Saturday, March 17, 2018

Spermatozoal RNAs in the Reproductive Clinic /Sperm RNA and Its Use as a Clinical Marker


ది స్ ఇస్ ఎక్ససర్' ప్టెడ్ ఫ్రామ్  ది బుక్ The Sperm Cell 2న్డ్ ఎడిషన్ 

Sperm RNA and Its Use as a Clinical Marker 
Meritxell Jodar, Ester Anton and Stephen A. Krawetz 

Introduction 
Infertility affects 12–30% of childbearing-age couples  [1], and it is expected that its prevalence will keep  rising [2]. This likely reflects the influence of several  lifestyle factors in developed countries, such as progressive  delay in maternal age, stress, obesity, exposure  to environmental hazards and the use of drugs.  It is suggested that upon presentation to an infertility  clinic half of these infertile couples involve a genetically  associatedmale factor. Nevertheless, a significant  proportion of male infertility (around 25%) remains  diagnosed as idiopathic [3].  The current standard of practice in assessing male  fertility in the andrology laboratory is usually through  semen analysis. This includes a microscopic analysis  in which sperm concentration, motility and morphology  are evaluated [4]. Although this test is widely  used in clinics to ascertain male reproductive potential,  it has been suggested that additional clinicalmarkers  are needed for more accurate assessments [5, 6].  In this context, several parameters have been proposed  as suitable fertility biomarker candidates, some  ofwhich have already been incorporated in clinics (e.g.  sperm FISH studies, sperm DNA integrity) [7]. The  recent development of new molecular methodologies  has been driven by genomics, transcriptomics, proteomics  and metabolomics. In this chapter, we present  a synopsis of the current state of the art of spermatozoal  RNAs, with a glimpse to the future.  Spermatozoal RNAs  Spermatogenesis is a complex biological process that  requires a highly regulated genetic program to orchestrate  a successful differentiation program. This process  occurs over at least 64 days in humans, beginning  as a primordial germ cell differentiates into a spermatogonium.  The proliferation and differentiation of  spermatogonia into spermatocytes, the progression of  spermatocytes through meiosis to form spermatids,  and final differentiation yield spermatozoa. Dramatic  changes in the shape of the round spermatid and  nucleus, as well as the formation of new organelles,  typify the mature sperm. This is exemplified by the  development of the flagellum and the acrosome, while  most cytoplasm is removed during the final steps of  spermatogenesis [8–10].These last steps are also characterized  by a general shutdown of transcription and  translation, as the genome is compacted by the progressive,  albeit incomplete replacement of themajority  of histones by protamines. Despite the resulting transcriptional  blockage and the loss of a large proportion  of RNAs during the final stages of spermatogenesis, a  small but complex population of RNAs is preserved in  mature sperm. Several studies have suggested that the  retention of these transcripts may not be random, but  rather reflects residues of spermatogenic processes or  the selection of molecules that will display their function  in early embryogenesis [11–15].  A single spermatozoon contains _50–90 fg of  long RNA (_200 nt), including coding and noncoding  RNAs [16]. This is about 200 times less than a  somatic cell [14]. An extensive and detailed catalogue  of sperm RNAs has been generated with the recent  application of Next Generation Sequencing (NGS) as  summarized in Figure 4.1, revealing the singularity of  RNAs retained in the sperm [14, 17].The most abundant  transcripts in sperm correspond to ribosomal  RNAs (rRNAs), but unlike those in somatic cells, these  rRNAs in sperm are selectively cleaved, most likely to  ensure the translational quiescent state of spermatozoa  [18]. A substantial portion of spermcoding RNAs also  The Sperm Cell, Second Edition, ed. Christopher J. De Jonge and Christopher L. R. Barratt. Published by Cambridge  University Press. _C Cambridge University Press 2017.  59  https:/www.cambridge.org/core/terms. https://doi.org/10.1017/9781316411124.006  Downloaded from https:/www.cambridge.org/core. Boston University Theology Library, on 28 May 2017 at 16:50:26, subject to the Cambridge Core terms of use, available at  Chapter 4: Sperm RNA and Its Use as a Clinical Marker  Figure 4.1 Characteristics of spermatozoal RNAs. The spermatozoa contain a complex population of coding and noncoding long RNAs  and small noncoding RNAs. Whereas the majority of coding RNAs are in a biologically fragmented state showing significant 3’ end profile bias  (i.e. RN2), a small percentage of transcripts are intact where all exons are well represented (i.e. DDX3X). A high percentage of sperm RNAs  correspond to long noncoding RNAs including rRNAs, annotated long noncoding RNAs, natural antisense transcripts and sperm-specific  RNAs named intronic elements. High levels of specific intronic sperm RNAs are also observed, while the coding regions of this transcript are  absent in sperm (i.e. QRICH1). Several types of small noncoding RNAs are also observed in sperm, including repeat-associated small RNAs,  piRNAs, transcription start sites/promoter associated RNAs, miRNAs, small nucleolar RNAs and small nuclear RNAs.  appear fragmented as remnants of spermatogenesis.  However, some transcripts are maintained intact, with  potential roles in sperm transit through the female  reproductive tract, fertilization and early embryogenesis  (Figure 4.1) [17].  