Abstract

BACKGROUND

Spermatozoa are highly differentiated, transcriptionally inert cells characterized by a compact nucleus with minimal cytoplasm. Nevertheless they contain a suite of unique RNAs that are delivered to oocyte upon fertilization. They are likely integrated as part of many different processes including genome recognition, consolidation-confrontation, early embryonic development and epigenetic transgenerational inherence. Spermatozoal RNAs also provide a window into the developmental history of each sperm thereby providing biomarkers of fertility and pregnancy outcome which are being intensely studied.

METHODS

Literature searches were performed to review the majority of spermatozoal RNA studies that described potential functions and clinical applications with emphasis on Next-Generation Sequencing. Human, mouse, bovine and stallion were compared as their distribution and composition of spermatozoal RNAs, using these techniques, have been described.

RESULTS

Comparisons highlighted the complexity of the population of spermatozoal RNAs that comprises rRNA, mRNA and both large and small non-coding RNAs. RNA-seq analysis has revealed that only a fraction of the larger RNAs retain their structure. While rRNAs are the most abundant and are highly fragmented, ensuring a translationally quiescent state, other RNAs including some mRNAs retain their functional potential, thereby increasing the opportunity for regulatory interactions. Abundant small non-coding RNAs retained in spermatozoa include miRNAs and piRNAs. Some, like miR-34c are essential to the early embryo development required for the first cellular division. Others like the piRNAs are likely part of the genomic dance of confrontation and consolidation. Other non-coding spermatozoal RNAs include transposable elements, annotated lnc-RNAs, intronic retained elements, exonic elements, chromatin-associated RNAs, small-nuclear ILF3/NF30 associated RNAs, quiescent RNAs, mse-tRNAs and YRNAs. Some non-coding RNAs are known to act as epigenetic modifiers, inducing histone modifications and DNA methylation, perhaps playing a role in transgenerational epigenetic inherence. Transcript profiling holds considerable potential for the discovery of fertility biomarkers for both agriculture and human medicine. Comparing the differential RNA profiles of infertile and fertile individuals as well as assessing species similarities, should resolve the regulatory pathways contributing to male factor infertility.

CONCLUSIONS

Dad delivers a complex population of RNAs to the oocyte at fertilization that likely influences fertilization, embryo development, the phenotype of the offspring and possibly future generations. Development is continuing on the use of spermatozoal RNA profiles as phenotypic markers of male factor status for use as clinical diagnostics of the father's contribution to the birth of a healthy child.

Introduction

Spermatogenesis is a highly regulated transcriptional, translational and posttranslational process. Transcription continues through the initial stages of spermiogenesis until development of the round spermatids. Those transcripts that are required to complete the transition to the spermatozoa are protected and maintained as ribonucleoproteins (RNPs). During this time, the majority of the cytoplasm with its RNA component is depleted as a cytoplasmic droplet, residual body (reviewed in Cooper, 2005) and phagocytosed by the Sertoli cells. At this point, nuclear compaction can be cytologically observed as the majority of sperm histones are replaced by protamines and the sperm nucleus assumes a highly condensed structure. This yields a cell which is transcriptionally inert and devoid of translational activity as ensured by the absence of intact rRNAs (Betlach and Erickson, 1976; Johnson et al., 2011b). The transcripts that do remain within the spermatozoa provide a select source of both coding and non-coding RNAs that include both fragmented and preferentially non-degraded mRNAs, si (small interfering), mi (micro), pi (Piwi-interacting) and lnc (long non-coding)-RNAs.

The very existence of spermatozoal RNAs was originally questioned based on the assumption that since transcription ceases in the round spermatid stage, with the cytoplasm destined to be expunged and thus void of the components necessary for translational activity, any remaining male haploid RNA would be inconsequential. This view was supported by the observed heterogeneity of the ejaculate, the presence of somatic cell contaminants which accounted for the majority of large RNAs in most samples and the absence of intact ribosomal RNAs. These caveats partly reflected the inadequacy of the methods that were available to purify spermatozoa and to detect low abundance RNAs (reviewed in Krawetz, 2005). The controversy was resolved when several laboratories independently identified specific sperm RNAs in plants (Rejon et al., 1988) and in mammals, including rat (Pessot et al., 1989), mouse (Wykes et al., 2000) and human (Kumar et al., 1993; Miller et al., 1994; Wykes et al., 1997) using RT–PCR and in situ hybridization.

To date the population of spermatozoal transcripts from the human, are the best characterized amongst all mammals. The RNA profile of human spermatozoa was initially attempted using a cDNA cloning and sequencing strategy (Miller et al., 1999) that was followed by select RT–PCR (Lambard et al., 2004; Wang et al., 2004). Both of these methods were able to survey only a small fraction of all potential transcripts. The first general spermatozoal RNA profiles obtained using microarrays suggested that human spermatozoa contain ∼3000–7000 different coding transcripts (Ostermeier et al., 2002). This was subsequently extended to the clinic with the assessment of specific transcripts in cases of asthenozoospermia (Jodar et al., 2012), teratozoospermia (Platts et al., 2007), oligozoospermia (Montjean et al., 2012) and idiopathic infertile males (Garrido et al., 2009). Their potential as biomarkers of fertility was highlighted. Transcript profiling of coding RNAs using microarrays in conjunction with RT–PCR has now broadly defined the abundance of known sperm transcripts in other mammals (Gilbert et al., 2007; Bissonnette et al., 2009; Yang et al., 2010) and non-mammalian species like plants (Borges et al., 2008) and Drosophila Melanogaster (Fischer et al., 2012).

In comparison to the aforementioned, RNA-seq has provided a much more complete picture of the population of human sperm transcripts, allowing for the identification, quantification and characterization of both known and previously unknown RNAs (Krawetz et al., 2011; Sendler et al., 2013). These studies highlighted the selective retention of a cadre of both coding RNAs and small non-coding RNAs in all individuals studied. Recently others have begun employing RNA-seq to examine the distribution of sperm RNAs in bovine (Card et al., 2013) and stallion (Das et al., 2013) and the small RNA population of mouse (Kawano et al., 2012; Peng et al., 2012).

The overall functional significance of many spermatozoal RNAs remains to be understood and their individual importance remains to be elucidated. Using the zona-free hamster oocyte/human sperm penetration assay, it has been established that sperm-specific transcripts (not present in the unfertilized oocyte) are transmitted to the oocyte upon fertilization (Ostermeier et al., 2004). They can also be translated into a functional protein as shown by the injection of the sperm borne PLCζ (1-phosphatidylinositol 4,5-bisphosphate phosphodiesterase zeta) transcript into the mouse oocyte, yielding a functional calcium oscillator (Sone et al., 2005). The distinctive landscape of non-coding RNAs that appears during the final stages of sperm maturation also strongly hints at their potential role in early post-fertilization and embryo development (Ostermeier et al., 2004; Liu et al., 2012). This has now been extended to the position that spermatozoal RNAs may epigenetically and transgenerationally affect phenotype (reviewed in Rando, 2012). These avenues remain to be explored.

A substantial number of spermatozoal transcripts appear compromised (Sendler et al., 2013), suggesting that they may simply be remnants echoing prior roles (Ostermeier et al., 2002). Even if only existing in mature sperm in this form, comparison of the differential transcript profiles between fertile and infertile patients has shown their utility as markers of fertility (reviewed in Waclawska and Kurpisz, 2012).

The primary focus of this review is to examine what a spermatozoal transcript profile may reveal with regard to the integrity of the spermatogenic pathway, characteristics of the mature sperm and their potential epigenetic and post-fertilization developmental functions. The potential use of spermatozoal RNAs as biomarkers impacting human clinical diagnosis and agriculture will also be discussed.

Methods

Previously published human RNA-seq results (Krawetz et al., 2011; Sendler et al., 2013) and recent results from RNA sequencing of four representative sperm and two testes samples from a larger dataset were utilized. This set of sperm samples (D1–D4) were from four of the National Institutes of Health (NIH)/Eunice Kennedy Shriver National Institute of Child Health and Human Development (NICHD), Reproductive Medicine Network for Assessment of Multiple Intrauterine Gestations from Ovarian Stimulation dataset, whose partner withdrew from the study. Sperm and testes libraries were prepared for sequencing using the SEQR RNA Amplification kit (Sigma-Aldrich) and ENCORE® NGS Library System (NUGEN Technologies, Inc.). Amplification begins using 2 ng of total RNA for both sperm and testes samples. Library construction continues using 200 ng of amplified material. All libraries were paired-end sequenced using Illumina Hi-Seq 2500 and aligned using Novoalign (V2.08.01, NovoCraft Technologies Sdn Bhd), with distribution of reads shown as UCSC genome browser tracks, similar to that described (Sendler et al., 2013). The main figures in this review show sperm transcript characteristics of interest as portrayed by a single sample, while the supplementary figures show similarities among the four sequenced donors for each of these characteristics.

Genomic distribution (exonic, intronic and intergenic) of the large sperm RNA fraction was obtained from total sperm sample AS062 (GEO accession GSM721696) using Genomatix RegionMiner (Genomatix, v 3.0426). RT–PCR of specific intronic RNAs was performed using primer pairs that spanned the complete intron length in both sense and anti-sense orientations and were separately performed on both sperm and testes RNA in order to determine both the direction and relative abundance of these elements in sperm. Distribution of snc-RNA reads across transposable elements LINE1 (Genbank accession M80343), variant forms of ERVL-MaLR (RepBase 16.09) and tRNAs were obtained from results of small RNA sequencing of sperm sample AS062 (GSM530234). The NCBI HomoloGene database (Coordinators, 2013) was mined to identify homologous human, stallion and bovine sequences. Gene abundance from stallion RNA-seq was based on the distribution the most abundant exon of mapped RNA sequence tags (Das et al., 2013). Literature was mined through i-HOP, PubMed, GoPubMed and GEPS (Dietze et al., 2009; Epple and Sherf, 2009; Leitner et al., 2009; Reavie, 2009).

Spermatozoal RNAs-characterization and potential roles

The most extensive sperm transcript profiling is available from human. RNA-seq has been performed for both small and large RNA fractions, with the latter examined as both total RNA and separate A+ and A fractions. The landscape of sperm RNAs evident from multiple normal individual samples has revealed a wealth of different classes of coding and non-coding transcripts (Fig. 1). Many relatively abundant RNAs that appear in all normal sperm samples are unannotated or of unknown function and appear uniquely in sperm, emphasizing the high level of complexity of the population of spermatozoal RNAs.

Figure 1

Composition of spermatozoal RNAs. The distribution of the various classes of RNAs as determined by RNA-seq is shown. The most abundant class is ribosomal RNAs followed by mitochondrial RNA (mitoRNAs), annotated coding transcripts, small non-coding RNAs (snc-RNAs), intronic retained elements, lnc-RNAs and Transcribed regions of Unknown Coding Potential (TUCP), short expressed regions, transposable elements and annotated non-coding RNAs, including snars, sno, pri-mir and RNU.

Considerations

A single spermatozoon contains ∼50 fg of long RNA (>200 nt) and 0.3 fg of small non-coding (snc) RNA (<200 nt; Goodrich et al., 2013). This amount is ∼200 times less than the quantity of RNA found in other cells (10 pg of long RNA and 1–3 pg of snc-RNAs), making somatic cell removal essential to resolve the unique pool of sperm transcripts. The absence of intact 28S and 18S sperm rRNA is often used to confirm the absence of somatic contamination (Johnson et al., 2011b; Goodrich et al., 2013). This was previously thought to be reflective of complete removal of rRNA with consequent enrichment of mRNA, but in fact was later resolved as the selective fragmentation of the majority of sperm rRNAs (Ostermeier et al., 2002; Johnson et al., 2011b). To overcome these challenges the protocol to isolate spermatozoal RNA has undergone several revisions (Goodrich et al., 2013).

Resolution of the RNA population has been optimized with the use of Next Generation Sequencing (NGS) strategies. Typical RNA-seq library construction uses poly(A+) selection to provide an enriched population of mRNAs excluding the otherwise overwhelming contribution of ribosomal and mitochondrial RNAs. While this provides an effective strategy to enrich for coding transcripts, it will exclude RNAs with short poly(A) tails and those that are not polyadenylated. In contrast, total RNA libraries are not subject to this limitation. This is an important consideration when characterizing sperm RNAs that have been observed to possess a large population of non-coding RNAs. However, sequencing the total population of RNAs comes at the cost of the increased representation of ribosomal and mitochondrial RNAs. Specific fractions of snc-RNAs (<200 nt) are typically lost during library construction but can be recovered and sequenced with modified protocols and size selection (Krawetz et al., 2011).

Coding RNAs

Both RNA-seq analysis in the human (Sendler et al., 2013) and RT–PCR of select bovine transcripts (Gilbert et al., 2007) suggests that the majority of coding RNA observed in sperm exists in a fragmented, or at the very least, atypical state. In contrast to equivalently selected testes RNA, RNA-seq profiles of the majority of poly(A+) selected sperm transcripts exhibit a marked 3′ bias. As illustrated in Fig. 2, this characteristic is indicative of transcript fragmentation in spermatozoal RNA. Ontological analysis of the relatively minor fraction of intact coding transcripts retained in sperm shows an enrichment of genes associated with male infertility, fertilization and early embryo development (Sendler et al., 2013). This is strongly suggestive of a functional role for these preferentially retained transcripts during the final stages of spermatogenesis or upon delivery. For example, INTS1 (Integrator complex subunit 1), involved in the transcription and processing of small-nuclear RNAs (snRNA) U1 and U2, is retained and appears by microarray analysis to increase after fertilization prior to zygotic genome activation (Vassena et al., 2011). INTS1 knockouts are embryonic lethal at the blastocyst stage (Hata and Nakayama, 2007). This is congruent with a potential role of this complex in the first steps of embryogenesis which could reflect the paternal contribution.

Figure 2

The degree of transcript fragmentation can influence the sequencing profile after Poly(A+) selection. (A) Intact RNA shows minimal 3′ bias, even with poly(A+) selection. However, transcripts which are biologically fragmented (i.e. prior to the typical fragmentation step of most RNA-seq protocols) show significant 3′ end profile bias as selection preferentially retains the 3′ poly(A+) containing ends. (B) For example, SPACA4, exhibits fairly even coverage across transcript length in testes, and marked 3′ bias in sperm.

As in the above, RNA-seq also affords the ability to identify novel transcripts or isoforms. The production of transcript variants through the use of alternative promoters and splicing has been described in testes (Freiman, 2009). Interestingly, mature sperm display isoforms that are distinct in a variety of ways from those found in whole testes, indicating that these modifications arise only in the final transcriptionally active stages of spermatogenesis. This includes the sperm-specific isoform of PKM2 (Pyruvate kinase isozymes M1/M2), a key enzyme regulating glucose metabolism (Sendler et al., 2013). Further, approximately one quarter of the sperm transcripts show alternative sites of polyadenylation (APA), which maintain the integrity of the coding region, but exhibit an abbreviated 3′ untranslated region (UTR) (Fig. 3). This trait is common in testes (Liu et al., 2007) and may serve to modulate transcript stability, localization and/or transport. Additionally, this modification may impact translation by affecting the ability of different regulatory proteins and miRNAs to bind to the alternative UTR (Di Giammartino et al., 2011). It has recently been reported that Bromodomain testis-specific protein (BRDT) actively modulates APA in testes (Berkovits et al., 2012). Male mice lacking the first bromodomain of BRDT are infertile (Shang et al., 2007) and spermatozoal transcripts from such knockouts possess longer 3′UTRs (Berkovits et al., 2012). This likely emphasizes the critical nature of APA observed in human sperm. Lastly, sperm RNA-seq has identified many examples of abundant predicted transcripts (such as ORFs) that are not observed in somatic cells and are of low abundance in testes. Together, variations in expression and form of coding RNAs found in sperm likely have a significant impact on both the regulation and function of this class of RNAs.

Figure 3

Alternative polyadenlation of sperm transcripts. GIGYF2 encodes a protein that interacts with GRB10 and may be involved in the regulation of tyrosine kinase receptor signaling. The 3′UTR region of GIGYF2 gene is highlighted (upper panel). RNA-seq (lower panel) of this specific region exhibits a truncated 3′UTR in sperm (green). This contrasts with coverage extending over most of the UTR observed in testes (black). (See Supplementary data, Fig. S1 for more details.)

