposes of the study. Men with a severe asthenozoospermia (SA) or a normal semen analysis, i.e. normozoospermia (NSP), were enrolled. Patients with known varicocele, genitourinary inflammation, seminal tract infections and smokers were excluded. Fresh semen samples were evaluated according to the 2010 guidelines of the World Health Organization (WHO, 2010; Cooper et al., 2010). Subjects were subdivided into three groups: a discovery cohort (five NSP men and six SA patients), a validation cohort (seven NSP and seven SA men) and the ‘glycodelin analysis’ cohort (20 NSP and 37 SA men). All the experiments were performed in accordance to the principles set out in the World Medical Association Declaration of Helsinki. The study was approved by the Institutional ethical committee and subjects provided written informed consent (Prot. 2016 - Mummy Sperm).

Isolation of exosomes from human seminal plasma

Fresh samples were obtained from every participant after at least two days of sexual abstinence (Cooper et al., 2010), allowed to liquefy at 37°C for 30 min and then processed immediately according to the 2010 guidelines of the WHO (WHO, 2010; Cooper et al., 2010). Exosomes were isolated by employing a protocol based on sequential centrifugation and ultracentrifugation, in order to separate them from microvesicles (MVs) (Murdica et al., 2019). Briefly, to isolate exosomes, samples were centrifuged at 800×g for 15 min at 25°C to separate spermatozoa. Seminal plasma was then centrifuged at 10 000×g for 30 min at 4°C to remove cell debris and other impurities like macroparticles. Supernatants were in turn centrifuged at 20 000×g for 90 min to pellet MVs. Remaining supernatants were ultra-centrifuged at 200 000×g for 120 min and exosome-containing pellets were washed with PBS, ultra-centrifuged again, then re-suspended in PBS and utilized.

Nanoparticle tracking analysis

To evaluate the size distribution and concentration of vesicles, nanoparticle tracking analysis (NTA) was performed on isolated vesicles using a NanoSight LM10-HS microscope (NanoSight Ltd., Amesbury, UK), as previously described (Zarovni et al., 2015). Briefly, three 60 s videos were recorded for each sample, with camera level set at 16 and detection threshold set at 10. Videos were analysed with NTA software version 2.3 that allows defining both the concentration and size of measured particles. The NanoSight system was calibrated with polystyrene latex microbeads of 50, 100 and 200 nm (Thermo Scientific Waltham, MA, USA) prior to analysis and auto-settings were used to define the blur, minimum track length and minimum expected particle size. Samples were used at ~1000-fold starting dilution with concentrations adjusted, if necessary, in order to specifically fit the optimal working range of the instrument particle tracking analysis (i.e. 20 particles per frame).

Transmission electron microscopy

Exosomes, freshly purified from seminal plasma, were absorbed on glow discharged carbon-coated formvar copper grids, washed with water, contrasted with 2% uranyl acetate and air-dried. Grids were observed with a Zeiss LEO 512 transmission electron microscope. Images were acquired by a 2k × 2k bottom-mounted slow-scan Proscan camera controlled by EsivisionPro 3.2 software.

Western blot analysis

Samples of 10–30 μg of isolated exosomes were lysed in reducing Laemmli buffer and boiled for 5 min at 95°C. In particular, 10 μg of isolated exosomes were used for the detection of tetraspanins, 20 μg was used for ALIX, TSG101, prostate-specific antigen (PSA) and flotillin and 30 μg was used for TGM4, CRISP1 and glycodelin, respectively. For tetraspanins detection, non-reducing sample buffer was used. Proteins were resolved by sodium dodecyl sulphate–polyacrylamide gel electrophoresis on the basis of equivalent quantity of proteins per lane and electro-transferred onto a nitrocellulose membrane. Non-specific sites were blocked with 5% (w/v) skimmed milk in T-TBS (Tris-buffered saline: 150 mM NaCl, 20 mM TrisHCl, pH 7.4 and 0.5% Tween 20). Membranes were incubated overnight at 4°C with the following antibodies: anti-CD63 (1:20000; BD Pharmingen, #556019, San Jose, CA, USA), anti-CD81(1:5000; BD Pharmingen, #555675, San Jose), anti-ALIX (1:500, Santa Cruz, #sc-271975, Santa Cruz, CA, USA), anti-TSG101 (1:500, Novus Bio, #NB200-112, Littleton, CO, USA), anti-PSA (1:1000), anti-flotillin (1:1000, GeneTex, #104769), anti-TGM4 (1:1000, ThermoFisher, #PA5-72495, Rockford, USA), anti-CRISP1(1:1000, Abcam, #ab74785, Abcam Inc., Cambridge, UK) and anti-glycodelin (1:1000, Novus Bio, #NBP1-89782, Littleton, CO, USA).

After washing with T-TBS, membranes were incubated with goat anti-mouse (1:10000–1:50000) or anti-rabbit (1:2000) IgG conjugated to horseradish peroxidase for 45 min. Positive immunoreactive bands were detected by the enhanced chemiluminescence method (ImmobilonTM HRP substrate, #WBKLS0500, Millipore Corp., Billerica, MA, USA).

After antibody detection, amido black (Sigma, #A8181) staining was performed following manufacturer’s indications, to detect total proteins on blot transfer membranes. Proteins appear as dark blue bands on membranes, which, after digitalization with a Kodak Image station 440 documentation system (Kodak), were quantified by using ImageJ v1.48 software (http://imagej.nih.gov).

Proteomics analysis

Exosomes from male seminal fluid of patients were lysed with 8 M Urea 100 mM Tris–HCl pH 8 buffer, proteins were quantified by BCA assay and 30 μg samples were in-solution digested (Osti et al., 2019). Samples were injected in duplicate on an nLC–ESI–MS-MS quadrupole Orbitrap QExactive-HF mass spectrometer (Thermo Fisher Scientific). Peptides separation was achieved on a linear gradient from 100% solvent A (2% ACN, 0.1% formic acid) to 30% solvent B (80% acetonitrile, 0.1% formic acid) over 62 min and from 30 to 60% solvent B in 5 min at a constant flow rate of 0.25 μl/min on UHPLC Easy-nLC 1000 (Thermo Fisher Scientific) connected to a 25 cm fused-silica emitter of 75 μm inner diameter (New Objective, Inc.), packed in-house with ReproSil-Pur C18-AQ 1.9 μm beads (Dr Maisch Gmbh).