A large proportion of human spermatozoal RNAs  correspond to noncoding RNAs. Some are annotated  in other cell types as long intergenic noncoding RNAs  (lincRNAs), transposable elements (LINE1, ERVLMaLR,  etc.), natural antisense transcripts (NAT)  and chromatin-associated (CAR) and small-nuclear  ILF3/NF30-associated (snaR) RNAs. Regulatory roles  have been ascribed to these long noncoding RNAs,  functioning both at the transcriptional and posttranscriptional  level. In addition to this group of  known RNAs, sperm-specific noncoding RNAs are  also observed [14, 17]. Typically they range in size  from 100 to 300 nt and overlap either the coding,  intronic or untranslated (UTR) regions of an otherwise  low-expressed or absent transcript (Figure 4.1).Their  specific retention in all samples sequenced (more than  100 individuals) and the presence of the corresponding  full transcript in testes suggests that they may be  part of a separate regulatory mechanism [17, 19].  Additionally, sperm contain approximately 0.3 fg  of small noncoding RNAs (sncRNAs) [14] which have  been described as playing an important regulatory role  throughout spermatogenesis [20, 21]. Their presence  in human spermatozoa was first described by Ostermeier  et al. [22], and subsequently, several studies have  evaluated their content in fertile and infertile males. To  date, 2,588 mature microRNAs (miRNAs) have been  described in humans (Sanger miRBase v.21;1) [23]. It  is estimated that they regulate the expression of up to  60% of the coding genes [24]. sncRNAs include several  classes of RNA transcripts with sizes ranging from  _21 to 30 nucleotides (nt) for miRNAs [25] and up  to _35 nt for piRNAs and tRNA fragments. The bestcharacterized  are the miRNAs, which are considered  to be highly relevant to regulating important processes  60  https:/www.cambridge.org/core/terms. https://doi.org/10.1017/9781316411124.006  Downloaded from https:/www.cambridge.org/core. Boston University Theology Library, on 28 May 2017 at 16:50:26, subject to the Cambridge Core terms of use, available at  Chapter 4: Sperm RNA and Its Use as a ClinicalMarker  that encompass germ cell development, differentiation  and proliferation [26]. The study of sperm sncRNAs  has been directed primarily towards miRNAs as new  potential fertility biomarkers [27]. This is reflected in  the enrichment of categories of processes modulated  by miRNAs. The canonical pathway of miRNA biogenesis  is highly regulated through their transcription,  processing, loading into effective ribonucleoprotein  complexes and turnover [28]. For example, the  polymerase II pri-miRNAs transcripts can be as large  as 3–4 kb, encoding multiple miRNAs [29]. The primiRNAs  are then processed by the RNase III Drosha  into shorter 41–180 nt (mode 83 nt) pre-miRNAs [30].  When exported to the cytosol by exportin 5, they are  primed for processing by the RNase III Dicer, yielding  semicomplementary double-stranded structures.  These are unwound through a helicase to yield two 16–  27 nt (mode 22 nt) mature miRNAs [30] denoted as  -5p and -3p, respectively, for their strand of origin.  Each mature miRNA is capable of regulating the translation  of several target mRNAs by forming imperfect  3’UTR complementary stem–loop structures, subsequent  to their assembly as part of an Argonaute (AGO)  family protein complex.  Another major class of sncRNAs is the piwiinteracting  RNAs (piRNAs).They have been described  as essential for mammalian spermatogenesis [31],  although their function and biogenesis are still not  fully understood. To date, _30,000 piRNAs have been  described in humans (piRNABank2) [32]. piRNAs can  be distinguished from miRNAs using several criteria.  These include their larger size (24–32 nt), their genesis  from single-strand precursors through an RNase  III-independent mechanism, and the formation of  effector complexes with PIWI proteins (germ-linespecific  subfamily members of AGO) [33, 34].The primary  functions of piRNAs are transposable element  silencing [35], epigenetic programming [36] and posttranscriptional  regulation of gene expression [37].  Recent studies have shown the existence of a turnover  mechanism that promotes the active degradation  of the ribonucleoprotein complex piRNAs-MIWI  (mouse PIWI protein) in the late stages of spermatogenesis  through the ubiquitin–proteasome pathway [38].  This turnover has been proposed to be part of transposon  silencing to avoid transmitting paternal piRNAs  to the embryo, where they could function in a similar  manner and may impede the zygote-to-embryo  transition [39]. Although the depletion of piRNAs in  the later stages of germ-cell development has been  observed [40–42], elimination may not be complete.  Recent data describe the presence of several piRNA  species in human sperm from fertile individuals [43,  44], along with a population of different intact mRNA  transcripts, in mature sperm cells [14]. The reduction  in piRNA abundance parallels that observed for  the population of mRNAs as the elongating spermatid  matures and the cytoplasm is expelled [45, 46].  