2,542
Views
19
CrossRef citations to date
0
Altmetric
Review and Hypothesis

The relationship among sperm global DNA methylation, telomere length, and DNA fragmentation in varicocele: a cross-sectional study of 20 cases

, , , , , , , , & show all
Pages 95-104 | Received 11 Jul 2018, Accepted 18 Nov 2018, Published online: 02 Jan 2019

ABSTRACT

Varicocele pathophysiology is related to increased oxidative stress, which might result in loss sperm DNA integrity as well as in genomic instability. Sperm telomere shortening and loss of global DNA methylation are the main features of genomic instability, leading to cell senescence and death, whereas sperm DNA fragmentation (SDF) characterizes the loss of chromatin integrity. We hypothesize that sperm genomic stability and DNA integrity is reduced in infertile men with moderate and large-sized varicoceles, thus being candidate markers of sperm quality in varicocele-related infertility. Here, we assessed the sperm global DNA methylation, telomere length, and SDF in men with and without clinically palpable varicoceles. While the rates of SDF and telomere length were not statistically different between varicocele patients and controls, global sperm DNA methylation seems to be lower in men with varicocele (49.7% ± 20.7%) than controls (64.7% ± 17.1%). A negative correlation between SDF and sperm motility and a positive correlation between sperm morphology and telomere length were observed. Our results suggest that varicocele may result in genomic instability, in particular, global DNA hypomethylation. However, a large sample size may confirm these findings. The understanding of the molecular mechanisms involved in the pathophysiology of varicocele-related infertility may help to better select candidates for varicocele repair.

Introduction

Varicocele is a major cause of male infertility affecting 19–41% of men at reproductive age (Jarow Citation2001). The disease is characterized by an abnormal dilatation of scrotal veins in the pampiniform plexus (Miyaoka and Esteves Citation2012) and it can impair spermatogenesis through several distinct pathophysiological mechanisms. Current evidence supports oxidative stress as a central element in the pathophysiology of varicocele-related infertility, although other mechanisms including scrotal hyperthermia, hormonal disturbances, testicular hypoperfusion and hypoxia as well as backflow of toxic metabolites are potential contributors (Naughton et al. Citation2001; Agarwal et al. Citation2012; Sheehan et al. Citation2014). The semen of infertile men with clinically palpable varicoceles is often abnormal, including alterations in sperm count, motility, and morphology (Xue et al. Citation2012; Kadioglu et al. Citation2014; Agarwal et al. Citation2016). Furthermore, an increase proportion of sperm with damaged chromatin is commonly seen in ejaculates of such men (Zini and Dohle Citation2011; Wang et al. Citation2012; Roque and Esteves Citation2018), presumably due to overproduction of reactive oxygen species (ROS) and decreased seminal antioxidant capacity (Shiraishi et al. Citation2012). It has been also suggested that the decrease in sperm chromatin integrity may result from loss of global DNA methylation and sperm DNA fragmentation (SDF) (Tavalaee et al. Citation2015). DNA methylation, along with other epigenetic markers, such as histone modification, non-coding RNAs, and telomere, play a critical role in maintaining chromatin stability (Ng and Bird Citation1999; O’Sullivan and Karlseder Citation2010).

A complex molecular pathway strictly related to environmental condition seems to contribute to the development of varicocele and may aggravate this condition (Benoff and Gilbert Citation2001; Santana et al. Citation2017; Hassanin et al. Citation2018). DNA methylation, in particular, is important for maintaining telomere integrity since hypomethylation of subtelomeric regions relates to telomere shortening as well as cell senescence and apoptosis (Vera et al. Citation2008; Yehezkel et al. Citation2008). Telomeres are one of the most important biological markers of genomic stability, protecting against DNA damage, and ensuring the correct chromosome alignment during DNA replication (Farr et al. Citation1991; O’Sullivan and Karlseder Citation2010). Excessive telomere erosion is associated with decreased sperm concentration, motility, and vitality (Ferlin et al. Citation2013; Cariati et al. Citation2016; Rocca et al. Citation2016), and telomere shortening has been suggested as an important marker of male infertility (Thilagavathi et al. Citation2013). DNA methylation and telomere integrity are not only important for sperm genomic stability but also for normal fertilization and embryo development (Dada et al. Citation2012; Jenkins and Carrell Citation2012; Thilagavathi et al. Citation2013).

In this pilot study, we postulate that sperm global DNA methylation and telomere length are altered in infertile men with varicocele (). Furthermore, we hypothesize that (epi)genomic stability lead to poor seminal quality and SDF in men with varicocele-related infertility.

Figure 1. An altered testicular environment with factors such as thermal and oxidative stress, hypoxia and toxic metabolite accumulation can lead to reduced testicular function and cause alterations in the sperm DNA, like telomere shortening and DNA methylation changes. Alterations in DNA are related to loss of genomic stability, which can directly affect seminal quality, fertilization, and correct embryonic development.

Figure 1. An altered testicular environment with factors such as thermal and oxidative stress, hypoxia and toxic metabolite accumulation can lead to reduced testicular function and cause alterations in the sperm DNA, like telomere shortening and DNA methylation changes. Alterations in DNA are related to loss of genomic stability, which can directly affect seminal quality, fertilization, and correct embryonic development.