Table I

Descriptive statistics table of the whole cohort of participants.

Discovery cohortValidation cohortGlycodelin analysis cohort
CharacteristicNSPSAP-value
NSP vs SA
NSPSAP-value
NSP vs SA
FERTNSPSAP-value
FERT vs SA
P-value
NSP vs SA
Whole samples (n)5667122037
Age (years)36.4 ± 9.939.0 ± 9.60.6737.3 ± 9.941.1 ± 9.20.533.4 ± 3.836 ± 9.935.8 ± 90.40.9
pH7.6 ± 0.37.8 ± 0.40.327.5 ± 0.37.7 ± 0.30.37.7 ± 0.37.7 ± 0.37.8 ± 0.30.30.06
Abstinence (day)3.6 ± 0.53.0 ± 1.10.304.5 ± 1.44.0 ± 1.60.62.8 ± 0.93.9 ± 1.34.1 ± 1.70.050.5
Volume (ml)3.7 ± 1.33.3 ± 0.70.564.2 ± 0.83.0 ± 1.20.13.2 ± 1.64.2 ± 1.43.4 ± 1.90.80.1
Motility (%)80.2 ± 5.311.8 ± 4.31.91E-09*72.2 ± 8.315.9 ± 8.29.2E-08*44.0 ± 12.365.7 ± 9.517.3 ± 8.41.4E-18*1.09E-26*
Viability (%)73.8 ± 8.067.0 ± 8.00.1980.7 ± 8.966.6 ± 16.50.166.4 ± 13.281.8 ± 6.354.8 ± 17.21.36E-03*4.73E-09*
Discovery cohortValidation cohortGlycodelin analysis cohort
CharacteristicNSPSAP-value
NSP vs SA
NSPSAP-value
NSP vs SA
FERTNSPSAP-value
FERT vs SA
P-value
NSP vs SA
Whole samples (n)5667122037
Age (years)36.4 ± 9.939.0 ± 9.60.6737.3 ± 9.941.1 ± 9.20.533.4 ± 3.836 ± 9.935.8 ± 90.40.9
pH7.6 ± 0.37.8 ± 0.40.327.5 ± 0.37.7 ± 0.30.37.7 ± 0.37.7 ± 0.37.8 ± 0.30.30.06
Abstinence (day)3.6 ± 0.53.0 ± 1.10.304.5 ± 1.44.0 ± 1.60.62.8 ± 0.93.9 ± 1.34.1 ± 1.70.050.5
Volume (ml)3.7 ± 1.33.3 ± 0.70.564.2 ± 0.83.0 ± 1.20.13.2 ± 1.64.2 ± 1.43.4 ± 1.90.80.1
Motility (%)80.2 ± 5.311.8 ± 4.31.91E-09*72.2 ± 8.315.9 ± 8.29.2E-08*44.0 ± 12.365.7 ± 9.517.3 ± 8.41.4E-18*1.09E-26*
Viability (%)73.8 ± 8.067.0 ± 8.00.1980.7 ± 8.966.6 ± 16.50.166.4 ± 13.281.8 ± 6.354.8 ± 17.21.36E-03*4.73E-09*

Characteristics from patients and semen samples from which exosomes were isolated are reported: discovery cohort (five NSP men and six SA patients), validation cohort (six NSP and seven SA men) and the ‘glycodelin analysis’ cohort (20 NSP, 37 SA and 12 FERT). Keys: NSP = normozoospermic; SA= severe asthenozoospermic; FERT= fertile men. Data presented as mean ± standard deviation. Asterisks indicate statistically significant differences (P-value < 0.001).

Table I

Descriptive statistics table of the whole cohort of participants.

Discovery cohortValidation cohortGlycodelin analysis cohort
CharacteristicNSPSAP-value
NSP vs SA
NSPSAP-value
NSP vs SA
FERTNSPSAP-value
FERT vs SA
P-value
NSP vs SA
Whole samples (n)5667122037
Age (years)36.4 ± 9.939.0 ± 9.60.6737.3 ± 9.941.1 ± 9.20.533.4 ± 3.836 ± 9.935.8 ± 90.40.9
pH7.6 ± 0.37.8 ± 0.40.327.5 ± 0.37.7 ± 0.30.37.7 ± 0.37.7 ± 0.37.8 ± 0.30.30.06
Abstinence (day)3.6 ± 0.53.0 ± 1.10.304.5 ± 1.44.0 ± 1.60.62.8 ± 0.93.9 ± 1.34.1 ± 1.70.050.5
Volume (ml)3.7 ± 1.33.3 ± 0.70.564.2 ± 0.83.0 ± 1.20.13.2 ± 1.64.2 ± 1.43.4 ± 1.90.80.1
Motility (%)80.2 ± 5.311.8 ± 4.31.91E-09*72.2 ± 8.315.9 ± 8.29.2E-08*44.0 ± 12.365.7 ± 9.517.3 ± 8.41.4E-18*1.09E-26*
Viability (%)73.8 ± 8.067.0 ± 8.00.1980.7 ± 8.966.6 ± 16.50.166.4 ± 13.281.8 ± 6.354.8 ± 17.21.36E-03*4.73E-09*
Discovery cohortValidation cohortGlycodelin analysis cohort
CharacteristicNSPSAP-value
NSP vs SA
NSPSAP-value
NSP vs SA
FERTNSPSAP-value
FERT vs SA
P-value
NSP vs SA
Whole samples (n)5667122037
Age (years)36.4 ± 9.939.0 ± 9.60.6737.3 ± 9.941.1 ± 9.20.533.4 ± 3.836 ± 9.935.8 ± 90.40.9
pH7.6 ± 0.37.8 ± 0.40.327.5 ± 0.37.7 ± 0.30.37.7 ± 0.37.7 ± 0.37.8 ± 0.30.30.06
Abstinence (day)3.6 ± 0.53.0 ± 1.10.304.5 ± 1.44.0 ± 1.60.62.8 ± 0.93.9 ± 1.34.1 ± 1.70.050.5
Volume (ml)3.7 ± 1.33.3 ± 0.70.564.2 ± 0.83.0 ± 1.20.13.2 ± 1.64.2 ± 1.43.4 ± 1.90.80.1
Motility (%)80.2 ± 5.311.8 ± 4.31.91E-09*72.2 ± 8.315.9 ± 8.29.2E-08*44.0 ± 12.365.7 ± 9.517.3 ± 8.41.4E-18*1.09E-26*
Viability (%)73.8 ± 8.067.0 ± 8.00.1980.7 ± 8.966.6 ± 16.50.166.4 ± 13.281.8 ± 6.354.8 ± 17.21.36E-03*4.73E-09*