In the human spermatozoa, the major classes of  sncRNAs identified by the first RNA-seq study [43]  included repeat-associated smallRNAs (65%), piRNAs  (_17%), transcription start sites/promoter-associated  RNAs (_11%), miRNAs (_7%), small nucleolar RNAs  (0.3%) and small nuclear RNAs (0.1%) [43]. Heterogeneity  among the three sperm samples reflected  20–60% donor specificity. Interestingly, only 35 miRNAs  were consistently present in the three samples  sequenced. This included several epi-miRNAs (miRNAs  that specifically target effectors of the epigenetic  machinery) and miRNAs with a potential exclusive  paternal origin for early embryonic development  (not detected in mouse oocytes but present in the  zygote). The most abundant miRNA was hsa-miR-34c,  which has been described as playing an important  role in spermatogenesis [47] and early embryogenesis  [48], although species-specific conflicting data have  been reported [49]. Interestingly, most potential targetmRNAs  of these ubiquitousmiRNAs have not been  detected in sperm [50] supporting their potential role  in translational suppression by degradation. In direct  contrast, 1,137 piRNAs were detected (the most abundant  of which was has-piR-020548), which preferentially  target MER, LINE1 and LTR elements.  More recently, Pantano et al. [44] used the same  technology to evaluate the sncRNA in two other normozoospermic  individuals. They observed 182 miRNAs  present in both samples with predicted targets  among sperm-specific genes.The most abundant  miRNA detected by these authors was hsa-miR-1246.  Discordance with respect to this new study and that  of Krawetz et al. [43] primarily reflects the exclusion  of miRNA with multiple alignments to the genome  by Krawetz et al. [43] and the rapid progress in technology.  The similarity between hsa-miR-1246 and U2  small nuclear RNAs is notable. However, the corresponding  immature form, pri-miR-1246, has been  identified in sperm, confirming its presence. Considering  only the unique aligned miRNAs, hsa-miR-34c  was also found as the most abundant. In their study,  Pantano et al. [44] also observed piRNAs as the most  61  https:/www.cambridge.org/core/terms. https://doi.org/10.1017/9781316411124.006  Downloaded from https:/www.cambridge.org/core. Boston University Theology Library, on 28 May 2017 at 16:50:26, subject to the Cambridge Core terms of use, available at  Chapter 4: Sperm RNA and Its Use as a Clinical Marker  abundant sncRNAs molecules (some of them processed  from pseudogenes), including 408 small RNA  clusters containing _1 known piRNA (34 clusters in  particular accounted for 13,509 piRNAs). The preferential  targets predicted for these piRNAs also included  LINE1 transposons, concordant with the study of  Krawetz et al. [43].  Discovering Potential Causes of  Male Infertility  The diagnosis of male infertility is currently based on  the study of seminal parameters such as sperm concentration,  motility and morphology. To date, a limited  number of causes associated with altered parameters  have been identified [51]. This is exemplified  by the general class of chromosomal abnormalities  that are detected in approximately 5% of patients  with fertility problems. Examples include numerical or  structural chromosomal aberrations, as in Klinefelter  syndrome (47,XXY), structural chromosome reorganizations  (e.g. balanced translocations or inversions)  and Y chromosome deletions. All can have a direct  negative influence on spermatogenesis, thereby affecting  sperm production, probably disrupting meiotic  pairing. As an adjunct, the study of ejaculated sperm  RNAs has emerged. It has provided insight into the  basicmolecularmechanisms that regulate production,  maturation and transit of sperm, as well as the pathogenesis  of male infertility.  Coding RNAs and Male Infertility  The initial approach to assessing male fertility by  spermatozoa RNAs relied on RT-PCR for quantification  of sperm coding RNAs known to be essential  during spermatogenesis. This was followed by  the first-generation high-throughput techniques, i.e.  microarrays, which revealed specific biological pathways  affecting the seminal parameters from the different  subtypes of male infertility [52–55]. The relationship  of spermatozoal coding RNAs to humanmale  fertility is presented in Table 4.1. The corresponding  summary of the published data highlighting the potential  causes of different infertility phenotypes associated  with semen parameter alterations follows.  Oligozoospermia  Oligozoospermia is characterized by a sperm concentration  or total number of ejaculate sperm below 15  or 39 million sperm cells, respectively [4], but excluding  the absence of spermatozoa (azoospermia).There  are many known causes for oligozoospermia, including  hormonal and chromosomal disorders, single  genetic defects, testicular and post-testicular factors,  such as varicocele and hypogonadism, and environment  insults [56]. However, even with this multitude  of correlative presentations, the primary causative factor  of oligozoospermia remains to be identified. The  observed reduction of sperm density in unexplained  oligozoospermia suggests that altered spermatogenesis  could reflect an altered transcript profile.  Gene knockout technology has shown that about  388 genes are critical for murine spermatogenesis  (Mouse Genome Informatics3). Down-regulation of  some of these genes has been shown to lead to  significantly decreased RNA levels in infertile men  presenting oligozoospermia. Examples include the  DEAD (Asp-Glu-Ala-Asp) Box Polypeptide 4 (DDX4)  [57], Ubiquitin-Conjugating Enzyme E2B (UBE2B)  [58] and some heat-shock proteins (HSPA4, HSF1and  HSF2) [59]. These transcripts encode proteins that  play an essential role in the early stages of spermatogenesis,  e.g. DDX4, which is key to the differentiation  of primordial germ cells and spermatogonia  [60] and UBE2B is involved in chromatin organization  of meiotic cells [61]. Microarray studies have  shown that the transcript profiles of oligozoospermic  patients also display a massive down-regulation  of transcripts involved in germ cell development and  spermatogenesis [54].  