Sperm DNA fragmentation

SDF has a negative correlation with sperm concentration, motility, and morphology, leading to poor seminal quality that may result in infertility (Shen et al. Citation2002; Peluso et al. Citation2013). The loss of sperm DNA integrity may be due to genetic factors, such as gene mutation, chromosomal abnormalities, and altered gene expression. Other important modulators of DNA damage are the environmental and lifestyle, including exposure to toxins and pollutants, lifestyle (cigarette smoking, use of illicit and licit drugs with gonadotoxic potential), exposure to chemo-radiation, conditions that increase the DNA fragmentation and alter cell cycle division (Andrabi Citation2007; Aitken et al. Citation2009).

Men with palpable varicocele may have increased SDF, especially those with poor seminal quality (Saleh et al. Citation2003; Enciso et al. Citation2006; Smith et al. Citation2006; Blumer et al. Citation2008; Zini and Dohle Citation2011; Santana et al. Citation2015). The increase in SDF is usually accompanied by changes in the markers of oxidative stress and sperm function (Roque and Esteves Citation2018).

The main processes associated with the physiopathology of varicocele (thermal and oxidative stress, hypoxia, and increased apoptosis) are likely interconnected (Hassanin et al. Citation2018) and SDF seems to be their end point (Esteves and Agarwal Citation2016). The major sources of ROS in men with varicocele are the epididymis cells, endothelial cells of the dilated pampiniform plexus, developing germ cells, Leydig cells, macrophages, and peritubular cells (Esteves and Agarwal Citation2016). Excessive ROS leads to peroxidation of the sperm plasma membranes (Aitken et al. Citation2014) and results in a cascade of events that generates nuclear and mitochondrial DNA damage, including base modification, cross linking, and chromatin remodeling (Aitken and De Iuliis Citation2010; Sakkas and Alvarez Citation2010).

Sperm DNA methylation

DNA methylation controls gene expression by histone modification, chromatin condensation, and gene silencing (Rice and Allis Citation2001). DNA methylation occurs throughout the course of development in multicellular organisms and ensures the maintenance of tissue and lifetime-specific gene expression patterns (Urdinguio et al. Citation2015).

In mammals, DNA methylation occurs especially in cytosines followed by guanines (Cytosine-phosphate-Guanine, CpG), and consists of the covalent addition of a methyl group at position 5 of cytosine (Lister et al. Citation2009). DNA methyltransferases (DNMTs) mediate the transfer of the methyl group to the DNA (Lan et al. Citation2010; Jin et al. Citation2011). The loss of DNMT activity or changes in DNA sequence may lead to global hypomethylation of the genome (Tunc and Tremellen Citation2009).

DNA methylation is a primary mechanism of developmental processes, such as genomic imprinting (Feinberg et al. Citation2002), X-Chromosome inactivation (Payer and Lee Citation2008), silencing of transposable elements (Doerfler Citation1991), DNA compaction and post-meiotic gene silencing (Oakes et al. Citation2007). Epigenetic reprogramming during gametogenesis is essential for the fertilization process and embryo development, in which a second round of reprogramming during embryo development establish epigenetic markers for embryo growth and maintain paternal imprinting (Hammoud et al. Citation2009). During spermatogenesis, DNA methylation remains stable, and the sperm genome contains remnant epigenetic signatures of this process, in addition to tags that influence fertilization and embryonic development (Jenkins et al. Citation2017).

The sperm DNA methylation pattern is a predictor of fertilization capacity and embryonic quality (Jenkins and Carrell Citation2012; Aston et al. Citation2015). Camprubí et al (Camprubí et al. Citation2016) identified 696 CpG islands related to spermatogenesis differentially methylated in sperm from fertile compared to infertile men. Altered DNA methylation has been associated with decreased reproductive capacity, mainly related to low sperm count (Benchaib et al. Citation2003; Houshdaran et al. Citation2007; Kobayashi et al. Citation2007; Marques et al. Citation2008; Hammoud et al. Citation2010; Aston et al. Citation2012; Krausz et al. Citation2012; Jenkins et al. Citation2015; Urdinguio et al. Citation2015) and reduced rates of pregnancy (Benchaib et al. Citation2003, Citation2005).

To our knowledge, only two studies evaluated sperm global DNA methylation in men with varicocele. Bahreinian et al. (Citation2015) found DNA hypomethylation in men with varicocele when compared to fertile individuals, which had a negative relation with SDF. The same group also investigated the sperm global DNA methylation levels before and 3 months after varicocelectomy. The authors found a tendency to higher rates of methylation after surgery, although differences were not significant (Tavalaee et al. Citation2015). With the categorization of individuals into subgroups, the increase in global DNA methylation appears to have been higher in oligozoospermic men (Tavalaee et al. Citation2015).

Telomeres

In mammals, telomeres consist of non-coding (5ʹ-TTAGGG-3 ‘) repeating DNA sequences associated with proteins, which form the ends of eukaryotic chromosomes (Zalenskaya et al. Citation2000). Telomeres prevent the recognition of the end of the strands as a double strand break, protecting against DNA degradation, and allowing the correct alignment of the chromosomes during the replication, playing an important role in the regulation of genomic stability (Farr et al. Citation1991; O’Sullivan and Karlseder Citation2010).

Telomeres, shortened at each cell division, are the main biological marker of aging and limit the proliferative time of cells (Bonetti et al. Citation2013). However, to avoid excessive loss, the maintenance of telomeres is guaranteed by an enzymatic complex, consisting of a reverse transcriptase and an RNA template, called telomerase (Blackburn Citation1991).