Characteristics from patients and semen samples from which exosomes were isolated are reported: discovery cohort (five NSP men and six SA patients), validation cohort (six NSP and seven SA men) and the ‘glycodelin analysis’ cohort (20 NSP, 37 SA and 12 FERT). Keys: NSP = normozoospermic; SA= severe asthenozoospermic; FERT= fertile men. Data presented as mean ± standard deviation. Asterisks indicate statistically significant differences (P-value < 0.001).

Figure 1

Characterisation of seminal fluid-derived exosomes from different patients. TEM images of exosomes isolated from NSP (A) and SA (B) men; scale bar: 150 nm. Representative NTA plots of exosomes from NSP (C) and SA (D) men. (E) Biochemical characterisation of seminal fluid-derived exosomes: western blot analysis shows the presence of different canonical exosomal markers (CD63, CD81 and ALIX) in NSP and SA populations.

Figure 2

Proteomic comparison of SEs anaysed in this and other studies. (A) Workflow for the processing of exosomes derived from seminal plasma of 11 donors for a comprehensive proteomic analysis. (B) Venn diagram depicting proteins detected in the present study and overlapping with ExoCarta and Vesiclepedia databases. (C) Venn diagram comparing our identified proteins with previously published data from other four studies.

Figure 3

The GO analysis of the 2138 proteins identified in SEs in the present study. (A) cellular component, (B) molecular function and (C) site of expression.

MS data were acquired using a data-dependent top 15 method for HCD fragmentation. Survey full scan MS spectra (300–1650 Th) were acquired in the Orbitrap with 60 000 resolution, AGC target 3e6, IT 20 ms. For HCD spectra, resolution was set to 15 000 at m/z 200, AGC target 1e5, IT 80 ms; NCE 28% and isolation width 1.2 m/z.

For quantitative proteomics, raw MS files were processed with MaxQuant (version 1.5.2.8) using label-free quantification, searching against the database Uniprot_cp_human_setting trypsin specificity, up to two missed cleavages, cysteine carbamidomethyl as fixed modification, methionine oxidation and protein N-terminal acetylation as variable modifications. The peptides and protein FDR were set to 0.01; the minimal length required for a peptide was six amino acids, and a minimum of two peptides and at least one unique peptide were required for high-confidence protein identification. The lists of identified proteins were filtered to eliminate reverse hits and known contaminants.

Label-free quantification (LFQ) intensity were imported into Perseus (version 1.5.0.31) applying imputation and Z-score transformation. Statistical analysis was performed using t-test, Benjamini–Hochberg correction, FDR 0.05 (Cox et al., 2014).

Gene Ontology, Funrich and String Network analysis

The Funrich software was used to verify statistical over-representation of pathways, networks and Gene Ontology (GO) terms, such as biological processes and cellular components of the proteins found. The Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway (http://www.genome.jp/kegg/pathway.html) was used to evaluate other categorical annotations. The threshold for significance was set at an adjusted P-value of 0.05 after correcting for multiple testing by Benjamini and Hochberg FDR correction to obtain enriched components. The PANTHER (version 9.0, http://www.pantherdb.org) and STRING (version 10, http://string-db.org) softwares were used to obtain functional annotation and characterisation of identified proteins.

ELISA

ELISA (BIOSERV DiagnCox c) was used for the quantitative determination of glycodelin, following manufacturer’s indications. SE samples (4 μg/50 μl) were loaded in duplicates and incubated for 90 min at room temperature, followed by the addition of 50 μl of the enzyme conjugate solution (monoclonal anti-glycodelin antibody conjugated to horseradish peroxidase). After 30 min incubation at room temperature, 100 μl of chromogenic substrate solution (solution of tetramethylbenzidine) were added for 15 min at room temperature. The reaction was stopped with 100 μl 0.25 M H2SO4 and absorbance at 450 nm was measured using a plate reader. Plates were washed five times with Milli-Q water after each step. A standard curve was established by a glycodelin standard ranging from 3 ng to 100 ng (provided by the kit).

Statistical analysis

For nLC–ESI–MS-MS data, statistical analysis was performed using Student’s t-test. Results were filtered using the Benjamini–Hochberg procedure for FDR correction (FDR <0.05) (Cox et al., 2014). Two-way ANOVA followed by Bonferroni multiple comparison post-tests were used to determine significance of western blot and ELISA data. Differences were considered statistically significant for P-values <0.05. All results are expressed as mean ± SEM.

