Article
Spermatid count as a predictor of response to FSH therapy

https://doi.org/10.1016/j.rbmo.2014.02.014Get rights and content

Abstract

This study evaluated the predictive power of spermatid count and cytology for assisted reproduction outcome after FSH therapy. A total of 174 men with severe oligozoospermia and normal plasma FSH concentration underwent semen analysis including spermatid count, TUNEL test, FISH analysis for sperm aneuploidies and testicular fine-needle aspiration cytology. Ninety-two men with hypospermatogenesis received FSH therapy for 3 months and 82 patients with maturative disturbance or partial obstruction served as controls. Semen was analysed at baseline, after FSH therapy and after 3- and 9-month follow up, and pregnancies were recorded. Subjects not reaching pregnancy at 3-month follow up were recommended assisted reproduction treatment. Spermatid count was related to testicular cytology: spermatid concentrations <0.01, 0.01–0.3 and >0.3 × 106/ml were predictive of partial obstruction, hypospermatogenesis and maturative disturbance. FSH therapy patients showed increases in sperm number and motility (both P < 0.001), allowing some couples to undergo intrauterine insemination instead of IVF. Cumulative pregnancy rate after 12 months was higher with FSH therapy (44.6%) than without (22.0%; P = 0.002). FSH therapy improved pregnancy rate and sometimes allowed less invasive assisted reproduction treatment in well-selected patients. Spermatid count could represent a new parameter to predict response to FSH therapy.

One-hundred seventy-four patients with severe reduction of sperm count and normal sex hormones plasma levels underwent semen analysis with spermatid count, and testicular fine needle aspiration cytologiy (FNAC). Ninety-two men infertile men with reduced sperm production (hypospermatogenesis) were treated with highly purified urofollitropin and 82 patients with sperm maturative defects or partial obstruction of the seminal tract served as controls. After treatment and after the following 3 and 9 months all subjects performed a new semen analysis and pregnancies were recorded. Subjects who had not reached spontaneous pregnancy were suggested to undergo assisted reproductive techniques (ARTs). Spermatid count was strongly related to testicular cytology: spermatid concentrations were predictive of partial obstruction, hypospermatogenesis and maturative disturbance respectively. Treated patients showed significant increase in sperm number and motility allowing some couples to undergo easier and less invasive assisted reproductive techniques. The number of pregnancies was significantly higher among treated (44.6%) than untreated couples (22.0%). Our data confirmed that FSH treatment can induce a significant improvemet of pergnancy rate and sometimes allows less invasive ARTs use in well selected severe oligozoospermic patients. Moreover, we suggest that spermatid count can be useful to define tubular status and could represent a new parameter to predict response to FSH therapy.

Introduction

Normal functioning of the gonadotrophic axis, in particular normal FSH secretion, plays crucial roles in spermatogenesis (Matsumoto, 1989, Sharpe, 1989), and several experimental and clinical studies have demonstrated the importance of FSH in regulating a normal quantitative spermatogenic process in animals and humans (Marshall et al., 1995, McLachlan et al., 1995, Moudgal et al., 1997). It has previously been demonstrated that FSH administration is able to increase sperm concentration, spermatogonal population and pregnancy rate in oligozoospermic subjects with normal plasma concentrations of gonadotrophins (Acosta et al., 1992, Bartoov et al., 1994, Glander and Kratzsch, 1997, Merino et al., 1996, Strehler et al., 1997). In addition, some authors reported that a significant improvement of sperm quality was evident after therapy. FSH therapy is useful in reducing the apoptotic process of spermatozoa and improving the qualitative properties of the axoneme, chromatin and acrosome (Baccetti et al., 1997).

Recently Colacurci et al. (2012) showed that the degree of sperm DNA fragmentation significantly decreases after 90 days of FSH therapy. However, reducing fragmentation cannot guarantee fertilization and embryo development success systematically. Some authors reported that genetic, in particular epigenetic causes, can lead to abnormal spermatogenesis, with potential implications for sperm quality, fertilization and embryo development (Jenkins and Carrell, 2012, Lalancette et al., 2009). In particular, Montjean et al. (2013) evaluated methylation profiles in mature sperm DNA from normozoospermic and oligozoospermic men by observing the presence of methylation changes in spermatozoa of the latter group.