The maintenance of the telomere length involves an interaction between factors that shorten it and those that extend it. In addition to cell division, telomere shortening can be intensified by extrinsic environmental factors, such as environmental pollutants, radiation, aging, lifestyle, and oxidative stress (von Zglinicki Citation2000; Zhang et al. Citation2013; Sharma et al. Citation2015). The increase of the inflammatory activity is related to shortening of the telomeres and occurs due to the increase of ROS that damages the telomeric DNA (O’Donovan et al. Citation2011; Wong et al. Citation2014a).

Changes in telomere homeostasis may affect the synapse of homologous chromosomes and recombination in spermatocytes that will undergo meiosis I, so that telomere impairment may be indicative of a phenotype of sterility (Thilagavathi et al. Citation2013; Reig-Viader et al. Citation2014). Studies found that the sperm telomere length is related to sperm count since oligozoospermic men have shorter telomeres than normozoospermic men (Ferlin et al. Citation2013; Cariati et al. Citation2016). The telomere length has also been reported associated positively with progressive motility and vitality, and negatively with SDF and protamination (Rocca et al. Citation2016).

However, to our knowledge, there are no studies that have sought to evaluate the role of telomeres in the pathophysiology of varicocele. As telomeric shortening can generate accelerated sperm aging and might be associated with DNA fragmentation, leading to infertility, we sought to investigate this association in infertile men with varicocele. Our results might help unravel a novel pathophysiology mechanism in varicocele, and introduce a potential sperm quality biomarker in these patients.

Hypothesis

Varicocele is a major cause of male infertility as it adversely impacts semen parameters, the molecular and ultrastructural characteristics of spermatozoa, and the testicular microenvironment. However, the mechanism by which varicocele impairs fertility is not fully elucidated. Since methylation levels were related to environmental changes and DNA damage, and this epigenetic mark has a regulatory role in telomere length (Wong et al. Citation2014b; Udomsinprasert et al. Citation2016), we hypothesized that alterations in DNA methylation and telomere length may occur in men with varicocele, being related to poor seminal quality and the phenotype of infertility and this was tested in a pilot study. This study tested the hypothesis that the decrease of fertility in men with varicocele may be due to alterations in the sperm global DNA methylation and telomere length, negative consequences for seminal quality and SDF. In this pilot study, 20 men with varicocele and 20 healthy controls without the disease were investigated. The seminal quality was evaluated by conventional semen analysis and the SDF was analyzed by sperm chromatin dispersion assay. The global DNA methylation was evaluated by enzyme-linked immunosorbent assay (ELISA) and telomere content by quantitative PCR (q-PCR) as described in Supplemental File 1 Materials and Methods.

Preliminary results

Of the 20 patients with varicocele included, 10 (50%) had grade 3 left-sided varicocele; 7 (35%) had grade 2 left-sided varicocele; and 3 (15%) had bilateral varicocele. The seminal parameters, SDF, telomere length and global DNA methylation of men with varicocele and controls are presented in .

Table 1. Comparative analysis of quantitative variables measured in the varicocele and control groups.

Sperm concentration, progressive motility, and sperm morphology were significantly lower in men with varicocele than controls (). SDF rates were higher in varicocele patients than and controls, albeit of not statistical significance (37.0 ± 20.0% vs. 26.0 ± 10.0%, p = 0.09) ()). Telomere length was not different between men with varicocele (3.56 ± 1.04) and controls (3.57 ± 0.8, p = 0.82) ()). As for global DNA methylation analysis, we observed reduced methylation level in the varicocele group (49.74% ± 20.75%) compared to the control group (64.66% ± 7.08%) ()), however we could not do statistical analysis due to limited sample size.

Figure 2. Variables analyzed in the varicocele and control groups. (A) DNA fragmentation index; (B) Telomere content (T/S ratio); (C) Percentage of global DNA Methylation (p > 0.05) .

Figure 2. Variables analyzed in the varicocele and control groups. (A) DNA fragmentation index; (B) Telomere content (T/S ratio); (C) Percentage of global DNA Methylation (p > 0.05) .

There was no statistical difference in the outcome measures mentioned above between men with grade 2 and 3 varicoceles ().

Table 2. Comparative analysis of quantitative variables among varicocele patients according to varicocele grades 2 and 3 (G2 and G3).

A negative correlation was observed between SDF and progressive motility (p = 0.04), whereas a positive correlation was noted between SDF and the percentage of immotile spermatozoa (p = 0.01). We also observed a positive correlation between sperm morphology and telomere length (p = 0.05). No other significant correlations were observed when evaluating other variables ().

Table 3. Spearman’s correlation matrix between the variables evaluated in the spermogram and SDF, GDM and telomere length in the group with varicocele.

The participants were young (mean age, 27.6 years) and within the reproductive age. Notably, the group of men with varicocele had lower sperm concentration, decreased percentage of sperm with progressive motility, and increased percentage of sperm with abnormal morphology than those without varicocele. These findings are in accordance with previous results regarding poor seminal quality in these patients (Xue et al. Citation2012; Kadioglu et al. Citation2014).

Although the effect of varicocele on conventional seminal parameters is well reported, the genetic and epigenetic alterations are still poorly understood. Recent evidence indicates that SDF is increased in sperm from varicocele, which might be related to genomic instability (Saleh et al. Citation2003; Smith et al. Citation2006; Blumer et al. Citation2008; Zini and Dohle Citation2011; Wright et al. Citation2014; Santana et al. Citation2015; Roque and Esteves Citation2018). Thus, the evaluation of important biological markers of genomic stability, such as SDF, telomere length, and global DNA methylation, is important to better understand the underlying mechanisms leading to infertility in men with varicocele. Telomeres were similar between varicocele and controls. However, the sperm chromatin integrity and global DNA methylation were reduced in men with varicocele, albeit not reaching statistical significance, due to the small sample size given our study was designed as a proof of concept pilot study.