Results

Isolation and characterisation of exosomes derived from the seminal plasma

Table I details the descriptive statistics of the whole cohort of participants. As expected, groups showed comparable features, only differing in terms of progressive sperm motility, which was significantly decreased in SA men. A comprehensive characterisation of purified exosomes was performed: NSP and SA samples analysed by transmission electron microscopy displayed the classica round-shape, electron-dense vesicles, of ~50–150 nm diameter (Fig. 1A–B). Nanoparticle tracking analysis confirmed that all vesicles fell into the dimensional range of exosomes with no significant differences between NSP and SA men. Moreover, the size distribution overlapped between the groups (Fig. 1C–D). Purified exosomes showed the typical protein markers CD63, CD81 and ALIX (Fig. 1E); these proteins were usually expressed in a comparable manner between groups. The total protein content was used as loading control (Supplementary Fig. S1).

Figure 4

Exosomes derived from the seminal fluid of NSP and SA men displayed significant differences in their proteomic profile. (A) Heat map of supervised hierarchical clustering of differentially expressed proteins detected in each individual sample. Blue, increased expression relative to SEs from SA men; red, increased expression relative to SEs from NSP men; white, no difference in expression. (B) Volcano plot showing significantly differentially expressed proteins. The data for all proteins are plotted as log2 fold change (FC) versus the −log10 of the adjusted P-value. Thresholds are shown as dashed lines. Proteins selected as significantly different are highlighted as blue dots if over-represented in exosomes from SA patients or red dots if over-represented in exosomes derived from NSP men. White dots: no difference in expression. Named proteins have been evaluated in the validation phase.

Comprehensive proteomics analysis of exosomal proteome

The flowchart in Fig. 2A details the analysis performed on semen samples. By employing nLC-MS/MS, the proteomic analysis identified a total of 2138 proteins for both groups. Of these, 1665 proteins overlapped between the ExoCarta and Vesiclepedia databases (Fig. 2B). Among the proteins shared between the two groups, several exosomal markers, including CD63, CD9, CD81, flotillin, ALIX and TSG101, were found to be quantitatively comparable in SEs of NPS and SA patients (Supplementary Table SI).

Figure 5

The GO analysis of the 89 differentially expressed proteins in SEs from SA and NSP men identified in the study. (A) Biological process and (C) protein class of the 37 overexpressed proteins in SEs from the NSP group. (B) Biological process and (D) protein class of the 52 overexpressed proteins in SEs from the SA group. STRING protein–protein interaction (with known and predicted functional partners) in SEs from NSP (E) and SA (F) groups are displayed.

Subcellular origins of the identified proteins were then evaluated by functional-enrichment analysis in order to determine cellular components (Fig. 3A), molecular function (Fig. 3B) and site of expression (Fig. 3C). Since exosomes derived from seminal plasma originate from several cell types throughout the male genital tract, we analysed the site of localisation of these proteins. Indeed, the top five sites of localisation were represented by semen, testis, prostate, epididymis and by spermatozoa themselves, though with a minor contribution for the latter (Fig. 3C).

Figure 6

Validation of the proteomic results by western blot analysis. The presence of TSG101, PSA, flotillin, TGM4, glycodelin and CRISP1 in SEs derived from seven NSP and seven SA men was analysed by western blot. Bands intensity of TGM4, CRISP1 and glycodelin were normalised to total protein content (revealed by amido black staining, see Supplementary Fig. S2) and quantified by using ImageJ software. Box and Whisker plot shows minimum, 25th percentile, 50th percentile (median), 75th percentile and maximum values of the quantified bands. Two-way ANOVA followed by Bonferroni post-tests were used to determine significance: **P < 0,01 and *P<0,05.

Proteomic profile of SEs derived from NSP and SA men

The comparative analysis of the SE proteins shared by NSP and SA patients revealed 89 proteins differentially expressed (Supplementary Table SII), of which 52 were increased and 37 were reduced in SA versus NSP samples, respectively (Fig. 4). In detail, every protein was evaluated in a technical replicate and a two-tailed, two-sample t-test was used to compare protein levels between SEs of the case (SA) and the control group (NSP). Results were filtered using the Benjamini–Hochberg procedure for FDR correction (FDR <0.05). The comparative visualisation of the levels of the differentially expressed proteins in each individual sample was displayed in heat maps (Fig. 4A). A different pattern of protein expression was evident between SEs of NSP and SA men (Fig. 4A). Figure 4B depicts the findings of the volcano plot analysis; the –Log10 of adjusted P-value was plotted against the differential protein expression (log2 of fold change) of the means between SEs of NSP and SA subjects. This plot showed significantly dysregulated proteins. Among all, cysteine-rich secretory protein-1 (CRISP1), sperm associated antigen 11B (SPAG11B) and defensin B126 (DEFB126), which are known to positively regulate sperm-specific functions, were among the proteins most strongly enriched in SE samples from NSP men; conversely, glycodelin (PAEP) and TGM4, were among the more represented proteins in SE from SA subjects. To gain further insight in the pathways that are represented by the 89 differentially expressed proteins, they were sorted into categories according to their ontology as determined with GO annotation terms, such as biological process (Fig. 5A–B) and protein class (Fig. 5C–D). The annotated biological processes in exosome-associated proteins in SA individuals were related to processes such as ‘response to stimulus’, ‘biological regulation’, ‘metabolic and developmental process’, ‘multicellular organismal process’ and ‘locomotion’ (Fig. 5B). On the other hand, one-third of exosomes-associated proteins in samples from NSP men were involved in reproduction, in particular in fertilisation and gamete generation processes (Fig. 5A).

The STRING database was used to identify protein–protein interactions (PPI) and to generate a network of interactions based on a variety of sources, thus including various interaction databases, text mining, genetic interactions and shared pathway interactions. The differentially expressed proteins were uploaded for the PPI network, and interaction with at least medium confidence (interaction score >0.4) were set by default. Among the proteins involved in reproduction (in particular in fertilisation and gamete generation processes) recovered in the 37 differentially expressed proteins of NSP group, CRISP1, SPAG11B and DEFB126 were found to be interacting (Fig. 5E). On the other hand, the 52 differentially expressed proteins in SEs from SA subjects were completely unassociated with any process promoting reproduction (Fig. 5F).