Small non-coding RNAs

It has been proposed that the germline genome is protected through paternal small non-coding miRNAs, siRNAs, piRNAs, qRNAs and repeat associated RNA mechanisms (reviewed in O'Donnell and Boeke, 2007; Bourc'his and Voinnet, 2010; Krawetz et al., 2011; Siomi et al., 2011). For function, the parental RNAs must be of sufficient quantity and quality to interact with their target for successful embryo development (Bourc'his and Voinnet, 2010). Recent sequencing of the small (18–24 nt) RNA population from multiple normal human donors has also shed light on the complexity of the snc-RNA population present in spermatozoa. The majority of sperm snc-RNAs correspond to four major classes: repetitive elements, transcription start sites (TSS)/promoter associated, piRNAs, and miRNAs, with other classes such as snRNAs, snoRNAs, mse-tsRNA and YRNAs representing a relatively minor portion (Krawetz et al., 2011). Additional snc-RNA sequencing reads correspond to unannotated regions of the genome and portions of coding and non-coding transcripts. Whether these short fragments serve a particular role, e.g. regulating their longer precursor elements, or are merely end-points of fragmentation is not yet known.

miRNAs

The most well-characterized non-coding sperm RNAs are miRNAs, which have been shown to modulate various stages of spermatogenesis (reviewed in Moazed, 2009). Along with siRNAs, these RNAs typically function to regulate expression by inhibiting or activating translation or targeting mRNAs for degradation usually by binding to a 3′UTR target sequence (reviewed in Gangaraju and Lin, 2009). They are typically transcribed by polymerase II as larger precursors that are then processed to an intermediate form by DROSHA (Ribonuclease 3) and DGCR8 (DiGeorge syndrome critical region 8). These precursors are subsequently transported to the cytoplasm and further matured by DICER, an RNase III endonuclease, to their mature 20–24 nt functional form. They are then incorporated into an Argonaute containing RNP forming RISC, RNA-induced silencing complex. Spermatogenic-specific DICER or DROSHA knockouts arrest spermatogenesis (Hayashi et al., 2008; Korhonen et al., 2011; Wu et al., 2012), confirming their essential role.

While many miRNAs are conserved among different species, some are species-specific (Curry et al., 2009; Krawetz et al., 2011; Govindaraju et al., 2012; Peng et al., 2012; Das et al., 2013). The majority of mature spermatozoal miRNAs are also observed in testes (Landgraf et al., 2007), but most of their computationally predicted 3′UTR targets are absent in mature sperm (Krawetz et al., 2011). Recent studies suggest that some miRNAs act as transcriptional regulators by targeting other regions, e.g. promoters (Kim et al., 2008; Place et al., 2008). Perhaps in the transcriptionally quiescent sperm, they provide a signal for early embryonic histone replacement (Johnson et al., 2011a) or transcriptionally poise the genome for early embryonic expression or affect epigenetic modification (Khraiwesh et al., 2010). Support for this notion has been gained from the observation that more than 10% of all snc-RNAs map to histone-enriched TSS and promoters. These novel RNAs, termed quiescent RNAs (qRNAs), are similar to tiny RNAs (tiRNAs). They are associated with the TSS region but not enriched in GC regions or correlated with histone modifications (Krawetz et al., 2011). The tiRNAs derived from regions adjacent to TSS may indirectly modulate local chromatin states through other binding factors (Taft et al., 2011). However, the function of qRNAs remains to be established (Krawetz et al., 2011).

The most abundant sperm miRNA in the human is miR-34c (Krawetz et al., 2011). It has also been identified in stallion and mouse (Peng et al., 2012; Das et al., 2013), and has been shown to be essential for the first cleavage division in mouse zygotes (Liu et al., 2012). Except for miR-34c-5p, where we have a glimpse, their mechanism of action and functional role in spermiogenesis and/or fertility remain to be fully delineated (Curry et al., 2011; Krawetz et al., 2011; Govindaraju et al., 2012; Das et al., 2013). For example, in mouse testes, miR-34c expression is p53 independent (Bouhallier et al., 2010), whereas miR-34c targets p53 in cancer cells (Corney et al., 2007). This is somewhat in line with their ability to influence growth status during periods of rapid growth like oncogenesis (reviewed in Shivdasani, 2006; Croce, 2009; Luningschror et al., 2013). Spermatozoa also contain several intact miRNA precursors (pri-miRNAs, 100–150 nt). Since the zygote has the capacity to process immature miRNAs (Liu et al., 2012), the potential role of the pri-miRNAs requires consideration. For example, pri-miRNA-181c is the most abundant immature miRNA in human spermatozoa. Predicted targets of this miRNA include those critical to early embryonic development and globally decrease at the 4–8 cell stage of human embryo development (Vassena et al., 2011; Sendler et al., 2013). One specific target of miR-181c is CARM1 (Coactivator-Associated aRginine Methyltransferase 1), an embryonic stem cell pluripotency factor. CARM1 directly catalyzes the methylation of H3 arginine in the promoters of POU5F1 (POU domain, class 5, transcription factor 1) and SOX2 (Transcription factor SOX-2). This forms an active chromatin mark coinciding with induction (Xu et al., 2013). At the 2-cell stage overexpression of CARM1 in one of the blastomeres predisposes its derivatives to contribute to the pluripotent cells of inner cell mass (Torres-Padilla et al., 2007). It is tempting to speculate that through the delivery by sperm miR-34c and pri-miR-181c, the division and partitioning of the targeted CARM1 are, respectively, ensured, thereby decreasing some pluripotency factors in one blastomere while pushing the other towards the trophoectoderm lineage.

piRNAs

Piwi-interacting RNA (piRNAs) are abundant in the mammalian male germline (reviewed in Girard et al., 2006) and their presence in spermatozoa has been confirmed in several species (Krawetz et al., 2011; Kawano et al., 2012; Peng et al., 2012). They are typically organized in the genome as clusters that range up to 100 kb in size. piRNAs precursors are processed to their 23–32 nt mature form by a PIWI protein-dependent mechanism (reviewed in Ishizu et al., 2012). Although not clearly articulated, several functions have been proposed for this class of transcripts. These include regulation of RNA stability and epigenetic states as well as protecting the germline genome from transposition (reviewed in Aravin and Hannon, 2008; Gangaraju and Lin, 2009). During spermatogenesis, the activation of mobile transposable elements is suppressed by piRNAs. The absence of these regulatory RNAs can induce spermatogenic arrest (Kuramochi-Miyagawa et al., 2004; Carmell et al., 2007). The piRNAs may act in a similar protective manner during early embryo development as the genome undergoes extensive demethylation and remethylation. They could protect genome integrity by binding to DNA and thus preventing the action of various classes of repetitive and transposable elements like SINE, LINE, MER and LTR at specific stages of embryogenesis (Krawetz et al., 2011).

Potentially novel classes of snc-RNAs, sperm RNAs (spR)-12 and -13, were recently identified in mouse spermatozoa. They are ∼20 nt in length and are likely derived from additional processing of mature piRNAs (Kawano et al., 2012). This has defined yet another snc-RNA biogenesis pathway. These abundant spRs are maintained post-fertilization until the blastocyst stage, suggestive of their potential role in early embryo development perhaps to ensure genome integrity at this critical stage.

Confrontation and consolidation

The classes of spermatozoal RNAs described above may play an integral role in the confrontation and consolidation mechanism that has been described in plants and animals (Bourc'his and Voinnet, 2010; Goring and Indriolo, 2010; Krawetz et al., 2011; Miller and Iles, 2013). When the sperm and oocyte meet, it is necessary to ensure the compatibility of the genomic contributions of each parent to ensure that their combination will be conducive to embryonic development. During confrontation, the pairing of paternal RNAs, such as repeat associated spermatozoal RNAs, with complementary maternal repetitive elements, may activate or suppress their partner. Once compatibility between gametes is assured, the RNA-based information could then be transferred to a chromatized state, i.e. consolidation, likely by modifying the epigenome. Interestingly, snc-RNAs have been associated with heterochromatization, perhaps consolidation (reviewed in Lippman et al., 2004; Lippman and Martienssen, 2004).

Examples of this surveillance pathway and its consequence are apparent when one considers the various outcomes that can occur when two different species or breeds are crossed to produce hybrid offspring. The potential consequences include failure at fertilization, inappropriate embryo development or compromised fertility of the offspring. The latter outcome is best characterized by the mule (mare and donkey hybrid) which is infertile/sterile (Short, 1975). Similarly, blastomere formation is halted at the 8-cell stage when hybrid embryos are created by in vitro fertilization of a water buffalo (Bubalus bubalis) oocyte with bovine (Bos taurus) spermatozoa. This parallels a failure to undergo zygotic genome activation leading to developmental arrest (Patil and Totey, 2003). Perhaps this reflects an incompatible paternal contribution in which the specific mechanism necessary to activate or suppress elements necessary for embryo development is absent.

Transposable elements

A large proportion of human spermatozoal snc-RNAs map to repetitive elements. The most abundant repeat classes represented in mature spermatozoa are the various members of the, LTR, SINE/ALU and LINE families of transposable elements (Krawetz et al., 2011). The role that transposable elements may play in the germline and early embryo remains controversial (Beraldi et al., 2006; Georgiou et al., 2009; van der Heijden and Bortvin, 2009). For example, LINE1 has a dynamic activity during early embryo development. Interrupting this activity results in embryonic arrest at the 2- or 4-cell stage (Beraldi et al., 2006). This may reflect the disruption of LINE1-associated reverse transcriptase (Pittoggi et al., 2003; reviewed in Spadafora, 2008). At this stage, LINE1 transcription is thought to be mediated by polypurine enriched LINE1 RNA fragments. These fragments form a triple helix within several regions of LINE1 potentially serving as a scaffolding that alters the association of chromatin modifiers and the transcriptional machinery (Fadloun et al., 2013) thereby promoting their own expression. Perhaps the large number of LINE1 fragments observed in and delivered by sperm activates this feedback loop. The distribution of short reads derived from RNA-seq of human spermatozoal snc-RNAs over the length of LINE1 exhibits very high enrichment of specific fragments, with some being homopurine polymers or near polypurine in sequence (Fig. 4). A similar distribution of highly enriched fragments is observed for other transposable elements in sperm such as ERVL-MaLR, for which early embryo function is also noted (Kigami et al., 2003; Inoue et al., 2012) and for which sperm-delivered fragments may play a similar activating role. It remains to be determined whether the transcribed transposable elements found in sperm such as SINE/ALU, which are complementary to coding and/or regulatory regions, may modulate host gene expression in early embryogenesis, since such behavior has been observed in other developmental processes (Polak and Domany, 2006). It appears though, that far from being ‘junk’ or simply an unintended consequence of global genomic demethylation, perhaps the abundant repeat associated sperm RNAs modulate other regulatory elements in the early embryonic stages of development.

Figure 4

Alignment of short RNAs (18–24 nt) from human sperm sample AS062 to the LINE1 repeat. Some LINE1 elements in the genome act as active transposable elements encoding both an ORF2 endonuclease and reverse transcriptase as well as the RNA-binding protein p40 encoded by ORF1. Specific LINE1 fragments are abundant in the small RNA fraction (blue peaks). Some of these fragments are purine rich sequences (red boxes), which may, through the formation of triplex structures, promote expression of complete LINE1 elements in the fertilized oocyte.

‘Other’ sperm RNAs

The distribution of poly(A+) selected sperm and testes sequencing reads demonstrates that while ∼10% of testes reads correspond to intergenic or intronic regions, more than two-thirds of sperm reads align to regions of unknown annotation or function (Sendler et al., 2013). Many of these unidentified yet prominent transcripts appear in all sperm samples from normal individuals and show little or no expression in other cell types. Although the majority of these RNAs have no known function, their abundance suggests that they may play a significant functional role. Whether this occurs during the final stages of sperm maturation, at delivery to the oocyte or during early preimplantation development remains to be determined. The observation that these transcripts correspond to genomic regions that retain histones, specifically H3K4me3, a histone modification correlated with transcriptional activity, suggests that sperm chromatin is uniquely structured to facilitate the transcription of these RNAs and that they are not artifacts (Hammoud et al., 2009; Sendler et al., 2013). Some classes of these elements with specific characteristics are described below.

Intronic retained elements

Non-coding RNAs contained within introns of coding mRNAs have been described in other systems (Hill et al., 2006). Both precursor miRNAs and snoRNAs often originate from these regions (Kiss et al., 1996; Lin et al., 2006; Li et al., 2007b). At least 200 distinctive non-coding sperm transcripts appear to be full-length introns specifically retained in sperm (Sendler et al., 2013). The mechanism by which they escape degradation after splicing remains to be defined. Interestingly, the corresponding mRNAs are often abundant in testes, while in sperm these coding segments show a marked reduced presence. Genes from which these intronic elements are derived do not classify into a distinct ontological category nor do they correlate with a specific pattern of early embryonic expression (Vassena et al., 2011). No evidence has been found to show that the intronic elements observed in sperm comply with the computationally predicted models for precursor elements of either sno- or mi-RNAs. Figure 5 shows an example of the sperm transcript DNAH1 (Dynein heavy chain 1, axonemal) in which several introns are retained. Several abundant intron spanning RNAs are apparent. Comparison of relative abundance of these intronic elements in sperm (A+) and (A) fractions shows that the vast majority are not polyadenylated. Examination by RT–PCR of three such elements found within transcripts, TRIM66 (Tripartite motif-containing protein 66), KAT8 (Histone acetyltransferase KAT8) and QRICH1 (Glutamine-rich protein 1), confirmed that they were transcribed in the same orientation as the transcript in which they are embedded. Interestingly, they are present in much higher levels in sperm than in testes (unpublished data). These observations suggest that they are retained in mature sperm perhaps as part of a separate regulatory mechanism. The recent suggestion that some intronic ncRNAs are specifically regulated by a drop in temperature (Heo and Sung, 2011) to target their host transcripts for rapid degradation is intriguing. Spermatogenesis is temperature-sensitive and this may act as a physiological monitor.

Figure 5

Sperm intronic retained elements. The structure of DNAH1 is shown in the upper panel. The sequence reads obtained from sperm (green) and testes (black) RNA-seq within the highlighted region are shown (lower panel). The levels of specific intronic sperm RNAs are enhanced, while the coding regions of this transcript are absent in sperm. In equivalently sequenced testes samples, these intronic regions are underrepresented and resemble levels observed across the complete transcript (note y-axis). (See Supplementary data, Fig. S2 for more details.)

Long non-coding RNAs

Long non-coding RNAs (lnc-RNAs) range in size from ∼200 to 10,000 nt and are scattered throughout the genome. They are generally classified as a function of their relative position to protein coding genes (reviewed in Ponting et al., 2009). This includes intronic or intergenic regions where strand orientation cannot be directly determined, exonic regions primarily derived from the reverse strand, or from pseudogenes and retrotransposons. Spermatogenesis is in part regulated through the action of lnc-RNAs (Nolasco et al., 2012) some of which are certain to be antisense. Specifically, the abundance of antisense transcripts in testes may add to the mechanisms strictly regulating expression and function during spermatogenesis (Lee et al., 2009a). Lnc-RNA mechanisms have been described to operate in somatic cells at both the transcriptional or posttranscriptional levels (reviewed in Mercer et al., 2009; Lee, 2012; Rinn and Chang, 2012). At the transcriptional level this is accomplished by promoting specific histone modifications. For example, HOTAIR (HOX transcript antisense RNA) can modulate transcription through chromatin structure by recruiting PRC2 (Polychrome Recruiting Complex) to the HoxD locus thereby repressively marking histone H3 (Tsai et al., 2010). Transcription can also be modulated through the interaction of an lnc-RNA with an associated promoter region as exemplified by DHFR (Dihydrofolate reductase). Transcription of this gene by an alternative promoter results in a regulatory transcript that targets the usual promoter via triplex formation repressing the expression of DHFR (Martianov et al., 2007). Lnc-RNAs can also function post-transcriptionally during splicing (reviewed in Yoon et al., 2012a). For example, MALAT1 (Metastasis-Associated Lung Adenocarcinoma Transcript 1) regulates the alternative splicing of a subset of transcripts through its interaction with splicing factors (Tripathi et al., 2010). Translation of RNAs can also be modulated through the interaction of lnc-RNAs with specific repressors (Yoon et al., 2012b) or by general interference with the translation initiation complex (reviewed in Kindler et al., 2005). Finally, a similar regulatory effect can be achieved by modifying RNA stability. This can be affected through sense–antisense pairing that protects the target from miRNA-mediated degradation (Faghihi et al., 2008).

A number of predicted lnc-RNAs (Trapnell et al., 2010; Cabili et al., 2011) correspond to both abundant poly (A+) and poly (A) sperm transcripts. In some cases, the sperm lnc-RNAs appear to represent different isoforms than the predicted forms. This is not unexpected given that the list of lnc-RNAs was primarily derived from expression in somatic cells. In many cases, the sperm lnc-RNAs appear more abundant in sperm than in testes (Fig. 6). The function of such lnc-RNAs in sperm maturation, fertilization and early embryo development remains to be explored.

Figure 6

Unique sperm lnc-RNAs isoforms. A 30 kb region of chromosome 3 containing a series of putative lnc-RNAs as identified by the Human Body Map lincRNA UCSC track (Trapnell et al., 2010; Cabili et al., 2011) is shown in upper panel. Although low- level expression of a number of identified lnc-RNAs is evident across this region in testes, a single highly expressed two exon RNA is observed in sperm (lower panel). Many junction reads, as measured by RUM (Grant et al., 2011) (box), confirm that these two exons are part of a single spliced transcript, which was not previously identified as a unique lnc-RNA isoform. (See Supplementary data, Fig. S3 for more details.)