Another potential mechanism of DNA damage that could be able to reduce the capability of spermatozoa to fertilize is related to DNA denaturation (Evenson and Wixon, 2008). It has been postulated that defective chromatin condensation during spermatogenesis could lead to spermatozoa unable to mature completely during spermiogenesis and therefore unable to repair DNA breaks during chromatin rearrangement occurring when histones are replaced by protamines (O’Brien and Zini, 2005, Zini and Libman, 2006). A positive effect on sperm DNA condensation after FSH therapy has been demonstrated in idiopathic infertile men (Kamischke et al., 1998).

These findings may explain the increase in oocyte fertilization and pregnancy rate, sometimes in absence of improved classic seminal parameters, that has been observed in couples undergoing assisted reproduction treatment in which the male partner received FSH (Acosta et al., 1991, Acosta et al., 1992, Ashkenazi et al., 1999). However, the improvement in seminal characteristics is variable and not much evident in many cases, and a proportion of patients do not respond to FSH therapy.

Oligozoospermia may be sustained by various alterations of the seminiferous epithelium that could explain the failure of FSH therapy reported by other studies (Attia et al., 2007, Bartoov et al., 1994, Foresta et al., 1995). Fine-needle aspiration cytology (FNAC) is a minimally invasive, rapid and effective procedure to evaluate the tubular status in oligozoospermic patients (Adhikari, 2009). This study group demonstrated that FSH therapy of oligozoospermic subjects with normal FSH plasma concentration, moderate hypospermatogenesis and absence of maturation disturbance is able to induce both a significant increase in sperm count (Foresta et al., 2002) and an improvement in pregnancy rate (Foresta et al., 2005). In contrast, when hypospermatogenesis is associated with maturative disturbance, FSH therapy shows no effect (Foresta et al., 2009). In the light of this consideration, selection criteria should be used to derive predictive information on the response to FSH therapy.

Some polymorphisms in the FSH receptor gene (FSHR) are able to influence the sensitivity of the receptor for the hormone and the reproductive parameters both in men and women (Aittomaki et al., 1995, Behre et al., 2005, Greb et al., 2005, Tüttelmann et al., 2012). On this basis, the current study group tested the hypothesis that FSHR sensitivity might be important in the response to FSH therapy and demonstrated that this treatment induces diverse improvement in seminal parameters according to defined FSHR genotypes; only subjects with at least one serine in position 680 (homozygotic AS/AS and heterozygotic TN/AS) had an improvement of seminal parameters (Selice et al., 2011). Again, molecular studies on the FSHB gene (coding for the β-subunit of FSH) showed that a G/T single-nucleotide polymorphism located in the FSHB gene promoter (−211 bp from the mRNA transcription start site; rs10835638) is responsible for the relative activity of endogenous FSH (Grigorova et al., 2007, Grigorova et al., 2008). Finally, research showed that oligozoospermic TT homozygotes for the FSHB −211 variant have a better response to FSH therapy in terms of sperm count and motility than patients with variants GG and GT (Ferlin et al., 2011).

Although the pharmacogenomic approach to male infertility seems to represent an important tool to predict the response to FSH therapy, it is still very expensive, time consuming in clinical situations and requires more and larger studies to determine tailored FSH dosage. This prospective controlled clinical study aimed to identify a new, simple, cheap and effective parameter to predict the response to FSH therapy in infertile oligozoospermic patients with normal FSH plasma concentrations in terms of sperm parameters and fertility outcome.

Section snippets

Patients and setting

This study was approved by the ethics committee of the Hospital-University of Padova (protocol number 2591P, approved 21 March 2010). Informed consent was obtained from each subject. All phases of the study were performed at the Infertility Centre of the University of Padova from January 2011 to December 2012. All men were informed about the FSH therapy protocol and in particular about the different phases of the study and the possibility of undergoing assisted reproduction treatment during

Results

Testicular cytology obtained by FNAC and seminal smears for spermatid count from patients in each group are shown in Figure 3.