Figure 7

Comparison of glycodelin levels in different groups of seminal fluid-derived exosomes from NSP and SA men. (A) Results are expressed as ng of glycodelin per μg of exosomes (orange line) and as percentage of non-motile sperm (blue histograms). (B) Box and Whisker plots showing minimum, 25th percentile, 50th percentile (median), 75th percentile and maximum values of glycodelin. Two-way ANOVA followed by Bonferroni post-tests were used to determine significance: ****P<0,0001.

Validation of the nLC–ESI–MS-MS data using a different cohort of patients

To confirm the differential expression of proteins of clinical relevance in SEs from NSP and SA populations, we focused on a subset of exosomal markers and selected protein candidates and assayed their levels in a different cohort of men (see Table I, validation cohort). As expected, several key proteins associated with exosomes biogenesis, including TSG101 and flotillin, were detected in a comparable manner in both groups (Fig. 6A and C). PSA did not differ between groups, as shown in Fig. 6B. We further analysed and quantified TGM4 and glycodelin (Fig. 6D and F), confirming that both were significantly higher in SEs of SA compared to NSP men. In addition, we validated the expression of CRISP1 in SEs from the NSP cohort and significantly reduced expression in the SA cohort (Fig. 6E). The abundance of the proteins of interest was normalized to the total protein content (Supplementary Fig. S2).

By ELISA, glycodelin was confirmed to be more abundant in SEs derived from SA compared to NSP men (Fig. 7).

Discussion

In this study, we have performed a comprehensive proteomic analysis of purified exosomes isolated from the seminal plasma of NSP and SA men by nLC–ESI–MS-MS, detecting 2136 proteins which have been matched both with exosomal proteins already registered in the ExoCarta and Vesiclepedia databases and with those previously found in four proteomic studies on seminal fluid EVs (Poliakov et al., 2009; Ronquist et al., 2013; Dubois et al., 2015; Yang et al., 2017). An overlap of 1665 proteins among the total exosomal proteome and the abovementioned databases was found; likewise, the overlap with previously published data from the four studies indicated that 733 proteins had been already identified but only 46 were common to all the studies. Of importance, 1403 non-overlapping proteins were found in the present study (Poliakov et al., 2009; Ronquist et al., 2013; Dubois et al., 2015; Yang et al., 2017). The higher number of SE proteins detected in our analysis as compared with those previously identified may be explained according to: (i) the evolution of mass spectrometry sensitivity over time, (ii) the type and the number of samples included in the studies and (iii) the method used to isolate exosomes. Moreover, those differences may be due in part to intersample-interindividual variations that some studies have tried to overcome using pools of different samples.

As expected, exosomal proteins are largely represented in our proteomic data along with cytosolic and lysosomal proteins. Exosomal markers CD63, CD9, CD81, flotillin, Alix and TSG101 were quantitatively similar in SEs from NSP and SA men, consistently with previously published western blot results both from our and other groups (Caballero et al., 2013; Lötvall et al., 2014; Murdica et al., 2019). Similarly, the male reproductive tract was the main site of localisation. Several proteins shown to be associated with epididymosomes, such as epididymal sperm-binding protein-1 (ELSPBP1), macrophage migration inhibitory factor, A-disintegrin and metalloproteinase (ADAM), glioma pathogenesis-related protein-1 (GLIPR1) and glutathione peroxidase family members (Sullivan and Saez, 2013), were detected in our proteomic analysis, thus confirming that epididymosomes are part of the complex mixture of exosomes we have isolated. Other bioinformatic analyses indicated that the retrieved proteins were mainly involved in processes of transportation, calcium ion binding and energy production (Ronquist et al., 2013).

Proteins such as aldo-keto reductase family 1, member B1 (AKR1B1), which are supposed to maintain epididymal sperm cells in a quiescent status (Frenette et al., 2010), dicarbonyl/L-xylulose reductase (DCXR), found to be important for the binding between sperm and the zona pellucida, PSA, which cleaves biological substrates in seminal fluid (including semenogelin I and II) into small peptides, and IZUMO, whose absence has been reported to compromise the fusion with oocytes (Inoue et al., 2005), have been also identified in SEs (Berube and Sullivan, 1994; Frenette and Sullivan, 2001; Sullivan et al., 2007). Overall, the presence of these proteins in SEs suggests that these vesicles may be an important source of proteins involved in the process of sperm maturation and acquisition of fertilisation capacity in humans (Caballero et al., 2011). In this context, it has been recently demonstrated that spermatozoa can uptake SEs in vitro (Park et al., 2011; Aalberts et al., 2013a; Aalberts et al., 2013b; Murdica et al., 2019). Different factors have been shown to influence sperm uptake of exosomes and pH has been reported to be important, since its variations can drastically reduce the cell capturing (Park et al., 2011; Aalberts et al., 2013a; Aalberts et al., 2013b). Further, exosomes uptake may be favoured or even impeded by other conditions, including sperm activity and capacitation status (Aalberts et al., 2013a; Aalberts et al., 2013b).

When the protein content of prostasomes isolated from seminal plasma of NSP and non-NSP men was previously compared, five proteins were found to be differentially expressed between the two groups, but none was implicated in the reproductive process (Garcia-Rodriguez et al., 2018). In contrast, our comparative analysis of SE proteins shared by NSP and SA patients revealed 89 proteins to be differentially expressed. Overall, the proteins overexpressed in NSP patients were particularly interesting. Among the 37 augmented proteins, we found SPAG11B, involved in sperm maturation (Radhakrishnan et al., 2009) and DEFB126, the lack of which have been associated with an impaired sperm function (Duan et al., 2015; Xin et al., 2016). In line with previous data from our group by western blot analysis (Murdica et al., 2019), SEs derived from NSP men displayed higher levels of CRISP1 compared to those derived from SA subjects. We have also previously demonstrated that CRISP1 can be transferred to spermatozoa through SEs (Murdica et al., 2019). CRISP1 is a calcium ion channel enabling the entry of Ca2+ and triggering a cascade of protein phosphorylation involved in sperm motility (Maldera et al., 2013; Da Ros et al., 2015; Ernesto et al., 2015; Pereira et al., 2017; Weigel Munoz et al., 2018). Moreover, Crisp−/− sperm in mice exhibit defects in acrosome reaction, motility and tyrosine phosphorylation (Weigel Munoz et al., 2018).