The global DNA methylation is closely related to SDF because genomic hypomethylation facilitates chromosomal rearrangements that lead to DNA damage, thus making it an important epigenetic marker of genomic stability and cell viability (Jones and Baylin Citation2002; Deaton and Bird Citation2011). In our preliminary results, we noted that men with varicocele exhibited a lower percentage of global DNA methylation in sperm. Only two studies have reported similar findings, both of which reported hypomethylation in men with varicocele (Bahreinian et al. Citation2015; Tavalaee et al. Citation2015). The loss of methylation might make the sperm DNA more susceptible to damage, creating a negative correlation between global DNA methylation and the SDF (Bahreinian et al. Citation2015). In this study, we observed a lower percentage of DNA methylation in men with grade 3 varicocele compared with men with grade 2 varicocele, suggesting that the disease severity could have a differential impact on sperm methylation.

To our knowledge, our preliminary data is the first attempting to evaluate sperm telomere length in men with varicocele. Telomeric changes can affect the recombination of homologous chromosomes in spermatocytes undergoing meiosis and may be indicative of a sterile phenotype (Reig-Viader et al. Citation2014). Spermatozoa with shorter telomeres exhibit increased DNA fragmentation (Moskovtsev et al. Citation2010) and excessive telomere shortening is a known trigger of apoptotic DNA fragmentation (Blasco Citation2003). As telomere shortening can lead to accelerated testicular aging and infertility, we believe that further studies of telomere length may be crucial to better understanding the mechanisms involved in seminal quality of patients with varicocele.

Current evidence supports the concept that oxidative stress plays a key role in the pathophysiology of varicocele-related infertility (Hamada et al. Citation2013). Excessive ROS leads to higher rates of DNA damage, consequently leading to poor sperm quality and lower fertility rates (Koksal et al. Citation2003; Agarwal et al. Citation2012; Muratori et al. Citation2015; Cho et al. Citation2016). Therefore, the semen from patients with varicocele contains a greater proportion of spermatozoa with abnormal and immature chromatin than in that obtained from men without the disease (Saleh et al. Citation2003; Smith et al. Citation2006; Blumer et al. Citation2008; Zini and Dohle Citation2011; González-Marín et al. Citation2012; Wright et al. Citation2014; Santana et al. Citation2015). Although our study revealed no statistical difference between the varicocele and control groups, the higher SDF values observed in the varicocele group should be investigated further.

The relationship between varicocele grade and the degree of sperm quality deterioration is controversial. Some studies have reported that grade 3 varicocele is associated with a more pronounced decrease in sperm quality (Ishikawa and Fujisawa Citation2005; Al-Ali, Marszalek, et al. Citation2010; Al-Ali, Shamloul, et al. Citation2013), while others have demonstrated that disease grade does not directly affect sperm quality (Diamond et al. Citation2007; Zargooshi Citation2007; Shiraishi et al. Citation2010). Our findings corroborate the hypothesis that varicocele, regardless of grade, negatively affects semen quality.

Despite no differences in the genomic instability parameters evaluated, a relationship between SDF and sperm motility, including a negative correlation with progressive motility and a positive correlation with the percentage of immobile spermatozoa, was observed. This finding is in agreement with published reports (Peluso et al. Citation2013), and this mechanism may be due to the presence of ROS or other types of agents which compromise the energetic metabolism of gametes in men with varicocele (Armstrong et al. Citation1999). Therefore, the present results also point toward a positive correlation between morphology and telomere length, and to our knowledge no other reports on this matter exists. Defects in sperm maturation are mirrored in sperm morphology (Ghasemian et al. Citation2015), and a correlation may exist between changes in genomic stability (e.g., that caused by telomere length modifications) and the formation of sperm with morphological abnormalities.

The variability in the clinical phenotypes of varicocele suggests that genetic and/or epigenetic factors may play an important role in the disease etiology, the mechanisms of which are still not well understood (Santana et al. Citation2017). Global DNA hypomethylation may be a candidate factor as it has been associated with shorter telomere length (Wong et al. Citation2014b; Udomsinprasert et al. Citation2016). By contrast, global hypermethylation in the subtelomeric regions confers protection to telomeres and helps maintain genomic stability (O’Sullivan and Karlseder Citation2010; Deaton and Bird Citation2011). Disturbances in these processes may lead to DNA fragmentation and, consequently, to decreased sperm quality and fertility in men with varicocele, a phenomenon that warrants further investigation.

The main limitation of our pilot study is the small sample size. Also, a few specimens had insufficient DNA quality to perform telomere length and global DNA methylation analyzes, thus we could not evaluate these parameters in all men included in the study. We used the Mann–Whitney test to compare the variables between the groups, but since the sample size in the global DNA methylation evaluation was very small, we elected to present the results descriptively only.

Our preliminary results suggest that sperm global methylation seems to be reduced in men with clinically palpable varicocele and might affect sperm chromatin integrity. Large cohorts and well-defined phenotypes are required to reveal the true picture of the association of varicocele with the analyzed parameters. However, it should be noted that these data are the first evaluation of sperm telomere length in men with varicocele. Further studies with larger sample size are essential to confirm this hypothesis and provide a better understanding of the underlying genetic and epigenetic mechanisms associated with varicocele, which may be influencing the infertility phenotype in these men.

