sperm proteomics: potential impact on male infertility treatment · ashok agarwala, ricardo pimenta...

12
REVIEW Sperm proteomics: potential impact on male infertility treatment Ashok Agarwal a , Ricardo Pimenta Bertolla b and Luna Samanta c a American Center for Reproductive Medicine, Department of Urology, Cleveland Clinic, Cleveland, OH, USA; b Department of Surgery, Division of Urology, Human Reproduction Section, Federal University of São Paulo, São Paulo, Brazil; c Department of Zoology, School of Life Sciences, Ravenshaw University, Cuttack, India ABSTRACT Spermatozoa are unique cells that have highly compact DNA, motility (and hypermotility) patterns, a specific morphology, localized mitochondria and an apical acrosome. They are the end product of a dynamic process termed spermatogenesis. Sperm are therefore produced with specific proteins in order to effect different traits, such as the presence of cysteine-rich protamines in DNA, which effectively compacts DNA. Moreover, specific proteins are transferred during epididymal maturation and after ejaculation in order to render sperm capable of undergoing post-ejaculatory alterations, generally termed capacitation, which confers capacity to fertilize a mature oocyte. In addition, sperm exhibit several post-translational modifications, which are fundamental to their function, such as SUMOylation and ubiquitination. Discussed in this review is the current knowledge of the sperm proteome in terms of its composition and the function that these proteins determine, as well as their post-translational modifications and how these alter sperm functional integrity. Studies are emphasized that focus on shotgun proteomics untargeted determination of the protein constituent of a cell in a given biological condition and technologies currently applied toward that end are reviewed. ARTICLE HISTORY Received 4 November 2015 Accepted 3 February 2016 KEYWORDS Sperm; proteome; male infertility; human; post- translational modifications; LC-MS/MS Introduction Infertility affects 15% of couples of reproductive age, and in 50% of these cases, there is a male component [1]. Various cellular mechanisms are related to? Cause? Male infertility, such as sperm functional alterations (i.e. DNA fragmentation, acrosome integrity, among others) related to failed fertilization [2], poor embryo development [3,4], and assisted reproduction outcome [3,5,6]. Recently, focus has been placed on investi- gating the molecular machineries that affect these mechan- isms, with a special focus on the protein composition of ejaculated sperm [79]. Spermatozoa are the product of spermatogenesis a com- plex process in which diploid spermatogonia undergo meiosis and extensive morphological remodeling within the seminifer- ous tubules in the testes [10]. In humans, the process of sperma- togenesis is responsible for generating between 20 and 240 million sperm per day and depends on tight regulation of cellular metabolism [10]. Moreover, a number of selective mechanisms within the testes and epididymides help ensure that high quality sperm are available for ejaculation [10]. Many studies have hinted at a specific proteome signature that allow for the differentiation of high quality from low quality sperm [9], as well as specific proteins that activate following ejaculation in order to orchestrate acquisition of specific motility patterns [11], exocytosis of the acrosomal content, [11,12] and fusion of the sperm membrane to the oolemma [13,14]. However, many proteins in sperm may be added after spermatogenesis via microvesicles within the epi- didymal lumen (epididymosomes) [1518] or even after ejacu- lation through prostatic microvesicles (prostasomes) [1921]. Moreover, different subsets of sperm seem to coexist within an ejaculate. Damours et al., for example, demonstrated that dead bovine sperm (but not live sperm) are tagged, via transfer by epididymosomes, with a protein containing multiple fibronectin II domains, which are known to bind to collagen Epididymal Sperm Binding Protein 1 (ELSPBP1) [15]. This protein has also been associated with increased sperm DNA fragmentation in humans [9]. Other bovine studies have localized Fas a membrane recep- tor of the tumor necrosis factor family that initiates apoptosis to at least three different sperm regions, indicating that these are indeed subsets with different activities [22]. This division of sperm into subsets poses important con- siderations in regards to study design because the whole proteome of an ejaculate may not reflect its true fertility potential. It has been demonstrated, for example, that sperm marked for specific processes, such as apoptosis, are indeed of lower reproductive potential than those unmarked [23]. Conversely, specific proteins, which may offer sperm resis- tance within the female reproductive tract may also mark a population of viable sperm [24]. The field of proteomics has greatly benefited from the development of mass spectrometry (MS)-based systems [25,26]. While initial studies relied on gel separation of pro- teins by their isoelectric point and their mass (two-dimen- sional (2D) gel electrophoresis) [27,28], the exponential rate at which mass spectrometric resolution and sensitivity, allied to the development of protein- and peptide-specific chroma- tographic separation techniques, have led to systems that can detect and quantify a greater number of proteins at a higher dynamic range [26,2931]. The latter is usually achieved via digestion of proteins into their constitutive peptides (this may CONTACT Ashok Agarwal [email protected] EXPERT REVIEW OF PROTEOMICS, 2016 VOL. 13, NO. 3, 285296 http://dx.doi.org/10.1586/14789450.2016.1151357 © 2016 Taylor & Francis Downloaded by [New York University] at 12:46 16 October 2017

Upload: buithien

Post on 09-Nov-2018

216 views

Category:

Documents


0 download

TRANSCRIPT

REVIEW

Sperm proteomics: potential impact on male infertility treatmentAshok Agarwala, Ricardo Pimenta Bertollab and Luna Samanta c

aAmerican Center for Reproductive Medicine, Department of Urology, Cleveland Clinic, Cleveland, OH, USA; bDepartment of Surgery, Division ofUrology, Human Reproduction Section, Federal University of São Paulo, São Paulo, Brazil; cDepartment of Zoology, School of Life Sciences,Ravenshaw University, Cuttack, India

ABSTRACTSpermatozoa are unique cells that have highly compact DNA, motility (and hypermotility) patterns, a specificmorphology, localized mitochondria and an apical acrosome. They are the end product of a dynamic processtermed spermatogenesis. Sperm are therefore produced with specific proteins in order to effect differenttraits, such as the presence of cysteine-rich protamines in DNA, which effectively compacts DNA. Moreover,specific proteins are transferred during epididymal maturation and after ejaculation in order to render spermcapable of undergoing post-ejaculatory alterations, generally termed capacitation, which confers capacity tofertilize a mature oocyte. In addition, sperm exhibit several post-translational modifications, which arefundamental to their function, such as SUMOylation and ubiquitination. Discussed in this review is the currentknowledge of the spermproteome in terms of its composition and the function that these proteins determine,as well as their post-translational modifications and how these alter sperm functional integrity. Studies areemphasized that focus on shotgun proteomics – untargeted determination of the protein constituent of a cellin a given biological condition – and technologies currently applied toward that end are reviewed.

ARTICLE HISTORYReceived 4 November 2015Accepted 3 February 2016

KEYWORDSSperm; proteome; maleinfertility; human; post-translational modifications;LC-MS/MS

Introduction

Infertility affects 15% of couples of reproductive age, and in50% of these cases, there is a male component [1]. Variouscellular mechanisms are related to? Cause? Male infertility,such as sperm functional alterations (i.e. DNA fragmentation,acrosome integrity, among others) related to failed fertilization[2], poor embryo development [3,4], and assisted reproductionoutcome [3,5,6]. Recently, focus has been placed on investi-gating the molecular machineries that affect these mechan-isms, with a special focus on the protein composition ofejaculated sperm [7–9].

Spermatozoa are the product of spermatogenesis – a com-plex process in which diploid spermatogonia undergo meiosisand extensive morphological remodeling within the seminifer-ous tubules in the testes [10]. In humans, the process of sperma-togenesis is responsible for generating between 20 and 240million sperm per day and depends on tight regulation of cellularmetabolism [10]. Moreover, a number of selective mechanismswithin the testes and epididymides help ensure that high qualitysperm are available for ejaculation [10].

Many studies have hinted at a specific proteome signaturethat allow for the differentiation of high quality from lowquality sperm [9], as well as specific proteins that activatefollowing ejaculation in order to orchestrate acquisition ofspecific motility patterns [11], exocytosis of the acrosomalcontent, [11,12] and fusion of the sperm membrane to theoolemma [13,14]. However, many proteins in sperm may beadded after spermatogenesis via microvesicles within the epi-didymal lumen (epididymosomes) [15–18] or even after ejacu-lation through prostatic microvesicles (prostasomes) [19–21].

Moreover, different subsets of sperm seem to coexist within anejaculate. D’amours et al., for example, demonstrated that deadbovine sperm (but not live sperm) are tagged, via transfer byepididymosomes, with a protein containing multiple fibronectinII domains, which are known to bind to collagen – EpididymalSperm Binding Protein 1 (ELSPBP1) [15]. This protein has also beenassociated with increased sperm DNA fragmentation in humans[9]. Other bovine studies have localized Fas – a membrane recep-tor of the tumor necrosis factor family that initiates apoptosis – toat least three different sperm regions, indicating that these areindeed subsets with different activities [22].

This division of sperm into subsets poses important con-siderations in regards to study design because the wholeproteome of an ejaculate may not reflect its true fertilitypotential. It has been demonstrated, for example, that spermmarked for specific processes, such as apoptosis, are indeed oflower reproductive potential than those unmarked [23].Conversely, specific proteins, which may offer sperm resis-tance within the female reproductive tract may also mark apopulation of viable sperm [24].

The field of proteomics has greatly benefited from thedevelopment of mass spectrometry (MS)-based systems[25,26]. While initial studies relied on gel separation of pro-teins by their isoelectric point and their mass (two-dimen-sional (2D) gel electrophoresis) [27,28], the exponential rateat which mass spectrometric resolution and sensitivity, alliedto the development of protein- and peptide-specific chroma-tographic separation techniques, have led to systems that candetect and quantify a greater number of proteins at a higherdynamic range [26,29–31]. The latter is usually achieved viadigestion of proteins into their constitutive peptides (this may

CONTACT Ashok Agarwal [email protected]

EXPERT REVIEW OF PROTEOMICS, 2016VOL. 13, NO. 3, 285–296http://dx.doi.org/10.1586/14789450.2016.1151357

© 2016 Taylor & Francis

Dow

nloa

ded

by [

New

Yor

k U

nive

rsity

] at

12:

46 1

6 O

ctob

er 2

017

be preceded by protein fractionation) and identification andquantification of these peptides by liquid chromatography–tandem mass spectrometry (LC–MS/MS) in an approach‘termed shotgun proteomics’ [25,26,29].