Asthenozoospermia  Asthenozoospermia is characterized by reduced  sperm motility or the absence of motile sperm in  greater than 35% of the spermatozoa examined.  Although a high percentage of oligozoospermic  patients also present low sperm motility, some infertile  patients have a normal sperm count but very poor  sperm motility. This can be caused by the presence  of ultrastructural anomalies, seminal infections and  antisperm antibodies. In addition, different habits  such as smoking, alcohol intake or a poor diet have  also been associated with asthenozoospermia [62–64].  In contrast to oligozoospermia, RT-PCR studies  revealed that asthenozoospermic patients present  alterations of RNAs associated with sperm maturation  or sperm metabolism in the latter stages of spermatogenesis.  For example, some of the transcripts  encode for sperm nuclear proteins such as protamines  62  https:/www.cambridge.org/core/terms. https://doi.org/10.1017/9781316411124.006  Downloaded from https:/www.cambridge.org/core. Boston University Theology Library, on 28 May 2017 at 16:50:26, subject to the Cambridge Core terms of use, available at  Chapter 4: Sperm RNA and Its Use as a ClinicalMarker  Table 4.1 Summary of studies showing altered spermatozoa transcripts and disrupted pathways associated with altered seminograms.  Male infertility  phenotype  GO biological altered  (microarray) Transcripts altered (RT-PCR) References  Oligozoospermic  Protein–lipid modification  mRNA transcription  Spermatogenesis and motility  Protein targeting and localization,  phospholipid metabolism  Nucleoside, nucleotide and  nucleic acid metabolism  DDX4  HSPA4, HSF1, HSF2  TPD52L3, PRM2, JMJD1A, NIPBL  Guo et al., 2007 [57]  Ferlin et al., 2010 [59]  Montjean et al., 2012 [54]  Asthenozoospermic UBE2B  PRM1, PRM2  HILS1, TNP1, TNP2  VDAC2  NFE2L2  Yatsenko et al., 2013 [58]  Kempisty et al., 2007 [65]  Jedrzejczak et al., 2007 [66]  Liu et al., 2010 [68]  Chen et al., 2012 [70]  Spermatid development  Ubiquinone biosynthesis pathway  Metabolic processes  Translation  Cell cycle  Localization  Protein transport  Meiosis  DNA repair  ANXA2, BRD2, OAZ3, PRM1, PRM2 Jodar et al., 2012 [53]  Bansal et al., 2015 [52]  Oligoasthenozoospermic CABYR  NTRK1  BDNF  Shen et al., 2015 [69]  Li et al., 2010 [79]  Zheng et al., 2011 [80]  Teratozoospermic Ubiquitin–proteasomal pathway  Apoptotic pathway  MAP kinase signalling pathway  Platts et al., 2007 [76]  Oligoteratozoospermic HSPA2 Cedenho et al., 2006 [78]  (PRM1 and PRM2) [65], transition nuclear proteins  (TNP1 and TNP2) and Histone Linker H1 Domain,  Spermatid-Specific 1 (HILS1) [66]. Changes in the  expression of sperm nuclear proteins are associated  with abnormal sperm chromatin condensation and  higher DNA damage, which might trigger apoptosis,  inactivating mitochondria and thereby affecting  sperm motility [67]. The expression of other transcripts  representative of processes associated with the  regulation of energy metabolism and sperm motility,  e.g. Voltage-Dependent Anion Channel 2 (VDCA2)  located in the mitochondrial outer membrane [68]  and Calcium-Binding Tyrosine-(Y)-Phosphorylation  Regulated (CABYR) [69], are also detected as altered.  Microarray-based discovery studies have confirmed  some of the trends brought forth by RT-PCR. In this  case, disturbances of later events of germ cell production  such as spermatid development were also  observed. Alterations in the energy production with  perturbations in ubiquinone biosynthesis pathways  and in different metabolic processes were also emphasized  [52, 53].  Different antioxidant trials have attempted to  negate the effects of reactive oxygen species known to  contribute to male infertility. It was hoped that seminal  parameters would improve, reflecting onmale fertility.  Consistent with the hypothesis, nuclear factorNFE2L2  RNA was down-regulated in asthenozoospermic  patients. Interestingly, this transcription factor regulates  the expression of several antioxidant enzymes  [70]. This is consistent with the beneficial effect of  antioxidant intake therapy on sperm motility [71].  Teratozoospermia  Teratozoospermia is diagnosed when less than 4%  of sperm have normal morphology evaluated using  Kruger’s strict criteria [72]. However, sperm morphology  has not been consistently predictive of fecundity,  and its etiology essentially remains unexplored  by conventional approaches. The one exception is  63  https:/www.cambridge.org/core/terms. https://doi.org/10.1017/9781316411124.006  Downloaded from https:/www.cambridge.org/core. Boston University Theology Library, on 28 May 2017 at 16:50:26, subject to the Cambridge Core terms of use, available at  Chapter 4: Sperm RNA and Its Use as a Clinical Marker  globozoospermia, which is characterized by altered  differentiation of the Golgi apparatus/acrosome. Diseases  such as Hodgkin’s disease and celiac disease,  lifestyle factors and habits may also be contributing  factors [73–75].  A single microarray-based study has been published  which examined the teratozoospermic sperm  RNA profile [76]. As in asthenzoospermia, teratozoospermic  patients also present a deficiency of  spermatocyte and spermatid transcripts, indicating a  disruption of the later stages of spermatogenesis.  