Supplemental material

Supplemental Material

Download MS Word (18.7 KB)

Acknowledgments

The authors are extremely grateful to the study participants and their families. We appreciated the technical assistance of Marilda Hatsumi Yamada Dantas and Cristiana C. Padovan Ribas.

Disclosure statement

No potential conflict of interest was reported by the authors.

Supplementary material

Supplemental data for this article can be accessed here.

Additional information

Funding

This work was supported by the Conselho Nacional de Desenvolvimento Científico e Tecnológico [478689/2013-6], Fundação de Amparo a Pesquisa do Estado de São Paulo (FAPESP), under the fellowship number 2012/11069-9 (CLMF), Coordenação de Aperfeiçoamento de Pessoal de Nível Superior (CAPES) - Programa de Excelência Acadêmica (PROEX) e Programa de Apoio à Pós-Graduação (PROAP).

Notes on contributors

Rosana Maria dos Reis

Conception and experiments design: VPS, CLM-F, RMR; Measurement of SDF and semen analysis: VPS, MACV; Performance and analysis of global DNA methylation: VPS, CLM-F, MCE; Measurement of telomere content: VPS, CLM-F, DCCP; Writing of the paper: VPS, CLM-F; Contribution with reagents and materials, and critical review of the manuscript: ESR, RTC, RAF, SCE, RMR. All authors have approved the final version and submission of this manuscript.

References

  • Agarwal A, Hamada A, Esteves SC. 2012. Insight into oxidative stress in varicocele-associated male infertility: part 1. Nat Rev Urol. 9(12):678–690.
  • Agarwal A, Sharma R, Harlev A, Esteves SC. 2016. Effect of varicocele on semen characteristics according to the new 2010 World Health Organization criteria: a systematic review and meta-analysis. Asian J Androl. 18(2):163–170.
  • Aitken RJ, De Iuliis GN. 2010. On the possible origins of DNA damage in human spermatozoa. Mol Hum Reprod. 16(1):3–13.
  • Aitken RJ, De Iuliis GN, McLachlan RI. 2009. Biological and clinical significance of DNA damage in the male germ line. Int J Androl. 32(1):46–56.
  • Aitken RJ, Smith TB, Jobling MS, Baker MA, De Iuliis GN. 2014. Oxidative stress and male reproductive health. Asian J Androl. 16(1):31–38.
  • Al-Ali BM, Marszalek M, Shamloul R, Pummer K, Trummer H. 2010. Clinical parameters and semen analysis in 716 Austrian patients with varicocele. Urology. 75(5):1069–1073.
  • Al-Ali BM, Shamloul R, Pichler M, Augustin H, Pummer K. 2013. Clinical and laboratory profiles of a large cohort of patients with different grades of varicocele. Cent Eur J Urol. 66(1):71–74.
  • Andrabi SM. 2007. Mammalian sperm chromatin structure and assessment of DNA fragmentation. J Assist Reprod Genet. 24(12):561–569.
  • Armstrong JS, Rajasekaran M, Chamulitrat W, Gatti P, Hellstrom WJ, Sikka SC. 1999. Characterization of reactive oxygen species induced effects on human spermatozoa movement and energy metabolism. Free Radic Biol Med. 26(7–8):869–880.
  • Aston KI, Punj V, Liu L, Carrell DT. 2012. Genome-wide sperm deoxyribonucleic acid methylation is altered in some men with abnormal chromatin packaging or poor in vitro fertilization embryogenesis. Fertil Steril. 97(2):285–292.
  • Aston KI, Uren PJ, Jenkins TG, Horsager A, Cairns BR, Smith AD, Carrell DT. 2015. Aberrant sperm DNA methylation predicts male fertility status and embryo quality. Fertil Steril. 104(6):1388–1397.e1381–e1385.
  • Bahreinian M, Tavalaee M, Abbasi H, Kiani-Esfahani A, Shiravi AH, Nasr-Esfahani MH. 2015. DNA hypomethylation predisposes sperm to DNA damage in individuals with varicocele. Syst Biol Reprod Med. 61(4):179–186.
  • Benchaib M, Ajina M, Lornage J, Niveleau A, Durand P, Guerin JF. 2003. Quantitation by image analysis of global DNA methylation in human spermatozoa and its prognostic value in in vitro fertilization: a preliminary study. Fertil Steril. 80(4):947–953.
  • Benchaib M, Braun V, Ressnikof D, Lornage J, Durand P, Niveleau A, Guerin JF. 2005. Influence of global sperm DNA methylation on IVF results. Hum Reprod. 20(3):768–773.
  • Benoff S, Gilbert BR. 2001. Varicocele and male infertility: part I. Preface. Hum Reprod Update. 7(1):47–54.
  • Blackburn EH. 1991. Structure and function of telomeres. Nature. 350(6319):569–573.
  • Blasco MA. 2003. Mammalian telomeres and telomerase: why they matter for cancer and aging. Eur J Cell Biol. 82(9):441–446.
  • Blumer CG, Fariello RM, Restelli AE, Spaine DM, Bertolla RP, Cedenho AP. 2008. Sperm nuclear DNA fragmentation and mitochondrial activity in men with varicocele. Fertil Steril. 90(5):1716–1722.
  • Bonetti D, Martina M, Falcettoni M, Longhese MP. 2013. Telomere-end processing: mechanisms and regulation. Chromosoma. 123(1–2):57–66
  • Camprubí C, Salas-Huetos A, Aiese-Cigliano R, Godo A, Pons MC, Castellano G, Grossmann M, Sanseverino W, Martin-Subero JI, Garrido N, et al. 2016. Spermatozoa from infertile patients exhibit differences of DNA methylation associated with spermatogenesis-related processes: an array-based analysis. Reprod Biomed Online. 33(6):709–719.
  • Cariati F, Jaroudi S, Alfarawati S, Raberi A, Alviggi C, Pivonello R, Wells D. 2016. Investigation of sperm telomere length as a potential marker of paternal genome integrity and semen quality. Reprod Biomed Online. 33(3):404–411.
  • Cho CL, Esteves SC, Agarwal A. 2016. Novel insights into the pathophysiology of varicocele and its association with reactive oxygen species and sperm DNA fragmentation. Asian J Androl. 18(2):186–193.
  • Dada R, Kumar M, Jesudasan R, Fernandez JL, Gosalvez J, Agarwal A. 2012. Epigenetics and its role in male infertility. J Assist Reprod Genet. 29(3):213–223.
  • Deaton AM, Bird A. 2011. CpG islands and the regulation of transcription. Genes Dev. 25(10):1010–1022.
  • Diamond DA, Zurakowski D, Bauer SB, Borer JG, Peters CA, Cilento BG, Paltiel HJ, Rosoklija I, Retik AB. 2007. Relationship of varicocele grade and testicular hypotrophy to semen parameters in adolescents. J Urol. 178(4 Pt 2):1584–1588.
  • Doerfler W. 1991. Patterns of DNA methylation–evolutionary vestiges of foreign DNA inactivation as a host defense mechanism. Biol Chem Hoppe Seyler. 372(8):557–564.
  • Enciso M, Muriel L, Fernández JL, Goyanes V, Segrelles E, Marcos M, Montejo JM, Ardoy M, Pacheco A, Gosálvez J. 2006. Infertile men with varicocele show a high relative proportion of sperm cells with intense nuclear damage level, evidenced by the sperm chromatin dispersion test. J Androl. 27(1):106–111.
  • Esteves SC, Agarwal A. 2016. Afterword to varicocele and male infertility: current concepts and future perspectives. Asian J Androl. 18(2):319–322.
  • Farr C, Fantes J, Goodfellow P, Cooke H. 1991. Functional reintroduction of human telomeres into mammalian cells. Proc Natl Acad Sci U S A. 88(16):7006–7010.
  • Feinberg AP, Cui H, Ohlsson R. 2002. DNA methylation and genomic imprinting: insights from cancer into epigenetic mechanisms. Semin Cancer Biol. 12(5):389–398.
  • Ferlin A, Rampazzo E, Rocca MS, Keppel S, Frigo AC, De Rossi A, Foresta C. 2013. In young men sperm telomere length is related to sperm number and parental age. Hum Reprod. 28(12):3370–3376.