In the first part of this review, we will discuss the technol-ogies that are currently used for sperm proteomic studies aswell as the challenges in data analysis. In the second part, wewill extensively review the data that have been generatedregarding the human sperm proteome.

Review criteria

A literature search was performed using PubMed and GoogleScholar electronic databases, using the following keywords:‘human’, ‘spermatozoa’, ‘proteome’, ‘protein–protein interac-tion network’, ‘shotgun proteomics’, ‘proteomics’, ‘spermato-genesis’, ‘epididymal maturation’, ‘post-translationalmodifications’, ‘infertility’, and ‘sperm function’. Only articlespublished in peer-reviewed journals were selected. The searchtargeted studies that were published in English-based journalswithin the past 10 years.

Methods for determining the sperm proteome

Proteomics is the identification and quantification of all theproteins in a given fluid or cell under a specific biologicalcondition [32]. Initial untargeted studies used 2D-gel electro-phoresis (2D-GE) in which proteins are separated initially bythe isoelectric points – the pH at which their net charge is zero– followed by separation by mass in conventional laemmli gelsand staining. Each protein thus forms a spot on a gel, and spotintensities are compared across gels in order to observe differ-ential expression levels. Spots of interest may then be identi-fied by MS (in the same principle that will be discussedfurther), and the relative quantification information isextracted from the gel (spot intensities) (for a review, see [27]).

Due to the variable nature of gel polymerization, thesestudies usually have high intra- and inter-assay variation[33,34]. To decrease this variability, difference gel electrophor-esis (DIGE) can be used. With DIGE, proteins of one sample aretagged with a fluorescent stain whereas proteins of anothersample are tagged with a different fluorescent stain. Thesesamples are then pooled and run through all the stepstogether. Fluorescent gel analysis then quantifies the fluores-cent signals from each sample to provide a more accuratedifferential analysis [27,35].

More recently, MS-based proteomics pipelines have beendeveloped, increasing dynamic range, quantification, and sen-sitivity with direct integration to chromatographic separation[26,29,31,36]. In these studies, proteins are digested into theirconstituent peptides, which are extensively analyzed andquantified. Proteins are therefore identified and quantifiedbased on in silico construction of their identified peptides(i.e. a bottom-up approach). This is achieved using MS equip-ment in an approach termed ‘shotgun proteomics’ [26,29,37].

Mass spectrometers analyze the masses of ionized mole-cules in a gas phase. The spectrometers contain an ionizationsource and one or more mass analyzers [31]. Ionization isachieved through the use of ‘soft’ ionization – usually matrix-

assisted laser desorption/ionization [38,39] and electrosprayionization (ESI) [40]. Because ESI ionizes molecules that are ina liquid phase, they are utilized for direct integration withliquid chromatography equipment in a setup (LC–MS) thatallows quantitative information to be derived from the con-structed chromatograms [36].

Following ionization, the mass analyzers select or analyzethe mass of the ionized molecules, utilizing different princi-ples. In general, quadrupole (Q) and ion trap mass analyzersfilter and accumulate the ions of interest, whereas time-of-flight and orbitrap mass analyzers detect mass values withhigh resolution [31]. Current shotgun proteomic platformsusually couple an ultraperformance liquid chromatographer(UPLC) to a hybrid mass spectrometer (MS/MS) in a setuptermed LC–MS/MS [7,9,41–43].

In most MS-based shotgun proteomics studies, an initialscan is generated in the high-resolution mass analyzer, andthe most important parent scan ions are determined. In sub-sequent scans, each parent ion selected in the first massanalyzer is fragmented in a collision chamber, and the frag-ment ions are identified in the high-resolution mass analyzer[31]. This approach is considered a data-dependent approachbecause an initial scan is necessary.

More recently, data-independent acquisition techniques havebeen generated, such as MSe [42,44] and sequential windowacquisition of all theoretical fragment ion spectra-MS [45–47].In data-independent acquisition, there is no precursor ion selec-tion: all ions are analyzed in their unfragmented and fragmentedforms. These techniques improve quantitative analysis of theidentified proteins as well as reproducibility of the data analyses[48].

Another important point is that coverage of the proteomedepends on the resolution of the chromatographic separation,because coelution of peptides decreases the odds that LC–MS/MS will be able to identify them, and this effect is moreimportant than low peptide quantity [49]. This ultimatelymeans that decreasing the sample complexity will allow for agreater number of peptides (and thus of proteins) to bedetermined. Fractionation of proteins before the LC–MS/MSexperiment has increasingly been used toward that end.Fractionation may be performed using a number of methods:running 1D gels to preseparate proteins based on their massand then slicing the generated protein lanes into mass frac-tions; in-solution isoelectric focusing, and prefractionation uti-lizing affinity columns [50–52].

Finally, in order to decrease inter-assay variability, many stu-dies have utilized differential mass tags. These mass tags, such asiTRAQ, iCAT, tandem mass tags [53], and dimethyl labels [54]allow for the differential marking of samples. These samples arethen pooled prior to the LC–MS/MS experiment. Software pro-cessing allows for relative quantification between different sam-ples in a same run, thus increasing reproducibility.

In studies focusing on sperm proteins, some other impor-tant points should be considered. If sperm are collected byejaculation, it is important to remove any contaminatinground cells [9]. Moreover, some authors have suggested thatdifferent subpopulations of sperm coexist in the human eja-culate [22,55,56]. For example, if this is the case, researchersmay want to select only viable sperm using density gradient or

286 A. AGARWAL ET AL.

Dow

nloa

ded

by [

New

Yor

k U

nive

rsity

] at

12:

46 1

6 O

ctob

er 2

017

swim-up techniques. Sperm selection based on functionaltraits could also generate important information on spermphysiology (such as selecting sperm with fragmented DNA orsperm bound to the zona pellucida of mature oocytes). Thisinformation is probably diluted in whole ejaculates, whichenhances the importance of planning sperm prefractionationeven before protein extraction.

Because sperm interaction with the surrounding media isfundamental in sperm physiology, approaches targeting spe-cific sperm regions are also important. Isolating membraneproteins, for example, may be useful in determining sperm–egg interacting proteins [57] or proteins transferred duringepididymal maturation [15,58]. Similarly, isolating mitochon-drial proteins could generate information regarding oxidativephosphorylation energy-producing pathways in mature spermunder diverse conditions [59].

There are many methods for determining the constituentsperm proteome as well as differential proteomes under dif-ferent biological conditions. It is important, however, to con-sider isolating specific subsets of sperm, or specific regionswithin these subsets, in order to focus on the intended organ-isms. Furthermore, technological considerations should bemade such as whether a data-independent approach is feasi-ble and if a label-free approach will suffice or if poolingsamples with different labels for comparison within a samerun will generate better information. Finally, studies may focuson differential proteome networks, as these will overexpressspecific functions when acting conjointly. Or, a functionalcomponent may be added to these studies by verifying reg-ulatory post-translational modifications in specific proteins orin a similar shotgun approach.

Differences in form and function: differentialproteome signatures and post-translationalmodifications

Technological advances in MS-based proteomics and inpeptide separation technologies have greatly increased cov-erage of the proteome [29,31]. As a result, studies haveidentified and quantified thousands of peptides and

proteins within biological samples [60,61]. This has led tothe understanding that proteins interact with one anotherto enrich specific functions and thus to produce a givenphenotype [62].

Proteins thus form protein–protein interaction (PPI) net-works and participate in specific pathways to produce spe-cific functions, such as fertilization (Figure 1). These PPInetworks are scale-free in that few proteins may lead tomultiple interactions (hub proteins) while many proteinsmay lead to few interactions (Figure 2) [63]. This is impor-tant to understand because proteins of more centrality (orwith more interactions), when altered or absent, will affectthe function of many different proteins and pathways.These are usually lethal gene products. More often thannot, however, mild phenotypic differences will be observeddue to altered expression of proteins with few interac-tions [64].

Another important point is that proteins, even when pre-sent, may have altered function due to post-translationalmodifications (PTMs) [65,66]. There are many different typesof PTMs, such as SUMOylation, phosphorylation, glycosylation,and ubiquitination [67]. In many cases, the proteome mapitself may not be altered but differences in PTMs may causea deviation in function. Sperm capacitation, for example,depends on an orchestrated cascade of protein phosphoryla-tion which, when finely regulated, leads to alteration in moti-lity patterns and eventually in acrosome exocytosis [68]. Inaddition, ubiquitination of sperm mitochondria prevents thepaternal mitochondrial DNA from being transmitted to theoffspring [69]. Finally, SUMOylation of specific proteins insperm such as topoisomerase-IIα has been associated withDNA damage, thus demonstrating an effect of PTM onsperm function [55].

In order to convincingly establish protein function, as afunction of their interaction networks, there is a necessity todesign studies that provides as complete a picture of theproteome as possible. Moreover, enriching PTMs for theircharacterization in shotgun studies may lend a functionalcomponent to the understanding of the sperm proteome.Downstream data analysis therefore must be able to deal

Figure 1. Protein-protein interaction (PPI) network (left) and the ‘Fertilization’ Reactome Pathway [75] effected by this PPI network.

EXPERT REVIEW OF PROTEOMICS 287

Dow

nloa

ded

by [

New

Yor

k U

nive

rsity

] at

12:

46 1

6 O

ctob

er 2

017

with a large number of inter-correlated variables to filter outunimportant data.