Even more interesting, among the 52 differentially expressed proteins augmented in SEs of SA men, the epididymal secretory protein E3-alpha, which has been never reported in exosomes before and is poorly characterized, is a protein predominantly expressed in the corpus region of the epididymis and moderately in the vas deferens, likely involved in sperm maturation (Kirchhoff et al., 1994). Aquaporins 2 and 5, expressed in different sections of the epididymis and testis of some species (Yeste et al., 2017), were found to be upregulated respectively in SEs of SA and NSP individuals. Both TGM4 and glycodelin, increased in SEs of SA men, have been associated with primary infertility (Chiu et al., 2005; Yeung et al., 2006; Landegren et al., 2015). Semen glycodelin (glycodelin-S) inhibits albumin-induced cholesterol efflux from spermatozoa (Chiu et al., 2005), and thereby may prevent capacitation, which is triggered by a cholesterol decrement in the sperm plasma membrane. After removal of glycodelin-S, cholesterol efflux and capacitation can occur. We herein confirmed previous findings from Ronquist et al. (2013) showing the presence of glycodelin in seminal EVs; moreover, we have demonstrated for the first time that the content of glycodelin depends on the exosome origin, presenting the highest level in SEs of SA men. Transfer of glycodelin from SEs to sperm has not been demonstrated yet but we cannot exclude a role for the exosomal protein in regulating cholesterol quantity in the membrane, and consequently the ability to undergo capacitation.

As a whole, our findings strongly support the evidence that the network of SE-associated proteins may be connected to the molecular mechanisms involved in sperm maturation and motility. Therefore, current observations, along with the demonstration that exosomes can be taken up by spermatozoa (Park et al., 2011; Murdica et al., 2019), prompt for a critical role of SEs in affecting sperm parameters and function. Further investigations are needed to gain a deeper understanding of the possible mechanisms that are involved in these phenomena.

Acknowledgements

Part of this work was carried out in the Advanced Light and Electron Microscopy BioImaging Center (ALEMBIC) of San Raffaele Scientific Institute and Vita-Salute University and in the Proteomics Facility at Cogentech.

Authors’ roles

V.M., G.C. and N.Z. performed the experiments and acquired and analysed data; P.V. and R.V. supervised and coordinated the project; A.S., V.M., P.V. and R.V. discussed the results; V.M., P.V. and R.V. drafted the manuscript. All authors read and approved the manuscript.

Funding

This study has been supported by the Italian Ministry of Health (5x1000 project).

Conflict of interest

The authors have declared that no conflict of interest exists.

References

Aalberts
M
,
Stout
TA
,
Stoorvogel
W
.
Prostasomes: extracellular vesicles from the prostate
.
Reproduction
2013a
;
147
:
R1
R14
.

Aalberts
M
,
Sostaric
E
,
Wubbolts
R
,
Wauben
MW
,
Nolte-’t Hoen
EN
,
Gadella
BM
,
Stout
TA
,
Stoorvogel
W
.
Spermatozoa recruit prostasomes in response to capacitation induction
.
Biochim Biophys Acta
2013b
;
1834
:
2326
35
.

Arienti
G
,
Carlini
E
,
Nicolucci
A
,
Cosmi
EV
,
Santi
F
,
Palmerini
CA
.
The motility of human spermatozoa as influenced by prostasomes at various pH levels
.
Biol Cell
1999
;
91
:
51
54
.

Bean
BP
.
Pharmacology and electrophysiology of ATP-activated ion channels
.
Trends Pharmacol Sci
1992
;
13
:
87
90
.

Bechoua
S
,
Rieu
I
,
Sion
B
,
Grizard
G
.
Prostasomes as potential modulators of tyrosine phosphorylation in human spermatozoa
.
Syst Biol Reprod Med
2011
;
57
:
139
148
.

Belleannée
C
,
Calvo
É
,
Caballero
J
,
Sullivan
R
.
Epididymosomes convey different repertoires of microRNAs throughout the bovine epididymis
.
Biol Reprod
2013
;
89
:
30
.

Berube
B
,
Sullivan
R
.
Inhibition of in vivo fertilization by active immunization of male hamsters against a 26kDa sperm glycoprotein
.
Biol Reprod
1994
;
51
:
1255
1263
.

Caballero
JN
,
Frenette
G
,
Belleannée
C
,
Sullivan
R
.
CD9-positive microvesicles mediate the transfer of molecules to Bovine Spermatozoa during epididymal maturation
.
PLoS One
2013
;
8
:
e65364
.

Caballero
J
,
Frenette
G
,
Sullivan
R
.
Post testicular sperm maturational changes in the bull: important role of the epididymosomes and prostasomes
.
Vet Med Int
2011
;
757194
:
1
13
.

Chiu
PC
,
Chung
MK
,
Tsang
HY
,
Koistinen
R
,
Koistinen
H
,
Seppala
M
,
Lee
KF
,
Yeung
WS
.
Glycodelin-S in human seminal plasma reduces cholesterol efflux and inhibits capacitation of spermatozoa
.
J Biol Chem
2005
;
280
:
25580
25589
.

Cooper
TG
,
Noonan
E
,
von Eckardstein
S
,
Auger
J
,
Baker
HW
,
Behre
HM
.
World Health Organization reference values for human semen characteristics
.
Hum Reprod Update
2010
;
16
:
231
245
.