  • Ghasemian F, Mirroshandel SA, Monji-Azad S, Azarnia M, Zahiri Z. 2015. An efficient method for automatic morphological abnormality detection from human sperm images. Comput Methods Programs Biomed. 122(3):409–420.
  • González-Marín C, Gosálvez J, Roy R. 2012. Types, causes, detection and repair of DNA fragmentation in animal and human sperm cells. Int J Mol Sci. 13(11):14026–14052.
  • Hamada A, Esteves SC, Agarwal A. 2013. Insight into oxidative stress in varicocele-associated male infertility: part 2. Nat Rev Urol. 10(1):26–37.
  • Hammoud SS, Nix DA, Zhang H, Purwar J, Carrell DT, Cairns BR. 2009. Distinctive chromatin in human sperm packages genes for embryo development. Nature. 460(7254):473–478.
  • Hammoud SS, Purwar J, Pflueger C, Cairns BR, Carrell DT. 2010. Alterations in sperm DNA methylation patterns at imprinted loci in two classes of infertility. Fertil Steril. 94(5):1728–1733.
  • Hassanin AM, Ahmed HH, Kaddah AN. 2018. A global view of the pathophysiology of varicocele. Andrology. 6:654–661.
  • Houshdaran S, Cortessis VK, Siegmund K, Yang A, Laird PW, Sokol RZ. 2007. Widespread epigenetic abnormalities suggest a broad DNA methylation erasure defect in abnormal human sperm. PLoS One. 2(12):e1289.
  • Ishikawa T, Fujisawa M. 2005. Effect of age and grade on surgery for patients with varicocele. Urology. 65(4):768–772.
  • Jarow JP. 2001. Effects of varicocele on male fertility. Hum Reprod Update. 7(1):59–64.
  • Jenkins TG, Aston KI, James ER, Carrell DT. 2017. Sperm epigenetics in the study of male fertility, offspring health, and potential clinical applications. Syst Biol Reprod Med. 63(2):69–76.
  • Jenkins TG, Aston KI, Trost C, Farley J, Hotaling JM, Carrell DT. 2015. Intra-sample heterogeneity of sperm DNA methylation. Mol Hum Reprod. 21(4):313–319.
  • Jenkins TG, Carrell DT. 2012. The sperm epigenome and potential implications for the developing embryo. Reproduction. 143(6):727–734.
  • Jin B, Li Y, Robertson KD. 2011. DNA methylation: superior or subordinate in the epigenetic hierarchy? Genes Cancer. 2(6):607–617.
  • Jones PA, Baylin SB. 2002. The fundamental role of epigenetic events in cancer. Nat Rev Genet. 3(6):415–428.
  • Kadioglu TC, Aliyev E, Celtik M. 2014. Microscopic varicocelectomy significantly decreases the sperm DNA fragmentation index in patients with infertility. Biomed Res Int. 2014:1–4.
  • Kobayashi H, Sato A, Otsu E, Hiura H, Tomatsu C, Utsunomiya T, Sasaki H, Yaegashi N, Arima T. 2007. Aberrant DNA methylation of imprinted loci in sperm from oligospermic patients. Hum Mol Genet. 16(21):2542–2551.
  • Koksal IT, Usta M, Orhan I, Abbasoglu S, Kadioglu A. 2003. Potential role of reactive oxygen species on testicular pathology associated with infertility. Asian J Androl. 5(2):95–99.
  • Krausz C, Sandoval J, Sayols S, Chianese C, Giachini C, Heyn H, Esteller M. 2012. Novel insights into DNA methylation features in spermatozoa: stability and peculiarities. PLoS One. 7(10):e44479.
  • Lan J, Hua S, He X, Zhang Y. 2010. DNA methyltransferases and methyl-binding proteins of mammals. Acta Biochim Biophys Sin. 42(4):243–252.
  • Lister R, Pelizzola M, Dowen RH, Hawkins RD, Hon G, Tonti-Filippini J, Nery JR, Lee L, Ye Z, Ngo QM, et al. 2009. Human DNA methylomes at base resolution show widespread epigenomic differences. Nature. 462(7271):315–322.
  • Marques CJ, Costa P, Vaz B, Carvalho F, Fernandes S, Barros A, Sousa M. 2008. Abnormal methylation of imprinted genes in human sperm is associated with oligozoospermia. Mol Hum Reprod. 14(2):67–74.
  • Miyaoka R, Esteves SC. 2012. A critical appraisal on the role of varicocele in male infertility. Adv Urol. 2012:597495.
  • Moskovtsev SI, Willis J, White J, Mullen JB. 2010. Disruption of telomere-telomere interactions associated with DNA damage in human spermatozoa. Syst Biol Reprod Med. 56(6):407–412.
  • Muratori M, Tamburrino L, Marchiani S, Cambi M, Olivito B, Azzari C, Forti G, Baldi E. 2015. Investigation on the origin of sperm DNA fragmentation: role of apoptosis, immaturity and oxidative stress. Mol Med. 21:109–122.
  • Naughton CK, Nangia AK, Agarwal A. 2001. Pathophysiology of varicoceles in male infertility. Hum Reprod Update. 7(5):473–481.
  • Ng HH, Bird A. 1999. DNA methylation and chromatin modification. Curr Opin Genet Dev. 9(2):158–163.
  • O’Donovan A, Pantell MS, Puterman E, Dhabhar FS, Blackburn EH, Yaffe K, Cawthon RM, Opresko PL, Hsueh WC, Satterfield S, et al. 2011. Cumulative inflammatory load is associated with short leukocyte telomere length in the health, aging and body composition study. PLoS One. 6(5):e19687.
  • O’Sullivan RJ, Karlseder J. 2010. Telomeres: protecting chromosomes against genome instability. Nat Rev Mol Cell Biol. 11(3):171–181.
  • Oakes CC, La Salle S, Smiraglia DJ, Robaire B, Trasler JM. 2007. Developmental acquisition of genome-wide DNA methylation occurs prior to meiosis in male germ cells. Dev Biol. 307(2):368–379.
  • Payer B, Lee JT. 2008. X chromosome dosage compensation: how mammals keep the balance. Annu Rev Genet. 42:733–772.
  • Peluso G, Palmieri A, Cozza PP, Morrone G, Verze P, Longo N, Mirone V. 2013. The study of spermatic DNA fragmentation and sperm motility in infertile subjects. Arch Ital Urol Androl. 85(1):8–13.
  • Reig-Viader R, Capilla L, Vila-Cejudo M, Garcia F, Anguita B, Garcia-Caldés M, Ruiz-Herrera A. 2014. Telomere homeostasis is compromised in spermatocytes from patients with idiopathic infertility. Fertil Steril. 102(3):728–738.e721.
  • Rice JC, Allis CD. 2001. Histone methylation versus histone acetylation: new insights into epigenetic regulation. Curr Opin Cell Biol. 13(3):263–273.