Data analysis in proteomics

Shotgun proteomic studies can identify and quantify hun-dreds to thousands of proteins at a time [7,9,41–43]. Resultsmatrices are therefore quite complex, containing thousandsof variables – many of which are intercorrelated. In addition,the variables are scaled differently from those used in con-ventional sperm analysis studies such as sperm concentra-tion, motility, morphology, DNA integrity, and acrosomeintegrity as other information (age, hormonal profile, etc.).Therefore, the statistical analysis must consider the intercor-relation observed between the quantified proteins as wellas filter unimportant variables to remove them from con-structed models [41]. Furthermore, because proteins interactwith one another, the data analysis must also include func-tional enrichment analysis [70,71].

In regards to statistical analysis, a univariate approach isoften used. However, in these cases, the analysis mustinclude adjustments for multiple comparisons, such as per-mutation-based false discovery rate determination forp-value adjustment [72]. A multivariate statistical designmay deal with these data more appropriately. Principalcomponent analysis, an unsupervised data multivariate ana-lysis, extracts components (highly correlated variables thatexplain patterns in the data) of independent variables. Thisis useful if general patterns are observed and if eventualsample outliers are present as confounding individuals inthe data [72]. Partial least squares discriminant analysis, asupervised multivariate analysis test, extracts componentsfrom both the independent and the dependent variablesin order to generate a model that best explains why twogroups of patients or samples differ from one another[41].Logistic regression models may also be constructed in orderto predict group participation based on protein quantifica-tion. These models will produce odds ratios for individualprotein quantities – variations of these quantities willincrease the odds of that individual being of one group(e.g. infertile) or another [41].

Proteins can be annotated in different ways usingonline databases. Gene ontology terms, for example, areannotated into three general categories: biological process,molecular function, and cellular component [73]. However,proteins may also be annotated with pathway terms (Kyotoencyclopedia of genes and genomes – KEGG [74],Reactome [75], PantherDB [70]), domains (InterPro [76]),interactions (IntAct [77], BioGRID [78], HPRD [79], STRING[80], among others. Because a single protein is usuallyannotated with many terms from each database, statisticaltests have been designed to observe if the frequency ofthese terms in the PPI network or in one of its clusters ishigher than the frequency of these terms in the wholedatabase or in reference samples [81]. This approach,termed functional enrichment analysis, will offer a func-tional inference as to what role that protein (and its coun-terparts in the PPI network) plays in determining aphenotype [71,81–83].

In this workflow, a PPI network is generated by searchingfor interaction information from the databases cited above.Thenceforth, functional enrichment analysis of the differen-tially expressed proteins generates information regarding thefunctions enriched by these clusters and possible mechanismsthat explain the biological state [9,41–43].

The proteomic study of spermatozoa has taken a big leapever since the historical discovery of protamines in salmonsperm by Miescher [84]. Interest in using proteomic studies toidentify potential diagnostic markers has grown in recentyears. In one study, 2045 proteins were identified from agroup of patients with idiopathic infertility – 21 proteinswere differentially expressed (>1.2-fold) in men whose spermresulted in a clinical pregnancy via ART versus those who didnot [85]. With the absence of physiologically active transcrip-tion and translation, spermatozoa could be regarded as out-standing candidates for proteomic study. This is furthersupported by the fact that they can be purified at high con-centrations and can be pharmacologically manipulated intovarious functional states such as capacitated, non-capacitated,and acrosome reacted [86,87]. Moreover, understanding thedifferences in the proteomic profile of developing spermpopulations – caput verses cauda epididymis and ejaculatedverses capacitated, for example–will help unravel the post-

Figure 2. Scale-free protein-protein interaction sub-network of reviewed human sperm proteins (Uniprot [83]) highlighting hub proteins (proteins with manyinteractions) in red.

288 A. AGARWAL ET AL.

Dow

nloa

ded

by [

New

Yor

k U

nive

rsity

] at

12:

46 1

6 O

ctob

er 2

017

translational modifications relevant to sperm maturation[88,89]. In addition, comparative proteomic analysis will notonly pave the way for understanding any ‘hidden’ spermfactors responsible for infertility but also provide a molecularbiomarker for diagnosis and prognosis of male factor infertility– thus helping to develop an appropriate therapeuticapproach to treat male infertility [90].

Origin and characterization of sperm proteins –spermatogenesis, epididymal maturation,and post-ejaculation

Spermatogenesis is a highly coordinated process that marksthe differentiation of diploid spermatogonia through haploidround spermatid cells into the highly specialized, elongated,terminally differentiated spermatozoon that is packaged todeliver the paternal genome to an oocyte. Spermatozoa areleft with a limited amount of cytosolic organelles and a com-plete loss of transcriptional ability during post-testicularmaturation, while during epididymal transit, spermatozoaundergo a series of post translational modifications andacquire new proteins, making it competent to fuse with theoocyte [91]. Most of our knowledge regarding human sperm isbased on proteomic analysis of ejaculated human spermato-zoa. However, data on the characterization of proteins respon-sible for spermatogenesis and epididymal maturation are as ofyet still unavailable.

According to a systematic compilation by Amaral et al.,approximately 6198 different sperm proteins have been iden-tified – about 30% are known to be expressed in the testis[92]. Gene ontology (GO) analysis revealed that these proteinswere involved in various functional pathways, such as meta-bolism, apoptosis, cell cycle, meiosis, and membrane traffick-ing, among others. Apart from the expected abundance ofcytoskeletal, mitochondrial, flagellar, and membrane proteins,GO analysis has also provided reports on the presence of alarge proportion of proteins involved in transcription, proteinsynthesis, and turnover [93,94].

Human sperm proteins of testicular and epididymal originwere characterized by Li et al. and his group [95,96]. In theirreport, 112 of 319 identified human sperm-located proteins(35%) were exclusively of testicular-origin, 152 (48%) wereexclusively of epididymal origin and 55 (17%) were commonto both organs [96]. The report suggested that 47% of theidentified proteins were intrinsic sperm proteins expressed atthe spermatid stage while 23% were extrinsic sperm proteinsthat were acquired during epididymal transit and had anepididymal origin [95].

It was further validated that these proteins had a specificpattern of localization in sperm based on their origin.The epididymal proteins were more specifically concentratedat the principal piece while less so in the acrosomal areain comparison to testicular proteins [96]. HSPA2, a novel,testis-specific member of the HSP70 family, is expressedin the sperm plasma membrane along with another memberHSPA5 [97]. Underexpression of HSPA2 in oligoteratozoosper-mic men [98] suggests that human HSPA2 may act as a marker

for sperm maturity [99]. It is postulated to play a secondaryrole in the remodeling of the sperm plasma membrane duringspermatogenesis [100] to facilitate the formation of the ZP-binding sites. Another study also found this major spermcomponent to be underexpressed in oxidatively stressedspermatozoa [101]. Recent evidence states that HSPs, includ-ing 60, 70, and 90, migrate to the sperm surface and undergophosphorylation during capacitation [102] since tyrosine-phosphorylated sperm heads were able to bind to the zonapellucida [103]. Wang et al identified about 4675 proteins inhuman spermatozoa of which only 227 proteins had a testi-cular origin [104].

Sperm proteomics and male infertility

In the field of male infertility, where conventional semenanalysis sometimes fails to report the underlying cause ofmale factor infertility, recent advances in proteomic researchhave provided important biomarkers for diagnosis and prog-nosis of male infertility. One of the initial reports in the studyof sperm defects through the use of 2D proteomics reportedthe proteomic mapping of a patient who experienced a failurein in vitro fertilization, where 20 different proteins were iden-tified as compared to controls [105]. Subsequently, a differen-tial expression profile of 17 proteins was observed inasthenozoospermia [106], and further reports demonstratedthat clusterin and semenogelin were increased in thesepatients [101,107]. In an independent study, oligoastheno-zoospermic (OAT) patients exhibited significantly higher inci-dence of heterozygosity for cytosine-adenine-guanine repeatsand a higher mtDNA content with a lower percentage ofsperm expressing polymerase ᵞ and transcription factor A,mitochondrial in the spermatozoa of OAT group [108].

In globozoospermic patients, 35 proteins were differentiallyexpressed, of which 9 proteins were upregulated and 26proteins were downregulated in round-headed spermatozoacompared with normal spermatozoa [109]. These differentiallyexpressed proteins were suggested to be involved in a varietyof cellular processes and structures, including spermatogen-esis, cell skeleton, metabolism, and motility of spermatozoa.

A focused study on proteomic signatures in infertilepatients whose semen parameters were normal but with failedin vitro fertilization revealed 24 proteins with altered expres-sion in the normozoospermic infertile group. These proteinswere reported to be involved in energy production, structureand movement, and cell signaling and regulation [90]. Inanother study on patients with idiopathic infertility, a compar-ison of sperm proteome in men whose sperm resulted in aclinical pregnancy via assisted reproductive technology (ART)from those who did not, revealed 21 proteins to be differen-tially expressed (>1.2-fold) [85].

Abnormal DNA fragmentation and/or motility is asso-ciated with four main protein groups, viz. (i) sperm nuclearproteins such as the sperm protein associated with nucleusin the X chromosome (SPANX) isoforms and several types ofhistones [110]; (ii) mitochondria-related functions and oxi-dative stress proteins including mitochondrial ferritin,

EXPERT REVIEW OF PROTEOMICS 289

Dow

nloa

ded

by [

New

Yor

k U

nive

rsity

] at

12:

46 1

6 O

ctob

er 2

017

mitochondrial single-stranded DNA binding protein, andseveral isoforms of Peroxiredoxins; (iii) proteins related tosperm motility such as microtubule-based flagellum andspindle microtubule, and (iv) proteins related to the ubiqui-tin–proteasome pathway.

In an attempt to detect the differential protein profile inmen with and without varicocele using 2D gel electrophoresis;15 consistent differences in protein expression were identifiedin patients with varicocele when compared with controls[111]. Heat shock proteins, mitochondrial proteins, and cytos-keleton proteins are the major proteins affected by varicocele.Particularly, a significant upregulation of heat shock proteins(HSP70 and HSP90) was observed in subjects with varicocelewhich was further validated by Western blot and immunocy-tochemistry [112].

In a recent study, 369 differentially expressed proteins(DEP) were detected in infertile patients with unilateral vari-cocele in comparison to fertile donors [111]. Of the totalreported DEPs, 120 proteins were unique to the fertilegroup, and 38 were unique to the unilateral varicocele group114 overexpressed and 97 underexpressed in the unilateralvaricocele group. The major functional pathways of the 359DEP involve metabolism, disease, immune system, geneexpression, signal transduction, and apoptosis.

In another study comparing sperm of men with bilateralvaricocele with fertile controls, 73 DEP were reported, themajority of which associated with metabolic processes, stressresponse, oxidoreductase activity, enzyme regulation, andimmune system processes [112]. Comparison of the spermproteomes of men with unilateral versus bilateral varicoceledemonstrated 64 proteins in the bilateral group and 31 pro-teins in the unilateral group [7]. Core functions of the topprotein interaction networks were post-translational modifica-tion, protein folding, free-radical scavenging, cell death, andsurvival.

In another study, the sperm proteome from groups ofmen with high levels of reactive oxygen species (ROS+) werecompared with that of men with low or physiological levelsof ROS (ROS–) [101,113]. In the first case [113], a 2D-DIGEwith differential fluorescence tagging and in gel digestion ofthe proteins followed by LC–MS detection. A total of 31spots were differentially expressed with 6 significantlydecreased and 25 increased in the ROS– sample comparedwith the ROS+ sample. The result showed overabundance offour antioxidant proteins in ROS– sperm that may exertessential cytoprotective effects against the build-up of ROSlevels: lactotransferrin isoform 2, lactotransferrin isoform 1precursor, peroxiredoxin-1, and Mn-SOD mitochondrial iso-form A precursor [113]. In the second study, the sampleswere subjected to LC–MS/MS analysis through in-solutiondigestion of proteins for peptide characterization [101]. Thefindings revealed a total of 74 proteins in spermatozoa, outof which 15 proteins with more than twofold differencewere overexpressed (ROS+ group) when compared to theROS– group. The overexpressed proteins comprised the his-tone cluster 1 H2ba (HIST1H2BA); mitochondrial malate

dehydrogenase precursor (MDH2); heat shock protein 90kDa beta, member 1 (HSP90B1); heat shock 70 kDa protein5(HSPA5); glutamine synthetase (GLUL); transglutaminase4(prostate) (TGM4); glutathione peroxidase 4 isoform A pre-cursor (GPX4); sperm acrosomal membrane protein4(SPACA4); olfactomedin 4 precursor (OLFM4); and chromo-some 20 open reading frame 3 (C20orf3), whereas theunderexpressed proteins included semenogelin II precursor(SEMG2); peroxiredoxin 6 (PRDX6); clathrin heavy chain 1(CLTC); eukaryotic translation elongation factor 2 (EEF2);and enolase 1 (ENO1). Furthermore, there was significantchange in localization profile of the differentially expressedproteins. While the overexpressed proteins were predomi-nantly found in the cellular compartments of intracellular,organelle, macromolecular complex region and mitochon-dria; the underexpressed proteins were dominated in thecytoplasm extracellular, plasma membrane, protein complex(e.g. enolase) and the vesicular region (e.g. clathrin heavychain). Remarkably, the endosome, lipid particle, membrane-bound organelles and the microtubules showed an exclusiveabundance of overexpressed proteins in ROS+ group; whilethe proteinaceous extracellular matrix was restricted only tothe underexpressed proteins. In a recent study, comparisonswere made between protein profiles of spermatozoa fromfertile donors and infertile men with varying levels of ROS.Identification of six DEPs (Calmegin, Tripeptidyl peptidase II,Dynein intermediate chain 2, axonemal, heat shock 70 kDaprotein 4 L, early endosome antigen 1, and plasma serineprotease inhibitor) that were present in all the three ROSgroups with varying expression levels suggests that theymay serve as potential candidates of oxidative stress mar-kers [114].

Many of the proteins in its precursor or preprotein formhave been identified in studies relating to male fertilityanomalies, and it may be speculated that the accumulationof these protein is an indication of generalized defects at thepost translational level. Assumptions are also made regardingthe down regulation of some of the downstream functionsrelated to these proteins [101,106,115]. Such differential altera-tion pattern may be due to PTM which could be used as aneffective diagnostic strategy valuable for understanding malefactor infertility.

Ubiquitination and acetylation were reported to be theprincipal PTM found in non-obstructive azoospermia (NOA).Impairment in production of different enzymes belonging toUPS, namely UBE2B protein (an ubiquitin-conjugatingenzyme) may cause defects in spermatozoa maturation andresult in NOA [116,117]. Besides, mutations in genes encod-ing for testis-specific ubiquitin proteases, namely USP26 [118]and USP9Y [119], as observed in azoospermic patients, eluci-dates the role of protein ubiquitination in azoospermia. Onthe other hand, hyperacetylation of histone H4 is reported inobstructive azoospermic spermatids [120]. USP might playsome regulatory role in oligozoospermic patients [121],Differential expression of ubiquitin-conjugating enzymeUBE2B [117,122], and protease USP26 [123] was noticed in

290 A. AGARWAL ET AL.

Dow

nloa

ded

by [

New

Yor

k U

nive

rsity

] at

12:

46 1

6 O

ctob

er 2

017

oligozoospermic semen samples suggesting its possibleinvolvement.

Potential involvement of altered protein phosphorylationevents, such as phosphorylation of the gamma–tubulincomplex associated protein 2 (GCP2) in the pathogenesisof asthenozoospermia (AZS) [124] suggests the role of thisPTM in sperm motility. Parte et al. [125] further establishedthis by identifying 66 differentially expressed phosphopro-teins in AZS sperm, which comprised predominantly of theHSPs, cytoskeletal proteins, proteins associated with thefibrous sheath, and those associated with energy.Furthermore, a correlation between deficiency of tyrosinephosphorylation of tail proteins, especially those related tohyperactivated motility and AZS was also reported [126].Nevertheless, decreased membrane fluidity is hypothesizedto be the cause of hypophosphorylation of proteins duringcapacitation in these groups [127]. In two separate proteo-mic studies, several proteins having S-nitrosylation siteswere identified to be differentially expressed in AZS sperm[106,128]. Similarly, testis-specific α-tubulin isoformsTUBA3 C and TUBA8, in their acetylated state, wasdecreased, and TUBA4A was increased in AZS as comparedwith normal spermatozoa [129]. A variation of ubiquitinprotease USP26 was also reported to be directly associatedwith human sperm motility [123]. Moreover, in some AZSpatients, a severe form of structural anomaly is found in thesperm known as dysplasia of the fibrous sheath (DFS). Astudy demonstrated that sperm with DFS were accompa-nied by increased ubiquitination of the sperm surface aswell as the sperm mitochondrial proteins [130]. In AZSpatients only, percentage of SUMO1-positive spermatozoawas found to be inversely correlated with total and progres-sive motility [56]. In addition, an association of AZS withanomalies in seminal glycome has also been documented[131,132].

Teratozoospermia patients (TZS) are documented to pre-sent differential tyrosine phosphorylation of six sperm pro-teins in comparison to normozoospermic men [133].Furthermore, spermatozoa from TZS males exhibited a com-promised ability to undergo tyrosine phosphorylation fol-lowing capacitation [134]. Significant negative correlationwith the phosphorylated levels of protein Stat3 was alsoobserved in TZS [134]. Similarly, ejaculated sperm of theTZS group demonstrated increased binding of anti-ubiquitinantibodies to the sperm surface, reflecting the occurrence ofenhanced sperm surface ubiquitination in nonsystematiccases of TZS [135].

A comparative study on assessment of tyrosine phos-phorylation status in spermatozoa of varicocele patientsrevealed a reduced level of phosphorylation [136], which isattributed to an alteration in plasma membrane dynamicsresulting in sperm dysfunction in these patients.Furthermore, enzymes linked to protein glycosylationnamely acid β-glucuronidase, α-mannosidase, α-glucosidase,α-galactosidase, β-galactosidase, and β-N-acetylglucosamini-dase showed overexpression and hyperactivity in seminalplasma of infertile men with varicocele [137]. As theseextracellular glycosidases are implicated to cause the mod-ification of sperm plasma membrane glycoproteins during

sperm maturation process, it could be postulated that suchoverexpression of acid glycosidase in the seminal plasmawould mask the carbohydrate-containing molecules presentin ejaculated spermatozoa of varicocele group, altering theirfertilization potential. One of the major pathways of PTMand protein folding found to be severely compromised invaricocele individuals is the UPS [7]. In addition, assessmentof the ubiquitination median, a marker for functionality ofthe UPS was also lower in varicocele group, where it wasreported to show a negative correlation with sperm mor-phology [138].

Expert commentary

In the post-genomic era, studies have mainly focused onthe identification of novel protein biomarkers in complexbiological systems. A biomarker is a distinctive biological orbiologically derived indicator of a process, event, or condi-tion, and is considered ideal if it serves the purpose ofscreening, diagnosis, and monitoring disease activity. Inaddition, they may hold responsible for targeted therapyor would assess therapeutic response [139]. With regard tomale factor infertility, the main objective of a biomarker isto evaluate in an accurate and minimally invasive manner,the male potential to father a child. Furthermore, use ofsemen for proteomic analysis is complicated because itcontains sperm as well as seminal plasma and the mostimportant challenge in sperm proteomics is the fact thatspermatozoa undergoes distinct physiological changes afterejaculation. The advantages of using proteomic over routinesemen analysis is that it confirms the qualitative and quan-titative measure of a protein within the spermatozoa.Nevertheless, identification of the ideal biomarker for clin-ical diagnosis in such a complex scenario to distinguish thepathophysiological state is one of the main challenges.Furthermore, in a clinical proteomics study one sample perindividual is used with the assumption that this sample, isrepresentative of a disease. However, it is more likely repre-sentative of the time of day it was drawn, the nutritionalstatus and life style of the patient, and their genetic back-ground rather than the pathology under investigation.Therefore, longitudinal sampling where samples are col-lected over time to determine how the proteome changesover time within an individual is necessary for discovery ofputative biomarker. Personalized reference ranges are wheremedicine will have a tremendous impact; discovery-basedproteomics using longitudinal sampling will be critical toelucidate predictive biomarkers.

As vide supra in the past two decades, studies on pro-teomic profiling of spermatozoa result in the generation ofan increasing amount of data. The information generatedmainly comprises of complex protein compositions and alarge number of isolated proteins whose implications inthe biological state is yet to be discovered. This limits theuse of proteomic approaches to elucidate the role of biolo-gical markers [140]. Furthermore, an additional confoundingfactor is the inherent complexity of the disease, therefore,the pathogenesis cannot be singled out by a protein, butmight indeed be a result of multiple interacting proteins

EXPERT REVIEW OF PROTEOMICS 291

Dow

nloa

ded

by [

New

Yor

k U

nive

rsity

] at

12:

46 1

6 O

ctob

er 2

017

across multiple pathways. Thus, employing a panel of bio-markers for identification of infertility is more likely to besuccessful. With relation to oxidative stress in the spermato-zoa, proteins, such as MDH2, TGM4, GPX4, GLUL, HSP90B1,and HSPA5 are suggested as possible biomarkers due totheir increased expression in samples with high levels ofROS compared with those without oxidative stress [141].Data from a number of independent laboratories suggeststhat an underrepresentation of a key molecular chaperone,HSPA2 is responsible for impaired sperm–egg recognition[142] which is reported to be modulated by oxidative stress(lipid aldehydes) [143]. While considering the use of multipleinteracting-markers to provide specific valid signatures, theproblems inherent to validation increases exponentially[144]. The question is how to test the validity. Is the changein expression of a candidate protein directly associated withthe disease or is it due to the presence of a PTM(s) or mightit only be an artefact resulting from technical variability?This fact is further complicated by the identification of pro-teins possessing PTM within the normal and abnormal sper-matozoa, specific to different pathological states, such asoxidative stress. Nevertheless, the current trends to identifyPTMs are only predictive [145]. Therefore, creation of newpipelines as well as analytical methods [146] that allow foranalysis of specific PTMs on sperm function will impact theunderstanding of protein function in healthy and infertilespermatozoa. A study that attested to the inherent technicalvariability present in proteomic profiling, as published bymembers of the Human Proteome Organization (HUPO)showed that when an equimolar protein sample containing20 human proteins was given to 27 different laboratories,only 7 out of 27 labs were able to correctly identify all the20 proteins in the sample. The discrepancies in the resultswere attributed to the databases used for analysis, as theycontain erroneous confidence levels, thereby missing severalof the 20 proteins present in the sample [147].

Another aspect of understanding the sperm proteome indifferent pathophysiological states is to identify theabnormality in structure and function of the protein so asto target them for development of reversible male contra-ception. This necessitates the identification of sperm pro-teins on different domains, which are acquired by spermmostly as they traverse the epididymis. The sperm pro-teome is dynamic due to the fact that proteins are trans-ported into the spermatozoa from the epididymal, seminal,or prostatic fluid. Epididymal proteins are preferred targets

for contraception development over testicular proteins, asthe blockage of sperm maturational events will not obstructspermatogenesis and testicular endocrine function, as wellas allow for reversibility [148,149]. Both pre- and postfertili-zation events are possibly affected by targeting the spermhead and the flagellar proteins. Evaluation of sperm pro-teins thus allows for a straightforward assessment of testi-cular homeostasis, and is an increasingly important area ofstudy in male infertility.

Five-year view

Among the main challenges that will need to be dealt with instudying the infertile male, sperm proteomics studies will greatlybenefit from increased sensitivity, specificity and reproducibility ofMS-based proteomics. Studying post-translational modificationswill further allow for determination of understanding how thedynamic equilibrium of fertile sperm, co-inhabiting with alteredsperm, may lead to positive or negative effects to fertility.Furthermore, the clinical impact of proteomics to sperm biologyand functional state may (i) allow for determination of the biolo-gical systems leading to the observed cellular integrity and (ii)allow for pathway specific intervention. In a recent report, Intasquiet al. demonstrated a number of proteins associated with spermDNA fragmentation, some of which are transferred specificallyduring epididymal transit through epididymosomes [9]. It is inter-esting that multiplatform studiesmay indeed enhance our currentknowledge of sperm biology, as well as of male fertility.

Acknowledgement

Financial support for this study was provided by the American Center forReproductive Medicine, Cleveland Clinic.

Financial & competing interests disclosure

The authors have no relevant affiliations or financial involvement with anyorganization or entity with a financial interest in or financial conflict with thesubject matter or materials discussed in the manuscript. This includesemployment, consultancies, honoraria, stock ownership or options, experttestimony, grants or patents received or pending, or royalties.

ORCID

Luna Samanta http://orcid.org/0000-0002-2969-0071

Key issues

● Seminal improvement in MS and development of bioinformatics tools together with a renaissance in wet-bench aspects, such as 2D-gel electrophoresis,gel-free, and off-gel separation techniques have allowed proteomics to achieve its zenith.

● Proteomics is more promising for the study of sperm biology than other omics as these cells are essentially silent in terms of transcription andtranslation.

● A large number of spermatozoa proteins have been cataloged in various case-control studies.● A set of proteins differentially expressed in spermatozoa of infertile patients point toward use of a set of biomarkers rather than a single protein.● The spermatozoa proteome is a dynamic one and changes during post testicular stages enable sperm to fertilize the egg.● Spermatozoal proteins undergo a series of post-translational changes during post-testicular maturation.● Some proteins in spermatozoa undergo multiple post-translational modifications.● Current trends that involve identifying PTMs are only predictive.● Studying protein interactions and the structures of multi-protein complexes may shed more light on the different proteins involved in sperm function.

292 A. AGARWAL ET AL.

Dow

nloa

ded

by [

New

Yor

k U

nive

rsity

] at

12:

46 1

6 O

ctob

er 2

017

References

Papers of special note have been highlighted as:• of interest•• of considerable interest

1. World Health Organization. WHO laboratory manual for the exam-ination and processing of human semen. 5th ed. Geneva: WHOPress; 2010.

2. Mahutte NG, Arici A. Failed fertilization: is it predictable?. Curr.Opin. Obstet. Gynecol. 2003;15(3):211–218.

3. Virro MR, Larson-Cook KL, Evenson DP. Sperm chromatin structureassay (SCSA®) parameters are related to fertilization, blastocystdevelopment, and ongoing pregnancy in in vitro fertilization andintracytoplasmic sperm injection cycles. Fertility and Sterility.2004;81(5):1289–1295.

4. Borini A, Tarozzi N, Bizzaro D, et al. Sperm DNA fragmentation:paternal effect on early post-implantation embryo development inART. Hum. Reprod. Oxf. Engl. 2006;21(11):2876–2881.

5. Zhao J, Zhang Q, Wang Y, et al. Whether sperm deoxyribonucleic acidfragmentation has an effect on pregnancy and miscarriage after invitro fertilization/intracytoplasmic sperm injection: a systematicreview and meta-analysis. Fertil. Steril. 2014;102(4):998–1005.e8.

6. Jin J, Pan C, Fei Q, et al. Effect of sperm DNA fragmentation on theclinical outcomes for in vitro fertilization and intracytoplasmicsperm injection in women with different ovarian reserves. Fertil.Steril. 2015;103(4):910–916.

7. Agarwal A, Sharma R, Durairajanayagam D, et al. Differential pro-teomic profiling of spermatozoal proteins of infertile men withunilateral or bilateral varicocele. Urology. 2015;85(3):580–588.

8. Barazani Y, Agarwal A, Sabanegh ES. Functional sperm testing andthe role of proteomics in the evaluation of male infertility. Urology.2014;84(2):255–261.

9. Intasqui P, Camargo M, Del Giudice PT, et al. Unraveling the spermproteome and post-genomic pathways associated with spermnuclear DNA fragmentation. J. Assist. Reprod. Genet. 2013;30(9):1187–1202.

10. Plant TM, Zeleznik AJ. Knobil and Neill’s Physiology ofReproduction: Two-Volume Set. Netherlands: Elsevier; 2015.

11. Gadella BM, Boerke A. An update on post-ejaculatory remodelingof the sperm surface before mammalian fertilization.Theriogenology. 2016;85(1):113–124.

12. Nagdas SK, Smith L, Mcnamara A, et al. Identification and charac-terization of a bovine sperm acrosomal matrix protein and itsmechanism of interaction with acrosomal hydrolases. Mol. Cell.Biochem. 2015;410:11–23.

13. Erikson DW, Way AL, Bertolla RP, et al. Influence of osteopontin,casein and oviductal fluid on bovine sperm capacitation. AnimReprod. 2007;4(3/4):103–112.

14. Gonçalves RF, Wolinetz CD, Killian GJ. Influence of arginine-glycine-aspartic acid (RGD), integrins (αV and α5) and osteopontin onbovine sperm–egg binding, and fertilization in vitro.Theriogenology. 2007;67(3):468–474.

15. D’Amours O, Frenette G, Bordeleau L-J, et al. Epididymosomestransfer epididymal sperm binding protein 1 (ELSPBP1) to deadspermatozoa during epididymal transit in bovine. Biology ofReproduction. 2012;87(4):94.

16. Girouard J, Frenette G, Sullivan R. Comparative proteome and lipidprofiles of bovine epididymosomes collected in the intraluminalcompartment of the caput and cauda epididymidis. Int. J.Andrology. 2011;34(5pt2):e475–486.