Cox
J
,
Hein
MY
,
Luber
CA
,
Paron
I
,
Nagaraj
N
,
Mann
M
.
Accurate proteome-wide label-free quantification by delayed normalization and maximal peptide ratio extraction, termed MaxLFQ
.
Mol Cell Proteomics
2014
;
13
:
2513
2526
.

Da Ros
VG
,
Muñoz
MW
,
Battistone
MA
,
Brukman
NG
,
Carvajal
G
,
Curci
L
,
Gómez-ElIas
MD
,
Cohen
DB
,
Cuasnicu
PS
.
From the epididymis to the egg: participation of CRISP proteins in mammalian fertilization
.
Asian J Androl
2015
;
17
:
711
715
.

Da Silveira
JC
,
de Avila
ACGFCM
,
Garrett
HL
,
Bruemmer
JE
,
Winger
QA
,
Bouma
GJ
.
Cell-secreted vesicles containing microRNAs as regulators of gamete maturation
.
J Endocrinol
2018
;
236
:
R15
R27
.

Duan
S
,
Shi
C
,
Chen
G
,
Zheng
JF
,
Wu
B
,
Diao
H
,
Ji
L
,
Gu
Y
,
Xin
A
,
Wu
Y
et al. 
Another functional frame-shift polymorphism of DEFB126 (rs11467497) associated with male infertility
.
J Cell Mol Med
2015
;
19
:
1077
1084
.

Dubois
L
,
Ronquist
KK
,
Ek
B
,
Ronquist
G
,
Larsson
A
.
Proteomic profiling of detergent resistant membranes (lipid rafts) of prostasomes
.
Mol Cell Proteomics
2015
;
14
:
3015
3022
.

Ernesto
JI
,
Weigel Muñoz
M
,
Battistone
MA
,
Vasen
G
,
Martínez-López
P
,
Orta
G
,
Figueiras-Fierro
D
,
De la Vega-Beltran
JL
, Moreno IA, Guidobaldi HA et al. .
CRISP1 as a novel CatSper regulator that modulates sperm motility and orientation during fertilization
.
J Cell Biol
2015
;
210
:
1213
1212
.

Florman
HM
,
Arnoult
C
,
Kazam
IG
,
Li
C
,
O’Toole
CM
.
A perspective on the control of mammalian fertilization by egg-activated ion channels in sperm: a tale of two channels
.
Biol Reprod
1998
;
59
:
12
16
.

Frenette
G
,
Sullivan
R
.
Prostasomes-like particles are involved in the transfer of P25b from the bovine epididymal fluid to the sperm surface
.
Mol Reprod Dev
2001
;
59
:
115
121
.

Frenette
G
,
Girouard
J
,
D’Amours
O
,
Allard
N
,
Tessier
L
,
Sullivan
R
.
Characterization of Two Distinct Populations of Epididymosomes Collected in the Intraluminal Compartment of the Bovine Cauda Epididymis
.
Biol Reprod
2010
;
3
:
473
480
.

Garcia-Rodroguez
A
,
de la Casa
M
,
Peinado
H
,
Gosalvez
J
,
Roy
R
.
Human prostasomes from normozoospermic and non-normozoospermic men show a differential protein expression pattern
.
Andrology
2018
;
6
:
585
596
.

Inoue
N
,
Ikawa
M
,
Isotani
A
,
Okabe
M
.
The immunoglobulin superfamily protein Izumo is required for sperm to fuse with eggs
.
Nature
2005
;
434
:
234
238
.

Kirchhoff
C
,
Pera
I
,
Rust
W
,
Ivell
R
.
Major human epididymis-specific gene product, HE3, is the first representative of a novel gene family
.
Mol Reprod Dev
1994
;
37
:
130
137
.

Landegren
N
,
Sharon
D
,
Shum
AK
,
Khan
IS
,
Fasano
KJ
,
Hallgren
Å
,
Kampf
C
,
Freyhult
E
,
Ardesjö-Lundgren
B
,
Alimohammadi
M
et al. 
Transglutaminase 4 as a prostate autoantigen in male subfertility
.
Sci Transl Med
2015
;
7
:
292ra101
.

Lötvall
J
,
Hill
AF
,
Hochberg
F
,
Buzás
EI
,
Di Vizio
D
,
Gardiner
C
.
Minimal experimental requirements for definition of extracellular vesicles and their functions: a position statement from the International Society for Extracellular Vesicles
.
J Extracell Vesicles
2014
;
3
:
26913
.

Machtinger
R
,
Laurent
LC
,
Baccarelli
AA
.
Extracellular vesicles: roles in gamete maturation, fertilization and embryo implantation
.
Hum Reprod Update
2016
;
22
:
182
193
.

Maldera
JA
,
Weigel Munoz
M
,
Chirinos
M
,
Busso
D
,
Raffo
FGE
,
Battistone
MA
.
Human fertilization: epididymal Hcrisp1 mediates sperm-zona pellucida binding through its interaction with ZP3
.
Mol Hum Reprod
2013
;
20
:
341
349
.

Murdica
V
,
Giacomini
E
,
Alteri
A
,
Bartolacci
A
,
Cermisoni
G
,
Zarovni
N
,
Papaleo
E
,
Montorsi
F
,
Salonia
A
,
Viganò
P
,
Vago
R
.
Seminal plasma of men with severe asthenozoospermia contain exosomes affecting spermatozoa motility and capacitation
.
Fertil Steril
2019
;
5
:
897
908
.

Neary
JT
,
Laskey
R
,
van Breemen
C
,
Blicharska
J
,
Norenberg
LO
,
Norenberg
MD
.
ATP-evoked calcium signal stimulates protein phosphorylation/dephosphorylation in astrocytes
.
Brain Res
1991
;
566
:
89
94
.

Osti
D
,
Del Bene
M
,
Rappa
G
,
Santos
M
,
Matafora
V
,
Richichi
C
,
Faletti
S
,
Beznoussenko
GV
,
Mironov
A
,
Bachi
A
et al.
Clinical significance of extracellular vesicles in plasma from glioblastoma patients
.
Clin Cancer Res
2019
1
:
266
276
.