Specifically, the proteasome is broadly downregulated,  likely affecting sperm capacitation, thereby  impeding the hyperactive motility of spermatozoa  and the ability to undergo the acrosome reaction [77].  Combined Phenotypes  The three seminal parameters, sperm count, motility  and morphology, are usually interrelated and appear  coordinated in the individual. Oligozoospermia is  often accompanied by poor motility (asthenozoospermia)  and abnormal morphology (teratozoospermia),  which leads to even lower fertility. Alterations of  sperm RNAs involved in early spermatogenesis, but  not in later events, are observed in these collective  phenotypes. For example, a transcript encoding  heat-shock protein (HSPA2), essential for the maintenance  of the synaptonemal complex during meiosis,  is down-regulated in oligoteratozoospermic patients  [78]. Additionally, a receptor (Neurotrophic Tyrosine  Kinase, Receptor, Type 1; NTRK1) [79] and a nuclear  factor (Brain-Derived Neurotrophic Factor; BDNF)  [80], involved in the paracrine regulation of spermatogenesis,  are down-regulated in sperm from oligoasthenozoospermic  individuals. These results suggest  that alterations of sperm RNAs resulting in reduced  sperm production are more obvious than these affecting  sperm motility and/or morphology.  sncRNAs and Male Infertility  Few studies have been published describing the  sncRNA content of infertile individuals. Abu-Halima  et al. [81] used microarrays to analyze the sperm  miRNA content in infertile males with asthenozoospermia  (n = 9) and oligoasthenozoospermia  (n = 9) and compared the results with those of normozoospermic  individuals (n = 9). They identified  27 down-regulated and 50 up-regulated miRNAs in  spermfromasthenozoospermicpatients and44downregulated  and 42 up-regulated miRNAs in sperm from  oligoasthenozoospermic patients. Among these differentially  expressed miRNAs, five were selected (hsamiR-  34b, hsa-miR-34b, hsa-miR-34c-5p, hsa-miR-  429 and hsa-miR-122) and evaluated in a larger population  of infertilemales [82]. Based on these results, the  authors suggest this panel of five miRNAs as potential  biomarkers of impaired spermatogenesis in affected  individuals.  Recently Salas-Huetos et al. used TaqMan arrays  to evaluate the miRNA profiles in sperm from fertile  (n = 10) [83] and infertile individuals with a single  seminal parameter affected: oligozoospermia (n=10),  teratozoospermia (n = 10) and asthenozoospermia  (n=10) [84].The results showed a stable population of  ontologically related miRNAs corresponding to spermatogenesis  and embryogenesis in sperm from fertile  individuals. In comparison, each group of infertile  individuals presented a differential pattern ofmiRNAs.  These ‘altered’ profiles included 18 differential miRNAs  in the oligozoospermic group, 19 in the teratozoospermic  group and 32 in the asthenozoospermic  group. Interestingly, ontological analysis of predicted  targets of these differential miRNAs showed a direct  relationship with biological processes involved in the  specific seminal alterations present in each population.  Interestingly, certain miRNAs were correlated with  specific demographic parameters such as age (miR-  34b-3p), sperm motility (hsa-miR-629–3p) and sperm  concentration (hsa-miR-335–5p, hsa-miR-885–5p and  hsa-miR-152–3p), indicating that these miRNAs may  act as biomarkers for these attributes. Unfortunately,  studies describing the sperm piRNA cargo in infertile  individuals have yet to be published, and their functional  significance potential as biomarkers of fertility  remains to be determined.  Integrated Analysis of mRNAs and sncRNAs  Altered in Patients with Altered Sperm  Parameters  Differential patterns of mRNAs and miRNAs in  patients presenting altered seminal parameters (oligozoospermia,  asthenozoospermia and teratozoospermia)  suggests a possible functional relationship  between different RNA molecules. It is well known  that miRNAs regulate gene expression through  translational repression and/or mRNA deadenylation  and degradation (Figure 4.2A). Integrated analysis  is complex because each miRNA can target different  64  https:/www.cambridge.org/core/terms. https://doi.org/10.1017/9781316411124.006  Downloaded from https:/www.cambridge.org/core. Boston University Theology Library, on 28 May 2017 at 16:50:26, subject to the Cambridge Core terms of use, available at  Chapter 4: Sperm RNA and Its Use as a ClinicalMarker  Pathways altered in teratozoospermia and asthenozoospermia  Pathways altered in oligozoospermia  A  B  5  3 5  3UTR  Mitosis  Mitosis  spermatogonia  Spermatogenesis  Spermatid  development  Sperm motility  Sperm capacitation  has-miR-139-5p  hsa-miR-143-3p  hsa-miR-370  hsa-miR-198, hsa-miR-1305,  hsa-miR-432-3p  PSMC5, PSMD14, GABRB1  TNP1  VDAC2  2n GMPS  n  Meiosis  Spermiogenesis  Sperm function  (motility and  capacitation)  TRANSLATION INHIBITION/ mRNA DEGRADATION/ mRNA DEADENYLATION  AGO  Protein-coding region AAAA  Figure 4.2 Integrated analysis ofmRNAs and sncRNAs altered in patients with altered seminal parameters. (A) The mature  miRNAs, in conjunction with Argonaute (AGO) proteins, form a complex able to regulate gene silencing by translational repression followed  by mRNA deadenylation and degradation. (B) Results of an integrated analysis of mRNA and miRNA altered in patients presenting altered  sperm parameters (oligozoospermia, asthenozoospermia and teratozoospermia). A correlation of some miRNAs and the corresponding  predicted mRNA targets is apparent. For example, guanine monophosphate synthase (GMPS), which is essential during mitosis of  spermatogonia, is post-transcriptionally regulated by hsa-miRNA-139–5p, and both RNAs are altered in oligozoospermia. Several miRNAs  (hsa-miRNA-143–3p, hsa-miRNA-370, hsa-miR-198, hsa-miR-1305 and hsa-miR-432–3p) and their corresponding targets Transition nuclear  protein 1 (TNP1), Voltage-Dependent Anion Channel 2 (VDAC2), Proteasome (Prosome, Macropain) 26S Subunit, ATPase, 5 (PSMC5) and  Proteasome (Prosome, Macropain) 26S Subunit, Non-ATPase, 14 (PSMD14) and Gamma-Aminobutyric Acid (GABA) A Receptor, Beta 1  (GABARB1) are affected in asthenozoospermia or teratozoospermia. These RNAs have important roles during spermatid development (TNP1),  sperm motility (VDAC2) and sperm capacitation (PSMC5, PSMD14 and GABARB1).  