Table 1 shows the baseline clinical, hormonal and seminal characteristics observed in 174 oligozoospermic patients divided into three groups on the basis of FNAC. There were 92 subjects who showed only hypospermatogenesis (group A), 61 who showed hypospermatogenesis with maturative disturbance (44 round and 17 elongated spermatidic arrest) (group B) and 21 who showed

Discussion

Many pathological conditions can lead to oligozoospermia, such as varicocele, cryptorchidism, orchitis, testicular trauma, karyotype abnormalities, Y chromosome microdeletions, cystic fibrosis gene abnormalities, triplet amplification or polymorphism or mutation in the LH or androgen receptor genes and idiopathic causes. Indeed, reduced sperm production may be associated with different testicular alterations, such as hypospermatogenesis and maturation disturbance at different levels, most

Acknowledgements

The authors thank Alberto Bottacin, Barbara Sartini and Laura Marino for technical assistance and all the staff of the Unit for Human Reproduction Pathology for helpful discussion. Special thanks to Karyn Ashley Cecchini for her kind and careful editing of English.

References (56)

  • A.A. Acosta et al.

    Possible role of pure human follicle-stimulating hormone in the treatment of severe male infertility by assisted reproduction: preliminary report

    Fertil. Steril.

    (1991)
  • A.A. Acosta et al.

    Pure human follicle stimulating hormone has a role in the treatment of severe male infertility by assisted reproduction: Norfolk’s total experience

    Hum. Reprod.

    (1992)
  • R.C. Adhikari

    Testicular fine needle aspiration cytology in azoospermic males

    Nepal Med. Coll. J.

    (2009)
  • K. Aittomaki et al.

    Mutation in the follicle-stimulating hormone receptor gene causes hereditary hypergonadotropic ovarian failure

    Cell

    (1995)
  • J. Ashkenazi et al.

    The role of purified follicle stimulating hormone therapy in the male partner before intracytoplasmic sperm injection

    Fertil. Steril.

    (1999)
  • A.M. Attia et al.

    Gonadotrophins for idiopathic male factor subfertility

    Cochrane Database Syst. Rev.

    (2007)
  • B. Baccetti et al.

    The effect of follicle stimulating hormone therapy on human sperm structure (Notulae seminologicae)

    Hum. Reprod.

    (1997)
  • M.A. Baker et al.

    Reactive oxygen species in spermatozoa: methods for monitoring and significance for the origins of genetic disease and infertility

    Reprod. Biol. Endocrinol.

    (2005)
  • B. Bartoov et al.

    Sperm quality of subfertile males before and after treatment with human follicle-stimulating hormone

    Fertil. Steril.

    (1994)
  • H.M. Behre et al.

    Significance of a common single nucleotide polymorphism in exon 10 of the follicle-stimulating hormone (FSH) receptor gene for the ovarian response to FSH: a pharmacogenetic approach to controlled ovarian hyperstimulation

    Pharmacogenet. Genom.

    (2005)
  • S. Bhasin

    Approach to the infertile man

    J. Clin. Endocrinol. Metab.

    (2007)
  • N. Colacurci et al.

    Recombinant human FSH reduces sperm DNA fragmentation in men with idiopathic oligoasthenoteratozoospermia

    J. Androl.

    (2012)
  • D.P. Evenson et al.

    Data analysis of two in vivo fertility studies using Sperm Chromatin Structure Assay-derived DNA fragmentation index vs. pregnancy outcome

    Fertil. Steril.

    (2008)
  • A. Ferlin et al.

    Toward a pharmacogenetic approach to male infertility: polymorphism of follicle-stimulating hormone beta-subunit promoter

    Fertil. Steril.

    (2011)
  • C. Foresta et al.

    Assessment of testicular cytology by fine needle aspiration as a diagnostic parameter in the evaluation of the oligospermia subject

    Fertil. Steril.

    (1992)
  • C. Foresta et al.

    Assessment of testicular cytology by fine needle aspiration as a diagnostic parameter in the evaluation of the azoospermic subject

    Fertil. Steril.

    (1992)
  • C. Foresta et al.