Park
KH
,
Kim
BJ
,
Kang
J
,
Nam
TS
,
Lim
JM
,
Kim
HT
,
Park
JK
,
Kim
YG
,
Chae
SW
,
Kim
UH
.
Ca2+ signaling tools acquired from prostasomes are required for progesterone-induced sperm motility
.
Sci Signal
2011
;
4
:
ra31
.

Pereira
R
,
R
,
Barros
A
,
Sousa
M
.
Major regulatory mechanisms involved in sperm motility
.
Asian J Androl
2017
;
19
:
5
14
.

Poliakov
A
,
Spilman
M
,
Dockland
T
,
Amling
CL
,
Mobley
JA
.
Structural heterogeneity and protein composition of exosome-like vesicles (prostasomes) in human semen
.
Prostate
2009
;
69
:
159
167
.

Pons-Rejraji
H
,
Artonne
C
,
Sion
B
,
Brugnon
F
,
Canis
M
,
Janny
L
,
Grizard
G
.
Prostasomes: inhibitors of capacitation and modulators of cellular signalling in human sperm
.
Int J Androl
2011
;
34
:
568
580
.

Radhakrishnan
Y
,
Hamil
KG
,
Tan
JA
,
Grossman
G
,
Petrusz
P
,
Hall
SH
,
French
FS
.
Novel partners of SPAG11B isoform D in the human male reproductive tract
.
Biol Reprod
2009
;
81
:
647
656
.

Ronquist
G
,
Brody
I
,
Gottfries
A
,
Stegmayr
B
.
An Mg2+ and Ca2+-stimulated adenosine triphosphatase in human prostatic fluid: part I
.
Andrologia
1978
;
10
:
261
272
.

Ronquist
KG
,
Ek
B
,
Morrell
J
,
Stavreus-Evers
A
,
Strom
HB
,
Humblot
P
,
Ronquist
G
,
Larsoon
A
.
Prostasomes from four different species are able to produce extracellular adenosine triphosphate (ATP)
.
Biochim Biophys Acta
2013
;
1830
:
4604
4610
.

Ronquist
G
,
Hedström
M
.
Restoration of detergent-inactivated adenosine triphosphatase activity of human prostatic fluid with concanavalin A
.
Biochim Biophys Acta
1977
;
483
:
483
486
.

Ronquist
G
,
Nilsson
BO
,
Hjertën
S
.
Interaction between prostasomes and spermatozoa from human semen
.
Arch Androl
1990
;
2
:
147
157
.

Simon
C
,
Greening
DW
,
Bolumar
D
,
Balaguer
N
,
Salamonsen
LA
,
Vilella
F
.
Extracellular vesicles in human reproduction in health and disease
.
Endocr Rev
2018
;
39
:
292
332
.

Sullivan
R
,
Frenette
G
,
Girouard
J
.
Epididymosomes are involved in the acquisition of new sperm proteins during epididymal transit
.
Asian J Androl
2007
;
9
:
483
491
.

Sullivan
R
,
Saez
F
.
Epididymosomes, prostasomes, and liposomes: their roles in mammalian male reproductive physiology
.
Reproduction
2013
;
146
:
R21
R35
.

Utleg
AG
,
Yi
EC
,
Xie
T
,
Shannon
P
,
White
JT
,
Goodlett
DR
,
Hood
L
,
Lin
B
.
Proteomic analysis of human prostasomes
.
Prostate
2003
;
56
:
150
161
.

Weigel Munoz
M
,
Battistone
MA
,
Carvajal
G
,
Maldera
JA
,
Curci
L
,
Torres
P
.
Influence af the genetic background on the reproductive phenotype of mice lacking Cystein-Rich Secretory Protein 1 (CRISP1)
.
Biol Reprod
2018
;
99
:
373
383
.

World Health Organisation
.
WHO Laboratory Manual for the Examination and Processing of Human Semen
, 5th edn.
Geneva, Switzerland
,
WHO
,
2010
.

Xin
A
,
Cheng
L
,
Diao
H
,
Wu
Y
,
Zhou
S
,
Shi
C
,
Sun
Y
,
Wang
P
,
Duan
S
,
Zheng
J
et al. 
Lectin binding of human sperm associates with DEFB126 mutation and serves as a potential biomarker for subfertility
.
Sci Rep
2016
;
6
:
20249
.

Yang
C
,
Guo
W
,
Zhang
W
,
Bian
J
,
Yang
J
,
Zhou
Q
,
Chen
M
,
Peng
W
,
Qi
T
,
Wang
C
et al. 
Comprehensive proteimics analysis of exosomes derived from human seminal plasma
.
Andrology
2017
;
5
:
1007
1015
.

Yeste
M
,
Morató
R
,
Rodríguez-Gil
JE
,
Bonet
S
,
Prieto-Martínez
N
.
Aquaporins in the male reproductive tract and sperm: functional implications and cryobiology
.
Reprod Domest Anim
2017
;
52
:
12
27
.

Yeung
WS
,
Lee
KF
,
Koistinen
R
,
Koistinen
H
,
Seppala
M
,
Ho
PC
,
Chiu
PC
.
Roles of glycodelin in modulating sperm function
.
Mol Cell Endocrinol
2006
;
250
:
149
156
.

Zarovni
N
,
Corrado
A
,
Guazzi
P
,
Zocco
D
,
Lari
E
,
Radano
G
et al. 
Integrated isolation and quantitative analysis of exosome shuttled proteins and nucleic acids using immunocapture approaches
.
Methods
2015
;
87
:
46
58
.

This article is published and distributed under the terms of the Oxford University Press, Standard Journals Publication Model (https://academic.oup.com/journals/pages/open_access/funder_policies/chorus/standard_publication_model)

Supplementary data