  • Rocca MS, Speltra E, Menegazzo M, Garolla A, Foresta C, Ferlin A. 2016. Sperm telomere length as a parameter of sperm quality in normozoospermic men. Hum Reprod. 31(6):1158–1163.
  • Roque M, Esteves SC. 2018. Effect of varicocele repair on sperm DNA fragmentation: a review. Int Urol Nephrol. 50(4):583–603.
  • Sakkas D, Alvarez JG. 2010. Sperm DNA fragmentation: mechanisms of origin, impact on reproductive outcome, and analysis. Fertil Steril. 93(4):1027–1036.
  • Saleh RA, Agarwal A, Sharma RK, Said TM, Sikka SC, Thomas AJ Jr. 2003. Evaluation of nuclear DNA damage in spermatozoa from infertile men with varicocele. Fertil Steril. 80(6):1431–1436.
  • Santana VP, Furtado CLM, Molina CAF, Nobre YTDA, Ferriani RA, Reis RM. 2015. A randomized clinical trial study of the e ffects of varicocelectomy on sperm clinical analysis and DNA fragmentation: a preliminary data. Gynecol Obstet Res Open J. 2(1):29–34.
  • Santana VP, Miranda-Furtado CL, de Oliveira-Gennaro FG, Dos Reis RM. 2017. Genetics and epigenetics of varicocele pathophysiology: an overview. J Assist Reprod Genet. 34(7):839–847.
  • Sharma R, Agarwal A, Rohra VK, Assidi M, Abu-Elmagd M, Turki RF. 2015. Effects of increased paternal age on sperm quality, reproductive outcome and associated epigenetic risks to offspring. Reprod Biol Endocrinol. 13:35.
  • Sheehan MM, Ramasamy R, Lamb DJ. 2014. Molecular mechanisms involved in varicocele-associated infertility. J Assist Reprod Genet. 31(5):521–526.
  • Shen HM, Dai J, Chia SE, Lim A, Ong CN. 2002. Detection of apoptotic alterations in sperm in subfertile patients and their correlations with sperm quality. Hum Reprod. 17(5):1266–1273.
  • Shiraishi K, Matsuyama H, Takihara H. 2012. Pathophysiology of varicocele in male infertility in the era of assisted reproductive technology. Int J Urol. 19(6):538–550.
  • Shiraishi K, Takihara H, Matsuyama H. 2010. Elevated scrotal temperature, but not varicocele grade, reflects testicular oxidative stress-mediated apoptosis. World J Urol. 28(3):359–364.
  • Smith R, Kaune H, Parodi D, Madariaga M, Rios R, Morales I, Castro A. 2006. Increased sperm DNA damage in patients with varicocele: relationship with seminal oxidative stress. Hum Reprod. 21(4):986–993.
  • Tavalaee M, Bahreinian M, Barekat F, Abbasi H, Nasr-Esfahani MH. 2015. Effect of varicocelectomy on sperm functional characteristics and DNA methylation. Andrologia. 47(8):904–909.
  • Thilagavathi J, Kumar M, Mishra SS, Venkatesh S, Kumar R, Dada R. 2013. Analysis of sperm telomere length in men with idiopathic infertility. Arch Gynecol Obstet. 287(4):803–807.
  • Tunc O, Tremellen K. 2009. Oxidative DNA damage impairs global sperm DNA methylation in infertile men. J Assist Reprod Genet. 26(9–10):537–544.
  • Udomsinprasert W, Kitkumthorn N, Mutirangura A, Chongsrisawat V, Poovorawan Y, Honsawek S. 2016. Global methylation, oxidative stress, and relative telomere length in biliary atresia patients. Sci Rep. 6:26969.
  • Urdinguio RG, Bayon GF, Dmitrijeva M, Torano EG, Bravo C, Fraga MF, Bassas L, Larriba S, Fernandez AF. 2015. Aberrant DNA methylation patterns of spermatozoa in men with unexplained infertility. Hum Reprod. 30(5):1014–1028.
  • Vera E, Canela A, Fraga MF, Esteller M, Blasco MA. 2008. Epigenetic regulation of telomeres in human cancer. Oncogene. 27(54):6817–6833.
  • von Zglinicki T. 2000. Role of oxidative stress in telomere length regulation and replicative senescence. Ann N Y Acad Sci. 908:99–110.
  • Wang YJ, Zhang RQ, Lin YJ, Zhang RG, Zhang WL. 2012. Relationship between varicocele and sperm DNA damage and the effect of varicocele repair: a meta-analysis. Reprod Biomed Online. 25(3):307–314.
  • Wong JY, De Vivo I, Lin X, Fang SC, Christiani DC. 2014a. The relationship between inflammatory biomarkers and telomere length in an occupational prospective cohort study. PLoS One. 9(1):e87348.
  • Wong JY, De Vivo I, Lin X, Grashow R, Cavallari J, Christiani DC. 2014b. The association between global DNA methylation and telomere length in a longitudinal study of boilermakers. Genet Epidemiol. 38(3):254–264.
  • Wright C, Milne S, Leeson H. 2014. Sperm DNA damage caused by oxidative stress: modifiable clinical, lifestyle and nutritional factors in male infertility. Reprod Biomed Online. 28(6):684–703.
  • Xue J, Yang J, Yan J, Jiang X, He LY, Wu T, Guo J. 2012. Abnormalities of the testes and semen parameters in clinical varicocele. Nan Fang Yi Ke Da Xue Xue Bao. 32(4):439–442.
  • Yehezkel S, Segev Y, Viegas-Péquignot E, Skorecki K, Selig S. 2008. Hypomethylation of subtelomeric regions in ICF syndrome is associated with abnormally short telomeres and enhanced transcription from telomeric regions. Hum Mol Genet. 17(18):2776–2789.
  • Zalenskaya IA, Bradbury EM, Zalensky AO. 2000. Chromatin structure of telomere domain in human sperm. Biochem Biophys Res Commun. 279(1):213–218.
  • Zargooshi J. 2007. Sperm count and sperm motility in incidental high-grade varicocele. Fertil Steril. 88(5):1470–1473.
  • Zhang X, Lin S, Funk WE, Hou L. 2013. Environmental and occupational exposure to chemicals and telomere length in human studies. Occup Environ Med. 70(10):743–749.
  • Zini A, Dohle G. 2011. Are varicoceles associated with increased deoxyribonucleic acid fragmentation? Fertil Steril. 96(6):1283–1287.

Reprints and Corporate Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

To request a reprint or corporate permissions for this article, please click on the relevant link below:

Academic Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

Obtain permissions instantly via Rightslink by clicking on the button below:

If you are unable to obtain permissions via Rightslink, please complete and submit this Permissions form. For more information, please visit our Permissions help page.