    Diagnostic and clinical features in azoospermia

    Clin. Endocrinol.

    (1995)
  • C. Foresta et al.

    High incidence of sperm sex chromosomes aneuploidies in two patients with Klinefelter’s syndrome

    J. Clin. Endocrinol. Metab.

    (1998)
  • C. Foresta et al.

    FSH in the treatment of oligozoospermia

    Mol. Cell. Endocrinol.

    (2000)
  • C. Foresta et al.

    Use of recombinant human follicle-stimulating hormone in the treatment of male factor infertility

    Fertil. Steril.

    (2002)
  • C. Foresta et al.

    Treatment of male idiopathic infertility with recombinant human follicle-stimulating hormone: a prospective, controlled, randomized clinical study

    Fertil. Steril.

    (2005)
  • C. Foresta et al.

    Hormonal treatment of male infertility: FSH

    Reprod. Biomed.Online

    (2007)
  • C. Foresta et al.

    Gonadotropin administration after gonadotropin-releasing-hormone agonist: a therapeutic option in severe testiculopathies

    Fertil. Steril.

    (2009)
  • L. Gandini et al.

    Immature germ cell separation using a modified discontinuous Percoll gradient technique in human semen

    Hum. Reprod.

    (1999)
  • H.J. Glander et al.

    Effects of pure human follicle-stimulating hormone (pFSH) on sperm quality correlate with the hypophyseal response to gonadotropin-releasing hormone (GnRH)

    Andrologia

    (1997)
  • R.R. Greb et al.

    A common single nucleotide polymorphism in exon 10 of the human follicle stimulating hormone receptor is a major determinant of length and hormonal dynamics of the menstrual cycle

    J. Clin. Endocrinol. Metab.

    (2005)
  • M. Grigorova et al.

    Haplotype structure of FSHB, the beta-subunit gene for fertility-associated follicle-stimulating hormone: possible influence of balancing selection

    Ann. Hum. Genet.

    (2007)
  • M. Grigorova et al.

    FSHB promoter polymorphism within evolutionary conserved element is associated with serum FSH level in men

    Hum. Reprod.

    (2008)
  • Cited by (20)

    • Male infertility and gonadotropin treatment: What can we learn from real-world data?

      2023, Best Practice and Research: Clinical Obstetrics and Gynaecology
      Citation Excerpt :

      According to the latest WHO manual for examining human semen, published in 2021, oligozoospermia is defined as a sperm concentration below the lower reference limit of 16 million/mL [1]. Several studies have explored the use of hormonal treatment with FSH for these patients (133,137-156). The characteristics of the most relevant trials published are summarized in Table 2.

    • Sperm DNA fragmentation index as a promising predictive tool for male infertility diagnosis and treatment management – meta-analyses

      2018, Reproductive BioMedicine Online
      Citation Excerpt :

      Overall, 383 men with idiopathic infertility or with abnormal semen analyses were treated with FSH for a maximum of 3 months (Table 2). Recombinant FSH (rFSH) was used in three studies and urinary-derived FSH (uFSH) (Colacurci et al., 2012; Ruvolo et al., 2013; Simoni et al., 2016) in the other three (Garolla et al., 2014, 2017; Palomba et al. 2011) (Table 2). FSH administration significantly improved sDF in infertile men by 4.24% (95% CI: 0.24–8.25%; P = 0.04) after 3 months of treatment (Figure 4), although no sDF differences were observed between study and control groups both before and after FSH administration (Supplementary Figures 6A and 6B).

    View all citing articles on Scopus

    Carlo Foresta is full Professor of Endocrinology and Director of Human Reproduction Pathology Unit at the University of Padova, Italy. During the last 30 years his research activities included different aspects related to male reproduction and spermatogenesis. Current research and clinical interests are in the genetics of male infertility, cryptorchidism and testicular tumours, endocrine regulation of spermatogenesis, sexual transmittable diseases and sexual disorders. He is author of more than 250 papers in International peer-reviewed journals, and invited speaker to many international and national scientific congresses. He is Past President of the Italian Society of Andrology and Sexual Medicine and member of the executive board of many scientific societies.

    View full text