mRNAs and a single mRNA can be regulated by  different miRNAs. The analysis showed that more  than 50% of predicted targets from the differentially  abundant miRNAs in one altered sperm parameter  were also potentially affected with other alterations of  seminal parameters. However, 278 predicted targets  were unique to oligozoospermia; 2,346 were unique  to asthenozoospermia and 1,531 were unique to  teratozoospermia. Only one known altered transcript  in oligozoospermia (Guanine Monophosphate  Synthase (GMPS)) was predicted to be targeted by  a miRNA known to be altered specifically in oligozoospermic  individuals (hsa-miR-139) [54, 83, 84].  GMPS is crucial for purine synthesis. Alterations  65  https:/www.cambridge.org/core/terms. https://doi.org/10.1017/9781316411124.006  Downloaded from https:/www.cambridge.org/core. Boston University Theology Library, on 28 May 2017 at 16:50:26, subject to the Cambridge Core terms of use, available at  Chapter 4: Sperm RNA and Its Use as a Clinical Marker  in purine content could alter DNA synthesis and  consequently, mitosis in spermatogonia (Figure 4.2B).  Five transcripts associated with asthenozooospermia  (Voltage-Dependent Anion Channel 2 (VDAC2),  Gamma-Aminobutyric Acid (GABA) A Receptor,  Beta 1 (GABARB1), Transition Nuclear Protein 1  (TNP1), C1D Nuclear Receptor Corepressor (C1D)  and Solute Carrier Family 25 (Mitochondrial Carrier;  Phosphate Carrier), Member 3 (SLC25A3)) are predicted  targets of four differentially expressed miRNAs  (hsa-miR-143, hsa-miR-370 and hsa-mir-432 and  has-miR-615) in asthenozoospermic individuals  (Figure 4.2B). hsa-miR-143 regulates both the nuclear  protein TNP1 and C1D, which is involved in the  recruitment of RNA to the exosome [52, 66, 83, 84].  Sperm motility is affected by and reflective of abnormal  chromatin condensation, but it is not known how  alterations of C1D could affect sperm function. Two  sperm channels involved in sperm metabolism and  capacitation (VDCA2 and GABRB1) are predicted to  be regulated by two differentially expressed miRNAs  in asthenozoospermic individuals (hsa-miR-370 and  hsa-mir-432, respectively) [53, 68, 83, 84]. Finally, one  differentially expressed miRNA in asthenozoospermia,  hsa-miR-615, regulates a solute mitochondrial  carrier (SCL25A3) associated with sperm motility  as identified using a microarray [52, 83, 84]. Two  subunits of the proteasome (Proteasome 26S Subunit,  ATPase, 5 (PSMC5) and Proteasome 26S Subunit,  Non-ATPase, 14 (PSMD14)) are highly disrupted in  teratozoospermia and regulated by two altered miRNAs  in teratozoospermic individuals (has-miR-198  and hsa-miR-1305) (Figure 4.2B). Functional analysis  of the potential targets of these seven miRNAs in the  specific phenotypes could elucidate the mechanism by  which miRNAs affect sperm concentration, motility  and morphology.  Spermatozoal RNAs in the  Reproductive Clinic  Reproductive treatments are typically recommended  to couples unable to conceive after one year of unprotected  intercourse.The appropriate fertility treatment  for each couple is established after both the female and  the male partner are evaluated. If any severe female  or male factor is identified, e.g. an ovulatory or tubular  disorder in females or a diagnosis of azoospermia  or severe oligoasthenozoospermia in males, assisted  reproductive technologies such as in vitro fertilization  (IVF) or intracytoplasmic sperm injection (ICSI)  are recommended. Less invasive treatments such as  the combination of ovarian hyperstimulation with  controlled timed intercourse (TIC) and intrauterine  insemination (IUI) are typically indicated as the first  treatment for couples with mild to moderate male or  female factor and unexplained infertility [85]. While  ART has proven to be invaluable for couples with  severely compromised semen parameters, the success  of TIC or IUI in infertile patients with normal seminal  parameters (normozoospermia) is unpredictable.  After three or four unsuccessful cycles, idiopathic  infertile couples are usually shunted to ART. Some  studies suggest the primary use ofART, even if seminal  parameters are normal, thereby avoiding failed fertilization  [86]. On one hand, the use of ART for treating  some idiopathic infertile couples might be costeffective  when compared to providing IUI followed by  ART in those cases that fail [87]. On the other hand,  a successful TIC or IUI cycle minimizes exposure of  the female partner to invasive treatments such as egg  collection.  