17. Sullivan R, Frenette G, Girouard J. Epididymosomes are involved inthe acquisition of new sperm proteins during epididymal transit.Asian J. Andrology. 2007;9(4):483–491.

• One of the key findings that establishes sperm proteomechanges during epididymal transit.

18. Suryawanshi AR, Khan SA, Joshi CS, et al. Epididymosome-mediatedacquisition of MMSDH, an androgen-dependent and developmen-tally regulated epididymal sperm protein. J. Androl. 2012;33(5):963–974.

19. Aalberts M, Van Dissel-Emiliani FMF, Van Adrichem NPH, et al.Identification of distinct populations of prostasomes that differen-tially express prostate stem cell antigen, annexin A1, and GLIPR2 inhumans. Biol. Reprod. 2012;86(3):82.

20. Aalberts M, Sostaric E, Wubbolts R, et al. Spermatozoa recruitprostasomes in response to capacitation induction. (BBA)Proteins Proteom. 2013;1834(11):2326–2335. * One importantfinding that showed the role of exosomes (prostasomes) insperm capacitation

21. Kanagawa K, Sugimura K, Kuratsukuri K, et al. Norepinephrineactivates P44 and P42 MAPK in human prostate stromal andsmooth muscle cells but not in epithelial cells. The Prostate.2003;56(4):313–318.

22. Meggiolaro D, Porcelli F, Carnevali A, et al. A possible role of Fasantigen in ejaculated spermatozoa of fertile bulls: an immunocyto-chemical quantitative approach. Acta Histochemica. 2006;107(6):463–468.

23. Said T, Agarwal A, Grunewald S, et al. Selection of nonapoptoticspermatozoa as a new tool for enhancing assisted reproductionoutcomes: an in vitro model. Biol. Reprod. 2006;74(3):530–537.

24. Porcelli F, Meggiolaro D, Carnevali A, et al. Fas ligand in bullejaculated spermatozoa: A quantitative immunocytochemicalstudy. Acta Histochemica. 2006;108(4):287–292.

25. Aebersold R, Mann M. Mass spectrometry-based proteomics.Nature. 2003;422(6928):198–207.

26. Zhang Y, Fonslow BR, Shan B, et al. Protein analysis by shotgun/bottom-up proteomics. Chem. Rev. 2013;113(4):2343–2394.

27. Westermeier R. Looking at proteins from two dimensions: a reviewon five decades of 2D electrophoresis. Arch. Physiol. Biochem.2014;120(5):168–172.

28. Hall JC, Killian GJ. Two-dimensional gel electrophoretic analysis ofrat sperm membrane interaction with cauda epididymal fluid. J.Androl. 1989;10(1):64–76.

29. Bantscheff M, Lemeer S, Savitski MM, et al. Quantitative massspectrometry in proteomics: critical review update from 2007 tothe present. Anal. Bioanal. Chem. 2012;404(4):939–965.

30. Mallick P, Kuster B. Proteomics a pragmatic perspective. Nat.Biotechnol. 2010;28(7):695–709.

•• An important review that describes the evolution of massspectrometry-based proteomic technologies for understandingof the complex and dynamic nature of proteomes and therobustness of the technology for single protocol proteomicstrategy to address all biological questions.

31. Scherl A. Clinical protein mass spectrometry. Methods. 2015;81:3–14.

32. Pennington SR, Wilkins MR, Hochstrasser DF, et al. Proteome ana-lysis: from protein characterization to biological function. TrendsCell Biol. 1997;7(4):168–173.

33. Bland AM, Janech MG, Almeida JS, et al. Sources of variabilityamong replicate samples separated by two-dimensional gel elec-trophoresis. J. Biomol. Tech. JBT. 2010;21(1):3–8.

34. Choe LH, Lee KH. Quantitative and qualitative measure of intrala-boratory two-dimensional protein gel reproducibility and theeffects of sample preparation, sample load, and image analysis.Electrophoresis. 2003;24(1920):3500–3507.

35. López JL. Two-dimensional electrophoresis in proteome expressionanalysis. J. Chromatogr. B Analyt. Technol. Biomed. Life. Sci.2007;849(1–2):190–202.

36. Whitehouse CM, Dreyer RN, Yamashita M, et al. Electrospray inter-face for liquid chromatographs and mass spectrometers. Anal.Chem. 1985;57(3):675–679.

37. Cox J, Mann M. Quantitative high-resolution proteomics for data-driven systems biology. Annu. Rev. Biochem. 2011;80:273–299.

38. Karas M, Bachmann D, Hillenkamp F. Influence of the wavelengthin high-irradiance ultraviolet laser desorption mass spectrometry oforganic molecules. Anal. Chem. 1985;57(14):2935–2939.

39. Tanaka K, Waki H, Ido Y, et al. Protein and polymer analyses up tom/z 100 000 by laser ionization time-of-flight mass spectrometry.Rapid Commun. Mass Spectrom. 1988;2(8):151–153.

EXPERT REVIEW OF PROTEOMICS 293

Dow

nloa

ded

by [

New

Yor

k U

nive

rsity

] at

12:

46 1

6 O

ctob

er 2

017

40. Fenn JB, Mann M, Meng CK, et al. Electrospray ionization for massspectrometry of large biomolecules. Science. 1989;246(4926):64–71.

41. Intasqui P, Antoniassi MP, Camargo M, et al. Differences in theseminal plasma proteome are associated with oxidative stresslevels in men with normal semen parameters. Fertil. Steril.2015;104(2):292–301.

42. Camargo M, Intasqui Lopes P, Del Giudice PT, et al. Unbiased label-free quantitative proteomic profiling and enriched proteomic path-ways in seminal plasma of adult men before and after varicoce-lectomy. Hum. Reprod. Oxf. Engl. 2013;28(1):33–46.

43. Intasqui P, Camargo M, Del Giudice PT, et al. Sperm nuclear DNAfragmentation rate is associated with differential protein expres-sion and enriched functions in human seminal plasma. BJU Int.2013;112(6):835–843.

44. Lo Turco EG, Souza GHMF, Garcia JS, et al. Effect of endometriosison the protein expression pattern of follicular fluid from patientssubmitted to controlled ovarian hyperstimulation for in vitro ferti-lization. Hum. Reprod. Oxf. Engl. 2010;25(7):1755–1766.

45. Gillet LC, Navarro P, Tate S, et al. Targeted data extraction of theMS/MS spectra generated by data-independent acquisition: a newconcept for consistent and accurate proteome analysis. Mol. Cell.Proteomics MCP. 2012;11(6):016717. O111.

•• One of the first papers to propose the targeted data extractionof tandem mass spectra by data-independent acquisition.

46. Xu Y, Bailey U-M, Schulz BL. Automated measurement of site-specific N-glycosylation occupancy with SWATH-MS. Proteomics.2015;15(13):2177–2186.

•• Describes a method for automated relative quantification ofsite-specific glycosylation that is useful for research with clin-ical samples

47. Zhang Y, Bilbao A, Bruderer T, et al. The Use of Variable Q1 IsolationWindows Improves Selectivity in LC-SWATH-MS Acquisition. J.Proteome Res. 2015;14(10):4359–4371.

• A novel concept of ‘scheduled SWATH’ that incorporates (i)variable isolation windows and (ii) precursor retention timesegmentation for further improvement of both peptide andmetabolite identifications.

48. Law KP, Lim YP. Recent advances in mass spectrometry: dataindependent analysis and hyper reaction monitoring. Expert Rev.Proteomics. 2013;10(6):551–566.

49. Schliekelman P, Liu S. Quantifying the effect of competition fordetection between coeluting peptides on detection probabilities inmass-spectrometry-based proteomics. J. Proteome Res. 2014;13(2):348–361.

• Explains the method for quantifying co-eluting peptides.50. Camerini S, Mauri P, The role of protein and peptide separation

before mass spectrometry analysis in clinical proteomics. J.Chromatogr. A. 2015;1381:1–12.

• Sets out the importance of protein and peptide separationbefore MS in clinical samples.

51. Zuo X, Speicher DW. Comprehensive analysis of complex proteomesusingmicroscale solution isoelectrofocusing prior to narrow pH rangetwo-dimensional electrophoresis. Proteomics. 2002;2(1):58–68.

52. Zuo X, Hembach P, Echan L, et al. Enhanced analysis of humanbreast cancer proteomes using micro-scale solution isoelectrofo-cusing combined with high resolution 1-D and 2-D gels. J.Chromatogr. B Analyt. Technol. Biomed. Life. Sci. 2002;782(1):253–265.

53. Chahrour O, Cobice D, Malone J. Stable isotope labelling methodsin mass spectrometry-based quantitative proteomics. J. Pharm.Biomed. Anal. 2015;113:2–20.

54. Boersema PJ, Raijmakers R, Lemeer S, et al. Multiplex peptide stableisotope dimethyl labeling for quantitative proteomics. Nat. Protoc.2009;4(4):484–494.

• One of the Nature Protocols for multiplexing quantitative pro-teomics by stable isotope dimethyl labeling.

55. Marchiani S, Tamburrino L, Ricci B, et al. SUMO1 in human sperm:new targets, role in motility and morphology and relationship withDNA damage. Reprod. Camb. Engl. 2014;148(5):453–467.

56. Marchiani S, Tamburrino L, Giuliano L, et al. Sumo1-ylation ofhuman spermatozoa and its relationship with semen quality. Int.J. Androl. 2011;34(6 Pt 1):581–593.

57. Kasvandik S, Sillaste G, Velthut-Meikas A, et al. Bovine spermplasma membrane proteomics through biotinylation and subcellu-lar enrichment. Proteomics. 2015;15(11):1906–1920.

58. D’Amours O, Bordeleau L-J, Frenette G, et al. Binder of sperm 1and epididymal sperm binding protein 1 are associated withdifferent bull sperm subpopulations. Reproduction. 2012;143(6):759–771.

59. Nagdas SK, Winfrey VP, Olson GE. Identification of a hamster sperm26-kilodalton dehydrogenase/reductase that is exclusively localizedto the mitochondria of the flagellum. Biol. Reprod. 2006;75(2):197–202.

60. Nagaraj N, Wisniewski JR, Geiger T, et al. Deep proteome andtranscriptome mapping of a human cancer cell line. Mol. Syst.Biol. 2011;7(1):548.

61. Beck M, Schmidt A, Malmstroem J, et al. The quantitative proteomeof a human cell line. Mol. Syst. Biol. 2011;7(1):549.

62. De Las Rivas J, De Luis A. Interactome data and databases: differenttypes of protein interaction. Comp. Funct. Genomics. 2004;5(2):173–178.

63. Safari-Alighiarloo N, Taghizadeh M, Rezaei-Tavirani M, et al. Protein-protein interaction networks (PPI) and complex diseases.Gastroenterol. Hepatol. Bed Bench. 2014;7(1):17–31.

64. Sevimoglu T, Arga KY. The role of protein interaction networks insystems biomedicine. Comput. Struct. Biotechnol. J. 2014;11(18):22–27.

65. Baker MA. Proteomics of post-translational modifications of mam-malian spermatozoa. Cell Tissue Res. 2016;363(1):279–287.

66. Cornwall GA, Role of posttranslational protein modifications inepididymal sperm maturation and extracellular quality control.Adv. Exp. Med. Biol. 2014;759:159–180.

• This paper explains the importance of post-translational pro-tein modifications of spermatozoa during epididymalmaturation.

67. Olsen JV, Mann M. Status of large-scale analysis of post-transla-tional modifications by mass spectrometry. Mol. Cell. ProteomicsMCP. 2013;12(12):3444–3452.

68. Signorelli J, Diaz ES, Morales P. Kinases phosphatases and proteasesduring sperm capacitation. Cell Tissue Res. 2012;349(3):765–782.

69. Al Rawi S, Louvet-Vallée S, Djeddi A, et al. Postfertilization autop-hagy of sperm organelles prevents paternal mitochondrial DNAtransmission. Science. 2011;334(6059):1144–1147.

70. Mi H, Muruganujan A, Thomas PD. PANTHER in 2013: modeling theevolution of gene function, and other gene attributes, in the con-text of phylogenetic trees. Nucleic Acids Res. 2013;41(D1):D377–D386.

71. Bindea G, Mlecnik B, Hackl H, et al. ClueGO: a Cytoscape plug-in todecipher functionally grouped gene ontology and pathway anno-tation networks. Bioinformatics. 2009;25(8):1091–1093.

72. Robles MS, Cox J, Mann M. In-vivo quantitative proteomics revealsa key contribution of post-transcriptional mechanisms to the circa-dian regulation of liver metabolism. PLoS Genet. 2014;10(1):e1004047.

73. Ashburner M, Ball CA, Blake JA, et al. Gene Ontology: tool for theunification of biology. Nat. Genet. 2000;25(1):25–29.

74. Kanehisa M, Goto S. KEGG: kyoto encyclopedia of genes and gen-omes. Nucleic Acids Res. 2000;28(1):27–30.

75. Croft D, Mundo AF, Haw R, et al. The reactome pathway knowl-edgebase. Nucleic Acids Res. 2014;42(Database issue):D472–477.

76. Mitchell A, Chang H-Y, Daugherty L, et al. The InterPro proteinfamilies database: the classification resource after 15 years.Nucleic Acids Res. 2015;43(D1):D213–D221.

77. Orchard S, Ammari M, Aranda B, et al. The MIntAct project–IntActas a common curation platform for 11 molecular interaction data-bases. Nucleic Acids Res. 2014;42(Database issue):D358–363.

78. Stark C, Breitkreutz B-J, Reguly T, et al. BioGRID: a general reposi-tory for interaction datasets. Nucleic Acids Res. 2006;34(Databaseissue):D535–539.

294 A. AGARWAL ET AL.

Dow

nloa

ded

by [

New

Yor

k U

nive

rsity

] at

12:

46 1

6 O

ctob

er 2

017

79. Keshava Prasad TS, Goel R, Kandasamy K, et al. Human proteinreference database–2009 update. Nucleic Acids Res. 2009;37(Database issue):D767–772.

80. Szklarczyk D, Franceschini A, Wyder S, et al. STRING v10: protein-protein interaction networks, integrated over the tree of life.Nucleic Acids Res. 2015;43(Database issue):D447–452.

81. Schmidt A, Forne I, Imhof A. Bioinformatic analysis of proteomicsdata. BMC Syst. Biol. 2014;8(Suppl 2):S3.

82. Maere S, Heymans K, Kuiper M. BiNGO: a Cytoscape plugin toassess overrepresentation of gene ontology categories in biologicalnetworks. Bioinforma. Oxf. Engl. 2005;21(16):3448–3449.

83. UniProt Consortium. UniProt: a hub for protein information. NucleicAcids Res. 2015;43(Database issue):D204–212.

84. Miescher F. Das Protamin—Eine neue organische Basis ausdenSamenf—den des Rheinlachses. Ber Dtsch Chem Ges. 1874;7(1):376.

85. Zhu Y, Wu Y, Jin K, et. al. Differential proteomic profiling in humanspermatozoa that did or did not result in pregnancy via IVF andAID. Proteomics Clin Appl. 2013;7(11–12):850–858.

86. Baker MA. The omics revolution and our understanding of spermcell biology. Asian. J Androl. 2011;13(1):6–10.

• One of the earliest reviews on the role of omics study inunderstanding sperm structure and function.

87. Oliva R, De Mateo S, Estanyol JM. Sperm cell proteomics.Proteomics. 2009;9(4):1004–1017.

88. Ijiri TW, Merdiushev T, Cao W, et al. Identification and validation ofmouse sperm proteins correlated with epididymal maturation.Proteomics. 2011;11(20):4047–4062.

89. Secciani F, Bianchi L, Ermini L, et al., et. al.. Protein profile ofcapacitated versus ejaculated human sperm. J Proteome Res.2009;8(7):3377–3389.

•• This paper shows a comparative proteome profiling of humancapacitated and ejaculated sperm demonstrating the dynamicnature of sperm proteome in absence of transcription andtranslation in the cell.

90. Xu W, Hu H, Wang Z, et. al.. Proteomic characteristics of sperma-tozoa in normozoospermic patients with infertility. J Proteomics.2012;75(17):5426–5436.

•• One of the key papers studied in depth for this article. Theproteomics analysis was carried out on spermatozoa of infer-tile patients with normal spermiogram.

91. Gatti JL, Castella S, Dacheux F, et. al.. Post-testicular sperm envir-onment and fertility. Anim Reprod Sci. 2004;82–83:321–339.

92. Amaral A, Castillo J, Ramalho-Santos J, et al. The combined humansperm proteome: cellular pathways and implications for basic andclinical science. Hum Reprod Update. 2014;20(1):40–62.

• One of the preliminary reports on human sperm proteome.93. Martínez-Heredia J, Estanyol JM, Ballesca JL, et al. Proteomic iden-

tification of human sperm proteins. Proteomics. 2006;6(15):4356–4369.

• One of the preliminary reports on human sperm proteome.94. De Mateo S, Martínez-Heredia J, Estanyol JM, et. al.. Marked corre-

lations in protein expression identified by proteomic analysis ofhuman spermatozoa. Proteomics. 2007;7(23):4264–4277. Erratumin: Proteomics.8:4833 (2008). Domíguez-Fandos, David [correctedto Domínguez-Fandos, David].

• One of the preliminary reports on human sperm proteome.95. Li J, Liu F, Wang H, et al. Systematic mapping and functional

analysis of a family of human epididymal secretory sperm locatedproteins. Mol Cell Proteomics. 2010;9(11):2517–2528.

96. Li J, Liu F, Liu X, et al. Mapping of the human testicular proteomeand its relationship with that of the epididymis and spermatozoa.Mol Cell Proteomics. 2011;10(3):M110.004630.

97. Naaby-Hansen S, Herr JC. Heat shock proteins on the human spermsurface. J Reprod Immunol. 2010;84(1):32–40.

98. Cedenho AP, Lima SB, Cenedeze MA, et al. Oligozoospermia andheat-shock protein expression in ejaculated spermatozoa. HumReprod. 2006;21(7):1791–1794.

99. Ergur AR, Dokras A, Giraldo JL, et al. Sperm maturity and treatmentchoice of in vitro fertilization (IVF) or intracytoplasmic sperm

injection: diminished sperm HspA2 chaperone levels predict IVFfailure. Fertil Steril. 2002;77(5):910–918.

100. Huszar G, Stone K, Dix D, et al. Putative creatine kinase M-isoformin human sperm is identified as the 70-kilodalton heat shockprotein HspA2. Biol Reprod. 2000;63(3):925–932.

101. Sharma R, Agarwal A, Mohanty G, et al. Proteomic analysis ofhuman spermatozoa proteins with oxidative stress. Reprod BiolEndocrinol. 2013;11:48. doi:10.1186/1477-7827-11-48

•• One of the first papers that reports the differentially expressedproteins in human spermatozoa in response to oxidative stress.

102. Baker MA, Hetherington L, Aitken RJ. Identification of SRC as a keyPKA-stimulated tyrosine kinase involved in the capacitation-asso-ciated hyperactivation of murine spermatozoa. J Cell Sci. 2006;119(pt 15):3182–3192.

103. Asquith KL. Tyrosine phosphorylation activates surface chaperonesfacilitating sperm-zona recognition. J. Cell Sci. 2004;117:3645–3657.

104. Wang G, Guo Y, Zhou T, et al., et. al.. In-depth proteomic analysis ofthe human sperm reveals complex protein compositions. JProteomics. 2013;79:114–122.

•• This report shows in depth proteomic analysis of humansperm.

105. Wolfsberg TG, White JM. ADAMs in fertilization and development.Dev Biol. 1996;180(2):389–401.

106. Martínez-Heredia J, De Mateo S, Vidal-Taboada JM, et al.Identification of proteomic differences in asthenozoospermicsperm samples. Hum Reprod. 2008;23(4):783–791.

107. Zhao C, Huo R, Wang F-Q, et al. Identification of several proteinsinvolved in regulation of sperm motility by proteomic analysis.Fertil Steril. 2007;87(2):436–438.

108. Amaral A, Ramalho-Santos J, St John JC. The expression of poly-merase gamma and mitochondrial transcription factor A and theregulation of mitochondrial DNA content in mature human sperm.Hum Reprod. 2007;22(6):1585–1596.

109. Liao -T-T, Xiang Z, Zhu W-B, et al. Proteome analysis of round-headed and normal spermatozoa by 2-D fluorescence differencegel electrophoresis and mass spectrometry. Asian J Androl. 2009;11(6):683–693.

110. Behrouzi B, Kenigsberg S, Alladin N, et al. Evaluation of potentialprotein biomarkers in patients with high sperm DNA damage. SystBiol Reprod Med. 2013;59(3):153–163.

111. Agarwal A, Sharma R, Durairajanayagam D, et al. Major protein altera-tions in spermatozoa from infertile men with unilateral varicocele.Reprod Biol Endocrinol. 2015;13:8. doi:10.1186/s12958-015-0007-2

112. Agarwal A, Sharma R, Durairajanayagam D, et al. Spermatozoaprotein alterations in infertile men with bilateral varicocele. AsianJ Androl. 2016;18(1):43–53.

113. Hamada A1, Sharma R, Du Plessis SS, et al. Two-dimensional differ-ential in-gel electrophoresis-based proteomics of male gametes inrelation to oxidative stress. Fertil Steril. 2013;99(5):1216–1226.

114. Ayaz A, Agarwal A, Sharma R, et al. Impact of precise modulation ofreactive oxygen species levels on spermatozoa proteins in infertilemen. Clin Proteomics. 2015;12(1):4.

115. De Yebra L, Ballescá JL, Vanrell JA, et al. Detection of P2 precursorsin the sperm cells of infertile patients who have reduced protamineP2 levels. Fertil Steril. 1998;69(4):755–759.

116. Mou L, Zhang Q, Diao R, et al. A functional variant in the UBE2Bgene promoter is associated with idiopathic azoospermia. ReprodBiol Endocrinol. 2015;13:79. doi:10.1186/s12958-015-0074-4

117. Huang I, Emery BR, Christensen GL, et al. Novel UBE2B-associatedpolymorphisms in an azoospermic/oligozoospermic population.Asian J Androl. 2008;10(3):461–466.

118. Paduch DA, Mielnik A, Schlegel PN. Novel mutations in testis-specific ubiquitin protease 26 gene may cause male infertility andhypogonadism. Reprod Biomed Online. 2005;10(6):747–754.

119. Sun C, Skaletsky H, Birren B, et al. An azoospermic man with a denovo point mutation in the Y-chromosomal gene USP9Y. NatGenet. 1999;23(4):429–432.

120. Kleiman SE, Bar-Shira Maymon B, Hauser R, et al. Histone H4acetylation and AZFc involvement in germ cells of specimens ofimpaired spermatogenesis. Fertil Steril. 2008;89(6):1728–1736.

EXPERT REVIEW OF PROTEOMICS 295

Dow

nloa

ded

by [

New

Yor

k U

nive

rsity

] at

12:

46 1

6 O

ctob

er 2

017

121. Agarwal A, Mulgund A, Sharma R, et al. Mechanisms of oligozoos-permia: an oxidative stress perspective. Syst Biol Reprod Med.2014;60(4):206–216.

122. Yatsenko AN, Georgiadis AP, Murthy LJ, et al. Matzuk MM. UBE2BmRNA alterations are associated with severe oligozoospermia ininfertile men. Mol Hum Reprod. 2013;19(6):388–394.

123. Shi Y-C, Wei L, Cui Y-X, et al. Association between ubiquitin-specificprotease USP26 polymorphism and male infertility in Chinese men.Clin Chim Acta. 2011;412(7–8):545–549.

124. Chan -C-C, Shui H-A, Wu C-H, et al. Motility and protein phosphor-ylation in healthy and asthenozoospermic sperm. J Proteome Res.2009;8(11):5382–5386.

125. Parte PP, Rao P, Redij S, et al.. Sperm phosphoproteome profilingby ultra performance liquid chromatography followed by dataindependent analysis (LC-MS(E)) reveals altered proteomic signa-tures in asthenozoospermia. J Proteomics. 2012;75(18):5861–5871.

•• This paper reveals the importance of differential protein phos-phorylation in asthenozoospermia.

126. Yunes R, Doncel GF, Acosta AA. Incidence of sperm-tail tyrosine phos-phorylation and hyperactivatedmotility in normozoospermic and asth-enozoospermic human sperm samples. Biocell. 2003;27(1):29–36.

127. Buffone MG, Calamera JC, Verstraeten SV, et al. Capacitation-asso-ciated protein tyrosine phosphorylation and membrane fluiditychanges are impaired in the spermatozoa of asthenozoospermicpatients. Reproduction. 2005;129(6):697–705.

128. Siva AB, Kameshwari DB, Singh V, et al.. Proteomics-based study onasthenozoospermia: differential expression of proteasome alphacomplex. Mol Hum Reprod. 2010;16(7):452–462.

•• A proteomic study of sperm proteasome expression in apathological condition such as asthenozoospermia.

129. Bhagwat S, Dalvi V, Chandrasekhar D, et al. Acetylated α-tubulin isreduced in individuals with poor sperm motility. Fertil Steril.2014;101(1):95–104.e3.

130. Rawe VY, Olmedo SB, Benmusa A, et al. Sperm ubiquitination inpatients with dysplasia of the fibrous sheath. Hum Reprod. 2002;17(8):2119–2127.

131. Johnson WL, Hunter AG, Jeyendran RS, et al. Association of asthe-nozoospermia with a deficiency of a seminal glycoprotein. Int JFertil. 1985;30(2):57–60.

132. Kratz EM, Kałuża A, Zimmer M, et al. The analysis of sialylation,N-glycan branching, and expression of O-glycans in seminal plasmaof infertile men. Dis Markers. 2015;2015:941871. doi:10.1155/2015/941871

133. Sepideh J, Reza SM, Mahdi AM, et al. Tyrosine phosphorylationpattern in sperm proteins isolated from normospermic and tera-tospermic men. J Reprod Infertil. 2009;10(3):185–191.

134. Silva JV, Freitas MJ, Correia BR, et al. Profiling signaling proteins inhuman spermatozoa: biomarker identification for sperm qualityevaluation. Fertil Steril. 2015;104(4):845–856.e8.

135. Sutovsky P. Sperm proteasome and fertilization. Reproduction.2011;142:1–14.

136. Buffone MG, Brugo-Olmedo S, Calamera JC, et al. Decreased pro-tein tyrosine phosphorylation and membrane fluidity in spermato-zoa from infertile men with varicocele. Mol Reprod Dev. 2006;73(12):1591–1599.

137. Corrales JJ, Burgo RM, Galindo P, et al. Abnormal expression of acidglycosidases in seminal plasma and spermatozoa from infertilemen with varicocele. Reproduction. 2002;123(3):411–417.

138. Hosseinpour E, Shahverdi A, Parivar K, et al. Sperm ubiquitinationand DNA fragmentation in men with occupational exposure andvaricocele. Andrologia. 2014;46(4):423–429.

139. Etzioni R, Urban N, Ramsey S, et al. The case for early detection. NatRev Cancer. 2003;3(4):243–252.

140. Sanchez V, Wistuba J, Mallidis C. Semen analysis: update on clinicalvalue, current needs and future perspectives. Reproduction.2013;146(6):R249-258.

141. Agarwal A, Durairajanayagam D, Halabi J, et al. Proteomics, oxidativestress and male infertility. Reprod Biomed Online. 2014;29(1):32–58.

142. Nixon B, Bromfield EG, Dun MD, et al. The role of the molecularchaperone heat shock protein A2 (HSPA2) in regulating humansperm-egg recognition. Asian J Androl. 2015;17:568–573.

143. Bromfield EG, Aitken RJ, Anderson AL, et al. The impact of oxidativestress on chaperone-mediated human sperm-egg interaction. HumReprod. 2015;30(11):2597–2613.

144. Fu Q, Schoenhoff FS, Savage WJ, et al. Multiplex assays for biomar-ker research and clinical application: translational science comingof age. PROTEOMICS Clin. Appl. 2010;4(3):271–284.

• A detailed review of the use of multiplex assays in biomarkerresearch and clinical proteomics.

145. Imai K, Koshiyama A, Nakata K. Towards clinical proteomics analy-sis. Biomed Chromatogr. 2011;25(1–2):59–64.

146. Margreitter C, Petrov D, Zagrovic B. Vienna-PTM web server: atoolkit for MD simulations of protein post-translational modifica-tions. Nucleic Acids Res. 2013;41(Web Server issue):W422–6.

• A toolkit for simulation of protein post-translational modifica-tion to further our understanding on protein modification andits consequences.

147. Bell AW, Deutsch EW, Au CE, et al.. HUPO Test Sample WorkingGroup. A HUPO test sample study reveals common problems inmass spectrometry-based proteomics. Nature Methods. 2009;6(6):423–430.

•• A crucial study on reproducibility of mass spectrometry-basedproteomic data.

148. Cooper TG, Yeung CH. Recent biochemical approaches to post-testicular, epididymal contraception. Hum Reprod Update. 1999;5(2):141–152.

149. Khole V. Epididymis as a target for contraception. Indian J Exp Biol.2003;41(7):764–772.

296 A. AGARWAL ET AL.

Dow

nloa

ded

by [

New

Yor

k U

nive

rsity

] at

12:

46 1

6 O

ctob

er 2

017