A specific class of lnc-RNAs overlaps coding transcripts and they are derived from the reverse strand (NAT: natural antisense transcripts). Several NAT have been described in mature spermatozoa (Ostermeier et al., 2005; Sendler et al., 2013). Roles in gene silencing, selective transcript editing, promoter inactivation and epigenetic modifications of the genome have been revealed for such RNAs (Lavorgna et al., 2004; Lapidot and Pilpel, 2006; Faghihi and Wahlestedt, 2009; Werner et al., 2009; Werner and Swan, 2010). For example, independent regulation of sense–antisense pairs of some nc-RNAs at specific stages of cellular development (Cawley et al., 2004; Katayama et al., 2005; Werner et al., 2007) results in the rapid processing of the longer transcripts into short ∼23 nt fragments (Borsani et al., 2005; Carlile et al., 2008). There are several examples of abundant of 100–300 nt sperm RNAs that overlap either the coding or UTR portion of an otherwise low-expressed or absent transcript. A striking example is the antisense transcript that overlaps ARFGEF1 (Brefeldin A-inhibited guanine nucleotide-exchange protein 1). This transcript is an ADP-ribosylation factor, required for maintenance of Golgi structure and function (Manolea et al., 2008). The corresponding fragment is abundant in sperm and corresponds to the middle of the 5′UTR. The full-length transcript is present in testes but virtually absent in sperm (Fig. 7). It would appear likely that this may be one example of a processing mechanism in sperm perhaps targeting specific transcripts for rapid degradation and which may be achieved throughout spermatogenesis through a variety of means. In addition to the possible action of NATs in the physiology of spermatozoa, some NATs present in mature spermatozoa overlap genes involved in early embryo development (Ostermeier et al., 2005) suggesting that such antisense RNAs may also have a role during fertilization and in the first steps of embryogenesis (Li et al., 2002).

Figure 7

Exonic sperm element. An overview of the structure of ARFGEF1 is provided in the upper panel. In testes, significant coverage of the complete transcript is observed (Supplementary data, Fig. S4). In contrast, sperm show virtually no transcript within the length of the coding region. A ∼100 nt sperm-specific element is observed however within the 5′UTR (highlighted region), but is virtually absent in equivalently prepared testes samples. This unique sperm element may serve to accelerate degradation of its containing transcript. (See Supplementary data, Fig. S4 for more details.)

Specific classes of lnc-RNAs

A number of elements from previously identified nc-RNA classes are abundant in the longer fraction of sperm RNA. These include Chromatin-associated-(CAR) and some small-nuclear ILF3/NF30 associated-(snaR) RNAs. CARs are found associated with chromatin and may act in cis or in trans to influence genomic architecture or regulate gene expression (Rodriguez-Campos and Azorin, 2007; Mondal et al., 2010). Three intronic and three intergenic regions in sperm, which show substantial sequencing coverage, overlapped with CARs recently identified in human fibroblast (HF) cells (Mondal et al., 2010). Perhaps a significant number of the unidentified lone elements in sperm are CARs and serve to aid the unique packaging requirements of the paternal genome. Several of the small NF90-associated RNAs (snaR) including snaR-G1 are also abundant. While their role remains uncertain, it is significant that snaR-G1 resides within the promoter region of the embryonic developmentally important human chorionic gonadotrophin (hCG1) (Parrott and Mathews, 2007). The level of this sperm snaR is elevated relative to that observed in testes, which is already at a level that is ∼100 times that of somatic tissues. This is certainly suggestive of a prominent functional role for these transcripts in mature spermatozoa.

tRNA-derived snc-RNAs and YRNAs

The majority of mouse and human mse-tsRNAs (mature-sperm-enriched tRNA-derived small RNAs) correspond to specific cleavage products (Krawetz et al., 2011; Peng et al., 2012). They typically represent 5′ end fragments between the D-loop and anticodon loop (Peng et al., 2012). However, as illustrated in Fig. 8, unlike mouse, human-specific mse-tsRNAs are also derived from the 3′ region (Krawetz et al., 2011; Peng et al., 2012). While initial inspection suggested that the fragmentation of rRNAs and tRNAs is to ensure translational silence (Johnson et al., 2011b), perhaps some of the mse-tsRNAs are functional. For example, their action as a stress responder appears conserved as far back as bacteria. In response to stress, an abundant 5′ end fragment of Val-tRNA in Haloferaz volcanii specifically targets and inhibits the translational machinery (Gebetsberger et al., 2012). This effect can be mimicked by the transfection of natural and synthetic tRNAs fragments (Ivanov et al., 2011). Knock-down of trF-1001, (a 3′ end tRNA-derived fragment from the Ser-TGA tRNA precursor) inhibits cancer cell proliferation (Lee et al., 2009b), suggesting a role in maintaining proliferation. Perhaps mse-tRNAs acts in a similar manner upon delivery to the oocyte (Peng et al., 2012).

Figure 8

Enrichment of mse-tRNA fragments in human sperm samples. Short read alignment to specific tRNAs is highlighted in the dark green box. Significant enrichment of fragments corresponding to particular regions of each tRNA is observed. The corresponding region within each folded structure is marked in green. The upper panel highlights the enrichment of the 3′ fragment of tRNA58Leu. Lower panel, enrichment of the 5′ fragment of tRNA122-Ala.

A minor portion of the snc-RNAs corresponds to YRNAs. YRNAs are a small cytoplasmic RNAs (85–115 nt) associated with Ro protein forming a RNP complex. In humans, specific YRNAs fragments that bind the Ro RNP complex have been observed (Krawetz et al., 2011). It has been proposed that this RNP complex participates in a quality control pathway for misfolded small RNAs (Stein et al., 2005). After ultraviolet irradiation, bacterial YRNAs and Ro protein increase suggesting that Ro RNP complex could have a role in the recognition or repair of DNA damage (Chen et al., 2003). Similarly, this RNP complex could act in the first steps of embryogenesis to initiate repair.

Spermatozoal RNA as epigenetic modifiers

Sperm-specific RNAs can influence fertilization and early embryo development but may also epigenetically modify the phenotype of the offspring (reviewed in Cuzin et al., 2008; Lalancette et al., 2008a; Johnson et al., 2011a; Hamatani, 2012; Rando, 2012). Following somatic cell nuclear transfer, some pathological changes in the placenta and congenital defects in the fetus as well as in the offspring are observed (reviewed in Shiels et al., 1999; Lanza et al., 2000; Xu and Yang, 2003). These changes reflect inappropriate epigenetic reprogramming of the donor and recipient cells leading to aberrant inner cell mass and trophectoderm formation (Niemann et al., 2008). It is possible that this mechanistic perturbation reflects the absence of the early effects of paternal elements (Krawetz, 2005; Krawetz et al., 2011) as somatic cells lack spermatozoal specific RNAs (Krawetz et al., 2011) that are likely to be integral to this pathway. This could involve targeting by epigenetic RNAs (Krawetz et al., 2011) that modify chromatin structure (Taft et al., 2011), e.g. through DNA methylation (Khraiwesh et al., 2010).

While the role of RNAs as modifiers of the epigenome altering gene expression is generally accepted, their transmission through the mammalian germline has been debated (Rassoulzadegan et al., 2006). The transgenerational epigenetic effect of paramutation is well established in the plant kingdom. Paramutation is the transfer of an epigenetic state to an unlinked homologous wild-type allele (paramutated allele) yielding a heritable phenotype in absence of an altered gene. Paramutation was first observed in maize (Brink, 1956) of the r1 gene that affects plant color. The most well-characterized paramutated gene in maize is b1, which employs the siRNA silencing pathway to modify methylation. At least three-repeat sequences upstream of the b1 gene are required to exceed a threshold to observe the effect conveyed by the siRNAs (reviewed in Arteaga-Vazquez and Chandler, 2010) for full penetrance and stability across generations. Paramutation in mammals seems to be reflected in complex processes like color, growth and disease, e.g. cardiac hypertrophy (Rassoulzadegan et al., 2006; Wagner et al., 2008; Grandjean et al., 2009). For example, while complete disruption of the mouse Kit (Mast/stem cell growth factor receptor Kit) gene was lethal, the heterozygote and paramutated animal presented a white tail and feet. Spermatozoa from heterozygote and paramutated progeny were enriched with truncated kit RNA. Interestingly, microinjection of heterozygote RNAs or miRNAs that target Kit (miR-221, miR-222) into fertilized oocyte induced the heterozygote phenotype (Rassoulzadegan et al., 2006). Other miRNAs, with a paramutation function that display a transgenerational effect have been described. These include miR-1 and miR-124 that paramutate Cdk9 (Cyclin-dependent kinase 9) and Sox9 (Transcription factor SOX-9), respectively. Although, the mechanism remains unknown, transactivation through methytransferases is being aggressively pursued.

Paramutation of human genes may reconcile familiar predisposition of some non-Mendelian genetic diseases. Transgenerational epigenetic effects could be a mechanism to confer increased competitiveness that allows the progeny to adapt to new environments to which the parents have been exposed. Alternatively, exposure to a toxic environment could hijack this response compromising the offspring. Transgenerational genetic effects occur when genetic factors in one generation affect the phenotype in the following generations without inheritance of the parental genetic factor. For example, daughters from genetically equal fathers but with a different Y chromosome differ in behavioral phenotype. This is remarkable, considering the low number of genes present on the Y chromosome (Nelson et al., 2010). On the one hand, it may simply reflect the high level of recombination during spermatogenesis yielding diverse gamete genomes (Lu et al., 2012; Wang et al., 2012). On the other hand, the variability may reflect epigenetic and/or transcript sharing among the maturing sperm cells through the cytoplasmic bridges (Caldwell and Handel, 1991). Evidence for the latter is provided by the apparent unequal distribution of transcripts among each sperm (Wykes et al., 2000).

Several reviews are available that provide examples of transgenerational epigenetic effects in mammals (reviewed in Jirtle and Skinner, 2007; Curley et al., 2011; Rando, 2012). For example, the progeny of mice receiving a high-fat diet during pregnancy are at increased risk of obesity and metabolic disease with subsequent passage through the paternal lineage (Dunn and Bale, 2011). Exposure to endocrine disruptors in female rats during gonadal sex determination increases the incidence of F1 male infertility. Some consequences of transgenerational inheritance are reflected by changes in the pattern of male germ cell methylation (Anway et al., 2005). Perhaps spermatozoal non-coding RNAs known to regulate DNA methylation and chromatin structure are components of transgenerational epigenetic mechanisms (reviewed in Lee, 2012; Rinn and Chang, 2012).

Models in agriculture

Animal models including cattle (Adams and Pierson, 1995; Burns et al., 2005) and equine (Carnevale, 2008) have proved essential to developing various assisted reproductive techniques (Bavister, 2002) and for providing a framework to study human reproductive disease (reviewed in Matsunari and Nagashima, 2009). While many critical sperm transcripts are conserved among different mammals (mouse, sheep, cattle, horse, pig and human) some appear species-specific (Card et al., 2013; Das et al., 2013; Sendler et al., 2013). Several orthologous spermatozoal transcripts observed among human, bovine and stallion are presented in Table I. Among the roles identified for these critical genes, are sperm DNA condensation (PRM1: Protamine 1), sperm motility (AKAP4: A-kinase anchor protein 4), sperm capacitation and sperm–oocyte interaction (CRISP2: Cysteine-rich secretory protein 2) and nucleocytoplasmic exchange during spermatogenesis (KIF5C: Kinesin heavy chain isoform 5C). Orthologous transcripts which appear at high levels in all species, but are of unknown function, like FAM71D (Family with sequence similarity 71, member D) warrant further study. As summarized in Supplementary data, Table SI and SII, many of the abundant human spermatozoal transcripts are orthologous to those present in either stallion or bull spermatozoa. Although some of these orthologous genes like GPX4 and DDX4 are involved in spermatogenesis (Phospholipid hydroperoxide glutathione peroxidase and Probable ATP-dependent RNA helicase DDX4, respectively), sperm function (CA2, Carbonic anhydrase 2) or embryo development (PAFAH1B1, Platelet-activating factor acetylhydrolase IB subunit alpha), the functional role for many others remains to be delineated. Understanding their role should prove valuable in elucidating factors regulating spermatogenesis and the underlying cause(s) of male infertility.

Table I

Orthologous spermatozoa transcripts identified among human, bovine and stallion.

Transcript name (percentile ranking in human)Transcript symbolReported functionaReferences
Protamine 1 (0.99)PRM1Sperm DNA condensationCho et al. (2001) (reviewed in Miller et al., 2005; Oliva, 2006)
Cysteine-rich secretory protein 2 (0.99)CRISP2Sperm capacitation and sperm–egg interactionBusso et al. (2007), Wang et al. (2004)
A-kinase anchor protein 4 (0.99)AKAP4Sperm motilityMiki et al. (2002) (reviewed in Turner, 2006)
Kinesin heavy chain isoform 5C (0.98)KIF5CNucleocytoplasmic exchange activities during spermatogenesisMannowetz et al. (2010)
Family with sequence similarity 71, member D (0.95)FAM71DFunctional role in sperm not reportedPlatts et al. (2007)
Involved in sperm morphogenesis with likely role in genome stability, cell division, survival and/or proliferationKittler et al. (2007), Paulsen et al. (2009), Chia et al. (2010)
Transcript name (percentile ranking in human)Transcript symbolReported functionaReferences
Protamine 1 (0.99)PRM1Sperm DNA condensationCho et al. (2001) (reviewed in Miller et al., 2005; Oliva, 2006)
Cysteine-rich secretory protein 2 (0.99)CRISP2Sperm capacitation and sperm–egg interactionBusso et al. (2007), Wang et al. (2004)
A-kinase anchor protein 4 (0.99)AKAP4Sperm motilityMiki et al. (2002) (reviewed in Turner, 2006)
Kinesin heavy chain isoform 5C (0.98)KIF5CNucleocytoplasmic exchange activities during spermatogenesisMannowetz et al. (2010)
Family with sequence similarity 71, member D (0.95)FAM71DFunctional role in sperm not reportedPlatts et al. (2007)
Involved in sperm morphogenesis with likely role in genome stability, cell division, survival and/or proliferationKittler et al. (2007), Paulsen et al. (2009), Chia et al. (2010)

The transcripts were selected and compared based on the RNA-seq data from human (Sendler et al., 2013), bovine (Card et al., 2013) and stallion (Das et al., 2013). The top 5% of the transcripts of the RNA-seq data from human were compared with bovine (FPKM > 100) and stallion (FPKM > 40).

Ortholgous genes were identified with Genomatix RegionMiner: Search for orthologous GeneIDs module.

Only stallion, bovine were compared for abundance, as these are the only species for which equivalent RNA-seq data are available.

aThe functions were verified in www.gopubmed.com, www.iHop-net.com and www.uniprot.org.

Table I

Orthologous spermatozoa transcripts identified among human, bovine and stallion.

Transcript name (percentile ranking in human)Transcript symbolReported functionaReferences
Protamine 1 (0.99)PRM1Sperm DNA condensationCho et al. (2001) (reviewed in Miller et al., 2005; Oliva, 2006)
Cysteine-rich secretory protein 2 (0.99)CRISP2Sperm capacitation and sperm–egg interactionBusso et al. (2007), Wang et al. (2004)
A-kinase anchor protein 4 (0.99)AKAP4Sperm motilityMiki et al. (2002) (reviewed in Turner, 2006)
Kinesin heavy chain isoform 5C (0.98)KIF5CNucleocytoplasmic exchange activities during spermatogenesisMannowetz et al. (2010)
Family with sequence similarity 71, member D (0.95)FAM71DFunctional role in sperm not reportedPlatts et al. (2007)
Involved in sperm morphogenesis with likely role in genome stability, cell division, survival and/or proliferationKittler et al. (2007), Paulsen et al. (2009), Chia et al. (2010)
Transcript name (percentile ranking in human)Transcript symbolReported functionaReferences
Protamine 1 (0.99)PRM1Sperm DNA condensationCho et al. (2001) (reviewed in Miller et al., 2005; Oliva, 2006)
Cysteine-rich secretory protein 2 (0.99)CRISP2Sperm capacitation and sperm–egg interactionBusso et al. (2007), Wang et al. (2004)
A-kinase anchor protein 4 (0.99)AKAP4Sperm motilityMiki et al. (2002) (reviewed in Turner, 2006)
Kinesin heavy chain isoform 5C (0.98)KIF5CNucleocytoplasmic exchange activities during spermatogenesisMannowetz et al. (2010)
Family with sequence similarity 71, member D (0.95)FAM71DFunctional role in sperm not reportedPlatts et al. (2007)
Involved in sperm morphogenesis with likely role in genome stability, cell division, survival and/or proliferationKittler et al. (2007), Paulsen et al. (2009), Chia et al. (2010)

The transcripts were selected and compared based on the RNA-seq data from human (Sendler et al., 2013), bovine (Card et al., 2013) and stallion (Das et al., 2013). The top 5% of the transcripts of the RNA-seq data from human were compared with bovine (FPKM > 100) and stallion (FPKM > 40).

Ortholgous genes were identified with Genomatix RegionMiner: Search for orthologous GeneIDs module.

Only stallion, bovine were compared for abundance, as these are the only species for which equivalent RNA-seq data are available.

aThe functions were verified in www.gopubmed.com, www.iHop-net.com and www.uniprot.org.

Differences between mice, ovine, bovine, equine, porcine and human RNA profiles may reflect basic interspecies differences in fertilization and early embryo development. In mouse, zygotic genome activation occurs just prior or at the 2-cell stage (reviewed in Schultz, 1993) whereas in bovine, ovine, porcine and human, genome activation appears to occur at the 4–8-cell stage (reviewed in Telford et al., 1990; Memili and First, 2000). The centrosome of the developing embryo is maternally derived in mouse (Schatten et al., 1986), whereas in other mammals, including human, it is paternally derived (Sathananthan et al., 1991, 1997; Manandhar et al., 2005). In mouse, pig and human, the male pronucleus is rapidly demethylated following fertilization whereas in bovine, sheep and rabbit, demethylation is comparatively delayed (Fulka et al., 2004). In part, this may reflect the degree of sperm chromatin condensation (Beaujean et al., 2004). In mammals, early developmental failure or altered phenotype has been associated with the perturbation of demethylation following in vitro fertilization (Yoshizawa et al., 2010), ovulation induction (Shi and Haaf, 2002), embryo culture (Zaitseva et al., 2007) or somatic cell nuclear transfer (reviewed in Morgan et al., 2005; Ma et al., 2012).

Artificial insemination (AI) can be viewed as one of the most important techniques devised for the genetic improvement of animals. In the dairy sector, improving reproductive efficiency is 5–10 times more economically important than any of the other production parameters including milk production and carcass quality (Wiltbank, 1994). Unexpectedly, the genetic selection of animals for higher milk production has lowered fertility (Veerkamp et al., 2003). One of the factors influencing fertility in the herd is the quality of the semen. However, sires with equivalent measurable semen parameters may produce vastly different pregnancy rates (reviewed in Kastelic and Thundathil, 2008). Since a single bull is used to inseminate hundreds of females, the use of semen from subfertile or infertile animals can have devastating consequences for the dairy industry. Selection of bulls or semen samples-based primarily on progressive forward motility invariably does not equally yield bulls of high or equal fertility (Selvaraju et al., 2008). Efficient semen evaluation methods including cellular and molecular approaches are required to predict fertility potential of a bull with high reproductive efficiency. In this regard, the potential of sperm transcripts to provide a marker of sperm quality and embryonic development in farm animals is of considerable interest. The levels of specific sperm RNAs associated with sperm functional parameters (Bissonnette et al., 2009; Curry et al., 2011) and conception (Lalancette et al., 2008b; Arangasamy et al., 2011; Kasimanickam et al., 2012) have been explored (Table II). These studies have now been extended to miRNAs in bovine (Govindaraju et al., 2012), porcine (Curry et al., 2009, 2011) and stallion (Das et al., 2013). As in the human (Krawetz et al., 2011), they may have critical roles both in mature sperm or after delivery to the oocyte where they may regulate fertilization and/or early embryonic development.

Table II

Mature spermatozoa RNAs associated with semen parameters and fertility in animals determined by RT–PCR.

StudyTechniquePhenotypeAltered specific RNAsa
Bissonnette et al. (2009)RTPCRHigh motility bovine sperm fractionTSSK6 and ADAM5P
Arangasamy et al. (2011)RTPCRHigh sire conception rate in bovineCRISP2
CCT8
Curry et al. (2011)RTPCRLow motility in porcine↑miR let 7d and 7e
Altered morphology in porcine↑miR let 7a, 7d,7e and miR22
Hwang et al. (2012)RTPCRLow porcine embryo cleavage after IVFMYC, CYP19, ADAM2, PRM1 and PRM2
High capacitated porcine spermatozoaMYC
Kasimanickam et al. (2013)RTPCRFertile males in bovine↑Adiponectine and receptors ADR1 and ADR2
Ganguly et al. (2013)RTPCRMotility impaired in bovinePRM1
StudyTechniquePhenotypeAltered specific RNAsa
Bissonnette et al. (2009)RTPCRHigh motility bovine sperm fractionTSSK6 and ADAM5P
Arangasamy et al. (2011)RTPCRHigh sire conception rate in bovineCRISP2
CCT8
Curry et al. (2011)RTPCRLow motility in porcine↑miR let 7d and 7e
Altered morphology in porcine↑miR let 7a, 7d,7e and miR22
Hwang et al. (2012)RTPCRLow porcine embryo cleavage after IVFMYC, CYP19, ADAM2, PRM1 and PRM2
High capacitated porcine spermatozoaMYC
Kasimanickam et al. (2013)RTPCRFertile males in bovine↑Adiponectine and receptors ADR1 and ADR2
Ganguly et al. (2013)RTPCRMotility impaired in bovinePRM1

aTranscript abundance: ↑ increased and ↓ decreased.

Table II

Mature spermatozoa RNAs associated with semen parameters and fertility in animals determined by RT–PCR.

StudyTechniquePhenotypeAltered specific RNAsa
Bissonnette et al. (2009)RTPCRHigh motility bovine sperm fractionTSSK6 and ADAM5P
Arangasamy et al. (2011)RTPCRHigh sire conception rate in bovineCRISP2
CCT8
Curry et al. (2011)RTPCRLow motility in porcine↑miR let 7d and 7e
Altered morphology in porcine↑miR let 7a, 7d,7e and miR22
Hwang et al. (2012)RTPCRLow porcine embryo cleavage after IVFMYC, CYP19, ADAM2, PRM1 and PRM2
High capacitated porcine spermatozoaMYC
Kasimanickam et al. (2013)RTPCRFertile males in bovine↑Adiponectine and receptors ADR1 and ADR2
Ganguly et al. (2013)RTPCRMotility impaired in bovinePRM1
StudyTechniquePhenotypeAltered specific RNAsa
Bissonnette et al. (2009)RTPCRHigh motility bovine sperm fractionTSSK6 and ADAM5P
Arangasamy et al. (2011)RTPCRHigh sire conception rate in bovineCRISP2
CCT8
Curry et al. (2011)RTPCRLow motility in porcine↑miR let 7d and 7e
Altered morphology in porcine↑miR let 7a, 7d,7e and miR22
Hwang et al. (2012)RTPCRLow porcine embryo cleavage after IVFMYC, CYP19, ADAM2, PRM1 and PRM2
High capacitated porcine spermatozoaMYC
Kasimanickam et al. (2013)RTPCRFertile males in bovine↑Adiponectine and receptors ADR1 and ADR2
Ganguly et al. (2013)RTPCRMotility impaired in bovinePRM1

aTranscript abundance: ↑ increased and ↓ decreased.

Biomarkers of human fertility

Infertility is a growing problem in contemporary society, affecting ∼10–15% of reproductive aged couples (reviewed in Evers, 2002). The evaluation of observable semen parameters is well suited to diagnosing some obvious forms of male infertility. However, even when the sample is deemed suitable based on external characteristics, fertilization potential is still in question. Hence, there is significant need for additional markers of sperm fertility status. Differences in the levels of individual or transcript groups between infertile patients and fertile controls may provide a means to assess the fidelity of past spermatogenic events and/or potential post-fertilization success (reviewed in Anton and Krawetz, 2012). Microarray analysis has identified altered mRNA profiles in infertile patients presenting suboptimal seminal parameters (Platts et al., 2007; Jodar et al., 2012; Montjean et al., 2012). These results identified some altered pathways allowing further insight into the pathogenic mechanisms involved in male infertility. For example, the ubiquitin–proteosome pathway is severely disrupted in teratozoospermic patients (Platts et al., 2007) and in oligozoospermic patients a decrease in the transcripts involved in DNA repair and oxidative stress regulation has been observed (Montjean et al., 2012). Because of the relatively high cost of microarrays, the use of real-time PCR has been explored.

Protamine transcripts are among those most strongly associated with the different seminal parameters such as sperm concentration and motility (Lambard et al., 2004; Kempisty et al., 2007) as well as with sperm fertilization ability and embryo quality (Depa-Martynow et al., 2007, 2012; Steger et al., 2008; Jodar et al., 2012; Rogenhofer et al., 2013). This is likely reflective of the relative abundance of the protamines and their requirement for chromatin packaging. Although the spermatozoa contain a heterogenous population of transcripts, some transcript pairs are proposed to have a stable correlation of expression among different fertile individuals (Lima-Souza et al., 2012). All of these reported RNA factors could provide a useful suite of fertility biomarkers, and are summarized in Table III.

Table III

Altered spermatozoa transcripts and pathways associated with human male infertility determined by microarray or RT–PCR.

StudyTechniquePhenotypeAltered specific RNAsaAltered pathways
Lambard et al. (2004)RTPCRLow motility sperm fractionPRM1, eNOS and nNOS
High capacitated spermatozoaMYC
Wang et al. (2004)RTPCRAsthenozoospermic patientsTPX1 and LDHC
Depa-Martynow et al. (2007)RTPCRIVF failure↓Fetilin beta, PRM1 and PRM2
Guo et al. (2007)RTPCROligozoospermic patientsVASA
Jedrzejczak et al. (2007)RTPCRAthenozoospermic patientsHILS1, TNP1, and TNP2
Kempisty et al. (2007)RTPCRAthenozoospermic patientsPRM1 and PRM2
Li et al. (2007a, b)RTPCRLow motility sperm fractionCatSper2 and CatSper3
Platts et al. (2007)ArrayTeratozoospermic patientsUbiquitin-proteosome pathway, apoptotic pathway and MAP kinase signaling
Steger et al. (2008)RTPCRInfertile patientsAberrant PRM1/PRM2
Bcl2
Avendano et al. (2009)RTPCRInfertile patientsPSG1 and HLA-E
Garrido et al. (2009)ArrayInfertile normozoospermic patientsSpermatozoa differentiation
RTPCRTRY1, GGF1 and CAB39L
Nguyen et al. (2009)ArrayCryptorchid maleGerm cell maturation and sperm tail formation
RTPCRTPX1
Ferlin et al. (2010)RTPCRVaricocele, oligozoospermiaHSPA4, HSF1 and HSF2
Varicocele, normozoospermiaHSFY
Oligozoospermic patientsHSP90
Garcia-Herrero et al. (2011)ArrayICSI failureTesticular function, spermatogenesis and sperm physiology
Zheng et al. (2011)RTPCROligoasthenozoospermic patientsBDNF
Depa-Martynow et al. (2012)RTPCRLow concentration, motility, morphology, fertilization ability, embryo qualityPRM1 and PRM2
Jodar et al. (2012)ArrayAsthenozoospermic patientsSpermatid development and the ubiquinone biosynthesis pathway
RTPCRAsthenozoospermic patientsANXA2, BRD2 and OAZ3
Infertile patientsPRM1 and PRM2
Montjean et al. (2012)ArrayOligozoospermic patientsSpermatogenesis, sperm motility, DNA repair and oxidative stress regulation
RTPCRPRM2, TPD52L3, JMJD1A and NIPBL
Rogenhofer et al. (2013)RTPCRLow fertilization capacity (IVF and ICSI)Aberrant PRM1/PRM2
StudyTechniquePhenotypeAltered specific RNAsaAltered pathways
Lambard et al. (2004)RTPCRLow motility sperm fractionPRM1, eNOS and nNOS
High capacitated spermatozoaMYC
Wang et al. (2004)RTPCRAsthenozoospermic patientsTPX1 and LDHC
Depa-Martynow et al. (2007)RTPCRIVF failure↓Fetilin beta, PRM1 and PRM2
Guo et al. (2007)RTPCROligozoospermic patientsVASA
Jedrzejczak et al. (2007)RTPCRAthenozoospermic patientsHILS1, TNP1, and TNP2
Kempisty et al. (2007)RTPCRAthenozoospermic patientsPRM1 and PRM2
Li et al. (2007a, b)RTPCRLow motility sperm fractionCatSper2 and CatSper3
Platts et al. (2007)ArrayTeratozoospermic patientsUbiquitin-proteosome pathway, apoptotic pathway and MAP kinase signaling
Steger et al. (2008)RTPCRInfertile patientsAberrant PRM1/PRM2
Bcl2
Avendano et al. (2009)RTPCRInfertile patientsPSG1 and HLA-E
Garrido et al. (2009)ArrayInfertile normozoospermic patientsSpermatozoa differentiation
RTPCRTRY1, GGF1 and CAB39L
Nguyen et al. (2009)ArrayCryptorchid maleGerm cell maturation and sperm tail formation
RTPCRTPX1
Ferlin et al. (2010)RTPCRVaricocele, oligozoospermiaHSPA4, HSF1 and HSF2
Varicocele, normozoospermiaHSFY
Oligozoospermic patientsHSP90
Garcia-Herrero et al. (2011)ArrayICSI failureTesticular function, spermatogenesis and sperm physiology
Zheng et al. (2011)RTPCROligoasthenozoospermic patientsBDNF
Depa-Martynow et al. (2012)RTPCRLow concentration, motility, morphology, fertilization ability, embryo qualityPRM1 and PRM2
Jodar et al. (2012)ArrayAsthenozoospermic patientsSpermatid development and the ubiquinone biosynthesis pathway
RTPCRAsthenozoospermic patientsANXA2, BRD2 and OAZ3
Infertile patientsPRM1 and PRM2
Montjean et al. (2012)ArrayOligozoospermic patientsSpermatogenesis, sperm motility, DNA repair and oxidative stress regulation
RTPCRPRM2, TPD52L3, JMJD1A and NIPBL
Rogenhofer et al. (2013)RTPCRLow fertilization capacity (IVF and ICSI)Aberrant PRM1/PRM2

aTranscript abundance: ↑ increased and ↓ decreased.

Table III

Altered spermatozoa transcripts and pathways associated with human male infertility determined by microarray or RT–PCR.

StudyTechniquePhenotypeAltered specific RNAsaAltered pathways
Lambard et al. (2004)RTPCRLow motility sperm fractionPRM1, eNOS and nNOS
High capacitated spermatozoaMYC
Wang et al. (2004)RTPCRAsthenozoospermic patientsTPX1 and LDHC
Depa-Martynow et al. (2007)RTPCRIVF failure↓Fetilin beta, PRM1 and PRM2
Guo et al. (2007)RTPCROligozoospermic patientsVASA
Jedrzejczak et al. (2007)RTPCRAthenozoospermic patientsHILS1, TNP1, and TNP2
Kempisty et al. (2007)RTPCRAthenozoospermic patientsPRM1 and PRM2
Li et al. (2007a, b)RTPCRLow motility sperm fractionCatSper2 and CatSper3
Platts et al. (2007)ArrayTeratozoospermic patientsUbiquitin-proteosome pathway, apoptotic pathway and MAP kinase signaling
Steger et al. (2008)RTPCRInfertile patientsAberrant PRM1/PRM2
Bcl2
Avendano et al. (2009)RTPCRInfertile patientsPSG1 and HLA-E
Garrido et al. (2009)ArrayInfertile normozoospermic patientsSpermatozoa differentiation
RTPCRTRY1, GGF1 and CAB39L
Nguyen et al. (2009)ArrayCryptorchid maleGerm cell maturation and sperm tail formation
RTPCRTPX1
Ferlin et al. (2010)RTPCRVaricocele, oligozoospermiaHSPA4, HSF1 and HSF2
Varicocele, normozoospermiaHSFY
Oligozoospermic patientsHSP90
Garcia-Herrero et al. (2011)ArrayICSI failureTesticular function, spermatogenesis and sperm physiology
Zheng et al. (2011)RTPCROligoasthenozoospermic patientsBDNF
Depa-Martynow et al. (2012)RTPCRLow concentration, motility, morphology, fertilization ability, embryo qualityPRM1 and PRM2
Jodar et al. (2012)ArrayAsthenozoospermic patientsSpermatid development and the ubiquinone biosynthesis pathway
RTPCRAsthenozoospermic patientsANXA2, BRD2 and OAZ3
Infertile patientsPRM1 and PRM2
Montjean et al. (2012)ArrayOligozoospermic patientsSpermatogenesis, sperm motility, DNA repair and oxidative stress regulation
RTPCRPRM2, TPD52L3, JMJD1A and NIPBL
Rogenhofer et al. (2013)RTPCRLow fertilization capacity (IVF and ICSI)Aberrant PRM1/PRM2
StudyTechniquePhenotypeAltered specific RNAsaAltered pathways
Lambard et al. (2004)RTPCRLow motility sperm fractionPRM1, eNOS and nNOS
High capacitated spermatozoaMYC
Wang et al. (2004)RTPCRAsthenozoospermic patientsTPX1 and LDHC
Depa-Martynow et al. (2007)RTPCRIVF failure↓Fetilin beta, PRM1 and PRM2
Guo et al. (2007)RTPCROligozoospermic patientsVASA
Jedrzejczak et al. (2007)RTPCRAthenozoospermic patientsHILS1, TNP1, and TNP2
Kempisty et al. (2007)RTPCRAthenozoospermic patientsPRM1 and PRM2
Li et al. (2007a, b)RTPCRLow motility sperm fractionCatSper2 and CatSper3
Platts et al. (2007)ArrayTeratozoospermic patientsUbiquitin-proteosome pathway, apoptotic pathway and MAP kinase signaling
Steger et al. (2008)RTPCRInfertile patientsAberrant PRM1/PRM2
Bcl2
Avendano et al. (2009)RTPCRInfertile patientsPSG1 and HLA-E
Garrido et al. (2009)ArrayInfertile normozoospermic patientsSpermatozoa differentiation
RTPCRTRY1, GGF1 and CAB39L
Nguyen et al. (2009)ArrayCryptorchid maleGerm cell maturation and sperm tail formation
RTPCRTPX1
Ferlin et al. (2010)RTPCRVaricocele, oligozoospermiaHSPA4, HSF1 and HSF2
Varicocele, normozoospermiaHSFY
Oligozoospermic patientsHSP90
Garcia-Herrero et al. (2011)ArrayICSI failureTesticular function, spermatogenesis and sperm physiology
Zheng et al. (2011)RTPCROligoasthenozoospermic patientsBDNF
Depa-Martynow et al. (2012)RTPCRLow concentration, motility, morphology, fertilization ability, embryo qualityPRM1 and PRM2
Jodar et al. (2012)ArrayAsthenozoospermic patientsSpermatid development and the ubiquinone biosynthesis pathway
RTPCRAsthenozoospermic patientsANXA2, BRD2 and OAZ3
Infertile patientsPRM1 and PRM2
Montjean et al. (2012)ArrayOligozoospermic patientsSpermatogenesis, sperm motility, DNA repair and oxidative stress regulation
RTPCRPRM2, TPD52L3, JMJD1A and NIPBL
Rogenhofer et al. (2013)RTPCRLow fertilization capacity (IVF and ICSI)Aberrant PRM1/PRM2

aTranscript abundance: ↑ increased and ↓ decreased.

Most of the high abundant transcripts in human sperm have a relationship with testicular function and spermatogenesis (Sendler et al., 2013). Microarray analysis of serially sectioned testes has produced transcript profiles from different stages of spermatogenesis (Chalmel et al., 2012). These data, in conjunction with the profiles obtained from mature spermatozoa, will be instrumental in correlating perturbations during spermattogenesis and specific forms of male infertility. For example, using sperm transcript profiling, the initial effect of teratozoospermia was traced to the pachytene spermatocyte (Platts et al., 2007). Identification of these disruptions by profiling sperm transcripts rather than invasive testicular biopsy offers obvious benefits to the patient (Yatsenko et al., 2006). These novel techniques are expected to be useful in identifying the origins, prognosis and treatment of various forms of what was previously considered to be male idiopathic infertility.

In recent years, there has been speculation about the phenotype and health of the offspring born out of assisted reproductive technologies (ART) especially after IVF and ICSI (reviewed in Batcheller et al., 2011; Savage et al., 2011). The children born with the use of ART have been reported, by some, to have a higher risks to health (e.g. fertility disorders) when compared with those naturally conceived. However, considerable controversy remains as to the composition of the appropriate ‘control’ group that would permit such comparisons. The infertility associated with a genetic defect (reviewed in Matzuk and Lamb, 2008), if any, would also be carried to the next generation along with the increased risk of epigenetic disorders (Kobayashi et al., 2009) that could be mirrored by changes in the pattern of DNA methylation (reviewed in Savage et al., 2011; Feuer et al., 2013; Hart and Norman, 2013).

Maternally, it is well established that advanced age increases the risk of cytogenetic abnormalities that can manifest as Down syndrome (Hassold et al., 1984). With the ability of ART to ‘bypass’ some of the boundaries that have limited conception, studies are now beginning to suggest an association between increasing paternal age at conception and neurological disorders like autism, with a noticeable effect between 30 and 39 years and a substantial effect at ≥50 years (Grether et al., 2009; Hultman et al., 2011; van Balkom et al., 2012). Recent data show that the autism spectrum of disorders are strongly associated with de novo mutations (Sanders et al., 2012) present in spermatozoa. This likely reflects the continuity of sperm production during the life of an adult male that arises from the ∼840 divisions from each stem cell that give rise to the mature spermatozoon. The resulting cumulative effect of mutations at each division (Kong et al., 2012) along with effects on chromatin integrity (Wyrobek et al., 2006) and methylation errors (Flanagan et al., 2006) may contribute to the growing prevalence of age-related effects as observed, e.g. autism.

Paternal factors are thought to underlie the etiology of infertility/subfertility in approximately half of the couples undergoing ART (Jarow et al., 2002). This may, to some extent, be reflective of the sperm transcript profile that often varies between infertile and fertile males (Platts et al., 2007; Garrido et al., 2009; Garcia-Herrero et al., 2010a; Jodar et al., 2012; Montjean et al., 2012). Thus, the use of the paternal transcriptome as a biomarker deserves consideration. Moreover, different transcript profiles have been suggested that may coincide with successful pregnancy in different fertility treatments (Garcia-Herrero et al., 2010b, 2011). This has further supported the notion (Ostermeier et al., 2002; Platts et al., 2007; Lima-Souza et al., 2012) of the potential use of the microarray strategy as a clinical diagnostic tool (Garrido et al., 2013). Perhaps, sperm transcript profiling of patients undergoing ART (Garcia-Herrero et al., 2011) will aid in identifying both specific paternal factors and pathways which are negatively affecting fertility outcomes (reviewed in Carrell, 2008). As costs continue to decline, it is very likely that sperm transcript sequencing will reach the clinic shortly. These data and their analysis provide several advantages compared with the microarray assays. These include a quantitative description of abundance, immediate assessment of the fidelity of the information content and allele-specific expression reflective of Expressed Quantitative Trait Loci and hence genotype. This truly heralds the beginning of Male Personalized Reproductive Medicine.

Conclusion

Rapid development of RNA assay technologies including RNA-seq has detailed the specific presence of a wide variety of spermatozoal transcripts. Such transcripts appear to reflect both the past course of spermatogenesis, yet also include factors critical to fertilization and successful embryo development (Fig. 9). This likely includes a significant epigenetic component that is mediated by the action of sperm-borne RNAs on both genotype and phenotype of the offspring. An increasing number of comparative animal models will be of use in identifying key transcripts, many of which may be unique to sperm, and in elucidating their functional role. With this understanding of the differentiative history of the pool of spermatozoal transcripts, the outcome of ART, and perhaps in the future the ability to both treat underlying causes of infertility and ensure the birth of a healthy child, will be optimized.

Figure 9

The potential actions of spermatozoal RNAs during early embryo development. Spermatozoal RNAs are delivered to the oocyte acting during the first steps of embryogenesis. Some intact paternal mRNAs like INST1 could be translated by maternal machinery. On one hand, paternal mature miRNAs like mouse miR-34c are essential for the first cell division. On the other hand, primicroRNAs like 181c, can be processed and thus activated by maternal DICER to their mature miRNAs regulating transcript stability, whereas others may target promoters. Interestingly, some non-coding RNAs act through triplex structures and perhaps are transcriptional regulators. For example, homopurine fragments of LINE1 provided by spermatozoa induce LINE1 transcription during the first divisions of the zygote. It has also been proposed that piRNAs, miRNAs and other potential RNAs may be the pathway to confrontation and consolidation.

Authors' roles

M.J., S.S. and E.S. analyzed the data, performed the literature searches and wrote the manuscript with M.P.D. and S.A.K. S.A.K. directed the data analysis, writing and editing of the manuscript. The authors alone are responsible for the content and writing of the paper.

Funding

This work was supported in part by National Institutes of Health (NIH)/Eunice Kennedy Shriver National Institute of Child Health and Human Development (NICHD) Grant U10 HD039005. S.S. is supported by a Cutting-edge Research Enhancement and Scientific Training Award, Department of Biotechnology, Government of India. M.P.D. and S.A.K. are recipients of an EMD Serono grant to Wayne State University. Otherwise the authors report no conflicts of interest. The content is solely the responsibility of the authors and does not necessarily represent the official views of the NICHD or NIH.

Acknowledgements

The authors would like to thank Mr G. Johnson for his critical review of this manuscript and Mr R. Sanchez Giones and Mr Yitzchok Sendler for their assistance in the preparation of the illustrations. We apologize to others that we were not able to include their work in this review. The authors would like to thank the members of RMN for their invaluable assistance and for providing some of the samples used to illustrate the properties of spermatozoal RNAs. Prerelease access to the SEQR Whole Transcription Amplification system from Sigma Chemical Corporation is gratefully acknowledged.

References

Adams
GP
Pierson
RA
Bovine model for study of ovarian follicular dynamics in humans
Theriogenology
1995
, vol. 
43
 (pg. 
113
-
120
)
Anton
E
Krawetz
SA
Spermatozoa as biomarkers for the assessment of human male infertility and genotoxicity
Syst Biol Reprod Med
2012
, vol. 
58
 (pg. 
41
-
50
)
Anway
MD
Cupp
AS
Uzumcu
M
Skinner
MK
Epigenetic transgenerational actions of endocrine disruptors and male fertility
Science
2005
, vol. 
308
 (pg. 
1466
-
1469
)
Arangasamy
A
Kasimanickam
VR
DeJarnette
JM
Kasimanickam
RK
Association of CRISP2, CCT8, PEBP1 mRNA abundance in sperm and sire conception rate in Holstein bulls
Theriogenology
2011
, vol. 
76
 (pg. 
570
-
577
)
Aravin
AA
Hannon
GJ
Small RNA silencing pathways in germ and stem cells
Cold Spring Harb Symp Quant Biol
2008
, vol. 
73
 (pg. 
283
-
290
)
Arteaga-Vazquez
MA
Chandler
VL
Paramutation in maize: RNA mediated trans-generational gene silencing
Curr Opin Genet Dev
2010
, vol. 
20
 (pg. 
156
-
163
)
Avendano
C
Franchi
A
Jones
E
Oehninger
S
Pregnancy-specific {beta}-1-glycoprotein 1 and human leukocyte antigen-E mRNA in human sperm: differential expression in fertile and infertile men and evidence of a possible functional role during early development
Hum Reprod
2009
, vol. 
24
 (pg. 
270
-
277
)
Batcheller
A
Cardozo
E
Maguire
M
DeCherney
AH
Segars
JH
Are there subtle genome-wide epigenetic alterations in normal offspring conceived by assisted reproductive technologies?
Fertil Steril
2011
, vol. 
96
 (pg. 
1306
-
1311
)
Bavister
BD
Early history of in vitro fertilization
Reproduction
2002
, vol. 
124
 (pg. 
181
-
196
)
Beaujean
N
Taylor
JE
McGarry
M
Gardner
JO
Wilmut
I
Loi
P
Ptak
G
Galli
C
Lazzari
G
Bird
A
, et al. 
The effect of interspecific oocytes on demethylation of sperm DNA
Proc Natl Acad Sci USA
2004
, vol. 
101
 (pg. 
7636
-
7640
)
Beraldi
R
Pittoggi
C
Sciamanna
I
Mattei
E
Spadafora
C
Expression of LINE-1 retroposons is essential for murine preimplantation development
Mol Reprod Dev
2006
, vol. 
73
 (pg. 
279
-
287
)
Berkovits
BD
Wang
L
Guarnieri
P
Wolgemuth
DJ
The testis-specific double bromodomain-containing protein BRDT forms a complex with multiple spliceosome components and is required for mRNA splicing and 3′-UTR truncation in round spermatids
Nucleic Acids Res
2012
, vol. 
40
 (pg. 
7162
-
7175
)
Betlach
CJ
Erickson
RP
28 s and 18 s ribonucleic acid from mammalian spermatozoa
J Exp Zool
1976
, vol. 
198
 (pg. 
49
-
55
)
Bissonnette
N
Levesque-Sergerie
JP
Thibault
C
Boissonneault
G
Spermatozoal transcriptome profiling for bull sperm motility: a potential tool to evaluate semen quality
Reproduction
2009
, vol. 
138
 (pg. 
65
-
80
)
Borges
F
Gomes
G
Gardner
R
Moreno
N
McCormick
S
Feijo
JA
Becker
JD
Comparative transcriptomics of Arabidopsis sperm cells
Plant Physiol
2008
, vol. 
148
 (pg. 
1168
-
1181
)
Borsani
O
Zhu
J
Verslues
PE
Sunkar
R
Zhu
JK
Endogenous siRNAs derived from a pair of natural cis-antisense transcripts regulate salt tolerance in Arabidopsis
Cell
2005
, vol. 
123
 (pg. 
1279
-
1291
)
Bouhallier
F
Allioli
N
Lavial
F
Chalmel
F
Perrard
MH
Durand
P
Samarut
J
Pain
B
Rouault
JP
Role of miR-34c microRNA in the late steps of spermatogenesis
RNA
2010
, vol. 
16
 (pg. 
720
-
731
)
Bourc'his
D
Voinnet
O
A small-RNA perspective on gametogenesis, fertilization, and early zygotic development
Science
2010
, vol. 
330
 (pg. 
617
-
622
)
Brink
RA
A genetic change associated with the R locus in maize which is directed and potentially reversible
Genetics
1956
, vol. 
41
 (pg. 
872
-
889
)
Burns
DS
Jimenez-Krassel
F
Ireland
JL
Knight
PG
Ireland
JJ
Numbers of antral follicles during follicular waves in cattle: evidence for high variation among animals, very high repeatability in individuals, and an inverse association with serum follicle-stimulating hormone concentrations
Biol Reprod
2005
, vol. 
73
 (pg. 
54
-
62
)
Busso
D
Goldweic
NM
Hayashi
M
Kasahara
M
Cuasnicu
PS
Evidence for the involvement of testicular protein CRISP2 in mouse sperm–egg fusion
Biol Reprod
2007
, vol. 
76
 (pg. 
701
-
708
)
Cabili
MN
Trapnell
C
Goff
L
Koziol
M
Tazon-Vega
B
Regev
A
Rinn
JL
Integrative annotation of human large intergenic noncoding RNAs reveals global properties and specific subclasses
Genes Dev
2011
, vol. 
25
 (pg. 
1915
-
1927
)
Caldwell
KA
Handel
MA
Protamine transcript sharing among postmeiotic spermatids
Proc Natl Acad Sci USA
1991
, vol. 
88
 (pg. 
2407
-
2411
)
Card
C
Anderson
EJ
Zamberlan
S
Krieger
KB
Kaproth
M
Sartini
BL
Cryopreserved bovine spermatozoal transcript profile as revealed by high-throughput ribonucleic acid sequencing
Biol Reprod
2013
Carlile
M
Nalbant
P
Preston-Fayers
K
McHaffie
GS
Werner
A
Processing of naturally occurring sense/antisense transcripts of the vertebrate Slc34a gene into short RNAs
Physiol Genomics
2008
, vol. 
34
 (pg. 
95
-
100
)
Carmell
MA
Girard
A
van de Kant
HJ
Bourc'his
D
Bestor
TH
de Rooij
DG
Hannon
GJ
MIWI2 is essential for spermatogenesis and repression of transposons in the mouse male germline
Dev Cell
2007
, vol. 
12
 (pg. 
503
-
514
)
Carnevale
EM
The mare model for follicular maturation and reproductive aging in the woman
Theriogenology
2008
, vol. 
69
 (pg. 
23
-
30
)
Carrell
DT
Contributions of spermatozoa to embryogenesis: assays to evaluate their genetic and epigenetic fitness
Reprod Biomed online
2008
, vol. 
16
 (pg. 
474
-
484
)
Cawley
S
Bekiranov
S
Ng
HH
Kapranov
P
Sekinger
EA
Kampa
D
Piccolboni
A
Sementchenko
V
Cheng
J
Williams
AJ
, et al. 
Unbiased mapping of transcription factor binding sites along human chromosomes 21 and 22 points to widespread regulation of noncoding RNAs
Cell
2004
, vol. 
116
 (pg. 
499
-
509
)
Chalmel
F
Lardenois
A
Evrard
B
Mathieu
R
Feig
C
Demougin
P
Gattiker
A
Schulze
W
Jegou
B
Kirchhoff
C
, et al. 
Global human tissue profiling and protein network analysis reveals distinct levels of transcriptional germline-specificity and identifies target genes for male infertility
Hum Reprod
2012
, vol. 
27
 (pg. 
3233
-
3248
)
Chen
X
Smith
JD
Shi
H
Yang
DD
Flavell
RA
Wolin
SL
The Ro autoantigen binds misfolded U2 small nuclear RNAs and assists mammalian cell survival after UV irradiation
Curr Biol
2003
, vol. 
13
 (pg. 
2206
-
2211
)
Chia
NY
Chan
YS
Feng
B
Lu
X
Orlov
YL
Moreau
D
Kumar
P
Yang
L
Jiang
J
Lau
MS
, et al. 
A genome-wide RNAi screen reveals determinants of human embryonic stem cell identity
Nature
2010
, vol. 
468
 (pg. 
316
-
320
)
Cho
C
Willis
WD
Goulding
EH
Jung-Ha
H
Choi
YC
Hecht
NB
Eddy
EM
Haploinsufficiency of protamine-1 or -2 causes infertility in mice
Nat Genet
2001
, vol. 
28
 (pg. 
82
-
86
)
Cooper
TG
Cytoplasmic droplets: the good, the bad or just confusing?
Hum Reprod
2005
, vol. 
20
 (pg. 
9
-
11
)
Coordinators
NR
Database resources of the national center for biotechnology information
Nucleic Acids Res
2013
, vol. 
41
 (pg. 
D8
-
D20
)
Corney
DC
Flesken-Nikitin
A
Godwin
AK
Wang
W
Nikitin
AY
MicroRNA-34b and MicroRNA-34c are targets of p53 and cooperate in control of cell proliferation and adhesion-independent growth
Cancer Res
2007
, vol. 
67
 (pg. 
8433
-
8438
)
Croce
CM
Causes and consequences of microRNA dysregulation in cancer
Nat Rev Genet
2009
, vol. 
10
 (pg. 
704
-
714
)
Curley
JP
Mashoodh
R
Champagne
FA
Epigenetics and the origins of paternal effects
Horm Behav
2011
, vol. 
59
 (pg. 
306
-
314
)
Curry
E
Ellis
SE
Pratt
SL
Detection of porcine sperm microRNAs using a heterologous microRNA microarray and reverse transcriptase polymerase chain reaction
Mol Reprod Dev
2009
, vol. 
76
 (pg. 
218
-
219
)
Curry
E
Safranski
TJ
Pratt
SL
Differential expression of porcine sperm microRNAs and their association with sperm morphology and motility
Theriogenology
2011
, vol. 
76
 (pg. 
1532
-
1539
)
Cuzin
F
Grandjean
V
Rassoulzadegan
M
Inherited variation at the epigenetic level: paramutation from the plant to the mouse
Curr Opin Genet Dev
2008
, vol. 
18
 (pg. 
193
-
196
)
Das
PJ
McCarthy
F
Vishnoi
M
Paria
N
Gresham
C
Li
G
Kachroo
P
Sudderth
AK
Teague
S
Love
CC
, et al. 
Stallion sperm transcriptome comprises functionally coherent coding and regulatory RNAs as revealed by microarray analysis and RNA-seq
PLoS One
2013
, vol. 
8
 pg. 
e56535
 
Depa-Martynow
M
Kempisty
B
Lianeri
M
Jagodzinski
PP
Jedrzejczak
P
Association between fertilin beta, protamines 1 and 2 and spermatid-specific linker histone H1-like protein mRNA levels, fertilization ability of human spermatozoa, and quality of preimplantation embryos
Folia Histochem Cytobiol
2007
, vol. 
45
 
Suppl 1
(pg. 
S79
-
85
)
Depa-Martynow
M
Kempisty
B
Jagodzinski
PP
Pawelczyk
L
Jedrzejczak
P
Impact of protamine transcripts and their proteins on the quality and fertilization ability of sperm and the development of preimplantation embryos
Reprod Biol
2012
, vol. 
12
 (pg. 
57
-
72
)
Di Giammartino
DC
Nishida
K
Manley
JL
Mechanisms and consequences of alternative polyadenylation
Mol Cell
2011
, vol. 
43
 (pg. 
853
-
866
)
Dietze
H
Alexopoulou
D
Alvers
MR
Barrio-Alvers
L
Andreopoulos
B
Doms
A
Hakenberg
J
Monnich
J
Plake
C
Reischuck
A
, et al. 
Krawetz
SA
GoPubMed: exploring PubMed with ontological background knowledge
Bioinformatics for Systems Biology
2009
New York, USA
Humana Press, Humana Press, c/o Springer Science + Business Media
(pg. 
385
-
399
)
Dunn
GA
Bale
TL
Maternal high-fat diet effects on third-generation female body size via the paternal lineage
Endocrinology
2011
, vol. 
152
 (pg. 
2228
-
2236
)
Epple
A
Sherf
M
Krawetz
SA
BiblioSphere—hypothesis generation in regulatory network analysis
Bioinformatics for Systems Biology
2009
New York, USA
Humana Press, Humana Press, c/o Springer Science +Business Media
(pg. 
401
-
412
)
Evers
JL
Female subfertility
Lancet
2002
, vol. 
360
 (pg. 
151
-
159
)
Fadloun
A
Le Gras
S
Jost
B
Ziegler-Birling
C
Takahashi
H
Gorab
E
Carninci
P
Torres-Padilla
ME
Chromatin signatures and retrotransposon profiling in mouse embryos reveal regulation of LINE-1 by RNA
Nat Struct Mol Biol
2013
, vol. 
20
 pg. 
7
 
Faghihi
MA
Wahlestedt
C
Regulatory roles of natural antisense transcripts
Nat Rev Mol Cell Biol
2009
, vol. 
10
 (pg. 
637
-
643
)
Faghihi
MA
Modarresi
F
Khalil
AM
Wood
DE
Sahagan
BG
Morgan
TE
Finch
CE
St Laurent
G
3rd
Kenny
PJ
Wahlestedt
C
Expression of a noncoding RNA is elevated in Alzheimer's disease and drives rapid feed-forward regulation of beta-secretase
Nat Med
2008
, vol. 
14
 (pg. 
723
-
730
)
Ferlin
A
Speltra
E
Patassini
C
Pati
MA
Garolla
A
Caretta
N
Foresta
C
Heat shock protein and heat shock factor expression in sperm: relation to oligozoospermia and varicocele
J Urol
2010
, vol. 
183
 (pg. 
1248
-
1252
)
Feuer
SK
Camarano
L
Rinaudo
PF
ART and health: clinical outcomes and insights on molecular mechanisms from rodent studies
Mol Hum Reprod
2013
, vol. 
19
 (pg. 
189
-
204
)
Fischer
BE
Wasbrough
E
Meadows
LA
Randlet
O
Dorus
S
Karr
TL
Russell
S
Conserved properties of Drosophila and human spermatozoal mRNA repertoires
Proc Biol Sci
2012
, vol. 
279
 (pg. 
2636
-
2644
)
Flanagan
JM
Popendikyte
V
Pozdniakovaite
N
Sobolev
M
Assadzadeh
A
Schumacher
A
Zangeneh
M
Lau
L
Virtanen
C
Wang
SC
, et al. 
Intra- and interindividual epigenetic variation in human germ cells
Am J Hum Genet
2006
, vol. 
79
 (pg. 
67
-
84
)
Freiman
RN
Specific variants of general transcription factors regulate germ cell development in diverse organisms
Biochim Biophys Acta
2009
, vol. 
1789
 (pg. 
161
-
166
)
Fulka
H
Mrazek
M
Tepla
O
Fulka
J
Jr
DNA methylation pattern in human zygotes and developing embryos
Reproduction
2004
, vol. 
128
 (pg. 
703
-
708
)
Gangaraju
VK
Lin
H
MicroRNAs: key regulators of stem cells
Nat Rev Mol Cell Biol
2009
, vol. 
10
 (pg. 
116
-
125
)
Ganguly
I
Gaur
GK
Kumar
S
Mandal
DK
Kumar
M
Singh
U
Kumar
S
Sharma
A
Differential expression of protamine 1 and 2 genes in mature spermatozoa of normal and motility impaired semen producing crossbred Frieswal (HFxSahiwal) bulls
Res Vet Sci
2013
, vol. 
94
 (pg. 
256
-
262
)
Garcia-Herrero
S
Garrido
N
Martinez-Conejero
JA
Remohi
J
Pellicer
A
Meseguer
M
Ontological evaluation of transcriptional differences between sperm of infertile males and fertile donors using microarray analysis
J Assist Reprod Genet
2010a
, vol. 
27
 (pg. 
111
-
120
)
Garcia-Herrero
S
Meseguer
M
Martinez-Conejero
JA
Remohi
J
Pellicer
A
Garrido
N
The transcriptome of spermatozoa used in homologous intrauterine insemination varies considerably between samples that achieve pregnancy and those that do not
Fertil Steril
2010b
, vol. 
94
 (pg. 
1360
-
1373
)
Garcia-Herrero
S
Garrido
N
Martinez-Conejero
JA
Remohi
J
Pellicer
A
Meseguer
M
Differential transcriptomic profile in spermatozoa achieving pregnancy or not via ICSI
Reprod Biomed Online
2011
, vol. 
22
 (pg. 
25
-
36
)
Garrido
N
Martinez-Conejero
JA
Jauregui
J
Horcajadas
JA
Simon
C
Remohi
J
Meseguer
M
Microarray analysis in sperm from fertile and infertile men without basic sperm analysis abnormalities reveals a significantly different transcriptome
Fertil Steril
2009
, vol. 
91
 (pg. 
1307
-
1310
)
Garrido
N
Garcia-Herrero
S
Meseguer
M
Assessment of sperm using mRNA microarray technology
Fertil Steril
2013
, vol. 
99
 (pg. 
1008
-
1022
)
Gebetsberger
J
Zywicki
M
Kunzi
A
Polacek
N
tRNA-derived fragments target the ribosome and function as regulatory non-coding RNA in Haloferax volcanii
Archaea
2012
, vol. 
2012
 pg. 
260909
 
Georgiou
I
Noutsopoulos
D
Dimitriadou
E
Markopoulos
G
Apergi
A
Lazaros
L
Vaxevanoglou
T
Pantos
K
Syrrou
M
Tzavaras
T
Retrotransposon RNA expression and evidence for retrotransposition events in human oocytes
Hum Mol Genet
2009
, vol. 
18
 (pg. 
1221
-
1228
)
Gilbert
I
Bissonnette
N
Boissonneault
G
Vallee
M
Robert
C
A molecular analysis of the population of mRNA in bovine spermatozoa
Reproduction
2007
, vol. 
133
 (pg. 
1073
-
1086
)
Girard
A
Sachidanandam
R
Hannon
GJ
Carmell
MA
A germline-specific class of small RNAs binds mammalian Piwi proteins
Nature
2006
, vol. 
442
 (pg. 
199
-
202
)
Goodrich
RJ
Anton
E
Krawetz
SA
Isolating mRNA and small noncoding RNAs from human sperm
Methods Mol Biol
2013
, vol. 
927
 (pg. 
385
-
396
)
Goring
D
Indriolo
E
Gene expression: How plants avoid incest
Nature
2010
, vol. 
466
 (pg. 
926
-
928
)
Govindaraju
A
Uzun
A
Robertson
L
Atli
MO
Kaya
A
Topper
E
Crate
EA
Padbury
J
Perkins
A
Memili
E
Dynamics of microRNAs in bull spermatozoa
Reprod Biol Endocrinol
2012
, vol. 
10
 pg. 
82
 
Grandjean
V
Gounon
P
Wagner
N
Martin
L
Wagner
KD
Bernex
F
Cuzin
F
Rassoulzadegan
M
The miR-124-Sox9 paramutation: RNA-mediated epigenetic control of embryonic and adult growth
Development
2009
, vol. 
136
 (pg. 
3647
-
3655
)
Grant
GR
Farkas
MH
Pizarro
AD
Lahens
NF
Schug
J
Brunk
BP
Stoeckert
CJ
Hogenesch
JB
Pierce
EA
Comparative analysis of RNA-Seq alignment algorithms and the RNA-Seq unified mapper (RUM)
Bioinformatics
2011
, vol. 
27
 (pg. 
2518
-
2528
)
Grether
JK
Anderson
MC
Croen
LA
Smith
D
Windham
GC
Risk of autism and increasing maternal and paternal age in a large North American population
Am J Epidemiol
2009
, vol. 
170
 (pg. 
1118
-
1126
)
Guo
X
Gui
YT
Tang
AF
Lu
LH
Gao
X
Cai
ZM
Differential expression of VASA gene in ejaculated spermatozoa from normozoospermic men and patients with oligozoospermia
Asian J Androl
2007
, vol. 
9
 (pg. 
339
-
344
)
Hamatani
T
Human spermatozoal RNAs
Fertil Steril
2012
, vol. 
97
 (pg. 
275
-
281
)
Hammoud
SS
Nix
DA
Zhang
H
Purwar
J
Carrell
DT
Cairns
BR
Distinctive chromatin in human sperm packages genes for embryo development
Nature
2009
, vol. 
460
 (pg. 
473
-
478
)
Hart
R
Norman
RJ
The longer-term health outcomes for children born as a result of IVF treatment. Part II: mental health and development outcomes
Hum Reprod Update
2013
, vol. 
19
 (pg. 
244
-
250
)
Hassold
T
Warburton
D
Kline
J
Stein
Z
The relationship of maternal age and trisomy among trisomic spontaneous-abortions
Am J Hum Genet
1984
, vol. 
36
 (pg. 
1349
-
1356
)
Hata
T
Nakayama
M
Targeted disruption of the murine large nuclear KIAA1440/Ints1 protein causes growth arrest in early blastocyst stage embryos and eventual apoptotic cell death
Biochim Biophys Acta
2007
, vol. 
1773
 (pg. 
1039
-
1051
)
Hayashi
K
Chuva de Sousa Lopes
SM
Kaneda
M
Tang
F
Hajkova
P
Lao
K
O'Carroll
D
Das
PP
Tarakhovsky
A
Miska
EA
, et al. 
MicroRNA biogenesis is required for mouse primordial germ cell development and spermatogenesis
PLoS One
2008
, vol. 
3
 pg. 
e1738
 
Heo
JB
Sung
S
Vernalization-mediated epigenetic silencing by a long intronic noncoding RNA
Science
2011
, vol. 
331
 (pg. 
76
-
79
)
Hill
AE
Hong
JS
Wen
H
Teng
L
McPherson
DT
McPherson
SA
Levasseur
DN
Sorscher
EJ
Micro-RNA-like effects of complete intronic sequences
Front Biosci
2006
, vol. 
11
 (pg. 
1998
-
2006
)
Hultman
CM
Sandin
S
Levine
SZ
Lichtenstein
P
Reichenberg
A
Advancing paternal age and risk of autism: new evidence from a population-based study and a meta-analysis of epidemiological studies
Mol Psychiatry
2011
, vol. 
16
 (pg. 
1203
-
1212
)
Hwang
JY
Mulligan
BP
Kim
HM
Yang
BC
Lee
CK
Quantitative analysis of sperm mRNA in the pig: relationship with early embryo development and capacitation
Reprod Fertil Dev
2012
, vol. 
25
 (pg. 
807
-
817
)
Inoue
A
Matoba
S
Zhang
Y
Transcriptional activation of transposable elements in mouse zygotes is independent of Tet3-mediated 5-methylcytosine oxidation
Cell Res
2012
, vol. 
22
 (pg. 
1640
-
1649
)
Ishizu
H
Siomi
H
Siomi
MC
Biology of PIWI-interacting RNAs: new insights into biogenesis and function inside and outside of germlines
Genes Dev
2012
, vol. 
26
 (pg. 
2361
-
2373
)
Ivanov
P
Emara
MM
Villen
J
Gygi
SP
Anderson
P
Angiogenin-induced tRNA fragments inhibit translation initiation
Mol Cell
2011
, vol. 
43
 (pg. 
613
-
623
)
Jarow
JP
Sharlip
ID
Belker
AM
Lipshultz
LI
Sigman
M
Thomas
AJ
Schlegel
PN
Howards
SS
Nehra
A
Damewood
MD
, et al. 
Best practice policies for male infertility
J Urol
2002
, vol. 
167
 (pg. 
2138
-
2144
)
Jedrzejczak
P
Kempisty
B
Bryja
A
Mostowska
M
Depa-Martynow
M
Pawelczyk
L
Jagodzinski
PP
Quantitative assessment of transition proteins 1, 2 spermatid-specific linker histone H1-like protein transcripts in spermatozoa from normozoospermic and asthenozoospermic men
Arch Androl
2007
, vol. 
53
 (pg. 
199
-
205
)
Jirtle
RL
Skinner
MK
Environmental epigenomics and disease susceptibility
Nat Rev Genet
2007
, vol. 
8
 (pg. 
253
-
262
)
Jodar
M
Kalko
S
Castillo
J
Ballesca
JL
Oliva
R
Differential RNAs in the sperm cells of asthenozoospermic patients
Hum Reprod
2012
, vol. 
27
 (pg. 
1431
-
1438
)
Johnson
GD
Lalancette
C
Linnemann
AK
Leduc
F
Boissonneault
G
Krawetz
SA
The sperm nucleus: chromatin, RNA, and the nuclear matrix
Reproduction
2011a
, vol. 
141
 (pg. 
21
-
36
)
Johnson
GD
Sendler
E
Lalancette
C
Hauser
R
Diamond
MP
Krawetz
SA
Cleavage of rRNA ensures translational cessation in sperm at fertilization
Mol Hum Reprod
2011b
, vol. 
17
 (pg. 
721
-
726
)
Kasimanickam
V
Kasimanickam
R
Arangasamy
A
Saberivand
A
Stevenson
JS
Kastelic
JP
Association between mRNA abundance of functional sperm function proteins and fertility of Holstein bulls
Theriogenology
2012
, vol. 
78
 (pg. 
2007
-
2019
)
Kasimanickam
VR
Kasimanickam
RK
Kastelic
JP
Stevenson
JS
Associations of adiponectin and fertility estimates in Holstein bulls
Theriogenology
2013
, vol. 
79
 (pg. 
766
-
777
e763
Kastelic
JP
Thundathil
JC
Breeding soundness evaluation and semen analysis for predicting bull fertility
Reprod Domest Anim
2008
, vol. 
43
 
Suppl 2
(pg. 
368
-
373
)
Katayama
S
Tomaru
Y
Kasukawa
T
Waki
K
Nakanishi
M
Nakamura
M
Nishida
H
Yap
CC
Suzuki
M
Kawai
J
, et al. 
Antisense transcription in the mammalian transcriptome
Science
2005
, vol. 
309
 (pg. 
1564
-
1566
)
Kawano
M
Kawaji
H
Grandjean
V
Kiani
J
Rassoulzadegan
M
Novel small noncoding RNAs in mouse spermatozoa, zygotes and early embryos
PLoS One
2012
, vol. 
7
 pg. 
e44542
 
Kempisty
B
Depa-Martynow
M
Lianeri
M
Jedrzejczak
P
Darul-Wasowicz
A
Jagodzinski
PP
Evaluation of protamines 1 and 2 transcript contents in spermatozoa from asthenozoospermic men
Folia Histochem Cytobiol
2007
, vol. 
45
 
Suppl 1
(pg. 
S109
-
S113
)
Khraiwesh
B
Arif
MA
Seumel
GI
Ossowski
S
Weigel
D
Reski
R
Frank
W
Transcriptional control of gene expression by microRNAs
Cell
2010
, vol. 
140
 (pg. 
111
-
122
)
Kigami
D
Minami
N
Takayama
H
Imai
H
MuERV-L is one of the earliest transcribed genes in mouse one-cell embryos
Biol Reprod
2003
, vol. 
68
 (pg. 
651
-
654
)
Kim
DH
Saetrom
P
Snove
O
Jr
Rossi
JJ
MicroRNA-directed transcriptional gene silencing in mammalian cells
Proc Natl Acad Sci USA
2008
, vol. 
105
 (pg. 
16230
-
16235
)
Kindler
S
Wang
H
Richter
D
Tiedge
H
RNA transport and local control of translation
Annu Rev Cell Dev Biol
2005
, vol. 
21
 (pg. 
223
-
245
)
Kiss
T
Bortolin
ML
Filipowicz
W
Characterization of the intron-encoded U19 RNA, a new mammalian small nucleolar RNA that is not associated with fibrillarin
Mol Cell Biol
1996
, vol. 
16
 (pg. 
1391
-
1400
)
Kittler
R
Pelletier
L
Heninger
AK
Slabicki
M
Theis
M
Miroslaw
L
Poser
I
Lawo
S
Grabner
H
Kozak
K
, et al. 
Genome-scale RNAi profiling of cell division in human tissue culture cells
Nat Cell Biol
2007
, vol. 
9
 (pg. 
1401
-
1412
)
Kobayashi
H
Hiura
H
John
RM
Sato
A
Otsu
E
Kobayashi
N
Suzuki
R
Suzuki
F
Hayashi
C
Utsunomiya
T
, et al. 
DNA methylation errors at imprinted loci after assisted conception originate in the parental sperm
Eur J Hum Genet
2009
, vol. 
17
 (pg. 
1582
-
1591
)
Kong
A
Frigge
ML
Masson
G
Besenbacher
S
Sulem
P
Magnusson
G
Gudjonsson
SA
Sigurdsson
A
Jonasdottir
A
Jonasdottir
A
, et al. 
Rate of de novo mutations and the importance of father's age to disease risk
Nature
2012
, vol. 
488
 (pg. 
471
-
475
)
Korhonen
HM
Meikar
O
Yadav
RP
Papaioannou
MD
Romero
Y
Da Ros
M
Herrera
PL
Toppari
J
Nef
S
Kotaja
N
Dicer is required for haploid male germ cell differentiation in mice
PLoS One
2011
, vol. 
6
 pg. 
e24821
 
Krawetz
SA
Paternal contribution: new insights and future challenges
Nat Rev Genet
2005
, vol. 
6
 (pg. 
633
-
642
)
Krawetz
SA
Kruger
A
Lalancette
C
Tagett
R
Anton
E
Draghici
S
Diamond
MP
A survey of small RNAs in human sperm
Hum Reprod
2011
, vol. 
26
 (pg. 
3401
-
3412
)
Kumar
G
Patel
D
Naz
RK
c-MYC mRNA is present in human sperm cells
Cell Mol Biol Res
1993
, vol. 
39
 (pg. 
111
-
117
)
Kuramochi-Miyagawa
S
Kimura
T
Ijiri
TW
Isobe
T
Asada
N
Fujita
Y
Ikawa
M
Iwai
N
Okabe
M
Deng
W
, et al. 
Mili, a mammalian member of piwi family gene, is essential for spermatogenesis
Development
2004
, vol. 
131
 (pg. 
839
-
849
)
Lalancette
C
Miller
D
Li
Y
Krawetz
SA
Paternal contributions: new functional insights for spermatozoal RNA
J Cell Biochem
2008a
, vol. 
104
 (pg. 
1570
-
1579
)
Lalancette
C
Thibault
C
Bachand
I
Caron
N
Bissonnette
N
Transcriptome analysis of bull semen with extreme non-return rate: use of suppression-subtractive hybridization to identify functional markers for fertility
Biol Reprod
2008b
, vol. 
78
 (pg. 
618
-
635
)
Lambard
S
Galeraud-Denis
I
Martin
G
Levy
R
Chocat
A
Carreau
S
Analysis and significance of mRNA in human ejaculated sperm from normozoospermic donors: relationship to sperm motility and capacitation
Mol Hum Reprod
2004
, vol. 
10
 (pg. 
535
-
541
)
Landgraf
P
Rusu
M
Sheridan
R
Sewer
A
Iovino
N
Aravin
A
Pfeffer
S
Rice
A
Kamphorst
AO
Landthaler
M
, et al. 
A mammalian microRNA expression atlas based on small RNA library sequencing
Cell
2007
, vol. 
129
 (pg. 
1401
-
1414
)
Lanza
RP
Cibelli
JB
Blackwell
C
Cristofalo
VJ
Francis
MK
Baerlocher
GM
Mak
J
Schertzer
M
Chavez
EA
Sawyer
N
, et al. 
Extension of cell life-span and telomere length in animals cloned from senescent somatic cells
Science
2000
, vol. 
288
 (pg. 
665
-
669
)
Lapidot
M
Pilpel
Y
Genome-wide natural antisense transcription: coupling its regulation to its different regulatory mechanisms
EMBO Rep
2006
, vol. 
7
 (pg. 
1216
-
1222
)
Lavorgna
G
Dahary
D
Lehner
B
Sorek
R
Sanderson
CM
Casari
G
In search of antisense
Trends Biochem Sci
2004
, vol. 
29
 (pg. 
88
-
94
)
Lee
JT
Epigenetic regulation by long noncoding RNAs
Science
2012
, vol. 
338
 (pg. 
1435
-
1439
)
Lee
TL
Pang
AL
Rennert
OM
Chan
WY
Genomic landscape of developing male germ cells
Birth Defects Res C Embryo Today
2009a
, vol. 
87
 (pg. 
43
-
63
)
Lee
YS
Shibata
Y
Malhotra
A
Dutta
A
A novel class of small RNAs: tRNA-derived RNA fragments (tRFs)
Genes Dev
2009b
, vol. 
23
 (pg. 
2639
-
2649
)
Leitner
F
Hoffmann
R
Valencia
A
Krawetz
SA
Biological knowledge extraction—a case study of iHOP and other language processing systems
Bioinformatics for Systems Biology
2009
New York, USA
Humana Press, c/o Springer Science +Business Media
(pg. 
413
-
433
)
Li
T
Vu
TH
Lee
KO
Yang
Y
Nguyen
CV
Bui
HQ
Zeng
ZL
Nguyen
BT
Hu
JF
Murphy
SK
, et al. 
An imprinted PEG1/MEST antisense expressed predominantly in human testis and in mature spermatozoa
J Biol Chem
2002
, vol. 
277
 (pg. 
13518
-
13527
)
Li
HG
Ding
XF
Liao
AH
Kong
XB
Xiong
CL
Expression of CatSper family transcripts in the mouse testis during post-natal development and human ejaculated spermatozoa: relationship to sperm motility
Mol Hum Reprod
2007a
, vol. 
13
 (pg. 
299
-
306
)
Li
SC
Tang
P
Lin
WC
Intronic microRNA: discovery and biological implications
DNA Cell Biol
2007b
, vol. 
26
 (pg. 
195
-
207
)
Lima-Souza
A
Anton
E
Mao
S
Ho
WJ
Krawetz
SA
A platform for evaluating sperm RNA biomarkers: dysplasia of the fibrous sheath—testing the concept
Fertil Steril
2012
, vol. 
97
 (pg. 
1061
-
1066
e1061–1063
Lin
SL
Miller
JD
Ying
SY
Intronic microRNA (miRNA)
J Biomed Biotechnol
2006
, vol. 
2006
 pg. 
26818
 
Lippman
Z
Martienssen
R
The role of RNA interference in heterochromatic silencing
Nature
2004
, vol. 
431
 (pg. 
364
-
370
)
Lippman
Z
Gendrel
AV
Black
M
Vaughn
MW
Dedhia
N
McCombie
WR
Lavine
K
Mittal
V
May
B
Kasschau
KD
, et al. 
Role of transposable elements in heterochromatin and epigenetic control
Nature
2004
, vol. 
430
 (pg. 
471
-
476
)
Liu
D
Brockman
JM
Dass
B
Hutchins
LN
Singh
P
McCarrey
JR
MacDonald
CC
Graber
JH
Systematic variation in mRNA 3′-processing signals during mouse spermatogenesis
Nucleic Acids Res
2007
, vol. 
35
 (pg. 
234
-
246
)
Liu
WM
Pang
RT
Chiu
PC
Wong
BP
Lao
K
Lee
KF
Yeung
WS
Sperm-borne microRNA-34c is required for the first cleavage division in mouse
Proc Natl Acad Sci USA
2012
, vol. 
109
 (pg. 
490
-
494
)
Lu
S
Zong
C
Fan
W
Yang
M
Li
J
Chapman
AR
Zhu
P
Hu
X
Xu
L
Yan
L
, et al. 
Probing meiotic recombination and aneuploidy of single sperm cells by whole-genome sequencing
Science
2012
, vol. 
338
 (pg. 
1627
-
1630
)
Luningschror
P
Hauser
S
Kaltschmidt
B
Kaltschmidt
C
MicroRNAs in pluripotency, reprogramming and cell fate induction
Biochim Biophys Acta
2013
, vol. 
1833
 (pg. 
1894
-
1903
)
Ma
JY
Liang
XW
Schatten
H
Sun
QY
Active DNA demethylation in mammalian preimplantation embryos: new insights and new perspectives
Mol Hum Reprod
2012
, vol. 
18
 (pg. 
333
-
340
)
Manandhar
G
Schatten
H
Sutovsky
P
Centrosome reduction during gametogenesis and its significance
Biol Reprod
2005
, vol. 
72
 (pg. 
2
-
13
)
Mannowetz
N
Kartarius
S
Wennemuth
G
Montenarh
M
Protein kinase CK2 and new binding partners during spermatogenesis
Cell Mol Life Sci
2010
, vol. 
67
 (pg. 
3905
-
3913
)
Manolea
F
Claude
A
Chun
J
Rosas
J
Melancon
P
Distinct functions for Arf guanine nucleotide exchange factors at the Golgi complex: GBF1 and BIGs are required for assembly and maintenance of the Golgi stack and trans-Golgi network, respectively
Mol Biol Cell
2008
, vol. 
19
 (pg. 
523
-
535
)
Martianov
I
Ramadass
A
Serra Barros
A
Chow
N
Akoulitchev
A
Repression of the human dihydrofolate reductase gene by a non-coding interfering transcript
Nature
2007
, vol. 
445
 (pg. 
666
-
670
)
Matsunari
H
Nagashima
H
Application of genetically modified and cloned pigs in translational research
J Reprod Dev
2009
, vol. 
55
 (pg. 
225
-
230
)
Matzuk
MM
Lamb
DJ
The biology of infertility: research advances and clinical challenges
Nat Med
2008
, vol. 
14
 (pg. 
1197
-
1213
)
Memili
E
First
NL
Zygotic and embryonic gene expression in cow: a review of timing and mechanisms of early gene expression as compared with other species
Zygote
2000
, vol. 
8
 (pg. 
87
-
96
)
Mercer
TR
Dinger
ME
Mattick
JS
Long non-coding RNAs: insights into functions
Nat Rev Genet
2009
, vol. 
10
 (pg. 
155
-
159
)
Miki
K
Willis
WD
Brown
PR
Goulding
EH
Fulcher
KD
Eddy
EM
Targeted disruption of the Akap4 gene causes defects in sperm flagellum and motility
Dev Biol
2002
, vol. 
248
 (pg. 
331
-
342
)
Miller
D
Iles
D
Carrell
DT
Has the renewed interest in sperm RNA led to fresh insights? A critical review and hypothesis
Paternal Influences on Human Reproductive Success
2013
Cambridgem, UK
Cambridge University Press, The Edinburgh Building
(pg. 
38
-
49
CB2 8RU
Miller
D
Tang
PZ
Skinner
C
Lilford
R
Differential RNA fingerprinting as a tool in the analysis of spermatozoal gene expression
Hum Reprod
1994
, vol. 
9
 (pg. 
864
-
869
)
Miller
D
Briggs
D
Snowden
H
Hamlington
J
Rollinson
S
Lilford
R
Krawetz
SA
A complex population of RNAs exists in human ejaculate spermatozoa: implications for understanding molecular aspects of spermiogenesis
Gene
1999
, vol. 
237
 (pg. 
385
-
392
)
Miller
D
Ostermeier
GC
Krawetz
SA
The controversy, potential and roles of spermatozoal RNA
Trends Mol Med
2005
, vol. 
11
 (pg. 
156
-
163
)
Moazed
D
Small RNAs in transcriptional gene silencing and genome defence
Nature
2009
, vol. 
457
 (pg. 
413
-
420
)
Mondal
T
Rasmussen
M
Pandey
GK
Isaksson
A
Kanduri
C
Characterization of the RNA content of chromatin
Genome Res
2010
, vol. 
20
 (pg. 
899
-
907
)
Montjean
D
De La Grange
P
Gentien
D
Rapinat
A
Belloc
S
Cohen-Bacrie
P
Menezo
Y
Benkhalifa
M
Sperm transcriptome profiling in oligozoospermia
J Assist Reprod Genet
2012
, vol. 
29
 (pg. 
3
-
10
)
Morgan
HD
Santos
F
Green
K
Dean
W
Reik
W
Epigenetic reprogramming in mammals
Hum Mol Genet
2005
, vol. 
14
 
Spec No 1
(pg. 
R47
-
58
)
Nelson
VR
Spiezio
SH
Nadeau
JH
Transgenerational genetic effects of the paternal Y chromosome on daughters' phenotypes
Epigenomics
2010
, vol. 
2
 (pg. 
513
-
521
)
Nguyen
MT
Delaney
DP
Kolon
TF
Gene expression alterations in cryptorchid males using spermatozoal microarray analysis
Fertil Steril
2009
, vol. 
92
 (pg. 
182
-
187
)
Niemann
H
Tian
XC
King
WA
Lee
RS
Epigenetic reprogramming in embryonic and foetal development upon somatic cell nuclear transfer cloning
Reproduction
2008
, vol. 
135
 (pg. 
151
-
163
)
Nolasco
S
Bellido
J
Goncalves
J
Tavares
A
Zabala
JC
Soares
H
The expression of tubulin cofactor A (TBCA) is regulated by a noncoding antisense Tbca RNA during testis maturation
PLoS One
2012
, vol. 
7
 pg. 
e42536
 
O'Donnell
KA
Boeke
JD
Mighty Piwis defend the germline against genome intruders
Cell
2007
, vol. 
129
 (pg. 
37
-
44
)
Oliva
R
Protamines and male infertility
Hum Reprod Update
2006
, vol. 
12
 (pg. 
417
-
435
)
Ostermeier
GC
Dix
DJ
Miller
D
Khatri
P
Krawetz
SA
Spermatozoal RNA profiles of normal fertile men
Lancet
2002
, vol. 
360
 (pg. 
772
-
777
)
Ostermeier
GC
Miller
D
Huntriss
JD
Diamond
MP
Krawetz
SA
Reproductive biology: delivering spermatozoan RNA to the oocyte
Nature
2004
, vol. 
429
 pg. 
154
 
Ostermeier
GC
Goodrich
RJ
Moldenhauer
JS
Diamond
MP
Krawetz
SA
A suite of novel human spermatozoal RNAs
J Androl
2005
, vol. 
26
 (pg. 
70
-
74
)
Parrott
AM
Mathews
MB
Novel rapidly evolving hominid RNAs bind nuclear factor 90 and display tissue-restricted distribution
Nucleic Acids Res
2007
, vol. 
35
 (pg. 
6249
-
6258
)
Patil
S
Totey
S
Developmental failure of hybrid embryos generated by in vitro fertilization of water buffalo (Bubalus bubalis) oocyte with bovine spermatozoa
Mol Reprod Dev
2003
, vol. 
64
 (pg. 
360
-
368
)
Paulsen
RD
Soni
DV
Wollman
R
Hahn
AT
Yee
MC
Guan
A
Hesley
JA
Miller
SC
Cromwell
EF
Solow-Cordero
DE
, et al. 
A genome-wide siRNA screen reveals diverse cellular processes and pathways that mediate genome stability
Molecular Cell
2009
, vol. 
35
 (pg. 
228
-
239
)
Peng
H
Shi
J
Zhang
Y
Zhang
H
Liao
S
Li
W
Lei
L
Han
C
Ning
L
Cao
Y
, et al. 
A novel class of tRNA-derived small RNAs extremely enriched in mature mouse sperm
Cell Res
2012
, vol. 
22
 (pg. 
1609
-
1612
)
Pessot
CA
Brito
M
Figueroa
J
Concha
II
Yanez
A
Burzio
LO
Presence of RNA in the sperm nucleus
Biochem Biophys Res Commun
1989
, vol. 
158
 (pg. 
272
-
278
)
Pittoggi
C
Sciamanna
I
Mattei
E
Beraldi
R
Lobascio
AM
Mai
A
Quaglia
MG
Lorenzini
R
Spadafora
C
Role of endogenous reverse transcriptase in murine early embryo development
Mol Reprod Dev
2003
, vol. 
66
 (pg. 
225
-
236
)
Place
RF
Li
LC
Pookot
D
Noonan
EJ
Dahiya
R
MicroRNA-373 induces expression of genes with complementary promoter sequences
Proc Natl Acad Sci USA
2008
, vol. 
105
 (pg. 
1608
-
1613
)
Platts
AE
Dix
DJ
Chemes
HE
Thompson
KE
Goodrich
R
Rockett
JC
Rawe
VY
Quintana
S
Diamond
MP
Strader
LF
, et al. 
Success and failure in human spermatogenesis as revealed by teratozoospermic RNAs
Hum Mol Genet
2007
, vol. 
16
 (pg. 
763
-
773
)
Polak
P
Domany
E
Alu elements contain many binding sites for transcription factors and may play a role in regulation of developmental processes
BMC Genomics
2006
, vol. 
7
 pg. 
133
 
Ponting
CP
Oliver
PL
Reik
W
Evolution and functions of long noncoding RNAs
Cell
2009
, vol. 
136
 (pg. 
629
-
641
)
Rando
OJ
Daddy issues: paternal effects on phenotype
Cell
2012
, vol. 
151
 (pg. 
702
-
708
)
Rassoulzadegan
M
Grandjean
V
Gounon
P
Vincent
S
Gillot
I
Cuzin
F
RNA-mediated non-mendelian inheritance of an epigenetic change in the mouse
Nature
2006
, vol. 
441
 (pg. 
469
-
474
)
Reavie
KT
Krawetz
SA
Mining the research literature in systems biology
Bioinformatics for Systems Biology
2009
New York, USA
Humana Press, Humana Press, c/o Springer Science + Business Media
(pg. 
369
-
383
)
Rejon
E
Bajon
C
Blaize
A
Robert
D
RNA in the nucleus of a motile plant spermatozoid: characterization by enzyme-gold cytochemistry and in situ hybridization
Mol Reprod Dev
1988
, vol. 
1
 (pg. 
49
-
56
)
Rinn
JL
Chang
HY
Genome regulation by long noncoding RNAs
Annu Rev Biochem
2012
, vol. 
81
 (pg. 
145
-
166
)
Rodriguez-Campos
A
Azorin
F
RNA is an integral component of chromatin that contributes to its structural organization
Plos One
2007
, vol. 
2
 pg. 
e1182
 
Rogenhofer
N
Dansranjavin
T
Schorsch
M
Spiess
A
Wang
H
von Schonfeldt
V
Cappallo-Obermann
H
Baukloh
V
Yang
H
Paradowska
A
, et al. 
The sperm protamine mRNA ratio as a clinical parameter to estimate the fertilizing potential of men taking part in an ART programme
Hum Reprod
2013
, vol. 
28
 (pg. 
969
-
978
)
Sanders
SJ
Murtha
MT
Gupta
AR
Murdoch
JD
Raubeson
MJ
Willsey
AJ
Ercan-Sencicek
AG
DiLullo
NM
Parikshak
NN
Stein
JL
, et al. 
De novo mutations revealed by whole-exome sequencing are strongly associated with autism
Nature
2012
, vol. 
485
 (pg. 
237
-
U124
)
Sathananthan
AH
Kola
I
Osborne
J
Trounson
A
Ng
SC
Bongso
A
Ratnam
SS
Centrioles in the beginning of human development
Proc Natl Acad Sci USA
1991
, vol. 
88
 (pg. 
4806
-
4810
)
Sathananthan
AH
Tatham
B
Dharmawardena
V
Grills
B
Lewis
I
Trounson
A
Inheritance of sperm centrioles and centrosomes in bovine embryos
Arch Androl
1997
, vol. 
38
 (pg. 
37
-
48
)
Savage
T
Peek
J
Hofman
PL
Cutfield
WS
Childhood outcomes of assisted reproductive technology
Hum Reprod
2011
, vol. 
26
 (pg. 
2392
-
2400
)
Schatten
H
Schatten
G
Mazia
D
Balczon
R
Simerly
C
Behavior of centrosomes during fertilization and cell division in mouse oocytes and in sea urchin eggs
Proc Natl Acad Sci USA
1986
, vol. 
83
 (pg. 
105
-
109
)
Schultz
RM
Regulation of zygotic gene activation in the mouse
Bioessays
1993
, vol. 
15
 (pg. 
531
-
538
)
Selvaraju
S
Ravindra
JP
Ghosh
J
Gupta
PS
Suresh
KP
Evaluation of sperm functional attributes in relation to in vitro sperm-zona pellucida binding ability and cleavage rate in assessing frozen thawed buffalo (Bubalus bubalis) semen quality
Anim Reprod Sci
2008
, vol. 
106
 (pg. 
311
-
321
)
Sendler
E
Johnson
GD
Mao
S
Goodrich
RJ
Diamond
MP
Hauser
R
Krawetz
SA
Stability, delivery and functions of human sperm RNAs at fertilization
Nucleic Acids Res
2013
, vol. 
41
 (pg. 
4104
-
4117
)
Shang
E
Nickerson
HD
Wen
D
Wang
X
Wolgemuth
DJ
The first bromodomain of Brdt, a testis-specific member of the BET sub-family of double-bromodomain-containing proteins, is essential for male germ cell differentiation
Development
2007
, vol. 
134
 (pg. 
3507
-
3515
)
Shi
W
Haaf
T
Aberrant methylation patterns at the two-cell stage as an indicator of early developmental failure
Mol Reprod Dev
2002
, vol. 
63
 (pg. 
329
-
334
)
Shiels
PG
Kind
AJ
Campbell
KH
Waddington
D
Wilmut
I
Colman
A
Schnieke
AE
Analysis of telomere lengths in cloned sheep
Nature
1999
, vol. 
399
 (pg. 
316
-
317
)
Shivdasani
RA
MicroRNAs: regulators of gene expression and cell differentiation
Blood
2006
, vol. 
108
 (pg. 
3646
-
3653
)
Short
RV
The contribution of the mule to scientific thought
J Reprod Fertil Suppl
1975
, vol. 
23
 (pg. 
359
-
364
)
Siomi
MC
Sato
K
Pezic
D
Aravin
AA
PIWI-interacting small RNAs: the vanguard of genome defence
Nat Rev Mol Cell Biol
2011
, vol. 
12
 (pg. 
246
-
258
)
Sone
Y
Ito
M
Shirakawa
H
Shikano
T
Takeuchi
H
Kinoshita
K
Miyazaki
S
Nuclear translocation of phospholipase C-zeta, an egg-activating factor, during early embryonic development
Biochem Biophys Res Commun
2005
, vol. 
330
 (pg. 
690
-
694
)
Spadafora
C
A reverse transcriptase-dependent mechanism plays central roles in fundamental biological processes
Syst Biol Reprod Med
2008
, vol. 
54
 (pg. 
11
-
21
)
Steger
K
Wilhelm
J
Konrad
L
Stalf
T
Greb
R
Diemer
T
Kliesch
S
Bergmann
M
Weidner
W
Both protamine-1 to protamine-2 mRNA ratio and Bcl2 mRNA content in testicular spermatids and ejaculated spermatozoa discriminate between fertile and infertile men
Hum Reprod
2008
, vol. 
23
 (pg. 
11
-
16
)
Stein
AJ
Fuchs
G
Fu
C
Wolin
SL
Reinisch
KM
Structural insights into RNA quality control: the Ro autoantigen binds misfolded RNAs via its central cavity
Cell
2005
, vol. 
121
 (pg. 
529
-
539
)
Taft
RJ
Hawkins
PG
Mattick
JS
Morris
KV
The relationship between transcription initiation RNAs and CCCTC-binding factor (CTCF) localization
Epigenetics Chromatin
2011
, vol. 
4
 pg. 
13
 
Telford
NA
Watson
AJ
Schultz
GA
Transition from maternal to embryonic control in early mammalian development: a comparison of several species
Mol Reprod Dev
1990
, vol. 
26
 (pg. 
90
-
100
)
Torres-Padilla
ME
Parfitt
DE
Kouzarides
T
Zernicka-Goetz
M
Histone arginine methylation regulates pluripotency in the early mouse embryo
Nature
2007
, vol. 
445
 (pg. 
214
-
218
)
Trapnell
C
Williams
BA
Pertea
G
Mortazavi
A
Kwan
G
van Baren
MJ
Salzberg
SL
Wold
BJ
Pachter
L
Transcript assembly and quantification by RNA-Seq reveals unannotated transcripts and isoform switching during cell differentiation
Nat Biotechnol
2010
, vol. 
28
 (pg. 
511
-
515
)
Tripathi
V
Ellis
JD
Shen
Z
Song
DY
Pan
Q
Watt
AT
Freier
SM
Bennett
CF
Sharma
A
Bubulya
PA
, et al. 
The nuclear-retained noncoding RNA MALAT1 regulates alternative splicing by modulating SR splicing factor phosphorylation
Mol Cell
2010
, vol. 
39
 (pg. 
925
-
938
)
Tsai
MC
Manor
O
Wan
Y
Mosammaparast
N
Wang
JK
Lan
F
Shi
Y
Segal
E
Chang
HY
Long noncoding RNA as modular scaffold of histone modification complexes
Science
2010
, vol. 
329
 (pg. 
689
-
693
)
Turner
RM
Moving to the beat: a review of mammalian sperm motility regulation
Reprod Fertil Dev
2006
, vol. 
18
 (pg. 
25
-
38
)
van Balkom
IDC
Bresnahan
M
Vuijk
PJ
Hubert
J
Susser
E
Hoek
HW
Paternal age and risk of autism in an ethnically diverse, non-industrialized setting: aruba
Plos One
2012
, vol. 
7
 pg. 
e45090
 
van der Heijden
GW
Bortvin
A
Transient relaxation of transposon silencing at the onset of mammalian meiosis
Epigenetics
2009
, vol. 
4
 (pg. 
76
-
79
)
Vassena
R
Boue
S
Gonzalez-Roca
E
Aran
B
Auer
H
Veiga
A
Izpisua Belmonte
JC
Waves of early transcriptional activation and pluripotency program initiation during human preimplantation development
Development
2011
, vol. 
138
 (pg. 
3699
-
3709
)
Veerkamp
RF
Beerda
B
van der Lende
T
Effects of genetic selection for milk yield on energy balance, levels of hormones, and metabolites in lactating cattle, and possible links to reduced fertility's
Livestock Production Sci
2003
, vol. 
83
 (pg. 
257
-
275
)
Waclawska
A
Kurpisz
M
Key functional genes of spermatogenesis identified by microarray analysis
Syst Biol Reprod Med
2012
, vol. 
58
 (pg. 
229
-
235
)
Wagner
KD
Wagner
N
Ghanbarian
H
Grandjean
V
Gounon
P
Cuzin
F
Rassoulzadegan
M
RNA induction and inheritance of epigenetic cardiac hypertrophy in the mouse
Dev Cell
2008
, vol. 
14
 (pg. 
962
-
969
)
Wang
H
Zhou
Z
Xu
M
Li
J
Xiao
J
Xu
ZY
Sha
J
A spermatogenesis-related gene expression profile in human spermatozoa and its potential clinical applications
J Mol Med (Berl)
2004
, vol. 
82
 (pg. 
317
-
324
)
Wang
J
Fan
HC
Behr
B
Quake
SR
Genome-wide single-cell analysis of recombination activity and de novo mutation rates in human sperm
Cell
2012
, vol. 
150
 (pg. 
402
-
412
)
Werner
A
Swan
D
What are natural antisense transcripts good for?
Biochem Soc Trans
2010
, vol. 
38
 (pg. 
1144
-
1149
)
Werner
A
Schmutzler
G
Carlile
M
Miles
CG
Peters
H
Expression profiling of antisense transcripts on DNA arrays
Physiol Genomics
2007
, vol. 
28
 (pg. 
294
-
300
)
Werner
A
Carlile
M
Swan
D
What do natural antisense transcripts regulate?
RNA Biol
2009
, vol. 
6
 (pg. 
43
-
48
)
Wiltbank
JN
Fields
M.J.
Sand
R.S.
Challenges for improving calf crop
Factors affecting calf crop
1994
Boca Raton, USA
CRC Press
(pg. 
1
-
22
)
Wu
Q
Song
R
Ortogero
N
Zheng
H
Evanoff
R
Small
CL
Griswold
MD
Namekawa
SH
Royo
H
Turner
JM
, et al. 
The RNase III enzyme DROSHA is essential for microRNA production and spermatogenesis
J Biol Chem
2012
, vol. 
287
 (pg. 
25173
-
25190
)
Wykes
SM
Visscher
DW
Krawetz
SA
Haploid transcripts persist in mature human spermatozoa
Mol Hum Reprod
1997
, vol. 
3
 (pg. 
15
-
19
)
Wykes
SM
Miller
D
Krawetz
SA
Mammalian spermatozoal mRNAs: tools for the functional analysis of male gametes
J Submicrosc Cytol Pathol
2000
, vol. 
32
 (pg. 
77
-
81
)
Wyrobek
AJ
Eskenazi
B
Young
S
Arnheim
N
Tiemann-Boege
I
Jabs
EW
Glaser
RL
Pearson
FS
Evenson
D
Advancing age has differential effects on DNA damage, chromatin integrity, gene mutations, and aneuploidies in sperm
Proc Natl Acad Sci USA
2006
, vol. 
103
 (pg. 
9601
-
9606
)
Xu
J
Yang
X
Will cloned animals suffer premature aging—the story at the end of clones' chromosomes
Reprod Biol Endocrinol
2003
, vol. 
1
 pg. 
105
 
Xu
Z
Jiang
J
Xu
C
Wang
Y
Sun
L
Guo
X
Liu
H
MicroRNA-181 regulates CARM1 and histone aginine methylation to promote differentiation of human embryonic stem cells
PLoS One
2013
, vol. 
8
 pg. 
e53146
 
Yang
CC
Lin
YS
Hsu
CC
Tsai
MH
Wu
SC
Cheng
WT
Seasonal effect on sperm messenger RNA profile of domestic swine (Sus Scrofa)
Anim Reprod Sci
2010
, vol. 
119
 (pg. 
76
-
84
)
Yatsenko
AN
Roy
A
Chen
R
Ma
L
Murthy
LJ
Yan
W
Lamb
DJ
Matzuk
MM
Non-invasive genetic diagnosis of male infertility using spermatozoal RNA: KLHL10 mutations in oligozoospermic patients impair homodimerization
Hum Mol Genet
2006
, vol. 
15
 (pg. 
3411
-
3419
)
Yoon
JH
Abdelmohsen
K
Gorospe
M
Posttranscriptional Gene Regulation by Long Noncoding RNA
J Mol Biol
2012a
 
in press
Yoon
JH
Abdelmohsen
K
Srikantan
S
Yang
X
Martindale
JL
De
S
Huarte
M
Zhan
M
Becker
KG
Gorospe
M
LincRNA-p21 suppresses target mRNA translation
Mol Cell
2012b
, vol. 
47
 (pg. 
648
-
655
)
Yoshizawa
Y
Kato
M
Hirabayashi
M
Hochi
S
Impaired active demethylation of the paternal genome in pronuclear-stage rat zygotes produced by in vitro fertilization or intracytoplasmic sperm injection
Mol Reprod Dev
2010
, vol. 
77
 (pg. 
69
-
75
)
Zaitseva
I
Zaitsev
S
Alenina
N
Bader
M
Krivokharchenko
A
Dynamics of DNA-demethylation in early mouse and rat embryos developed in vivo and in vitro
Mol Reprod Dev
2007
, vol. 
74
 (pg. 
1255
-
1261
)
Zheng
L
Li
C
Sun
Y
Liu
Z
Zhou
X
Expression of brain-derived neurotrophic factor in mature spermatozoa from fertile and infertile men
Clin Chim Acta
2011
, vol. 
412
 (pg. 
44
-
47
)

Author notes

Reproductive Medicine Network.

Supplementary data