NGS of spermatozoal RNAs has revealed a set  of sperm RNA elements that may indicate the best  fertility treatment for idiopathic infertile couples  [19]. RNA-seq can provide a greater resolution than  microarrays, as the distribution of sequencing reads,  not the individual probe, reflects transcription from  that region. Each individual annotated or unannotated  transcribed genomic region can be defined as a sperm  RNA element.This strategy allowed the identification  of 648 abundant sperm RNA elements (SREs) that  were essentially at equivalent levels across seven fertile  individuals, indicative of a natural conception. It  was observed that patients expressing all SREs were  more likely to achieve live birth (LB) by TIC/IUI (73%;  22 of 30 individuals) than those with one or more  SREs absent (27%; 3 of 11 males; two-tailed Fisher’s  exact test, P = 0.012).These findings suggested that in  those patients lacking at least one of the SREs, the earlier  use of ART would avoid unsuccessful IUI cycles.  Approximately one-third of the idiopathic infertile  males included in the study (19 of 56 infertile patients)  did not present the complete set of SREs, suggestive of  amale factor underlying the couple’s infertility. In contrast,  the presence of all SREs in the remaining 37 idiopathic  infertilemales could indicate the presence of an  unknown female or couple’s factor.  About 40% of the male fertility SREs were located  within exonic regions of genes known to be involved in  66  https:/www.cambridge.org/core/terms. https://doi.org/10.1017/9781316411124.006  Downloaded from https:/www.cambridge.org/core. Boston University Theology Library, on 28 May 2017 at 16:50:26, subject to the Cambridge Core terms of use, available at  Chapter 4: Sperm RNA and Its Use as a ClinicalMarker  Table 4.2 Sperm RNA elements (SRE) required for natural  conception (described by Jodar et al., 2015) located within  transcripts altered in different infertile phenotypes (see  Table 4.1).  Altered seminogram  Transcripts altered (number  of containing SRE)  Oligozoospermia DDX3X (1)  TAF10 (1)  TPT1 (1)  Asthenozoospermia OAZ3 (1)  TNP1 (2)  CANX (3)  BRD2 (7)  HSP90AB1 (1)  RPS24 (2)  EIF4G2 (2)  HNRNPC (1)  TBC1D3C (4)  CABYR (2)  Teratozoospermia SKP1 (1)  ODF2 (9)  RERE (4)  spermatogenesis, spermmotility, fertilization and processes  that precede implantation. As expected, some of  the altered transcripts detected by RT-PCR ormicroarrays  associated with alteration of one seminal parameter  (oligozoospermia, asthenozoospermia and teratozoospermia;  see Section 3.1) contain some of the  required SREs for natural conception (Table 4.2).  New Perspectives  The ability of sperm RNAs to inform male factor status  in idiopathic infertile couples, together with the  rapidly decreasing cost of NGS, suggests that deep  sequencing of sperm RNA could change the counselling  approach for couples seeking reproductive care.  The routine use of this technique in reproductive clinics  might also make it possible to evaluate the quality  of the genetic contribution from the male to the  embryo.With the use of suitable computational tools,  it may be possible to identify genetic variants reflected  in the RNA-seq data [88].This strategy may be useful  to evaluate the presence of single-nucleotide polymorphisms  associated with male infertility, as well as different  monogenic diseases or allelic imbalance.  It is well known that environmental contaminants  and lifestyle factors influence human fertility, which  may be reflected to different degrees in the spermatozoa  [89]. These changes can be transmitted to future  generations and may affect their health and fertility.  Several examples of epigenetic transgenerational  inherence are described in different animal models,  e.g. altered metabolism of rodent offspring due to  paternal diet (low protein or high fat diet) [90, 91]  or presence of cardiac malformations in zebrafish offspring  after paternal exposure to bisphenol A similar  to those after direct exposure [92]. Some authors suggest  that transgenerational epigenetic inherence could  reconcile the adaptation of species to new environments  to which the parents were exposed. This is  exemplified in the case of the innate defence traumatic  mechanism against predators transmitted by  olfactory receptors [93]. The concept is controversial,  as it implies transference of hereditary information  from soma to germline. A recent study from Cossetti  et al. (2014) proposed that somatic-cell-derived  RNAs can be transferred to epididymal spermatozoa,  likely through exosomes [94]. Growing evidence  points to spermatozoal RNAs, e.g. miRNAs, as causing  phenotypic variations in the progeny reflective of  the father’s life experience [95, 96]. One example is  the observation of a decreased fear response and the  presence of depressive symptoms in the offspring (F2)  of traumatized male mice [95]. Mice exposed to traumatic  early postnatal events showed a dysregulation of  several sperm miRNAs that target genes involved in  DNA, RNA and epigenetic regulation. The injection  of sperm RNAs from these stressed males into wild  type fertilized oocytes resulted in offspring with similar  behavioural disorders, suggesting that sperm RNAs  participate in epigenetic transgenerational inheritance.  Although F3 also showed similar behavioural  disorders, the population of sperm miRNA from F2  did not present any alterations. This suggests that  the information provided by altered miRNA in F1 is  reversibly rooted in the F2 genome, i.e. by an epigenetic  mechanism, possibly DNA methylation or chromatin  organization. Interestingly, it was observed that  diet-induced paternal obesity in humans and rodents  could disturb metabolic processes in female offspring  [91, 97]. Obese males also exhibit changes in sperm  miRNAs as well as altered sperm DNA methylation.  However, intervention through a diet and exercise  program during two complete spermatogenic cycles  appears to transmit a normal metabolic profile to  female offspring [98]. These findings suggest that the  use of spermatozoal RNAs in clinics may assist the  clinical care of male infertility and serve as a predictor  of childhood outcomes.  All the studies reviewed in this chapter are based  on the study of sperm RNAs. However, spermatozoa  67  https:/www.cambridge.org/core/terms. https://doi.org/10.1017/9781316411124.006  Downloaded from https:/www.cambridge.org/core. Boston University Theology Library, on 28 May 2017 at 16:50:26, subject to the Cambridge Core terms of use, available at  Chapter 4: Sperm RNA and Its Use as a Clinical Marker  account for only 5% of the ejaculate, while the remaining  95% corresponds to secretions from the epididymis,  prostate and seminal vesicles. These accessory  sex glands release a substantial number of exosomes,  containing a large repertoire of RNAs and proteins,  into the seminal fluid [99].The majority of seminal  fluid (_70% by volume) is produced by the seminal  vesicles, with these secretions being rich in fructose,  a sugar essential for the nutrition of the spermatozoa  during the transit to the oocyte. The secretions  fromthe prostate,which constitute approximately 20%  by volume, contain proteins required for the coagulation  and liquefaction of semen aswell as immune components.  These classes of protein typically serve varying  roles in intercellular interaction and determination  of immune properties. Seminal fluid plays amuch  greater role than simply being a medium to carry the  spermatozoa through the female reproductive tract.  New perspectives suggest that the seminal fluid also  provides an optimal environment for the development  and successful implantation of the preimplantation  embryo, and that its alteration may impact the success  of the early pregnancy [100].The integrative analysis of  spermand seminal fluid transcriptomic and proteomic  high-throughput data has revealed the active transit of  seminal fluid proteins required for sperm maturation  [101], promoting the inclusion of seminal fluid RNAs  in future studies ofmale infertility. Some seminal fluid  RNAs appear to provide additional molecular markers  of foci of spermatogenesis in azoospermic patients  thatmay foreshadow the likelihood of testicular sperm  extraction [102].  Conclusion  When a couple first visit a reproductive clinic, they initiate  a well-defined clinical protocol with the goal of  determining the etiology of their infertility and receive  advice on the treatment options, as well as the success  rate and costs. Currently, the evaluation of the male  partner is principally based on the analysis of seminal  parameters in order to exclude a severe male factor,  defined as the absence or immobility of spermatozoa.  ART is recommended to those patients with a  severe male or female factor. The counselling of 15–  30% of the patients diagnosed with unexplained infertility  [103] presents a unique challenge. Despite ARTs  having a high success rate in cases of unexplained  infertility, they present an increased risk to the female  patient when the use of less invasive techniques such  Deep sequencing of  spermatozoal RNAs and  miRNA may predict future  health problems in offspring  ART  ART  NO FEMALE FACTOR TIC/IUI  FEMALE FACTOR OR SEVERE MALE FACTOR  COMPLETE SET OF SRE  Sperm RNA Elements  analysis  Extensive physical and  molecular evaluation  Physical and seminal  parameters evaluation  MILD OR NO MALE  FACTOR  INCOMPLETE SET OF  NO FEMALE FACTOR SRE  Figure 4.3 Proposed practice for reproductive counselling in  couples displaying infertility. Reproductive counselli




ng of  infertile couples begins with an extensive physical and molecular  evaluation of female and a basic physical analysis and seminal  parameters evaluation in males. If any severe female or male factor  is discovered, assisted reproductive techniques such as in vitro  fertilization (IVF) or intracytoplasmic sperm injection (ICSI) are  recommended. In contrast, less invasive techniques such as  controlled timed intercourse (TIC) and intrauterine insemination  (IUI) are usually indicated as the first treatment for couples with mild  male factor or unexplained infertility but with unpredictable results.  RNA-seq has revealed a set of 648 sperm RNA elements (SREs) that  are able to predict the success rate of TIC and IUI in idiopathic  infertile couples. The absence of at least one required SRE suggests  the primary use of ART, thereby avoiding unsuccessful IUI cycles.  The future health of offspring may also be assessed with the use of  RNA-seq based on the ability to evaluate the genetic contribution.  as TIC or IUI may be successful. The current dilemma  faced by the couple is that there is no accepted clinical  screen that avoids unsuccessful IUI cycles. Spermatozoal  RNAs including long and sncRNAs, in tandem  with the seminal fluid RNAs, opens a window to counselling  idiopathic infertile patients (Figure 4.3). At  present, the 648 sperm RNAelements required for natural  conception appears to discern which idiopathic  infertile patients have a high likelihood of achieving  pregnancy using TIC or IUI while advising others to  directly undergo ART [19]. However, it remains to be  established whether sperm RNA can predict ART outcome  and provide information about the future health  of offspring.  

No comments: