ISI Impact Factor (2004): 1.096


   
 

Editor-in-Chief
Prof. Yi-Fei WANG,

 
     

   

    Asian J Androl 2006; 8 (2): 143-157

กก

This web only provides the extract of this article. If you want to read the figures and tables, please reference the PDF full text on Blackwell Synergy. Thank you.

- .Review . -

Male idiopathic oligoasthenoteratozoospermia

Giorgio Cavallini

Operative Unit of Andrology, Societ+Italiana di Medicina della Riproduzione (SISMER), Via Mazzini 12, 40138 Bologna, Italy

Abstract

Idiopathic oligoasthenoteratozoospermia (iOAT) affects approximately 30% of all infertile men. This mini-review discussed recent data in this field. Age, non-inflammatory functional alterations in post-testicular organs, infective agents (Chlamydia trachomatis, herpes virus and adeno-associated viruses), alterations in gamete genome, mitochondrial alterations, environmental pollutants and “subtle” hormonal alterations are all considered possible causes of iOAT. Increase of reactive oxygen species in tubules and in seminal plasma and of apoptosis are reputed to affect sperm concentration, motility and morphology. iOAT is commonly diagnosed by exclusion, nevertheless spectral traces of the main testicular artery may be used as a diagnostic tool for iOAT. The following can be considered therapies for iOAT: 1) tamoxifen citrate (20mg/d)+testosterone undecanoate (120mg/d) (pregnancy rate per couple/month [prcm]: 3.8%); 2) folic acid (66mg/d)+Zinc sulfate (5mg/d); 3) L-carnitine (2g/d) alone or in combination with acetyl-L-carnitine (1g/d) (prcm: 2.3%); and 4) both carnitines+one 30mg cinnoxicam suppository every 4 days (prcm: 8.5%). Alpha-blocking drugs improved sperm concentration but not morphology, motility or pregnancy rate. Tranilast (300mg/d) increased sperm parameters and pregnancy rates in an initial uncontrolled study. Its efficacy on sperm concentration (but not on sperm motility, morphology or prcm) was confirmed in subsequent published reports. The efficacy of tamoxifen+testosterone undecanoate, tamoxifen alone, and recombinant follicle-stimulating hormone is still a matter for discussion. (Asian J Androl 2006 Mar; 8: 143-157)

Keywords: idiopathic oligoasthenoteratozoospermia; male infertility; diagnosis; pathogenesis

Correspondence to: Dr Giorgio Cavallini, Via Mascheraio 46, 44100 Ferrara, Italy.
Tel: +39-532-200-847, Fax: +39-532-246-496
E-mail: giorgiocavallini@libero.it
Received 2005-02-29 Accepted 2005-11-29


1 Introduction

Idiopathic oligoasthenoteratozoospermia (iOAT) is defined as defective spermatogenesis of unknown etiology and is regarded as undetectable by the common laboratory methods [1]. This mini-review presented recent data in the field. database, EMBase and Cochrane reports. Prospective studies were considered [2]. The efficacy of drugs was evaluated using specific parameters (see the section “Therapy”). A prospective/longitudinal research plan could not always be used for iOAT study (see subsections “Infective agents” and “Environmental pollutants” under the section “Etiology”), in these cases it was specified in the text. Reviews and meta-analyses were chosen within high-level journals (impact factor higher than 1.060).

Databases that were used to find relevant published reports were: Medline

2 Definition

Infertility means that a couple has failed to achieve a pregnancy within one year of regular (at least three times per month) unprotected intercourse. One objection to the general use of the term “infertility” is that the condition which is actually meant may instead be that of subfertility, as the ability to father or to conceive may exist with a different partner. However, here too it is difficult to delineate clearly between definitions. Infertility pertains to approximately 15% of sexually active couples [3]. A male factor is present in approximately 40% of infertility cases. It is considered a male factor when an alteration in sperm concentration and/or motility and/or morphology could be found in at least one sample of two sperm analyses which comply with World Health Organization (WHO) 1999 guidelines collected between 1-4 weeks apart. “Idiopathic” means that no etiological factor could be found with the common clinical, instrumental or laboratory methods. Approximately 30% of OAT infertile men are diagnosed as “idiopathic”. iOAT is classified as: isolated asthe-no±teratospermia (no alteration in sperm con-cen-tration); moderate (sperm concentration (20×106/mL and)×106/mL); or severe (sperm concentration (5×106/mL) [4].

3 Etiology

Descriptions of reputed etiologies of iOAT have at least two biases: 1) two patterns have been described whose alterations are linked to male infertility with normal sperm parameters [5, 6]; 2) the sum of the percentages of patients with different etiologies of iOAT gave a result much higher than 100%. This means that etiologies overlap and/or that the primary cause (if any) of iOAT is still unknown and/or that more than one cause is needed to affect sperm patterns.

Nevertheless, the following factors are accepted etiologies of iOAT, therefore the concept that iOAT is composed of an assortment of infertilities is confirmed [7].

3.1 Age

Semen volume and sperm motility, but not sperm concentration, continuously decrease between the ages of 22 years and 80 years, with no evidence of a thre-shold [8].

3.2 Non-inflammatory functional alterations in post-testicular organs

Low seminal concentration of prostate-specific antigen, zinc, fructose [9] and prostatic acid phospha-tase [10], and low seminal activity of neutral alpha-glucosidase are linked to isolated asthenospermia as well as to increased viscoelasticity [11] and osmolarity of seminal plasma [12].

Three spermatogenesis-specific genes were found to be unmethylated in adult spermatogenetic cells in the testis, but were then found to be remethylated in mature spermatozoa in the vas deferens. A role of post-testicular organs (epididymis+) on DNA male gamete methylation might be suspected [13]. DNA methylation is involved in imprinted gene expression in animal cells [14]. The methylation occurs at cytosine-guanine (CpG) dinucleotides at which methyl groups become covalently bound to cytosine residues [15]. Methylation mediates transcriptional repression by recruiting histone deacetylase [16]. Most tissue-specific genes are fully methylated in sperm and in almost all somatic cells of the adult organism. In tissue expression the same genes undergo a striking demethylation that is necessary for gene transcription. Specific genes that are expressed in somatic cell types have distinctive patterns of DNA methylation that have been correlated to the expression profile [17], even though more recent studies suggest that methylation is not necessary for transcription repression in many organs [18]. Even some germ cell-specific genes are regulated by (de)methylation [19] at CpG dinu-cleotides. Changes in CpG methylation are involved in senescence (even at the epydidymal level [20]) and tumoringenesis [21].

3.3 Infective agents

Longitudinal observational studies to determine the role, if any, of Chlamydia trachomatis (CT) asymptomatic infection in the aetiology of iOAT look extremely difficult. The alternative is to contrast and compare between fertile and infertile patients with regards to the detection and occurrence of CT, CT products, and/or of an immune response towards the CT products.

Some researchers estimate the prevalence of asymptomatic CT infection at approximately 20% in men with iOAT. As this percentage is higher than in the control population, asymptomatic CT infection has been regarded by some as a cause of iOAT [22, 23]. On the contrary, other researchers have maintained that CT prevalence in iOAT and in fertile subjects is similar (approximately 5%) [24]. Different detection techniques have been regarded as an explanation for this difference [25]. Furthermore, CT antibodies were not significantly related to the outcome of seminal analyses, including an anti-sperm antibodies analysis and an analysis of several parameters of sperm function [26]. Therefore, proof of the role asymptomatic CT infection has in infertility is considered inconclusive.

Herpes virus and adeno-associated viruses have been linked to OAT without significant leukospermia [27, 28].

3.4 Gamete genome

Y chromosome microdeletion [29-31], androgen receptor gene defects, and/or somatic karyotype aberration [30] are rarely found in iOAT patients (1%-3%).

Of the 11 955 rat loci investigated, 1 268 were identified as having been specifically transcribed in germ cells during the course of gametogenesis; 200 of these genes are important for the male germ cell development [32]. Approximately 4% of the mouse genome is dedicated to the expression of post-meiotic male germ cells. Targeted disruption of 19 of these genes has displayed their critical roles in fertility [33]. In order to be considered a key for iOAT, a gene must display three characteristics: 1) it should be specifically expressed in the germ cell line; 2) it should have an essential role in spermatogenesis; and 3) its altered expression should be associated with iOAT [34].

For example, cyclic adenosine monophosphate-responsive element modulator (CREM) is a transcription factor which is highly expressed in testicular post-meiotic germ cells and which regulates the expression of several germ cell-specific genes by controlling the imbalance between apoptosis and development. Its activation depends on follicle-stimulating hormone (FSH) and luteinizing hormone (LH) [35]. In histological patterns of the testicle, CREM expression was found significantly reduced in severe OAT patients who had round spermatid maturation arrest or mixed atrophy in combination with normal blood levels of testosterone, FSH and LH (i.e., iOAT patients) [36, 37]. However, azoospermic patients lacking spermatid elongation but who had spermatid maturation arrest were not found to necessarily have defective CREM expression [38].

The BOULE gene (a member of the deleted in azoospermia [DAZ] gene family) encodes a key factor (a cdc 25 phosphatase protein) which promotes progression through meiosis. A major group of azoospermic infertile men with meiotic arrest completely lack the BOULE gene and its target (cdc 25 phosphatase) expression [39]. Furthermore, several patients with a defective expression of BOULE protein and of cdc 25 phosphatase could be correctly classified as iOAT because their testicle displayed a histological picture of mixed atrophy, their sperm analyses displayed OAT and their serum displayed normal levels of testosterone, FSH and LH [39].

Pyridoxal-kinase mRNA splice variant (PKH-T) gene is expressed in the adult testes and in spermatozoa, but it has no expression in the testis of men with Sertoli cell-only syndrome. PKH-T was found to be expressed in three out of four patients with spermatogenetic arrest, and in one out of two patients with spermatid arrest. It was unexpressed in a minority of OAT patients with normal serum levels of testosterone, FSH and LH [40].

3.5 Mitochondrial alterations

In asthenospermia, both mitochondrial membrane potential [41] and DNA mitochondrial [42] content are impaired. Mitochondrial DNA oxidative damage has been observed in asthenospermic infertile men [43] and has been confirmed in experimental models [44]. These alterations are very similar to the peculiar mitochondrial modifications found in apoptotic sperm cells [45-49].

3.6 Environmental pollutants

The possible effect of environmental pollutants on sperm quality is a matter of discussion.

The hypothesis that environmental hormonally-active compounds of industrial origin (endocrine disrup-ters) may affect semen quality was first synthesized in 1992 [50].

Several laboratories published a decline in sperm concentration, motility, and morphology using archival data [51-56], yet there was no detectable deterioration in sperm analyses in areas that are similar in terms of human pollutant 3. 7 “Subtle” hormonal abnormalities

A decreased LH pulse frequency has been found to occur in iOAT men whose amplitude parallels the severity of the disorder [77].

The Gly102Ser variant of LH might be implicated in iOAT [78].

iOAT displays a shift toward lower testosterone (T) serum levels, lower calculated free T index, and lower T/LH ratio, and a shift toward higher serum LH levels, higher estradiol (E2) levels, and higher E2/T levels [79]. Also, significantly lower inhibin (I) levels and significantly higher FSH levels have been found in i(O)ATs when compared to the levels in fertile men [82].

All of these etiologies can be correctly classified as etiologies of iOAT. The causal link between these pathologies and iOAT is not always evident, nevertheless, the common final result is the impairment of spermatogenesis.

Despite this range of possible etiologies, male infertility also appears to have a familial occurrence especially among brothers and maternal uncles, who often have normal blood levels of FSH and LH [56]. Observations were also consistent with an autosomal recessive mode of inheritance in over half of the cases [57]. These published reports fit with a genetic etiology of iOAT which of late has received great consensus. Such a consensus is lacking, however, in the case of pollution and infection. On one hand, genetic etiology is consistent with iOAT being composed of an assortment of infertilities; on the other hand, it provides a holistic approach to the disease. Therefore, even though in the future genetic etiology will not prove to be the most probable cause of iOAT, it is currently the most convenient to accept.

4 Pathogenesis

The above etiologies are regarded to affect spermatogenetic process. Impaired spermatogenesis leads to increased ROS concentration and to unbalanced germ cell apoptosis. The result of these processes is an affected sperm concentration, motility and morphology.

4.1 Increased ROS

Increased ROS concentration and reduced total antioxidant capacity (TAC) were found in the tubula and the seminal plasma of the majority of iOAT. ROS originates from cellular physiological metabolism of O2 in aerobic conditions. Seminal cells include mature and immature gametes, leukocytes, epithelial cells, and red blood cells. ROS are mainly produced by leukocytes and immature gametes, therefore an increase in one or both cell types increases ROS and reduces TAC [72]. Leukocytes and/or immature gametes can be found in several types of OAT (hormonal alterations [73], inflammation [74], and varicocele [75]), therefore increased ROS and reduced TAC are not characteristic of iOAT. Physiological ROS concentration has positive effects. ROS are metabolic intermediates in metabolism of prostanoid, in the regulation of vasotonous, in gene regulation, and in facilitating sperm capacitation and acrosome reaction, but at a higher concentration they exert negative effects [74]. WHO [4] defines a 106 leukocytes/mLthreshold above which leukocytes and/or their product(s)/ROS impair(s) fertility. This threshold is commonly accepted today [4] but is still under discussion [76-79]. Therefore it may be assumed that the main source of ROS in iOAT is immature gametes.

ROS are short-lived chemical intermediates which contain one or more electrons with unpaired spin. In order to overcome this state of unpaired electrons, they are highly and unspecifically reactive molecules able to interact with lipids, amino acids and nucleic acids [74]. Seminal plasma is the human fluid with the highest concentration of anti-oxidants to protect gametes from ROS. Seminal plasma TAC is the sum of the potential anti-ROS activities: 1) of enzymes (superoxide dismutase, catalase, glutathione peroxidase); 2) of low molecular weight substances (+tocopherols, +carotene, ascorbate, urate); and 3) of the transition metal chelators (trans-ferring, lactoferrin, ceruloplasmin) [80].

ROS exert their activity on all cellular structures du-ring the course of tubular spermatogenesis and sperm maturation during the migration through the seminiferous tubules and epididymis [74].

ROS attack nuclear and mitochondrial DNA, indu-cing fragmentation, aberrant recombination, and/or defective packaging [81]. 2-Deoxyguanosine, a normal DNA base, is oxidized to 8-OH-deoxyguanosine. Whereas the former nucleotide binds to cytosine, the latter will form a base pair with thymine during DNA replication. Thymine will bind to adenosine so that a point mutation is introduced in the DNA [82]. The extent of DNA damage depends on the degree of oxidative stress [83].

ROS irreversibly damages cell membranes. Mam-malian sperm cell membranes have a highly specific lipid composition: a high content of polyunsaturated fatty acids (PUFA), plasmalogens and sphingomyelins. The unusual structure of their membrane is responsible for the flexi-bility and the functional ability of spermatozoa. ROS-dependent peroxidation of PUFA is an autocatalytic self-propagating reaction. The first step (initiation) is the extraction of a hydrogen atom from an unsaturated fatty acid. The second step (propagation) is the formation of a lipid-alkyl radical followed by its rapid reaction with oxygen to form a lipid-peroxyl radical. The peroxyl radical can extract a hydrogen atom from a PUFA with the concomitant formation of a lipid radical and lipid hydro-peroxide. As the peroxyl and alkyl radicals are regenerated, the cycle of propagation could continue indefinitely or until one of the substrates is consumed or terminated in the radical-radical reaction [84-86].

An increase in ROS changes the tertiary structure and expression of proteins, protein membrane receptors, and membrane transport proteins, which in turn results in disturbances in the ionic balance. The function of enzymes with thiolic (SH) groups may also be altered by ROS [87].

Sperm epididymal maturation involves extensive protamination and chromatin condensation [88, 89]. Increased ROS concentration has been linked to DNA denaturation in infertile men [90]. DNA denaturation is negatively correlated with semen parameters [91] but is positively correlated with cytoplasmic residues [92], that is, with the morphological markers of immature gametes.

Death of gametes (necrosis and/or apoptosis, i.e., oligo-/terato-spermia), reduction in the percentage of sperm typical forms, and impairment of sperm motility (i.e., asthe-nospermia) are resulting patterns. Asthenospermia is caused by axonemal damage mostly as a result of adenosine triphosphate depletion [93, 94].

Despite the progress in our knowledge of ROS activity, the causal link between these alterations and iOAT is not always evident.

4.2 Modified apoptosis

Apoptosis or programmed cell death (PCD) is critical for mammalian tissue morphogenesis because it eliminates unwanted or defective cells through an orderly process of cellular disintegration without inflammation, thus ensuring the production of intact functional spermatozoa [95]. Therefore, as a general rule, PCD is involved in the removal of arrested germ cells from the testicles of patients with spermatogenetic failures [96]. PCD is an active process; it involves specific gene expression and requires mRNA and protein synthesis. Apoptosis of type A2, A3 and A4 spermatogonia reduces the number of germ cells produced to approximately 25% of that expected if all A1 spermatogonial progeny were to survive. It has been proposed that individual spermatogenesis is the result of a framework involving the imbalance between pro-apoptotic and pro-survival factors [45]. In fact, the BCL-2 protein family can either support cell survival (BCL-2, BCL-XL, BCL-W, MCL-1, A1/BFL-1) or promote cell death (BAX, BAK, BCL-XS, BAD, BID, BIK, BOK, DIVA, HRK). BCL-2 family members interact competitively, thereby regulating activation of the proteases (cysteinyl aspartate-specific proteinases [caspases]) which dismantle the germ cell [97]. Furthermore, the C-kit stem cell factor system, bone morphogenetic protein 8B [45], lactate [98], and sphingosine-1-phosphate [99] inhibit male germ cell apoptosis, whereas the fibroblast-associated death receptor (Fas)/Fas-ligand (Fas-L) system is regarded as a pro-apoptotic factor [45].

Interestingly, toxicants and increased concentration of ROS from activated leukocytes or from immature spermatozoa trigger PCD [45-47]. These substances may [100] or may not [101] interact with the Fas/Fas-L system to activate caspase [48, 101], which leads to cellular morphological and biochemical alterations and finally to death [45-49].

The evidence of ethnic differences in the susceptibility of germ cells to PCD [102] may indicate that alterations of PCD may occur in the absence of an exogenous triggering. The degree of decreased expression of survivin (a PCD inhibitor) parallels the severity of iOAT [102], just as the overexpression of the Fas gene has been associated with altered meiotic and post-meiotic sperm cell maturation [103].

Ejaculated spermatozoa from infertile men display a number of apoptosis markers: various degrees of plasma membrane translocation of phosphatidylserine; DNA fragmentation; and active capsase-3 (the main executor of apoptosis) with an apparent exclusive cellular location in the mid-piece [104]. The apoptotic process is proba-bly set in motion before ejaculation [105] because healthy human ejaculated spermatozoa cannot initiate apoptosis, at least not under in vitro conditions [106].

Apoptosis has been inversely linked with sperm motility [107], morphology, vitality and concentration [108].

Increased apoptosis was found in a variety of infertilities , such as hormonal infertilities [109], anti-sperm auto-antibodies (ASA)-associated infertility, varicocele, testicle torsion [110] and inflammation [111]. Therefore, increased PCD is not atypical of iOAT.

In conclusion, the pathogenetic mechanisms of iOAT are not peculiar to this kind of disease and can be found in several types of OAT.

5 Diagnosis

iOAT is commonly diagnosed by exclusion; the differential diagnosis is presented in Table 1.

Impaired spermatogenesis of iOAT was assayed with testicular arteries duplex examination and inhibin B serum levels.

iOAT displayed peak systolic velocity (PSV) levels significantly lower than those of normospermic fertile men, inflammatory OAT patients, and varicocele OAT patients [121]. An echo-colour Doppler semi-quantitative score has been used to distinguish obstructive azoospermic patients from non-obstructive azoospermic patients affected by primary testicular pathology [122]. The intratesticular arterial blood flow and maximum blood flow velocity were significantly lower in patients with germ cell hypoplasia or maturation arrest [123]. The pulsatility index of the testicular transmediastinal artery was significantly higher in patients with obstructive azoospermia than in those with non-obstructive azoospermia [124]. Arterial impedence of undescended testes in adults may have a predictive value and provide more accurate information about histology than testicular volume [125]. These data link blood arterial supply to spermatogenesis [121]. Currently there is no direct explanation for this phenomenon. Testicular arteries are target organs for androgens [126] and in infertile men testicular arteries have a narrow lumen caused by enlarged endothelial cells, a thickened subendothelial layer, and an abundant adventitia rich in connective fibres and ground substance [127].

The clinical usefulness of a blood level measurement of inhibin B is considered rather low [128]; inhibin B serum levels are regarded as markers of Sertoli cell function, but the prediction of the quality of spermatogenesis is not higher than FSH [129]. iOAT men who had been exposed to phthalate displayed increased levels of inhibin B, lowered levels of FSH, and phthalate urinary excretion [65].

There are two pathologies that may confound the practitioner: sperm auto-immunity and male accessory gland inflammation (MAGI).

Disruption of the epithelial (i.e., Sertoli cell) blood barrier elicits ASA. Some reputed causes of ASA are vasectomy, vas obstruction, testicular trauma, torsion, malignancy and infection of the urogenital tract [130]. ASA are found in 26%-55% of infertile couples, in up to 19% of fertile men, and in 43% of fertile women, which means that not all ASA cause infertility [131].

In order to be defined as an “autoimmune disease”, three criteria must be met: direct proof, indirect evidence and circumstantial evidence [132]. The direct proof is easy to obtain because ASA can be transferred to the sperm of healthy persons and an impairment of functions can be demonstrated in the receiving cells [132]. Indirect proof (i.e., animal models) is difficult to obtain. Spermatozoa are not amenable to conventional molecular biology techniques because of the lack of much functional mRNA and the lack of many post-translational modifications of germ cells during maturation [132, 133]. Circumstantial evidence (i.e., the favourable response to immuno-suppression) was not demonstrated because it is impossible to treat infertile patients with cytotoxic drugs [132].

ASA are made of numerous antibodies interacting with multiple sperm components [134]. These findings support the hypothesis there might be a relatively non-specific binding of antibodies to the surface of spermatozoa [135] probably through crystalizable fragment (FC) or disulphide bonds [131].

On the other hand, ASA are reputed to affect the following: cervical mucus penetration [135], acrosome reaction [132], zona binding [136], zona penetration [137], oolemma binding [138] and pronucleus formation [139]. The effects of ASA on motility [140, 141] and on in vitro fertilization [139, 142, 143] are questionable. The WHO recommends direct testing for ASA on the surface of spermatozoa [4], nevertheless only some trials on iOAT therapy look for sperm autoimmunity [144-146], therefore underestimating the role of ASA in infertility. Our opinion is that ASA detection may be a prudent technique for interpreting results in infertility management.

Seminal leukocyte concentration greater than 106/mL is considered the lowest threshold of inflammation. Controversial results have been published about the efficacy of antimicrobial therapy on sperm quality and pregnancy rate in MAGI, even though seminal leukocyte concentration was found to fall below 106/mL after therapy [147, 148].

Some reasons for discrepancy may be: poor patient selection [147], inadequate drug administration [148], persistence of leukocytes [149] and/or of their products (ROS) [150], elastase [151], interleukin (IL)-8 and IL-6 [151].

Regardless, the vast majority of trials for idiopathic oligoasthenoteratozoospermia consider 106/mL leukocytes to be the threshold for the diagnosis of MAGI.

6 Indications for therapy

If sperm concentration exceeds 20×106/mL, the probability of spontaneous conception depends only to a minor extent on sperm motility, viability, and morphology. The lack of prognostic significance contrasts with the fact that these characteristics discriminate well between the semen of fertile and subfertile men. Men whose sperm concentration is over 20×106/mL but who have abnormal motility or morphology of idiopathic origin have a 40% higher probability of achieving spontaneous conception than men in whom the sperm alterations are related to a demonstrable cause [130]. Therefore it is difficult to justify the therapy of an iOAT patient with a sperm concentration of 40×106/mL spermatozoa and a class A WHO motility of 24%.

7 Therapy

Evidence-based medicine indicates that studies without a placebo-treated group should not be taken into consideration. “Placebo treatment” differs from “no treatment” because a placebo can improve sperm parameters due to a psychological mechanism [144, 145]. It is mandatory to establish whether an improvement in sperm count and quality is statistically or clinically significant, because the latter involves the former, but not vice versa. Spontaneous pregnancy rate when the partner is free from any detectable factor of infertility is an accepted method, even though 11%-17% of infertile couples do not display any cause of infertility as detected by the common techniques [130]. Therefore to define a drug as active, it should improve sperm patterns and pregnancy rates in at least one blind, prospective, placebo-controlled trial; more of these trials from independent groups are necessary in order to define a drug as unquestionably active [2].

Despite these instruments, difficult-to-interpret data still materialize. Two typical examples are recombinant FSH (rFSH) and tamoxifen citrate (TC). rFSH was reputed effective when compared to a “no treatment” group [152, 153], or ineffective when compared to a “placebo-treated” group [154]. A recent meta-analysis showed that rFSH given to men with iOAT increases the clinical pregnancy rate, the fertilization rate, and the rate of doubling sperm count significantly [155].

OAT decreases the monthly chance of conception in a dose-dependent manner, adding its effect to the duration of infertility [156]; an increased sperm concentration in OAT infertile males has been associated with disproportionately higher fecundability [157]. Further pregnancies can be expected on the basis of hyperbolic regression between sperm concentration and fecundability [157]. Therefore, treatments which double sperm concentration will have a stronger effect on the probability of conception when the initial sperm count is low, rather than when the initial sperm count is high [158]. The TC data were principally reviewed by Vanderkerckove et al. [159] and Comhaire [158]. Vanderkerckove maintained that there is not enough evidence to use TC to increase the fertility of iOATs [159]. Comhaire showed that, because a lower sperm concentration is related with a lower fecundability, the pregnancy rate with TC treatment is stronger in trials containing lower treatment-independent pregnancy rates [158]. In addition, Comhaire [158] also found an increased sperm concentration in trials that were discharged by Vanderkerckove [159].

The effects on seminal parameters of TC (10mg×+/d) combined with testosterone undecanoate (TU) (40mg×+/d) were studied in men with iOAT. The results were that TC+TU improved total sperm count, motility and functional sperm fraction after 3 and 6 months [160]. A further report showed a 33.9% (pregnancy rate per couple per month [prcm]=3.8%) rate of incidence of spontaneous pregnancy in the TC+TU treatment group at 9 months, and a 10.3% (prcm=1.1%) rate of spontaneous pregnancy in the placebo group at 9 months [161]. TC was introduced as an empirical treatment for iOAT [162] because of its stimulatory action on gonadotropin secretion [163], its direct effect on Leydig cell function [126], and because of its production of 5a-dihydrotes-tosterone in tubules and epididymis [164]. To overcome the putative inferior androgenic environment in the reproductive tract of men with iOAT, androgen-dependent epididymal and accessory gland function [165] were boosted by androgen administration [160].

The effects of folic acid and zinc sulfate treatment on semen variables in fertile and subfertile men were studied. One hundred and eight fertile and 103 subfertile men were randomly assigned to receive one of four treatments for 26 weeks: folic acid and placebo; zinc sulfate and placebo; zinc sulfate and folic acid; and two placebos. Folic acid was given at a daily dose of 5mg, and zinc sulfate was given at a daily dose of 66mg. Subfertile men demonstrated a significant 74% increase in total normal sperm count and a minor increase of 4% in abnormal spermatozoa. A similar trend was observed in fertile men. The beneficial effect on fertility, if any, remains to be established. Despite a putative role of zinc on post-testicular organs, a direct anti-oxidant effect of both drugs on tubules was sustained [166].

Although there is no clear pathophysiological hypothesis for the use of a-blocking agents in the treatment of iOAT, studies have been performed using these substances [167]. Two placebo-controlled studies claimed an improvement in sperm concentration but not of ejaculated volume, morphology, motility or pregnancy rate when compared to placebo [168, 169].

In bioptic testicle specimens, mast cell counts were higher in idiopathic infertile men than in normospermic men [170]. Tranilast (a mast cell blocker) was first used in an uncontrolled study and demonstrated a clinical benefit on semen parameters and conception rates (prcm=9.5%) [171]. A further double-blind controlled trial showed that tranilast significantly increased sperm concentration, but not motility, morphology or pregnancy rate when compared to placebo [172]. It has been concluded that tranilast demonstrates a certain clinical benefit in terms of improvement in semen parameters involving severe iOAT, but it does not appear to afford clinical benefit in the long term [173]. A second mast cell blocker (fexofenadine) was found ineffective [174].

Acetyl-L-carnitine (ALC) 500×+ g/d + L-carnitine (LC) 1×+ g/d have been shown to increase sperm count, sperm quality and pregnancy rate in patients with an ultrasound picture of genital inflammation and leukocyte sperm concentration (106/mL [175]. LC alone 2g/d was shown to increase sperm concentration and motility [144] in iOATs, although a higher activity on iOAT was found using LC 2g/d+ALC 1g/d [136]. Another therapy studied was intermittent administration of a non-steroidal anti-inflammatory drug (NSAID). Cinnoxicam (C) (one 30mg suppository every 4 days) was found to increase sperm concentration and quality in low grade varicoceles [176]. Therefore one 30mg C suppository every 4 days was used in combination with LC 2g/d+ALC 1g/d. This three-drug association significantly increased sperm concentration, motility, morphology, and pregnancy rates when compared to placebo and to the two carnitines alone [146]. Although more effective than placebo, a recent analysis of literature indicates that the spontaneous prcm following carnitine therapy is 2.3% both in placebo-controlled and open studies [177]. The use of LC+ALC+C gave a prcm of 8.5% in iOAT patients (51% at 6 months) [146].

Carnitines have several activities which might be useful for the male gamete: free-radical production is reduced by depleting fatty acid peroxidation; carnitines restore the phospholipid composition of mitochondrial membranes; carnitines enhance cellular energetics in mitochondria which increases cytoplasmic acetyl-coenzyme A concentration through the higher availability of acetyl groups, which in turn results in an increase in mitochondrial respiration and monoamine-oxidase activity and thus increasing the metabolism of histamine; carnitines stabilize cell membrane fluidity by regulating phospholipid levels and reducing ceramide production and insulin-like growth factor 1. Carnitines prevent cellular death and apoptosis. Most of their activities take place in the tubular microenvironment rather than in epididymal functions [144-146]. No biochemical study was performed regarding the role of C suppositories in increasing the efficacy of carnitines. The effect of oral and injected NSAIDs on sperm analyses was lower than that of suppositories. A more direct effect of suppositories on seminal plasma may be presumed because of the rectal-prostatic lymphatic pathways. Hydrophilic NSAIDs were less active than C, whose lipophilia facilitate absorption [176]. An animal model showed that chronic treatment with NSAIDs at low doses improved sperm quality and fertility [178]. Carnitine administration was found to increase E2 prostaglandin concentration [179], which affects sperm count [180, 181]. Finally, polyzoo-spermia has been associated with lowered prostaglandin content of tubuloseminal plasma and OAT has been associated with increased prostaglandin content of tubuloseminal plasma [180, 181]. NSAIDs stabilize lysosomal membranes, thereby partially preventing apoptosis [176].

The efficacy of conventional anti-ROS drugs (vitamin E, vitamin C and essential fatty acids) is controversial. An uncontrolled study found that N-acetyl-cysteine or the combination of vitamins A+E with essential fatty acids improved sperm concentration, and that acrosome reaction reduced ROS and sperm oxidized DNA [182], but no significant difference was found in terms of sperm motility, morphology, or round cells. A double-blind placebo-controlled study maintained that vitamin E+Celenium was effective because they combination improved sperm motility and lipid peroxidation markers. The number of patients who dropped out (n=34) is much higher than the actual group studied (n=20 patients), therefore its conclusions might be reputed as unreliable [183]. Vitamin E+C was ineffective in a prospective double-blind controlled trial [184].

Finally, T+TC and (C+)ALC+LC may be regarded as active drugs, whereas TC and FSH are considered to most likely be active drugs. As yet, no drug can be defined as unquestionably effective. Actually C+ALC proved significantly more active than placebo in two blind prospective controlled trials [145, 146], therefore it is likely that these drugs are going to be regarded as unquestionably effective.

8 Conclusion

The seasonal [185], regional [186], and racial [187] differences in sperm count and quality make it difficult to consider iOAT data as absolutely valid, therefore further multicentric studies should be performed in this field.

Acknowledgment

I would like to thank Mr Luca Gianaroli, M.D. (Societ+Italiana di Medicina della Riproduzione[SISMER] Scientific Director), Mrs Anna Pia Ferraretti, M.D. (SISMER Clinical Director), and Mrs Maria Cristina Magli, B.D. (SISMER Laboratory Director) for their active encouragement of my research over the years. A special acknowledgement to Ms Tracy Hammerstrom for her technical and editing contribution.

References

1 Bonanomi N, Lucente G, Silvestrini B. Male fertility: core chemical structure in pharmacological research. Contraception 2002; 65: 317-20.

2 Altman DG, Schultz KF, Moher D, Egger M, Davidoff F, Elbourne D, et al. The revised consort statement for reporting randomized trials: explanation and elaboration. Ann Intern Med 2001; 134: 663-94

3 Nieschlag E, Behre HM, eds. Andrology. Male reproductive health and dysfunction. Berlin: Springer; 1996. p4-18

4 World Health Organization. WHO laboratory manual for the examination of human semen and semen-cervical mucus interaction, 4th ed. Cambridge: Cambridge University Press; 1999. p1-86.

5 Saleh RA, Agarwal A, Nelson DR, Nada EA, El-Tonsi MH, Alvarez JG, et al. Increased sperm nuclear DNA damage in normozoospermic infertile men: a prospective study. Fertil Steril 2002; 78: 313-8.

6 Jensen M, Leffers H, Petersen JH, Nyboe Andersen A, Jorgensen N, Carlsen E, et al. Frequent polymorphism of the mitochondrial DNA polymerase gamma gene (POLG) in patients with normal spermiograms and unexplained subfertility. Hum Reprod 2004; 19: 65-70.

7 Silber SJ. Evaluation and treatment of male infertility. Clin Obstet Gynecol 2000; 43: 854-88.

8 Eskenazi B, Wyrobeck A.J, Sloter E, Kidd SA, Moore L, Young S, et al. The association of age and semen quality in healthy men. Hum Reprod 2003; 18: 447-54.

9 Elzanaty S, Richthoff J, Malm J, Giwerkman A. The impact of epididymal and accessory gland sex function on sperm motility. Hum Reprod 2002; 17: 2904-11.

10 Carpino A, Sisci D, Aquila S, Salerno R, Siciliano L, Sessa M, et al. Adnexal gland secretion markers in unexplained asthenozoospermia. Arch Androl 1994; 32: 37-43.

11 Elzanaty S, Malm J, Giwerkman A. Visco-elasticity of seminal fluid in relation to the epididymal and accessory sex gland function and its impact on sperm motility. Int J Androl 2004; 27: 94-100.

12 Rossato M, Balercia G, Lucarelli G, Foresta C, Mantero F. Role of seminal plasma osmolarity in the reduction of human sperm motility. Int J Androl 2002; 25: 230-5.

13 Ariel M, Cedar H, McCarrey J. Developmental changes in methylation of spermatogenesis-specific genes include reprogramming in the epididymis. Nat Gen 1994; 7: 59-63.

14 Reik W, Walter J. Genomic imprinting: parental influence on the genome. Nat Rev Genet 2001; 2: 21-32.

15 Zhang LP, Stroud JC, Walter CA, Adrian GS, McCarrey JR. A gene specific promoter in transgenic mice directs testis-specific demethylation prior to transcriptional activation in vivo. Biol Reprod 1998: 59: 284-92.

16 Nan X, Campoy FJ, Bird A. MeCP2 is a transcriptional repressor with abundant binding sites in genomic chromatin. Cell 1997; 88: 471-8.

17 Ariel M, McCarrey J, Cedar H. Methylation patterns of testis specific genes. Proc Natl Acad Sci 1991; 88: 2317-21.

18 Muller C, Readhead C, Diederichs S, Idos G, Yang I, Tidow N, et al. Methylation of the cyclin A1 promoter correlates with gene silencing in somatic cell lines, while tissue specific expression of cyclin A1 is methylation independent. Mol Cell Biol 2000; 20: 3316-29.

19 Xie W, Han S.Y, Khan M, DeJong G. Regulation of ALF gene expression in somatic and male germ line tissues involves partial specific and site specific patterns of methylation. J Biol Chem 2002; 277: 17765-74.

20 Hafez ESE, Hafez B, Hafez SD. An Atlas of reproductive physiology in men. Boca Raton (USA): Parthenon Publishing; 2004. p25-46.

21 Cameron EE, Bachman KE, Myohanen J, Herman JG, Baylin SB. Synergy of demethylation and histone deacetylase inhibition in the re-expression of genes in silenced cancer. Nat Genet 1999; 21: 103-7.

22 Penna Videau S, Cermeno Vivas J, Salazar N. IgA antibodies to Chlamydia trachomatis and seminal parameters in asymptomatic infertile males. Arch Androl 2001; 46: 189-95.

23 Veznik Z, Pospisil L, Svecova D, Zajicova A, Unzeitig V. Chlamydiae in the ejaculate: their influence on the quality and morphology of sperm. Acta Obstet Scand 2004; 83: 656-60.

24 Bollman R, Engel S, Petzoldt R, Gobel UB. Chlamydia trachomatis in andrologic patients. Direct and indirect detection. Infection 2001; 29: 113-8.

25 Krause M, Bohring C. Male infertility and genital chlamydial infection: victim or perpetrator+ Andrologia 2003; 35: 209-16.

26 Eggert-Kruse W, Rohr G, Demirakca T, Rusu R, Naher H, Petzoldt D, et al. Chlamydial serology in 1303 asymptomatic subfertile couples. Hum Reprod 1997; 12: 1464-75.

27 Erles K, Rohde V, Thaele M, Roth S, Edler L, Schlehofer JR. DNA of adeno associated virus (AAV) in testicular tissue and in abnormal semen samples. Hum Reprod 2001; 16: 2333-7.

28 Kapranos N, Petrakou E, Anastasiadou C, Kotronias D. Detection of herpes simplex virus, cytomegalovirus, and Epstein-Barr virus in the semen of men attending an infertility clinic. Fertil Steril 2003; 71: 1556-70.

29 El Awady MK, El Shater FF, Ragaa E, Atek K, Shaheen IM, Megiud NA. Molecular study on Y chromosome microdeletions in Egyptian males with idiopathic infertility. Asian J Androl 2004; 6: 53-7.

30 Dada R, Gupta NP, Kucheria K. AZF microdeletions associated with idiopathic and non-idiopathic case with cryptorchi-dism and varicocele. Asian J Androl 2002; 4: 259-63.

31 Shimizu A, Ichikawa T, Suzuki N, Yamazaki T, Imamoto T, Kojima S, et al. Microdeletions in the Y chromosome of patients with idiopathic azoospermia. Asian J Androl 2002; 4: 111-5.

32 Schlecht U, Demougin P, Koch R, Hermida L, Wiederker C, Descombes P, et al. Expression profiling of mammalian cell meiosis and gametogenesis identifies novel candidate genes for roles in the regulation of fertility. Mol Biol Cell 2004; 15: 1031-43.

33 Schultz N, Hamra KF, Garbers DL. A magnitude of genes expressed solely in meiotic or postmeiotic spermatogeneic cells offers a myriad of contraceptive targets. Proc Natl Acad Sci USA 2003; 100: 12201-6.

34 Makela S, Eklund R, Lahdetie J, Mikola A, Hovatta O, Kere J. Mutational analysis of the human SLC26A8 gene: exclusion as a candidate for male infertility due to primary spermatogenic failure. Mol Hum Reprod 2005; 11: 129-32.

35 Sassone-Corsi P. CREM: a master switch regulating the balance between differentiation and apoptosis in male germ cells. Mol Reprod Dev 2000; 56: 228-9.

36 Wienbauer GF, Behr R, Bergaman M, Nieschlag E. Testicular cAMP responsive element modulator (CREM) protein is expressed in round spermatids but is absent or reduced in men with round spermatid maturation arrest. Mol Hum Reprod 1998; 4: 9-15.

37 Peri A, Krausz C, Cioppi F, Granchi S, Forti G, Francavilla S, et al. Cyclic adenosine 3',5'-monophosphate-responsive element modulator gene expression in germ cells of normo- and oligoazoospermic men. J Clin Endocrinol Metab 1998; 83: 3722-6.

38 Palermo I, Arcidiacono G, Barone N, Italia F, Arizzi M, Spera G, et al. Normal expression of isoforms activating cyclic adenosine monophosphate responsive element modulator in patients with spermatid maturation arrest. Fertil Steril 2004; 82: 1072-6.

39 Luetjens CM, Xu EY, Rejo Pera MA, Kamischke A, Nieschlag E, Gromoll J. Association of meiotic arrest with lack of BOULE expression in infertile men. J Clin Endocrinol Metab 2004; 89: 1926-33.

40 Fang X, Zhou ZM, Lu L, Yin LL, Li JM, Zhen J, et al. Expression of a novel pyridoxal kinase mRNA splice variant, PKH-T, in human testis. Asian J Androl 2004; 6: 83-91.

41 Kasai T, Ogawa K, Mizuno K, Nagai S, Uccida Y, Ohta S, et al. Relationship between sperm mitochondrial membrane potential, sperm motility and fertility potential. Asian J Androl 2002; 4: 97-103.

42 Kao SH, Chao HT, Liu HW, Liao TL, Wei YH. Sperm mitochondrial DNA depletion in men with asthenospermia. Fertil Steril 2004; 82: 66-73.

43 Sharma RK, Said T, Agarwal A. Sperm DNA damage and its clinical relevance in assessing reproductive outcome. Asian J Androl 2004; 6: 139-48.

44 Paisley D, Banks S, Selfridge J, McLennan NF, Ritchie AM, McEwan C, et al. Male infertility and DNA damage in Doppel knockout and prion protein/Doppel double-knockout mice. Am J Pathol 2004; 164: 2279-88.

45 Boekelheide K, Fleming SL, Johnson JK, Patel SR, Schoenfeld HA. Role of Sertoli cells in injury-associated testicular germ cell apoptosis. Proc Soc Exp Biol Med 2000; 225: 105-15.

46 Villegas J, Schultz M, Soto L, Iglesias T, Miska W, Sanchez R. Influence of reactive oxygen species produced by activated leukocytes at the level of apoptosis in mature human spermatozoa. Fertil Steril 2005; 83: 635-42.

47 Henkel R, Kierspel E, Stalf T, Mehnert C, Menkveld R, Tinneberg HR, et al. Effect of reactive oxygen species produced by spermatozoa and leukocytes on sperm functions in non-leukocytospermic patients. Fertil Steril 2005; 83: 635-42.

48 Said TM, Paasch U, Glander HJ, Agarwal A. Role of caspase in male infertility. Hum Reprod Update 2004; 10: 39-51.

49 Erkkila K, Pentikainen V, Wikstrom M, Parvinen M, Dunkel L. Partial oxygen pressure and mitochondrial permeability transition affect germ cell apoptosis in the human testis. J Clin Endocrinol Metab 1999; 84: 4253-9.

50 Colborn T, Clement C, eds. Chemical induced alterations is sexual and functional development and functional development: the wildlife/human connection. Princeton: Princeton Scientific Publishing; 1992. p1-386.

51 Adamopoulos D, Pappa A, Nicopoulou S, Andreou E, Karamertzanis M, Michopoulos J, et al. Seminal volume and total sperm number trends in men attending subfertility clinics in the greater Athens area during the period 1977-1993. Hum Reprod 1996; 11: 1936-41.

52 Auger J, Kunstmann JN, Czyglic F, Jouannet P. Decline in semen quality among fertile men in Paris during the past 20 years. N Engl J Med 1995; 332: 281-5.

53 Irvine S, Cawood E, Richardson D, Macdonald E, Aitken J. Evidence of deteriorating semen quality in the United Kingdom: birth cohort study in 577 men in Scotland over 11 years. BMJ 1996; 312: 467-71.

54 Menchini-Fabris S, Rossi P, Palego P, Simi S, Turchi P. Declinining sperm counts in Italy during the past 20 years. Andrologia 1996; 28: 304.

55 Van Waeleghem K, De Clercq N, Vermeulen L, Shoonians F, Comhaire F. Deterioration of seprm quality in young healthy Belgian men. Hum Reprod 1996; 11: 325-9.

56 Younglai E, Collins JA, Foster WG. Canadian semen quality: an analysis of sperm density among eleven academic fertility centers. Fertil Steril 1998; 70: 76-80.

57 Bujan L, Mansat A, Pontonnier F, Mieusset R. Time series analysis of sperm concentration in fertile men in Toulouse (France) between 1977 and 1992. BMJ 1996; 312: 471-2.

58 Fisch H, Goluboff ET, Olson JH, Feldshuh J, Broder SJ, Barad DH. Semen analyses in 1,283 men from the United States over a 25-year period: no decline in quality. Fertil Steril 1996; 65: 1009-14.

59 Itoh N, Kayama F, Tatsuki J, Tsukamoto T. Have sperm counts deteriorated over the past 20 years in healthy young Japanese men+ Results from the Sapporo area. J Androl 2001; 22: 40-4.

60 Paulsen CA, Berman NG, Wang C. Data from men in greater Seattle area reveals no downward trend in semen quality: further evidence that detrioration of semen quality is not geographically uniform. Fertil Steril 1996; 65: 1015-20.

61 Rasmussen PE, Erb K, Westergaard LG, Laursen SB. No evidence for decreasing semen quality in four birth cohorts of 1,055 Danish men born between 1950 and 1970. Fertil Steril 1997; 68: 1059-64.

62 Vierula M, Niemi M, Keiski A, Saarinen M, Saarikoski S, Suominen J. High and unchanged sperm counts of Finnish men. Int J Androl 1996; 19: 11-7.

63 Nilsson R. Endocrine modulators in the food chain and environment. Toxicol Pathol 2000; 28: 420-31.

64 Comhaire FH, Mahmoud A. The role of food supplements in the treatment of the infertile man. Reprod Biomed Online 2003; 7: 385-91.

65 Duty SM, Calafat AM, Silva MJ, Ryan L, Hauser R. Phthalate exposure and reproductive hormones in adult men. Hum Reprod 2005; 20: 604-10.

66 Odabas O, Atilla MK, Yilmaz Y, Sekeroglu MR, Sengul E, Aydin S. Luteinizing hormone pulse frequency and amplitude in azoospermic, oligozoospermic and normal fertile men in Turkey. Asian J Androl 2002; 4: 156-8.

67 Ramanujan LN, Liao WX, Roy AC, Ng SC. Association of molecular variant of luteinizing hormone with male infertility. Hum Reprod 2000; 15: 925-8.

68 Andersson AM, Jorgensen N, Frydelund-Larsen L, Rajpert-De Meyts E, Skakkebaek NE. Impaired Leydig cell function in infertile men: a study of 357 idiopathic infertile men and 318 proven controls. J Clin Endocrinol Metab 2004; 89: 3158-60.

69 Andersson AM, Petersen JH, Jorgensen M, Jensen TK, Skakkebaek NE. Serum inhibin B and follicle-stimulating hormone levels as tools in the evaluation of infertile men: significance of adequate reference values from proven fertile men. J Endocrinol Clin Metab 2004; 89: 2873-9.

70 van Golde RJ, van der Avoort IA, Tuerlings JH, Kiemeney LA, Meuleman EJ, Braat DD, et al. Phenotypic characteristics of male subfertility and its familial occurrence. J Androl 2004; 25: 819-23.

71 Lilford R, Jones AM, Bishop DT, Thornton J, Mueller R. Case-control study of whether subfertility in men is familial. BMJ 1994; 309: 570-3.

72 Agarwal A, Saleh RA, Bedaiwy MA. Role of reactive oxygen species in the pathophysiology of human reproduction. Fertil Steril 2003; 79: 829-43.

73 McLachlan RI, Wreford NG, O’Donnel L, De Krester DM, Robertson DM. The endocrine regulation of spermatogenesis: independent roles for testosterone and FSH. J Endocrinol 1996; 148: 1-9.

74 Ochendorf FR. Infections in the male genital tract and reactive oxygen species. Hum Reprod Update 1999; 5: 399-420.

75 Aitken RJ, Krausz C. Oxidative stress, DNA damage and the Y chromosome. Reproduction 2001; 122: 497-506.

76 Depuydt C, Zalata A, Christophe A, Mahmoud A, Comhaire F. Mechanisms of sperm deficiency in male accessory gland infection. Andrologia 1998; 30: 29-33.

77 Sharma RK, Pasqualotto AE, Nelson DR, Thomas AJ, Agarwal A. Relationship between seminal white blood cell count and oxidative stress in men treated at an infertility clinic. J Androl 2001; 22: 575-83.

78 Kaleli S, Ocer F, Irez T, Budak K, Aksu MF. Does leukocyto-spermia associate with poor sperm parameters and sperm functions in male infertility+ The role of different seminal leukocyte concentrations. Eur J Gynecol Reprod Biol 2000; 89: 185-91.

79 Yanushpolsky EH, Politch JA, Hill JA, Anderson DJ. Is leukocytospermia clinically relevant+. Fertil Steril 1996; 66: 822-5.

80 Sanocka D, Kurpisz M. Reactive oxygen species and sperm cells. Reprod Biol Endocrinol 2004; 2: 12-9.

81 Hughes CM, Lewis SE, McKelvey-Martin VJ, Thompson W. A comparison and baseline induced DNA damage in human spermatozoa from fertile and infertile men, using a modified comet assay. Mol Hum Reprod 1996; 2: 613-9.

82 Cheng KH, Cahill DS, Kasai H, Nishimura S, Loeb LA. 8-Hydroxyguanine, an abundant form of oxidative damage, causes G-T and A-C substitutions. J Biol Chem 1992; 267: 166-72.

83 Fraga CG, Motchnik PA, Shigenaga K, Helbock H, Jacob RA, Ames BN. Ascorbic acid protects against endogenous oxidative DNA damage in human sperm. Proc Natl Acad Sci USA 1991; 88: 11003-6.

84 Gil-Guzman E, Ollero M, Lopez MC, Sharma RK, Alvarez JG, Thomas AJ Jr, et al. Differential production of reactive oxygen species by subsets of human spermatozoa at different stages of maturation. Hum Reprod 2001; 16: 1922-30.

85 Kumagai A, Kodama I, Kumagai J, Fukuda J, Kawamura K, Tanikawa H, et al. Xantine oxidase inhibitors suppress testicular germ cell apoptosis induced by experimental cryptorchidism. Mol Hum Reprod 2002; 8: 118-23.

86 Buttke TM, Sandstrom PA. Oxidative stress as a mediator of apoptosis. Immunol Today 1994; 15: 7-10.

87 Comhaire FH, Mahmoud AM, Depuydt CE, Zalata AA, Christophe AB. Mechanisms and effects of male genital tract infection on sperm quality and fertilizing potential: the andrologist’s viewpoint. Hum Reprod Update 1999; 5: 393-8.

88 Sharma RK, Pasqualotto F, Nelson DR, Thomas AJ, Agarwal A. The reactive oxygen species-total antioxidant capacity score is a new measure of oxidative stress to predict male infertility. Hum Reprod 1999; 14: 2801-7.

89 Said TM, Agarwal A, Sharma RK, Thomas AJ, Sikka SC. Impact of sperm morphology on DNA damage caused by oxidative stress induced by beta-nicotinamide adenine dinucle otide phosphate. Fertil Steril 2005; 83: 95-103.

90 Zini A, Fischer MA, Sharir S, Shayegan B, Phang B, Jarvi K. Prevalence of abnormal sperm DNA denaturation in fertile and infertile men. Urology 2002; 60: 1069-72.

91 Fischer MA, Willis J, Zini A. Human sperm DNA integrity: correlation with sperm cytoplasmic droplets. Urology 2003; 61: 207-11.

92 Agarwal A, Prabakaran SA, Said TM. Prevention of oxidative stress injury to sperm. J Androl. 2005; 26:654-60.

93 de Lamirande E, Gagnon C. Reactive oxygen species and human spermatozoa. I. Effects on the motility of intact spermatozoa and on sperm axonemes. J Androl 1992; 13: 368-78.

94 de Lamirande E, Gagnon C. Reactive oxygen species and human spermatozoa. II. Depletion of adenosine triphosphate plays an important role in the inhibition of sperm motility. J Androl 1992; 13: 379-86.

95 Kwon J, Mochida K, Wang YL, Sekiguchi S, Sankai T, Aoki S, et al. Ubiquitin C-terminal hydrolase L-1 is essential for the early apoptotic wave of germinal cells and for sperm quality control during spermatogenesis. Biol Reprod 2005; 73: 29-35.

96 Tesarik J, Greco E, Cohen-Bacrie P, Mendoza C. Germ cell apoptosis in men with complete and incomplete spermatogenesis failure. Mol Hum Reprod 1998; 8: 757-62.

97 Yan W, Huang JX, Lax AS, Pelliniemi L, Salminen E, Poutanen M, et al. Overexpression of Bcl-w in the testis disrupts spermatogenesis: revelation of a role of BCL-W in male germ cells cycle control. Mol Endocrinol 2003; 17: 1868-79.

98 Erkkila K, Aito H, Aalto K, Pentikainen V, Dunkel L. Lactate inhibits germ cell apoptosis in the human testis. Mol Hum Reprod 2002; 8: 109-17.

99 Suomalainen L, Hakala JK, Pentikainen V, Otala M, Erkkila K, Pentikainen MO, et al. Sphingosine-1-phosphate in inhibition of male germ cell apoptosis in the human testis. J Clin Endocrinol Metab 2003; 88: 5572-9.

100 Koji T, Hishikawa Y, Ando H, Nakanishi Y, Kobayashi N. Expression of FAS and FAS ligand in normal and ischemia reperfusion testes: involvement of the Fas system in the induction of the germ cell apoptosis. Biol Reprod 2001; 64: 946-54.

101 Grunenwald S, Paasch U, Said TM, Sharma RK, Glader HJ, Agarwal A. Caspase activation in human spematozoa in response to physiological and pathological stimuli. Fertil Steril 2005; 83: 1106-12.

102 Hikim AP, Wang C, Lue Y, Johnson L, Wang XH, Swerdloff RS. Spontaneous germ cell apoptosis in humans: evidence for ethnic differences in the susceptibility of germ cells to programmed cell death. J Clin Endocrinol Metab 1998; 83: 152-6.

103 Francavilla S, D’Abrizio P, Cordeschi G, Pelliccione F, Necozione S, Ulisse S, et al. Fas expression correlates with human germ cell degeneration in meiotic and post-meiotic arrest of spermatogenesis. Mol Hum Reprod 2002; 8: 213-20.

104 Oheninger S, Morshedi M, Weng SL, Taylor S, Duran H, Beebe S. Frequence and significance of somatic cell apoptosis in human ejaculated spermatozoa. Reprod Biomed Online 2003; 7: 469-79.

105 Oosterhuis GJ, Vermes I. Apoptosis in human ejaculated spermatozoa. J Biol Regul Homeost Agents 2004; 18: 115-9.

106 Lachaud C, Tesarik J, Canadas M, Mendoza C. Apoptosis and necrosis in human ejaculated spermatozoa. Hum Reprod 2004; 19: 607-10.

107 Barroso G, Morshedi M, Oehninger S. Analysis of DNA fragmentation, plasma membrane translocation of phosphatidylserine and oxidative stress in human spermatozoa. Hum Reprod 2000; 15: 1338-44.

108 Shen HM, Dai J, Chia SE, Ong CN. Detection of apoptoic alterations in sperm in subfertile patients and their correlations with sperm quality. Hum Reprod 2002; 17: 1266-73.

109 Sinha Hikim AP, Swerdloff RS. Hormonal and genetic control of germ cells apoptosis in the testis. Rev Reprod 1999; 4: 38-47.

110 Said TM, Paasch U, Glander HJ, Agarwal A. Role of capases in male infertility. Hum Reprod Update 2004; 10: 39-51.

111 Shang XJ, Huang YF, Xiong CL, Xu JP, Yin L, Wang CC. Uraplasma urealyticum infection and apoptosis of spermatogenetic cells. Asian J Androl 1999; 1: 127-9.

112 Rowe PJ, Comhaire FH, Hargreave TB, Mahmoud AMA. WHO manual for the standardized investigation, diagnosis and management of the infertile male. Cambridge: Cambridge University Press, 2000.

113 Diemer T, Desjardins C. Developmental and genetic disorders in spermatogenesis. Hum Reprod Update 1999; 5: 120-40.

114 Huynh T, Mollard R, Trouson A. Selected genetic factors associated with male infertility. Hum Reprod Update 2002; 8: 183-92

115 Forti G, Krausz C. Clinical review 100: Evaluation and treatment of the infertile couple. J Clin Endocrinol Metab 1998; 83: 4177-88.

116 Layman LC. Genetic causes of human infertility. Endocrinol Metab Clin North Am 2003; 32: 549-72.

117 Weidner W, Krause W, Ludwig M. Relevance of male accessory gland infection for subsequent fertility with special focus on prostatitis. Hum Reprod Update 1999; 5: 421-32.

118 Jarow JP. Endocrine causes of male infertility. Urol Clin North Am 2003; 30: 83-90.

119 Islam N, Trainer PJ. The hormonal assessment of the infertile male. BJU Int 1999; 82: 69-75. Tanriverdi F, Silveira LGF, MacColl GS, Bouloux PMG. The hypothalamic-pituitary-gonadal axis: immune function and autoimmunity. J Endocrinol 2003; 176: 293-304.

120 Hafez ESE, Hafez B, Hafez SD. An atlas of reproductive physiology in men. Boca Raton: Parthenon Publishing, 2004, p97-111.

121 Biagiotti G, Cavallini G, Modenini F, Vitali G, Gianaroli L. Spermatogenesis and spectral echo-colour Doppler traces from the main testicular artery. BJU Int 2002; 90: 903-8.

122 Foresta C, Garolla A, Bettella A, Ferlin A, Rossato A, Candiani F. Doppler ultrasound of the testis in azoospermic subjects as a parameter of testicular function.. Hum Reprod 1998; 13: 3090-3.

123 Battaglia C, Giulini S, Regnani G. Seminal plasma nitrite/nitrate and intratesticular Doppler flow in fertile and infertile subjects. Hum Reprod 2000; 15: 2554-8.

124 Battaglia C, Giulini S, Regnani G, Magdar I, Facchinetti F, Volpe A. Intratesticular Doppler flow, seminal plasma nitrites/nitrates and nonobstructive sperm extraction from patients with obstructive and nonobstructive azoospermia. Fertil Steril 2001; 75: 1088-94.

125 Atilla MK, Sargin H, Yilmaz Y, Odabas O, Keskin A, Aydin S. Undescended testes in adults: clinical significance of resistive index values of the testicular artery measured by Doppler ultrasound as a predictor of testicular histology. J Urol 1997; 158: 841-3.

126 Hsu TH, Huang JK, Ho DM, Liu RS, Chen MT, Chang LS. Role of the spermatic artery in spermatogenesis and sex hormone synthesis. Arch Androl 1993; 31: 191-7.

127 Jezek D, Schulze W, Rogatsch H, Hittmair A. Structure of small blood vessels in the testes of infertile men. Int J Androl 1996; 19: 299-306.

128 Meachem SJ, Nieschlag E, Simoni M. Inhibin B in male reproduction: pathophysiology and clinical relevance. Eur J Endocrinol 2001; 145; 561-71.

129 Bohring C, Krause W. Serum levels of inhibin B in men with different causes of spermatogenic failure. Andrologia 1999; 31: 137-41.

130 Comhaire FH, ed. Male Infertility: Clinical investigation, cause evaluation and treatment, 2nd ed. London: Chapman & Hall Medical, 1997, pp.123-31.

131 Chiu WW, Chamley LW. Use of antisperm antibodies in differential display Western blotting to identify sperm proteins important in fertility. Hum Reprod 2002; 17: 984-9.

132 Bohring C, Krause W. Immune infertility: towards a better understanding of sperm (auto)-immunity. The value of proteomic analysis. Hum Reprod 2003; 18: 915-24.

133 Clayton R, Moore H. Experimental models to investigate the pathology of antisperm antibodies: approach and problems. Hum Reprod Update 2001; 7: 457-9.

134 Koide SS, Wang L, Kamada M. Antisperm antibodies associated with infertility: properties and encoding genes of target antigens. Proc Soc Exp Biol Med 2000; 224: 123-32.

135 Jager S, Kremer J, Kuiken J, Mulder I. The significance of the Fc part of antispermatozoal antibodies for the shaking phenomenon in the sperm-cervical mucus contact test. Fertil Steril 1981; 36: 792-7.

136 Zouari R, De Almeida M. Effect of sperm associated antibodies on human sperm ability to bind to zona pellucida and to penetrate zona-free hamster oocytes. J Reprod Immunol 1993; 24: 175-86.

137 Clarke GN. Induction of the shaking phenomenon by IgA class antispermatozoal antibodies from serum. Am J Reprod Immunol 1985; 9: 12-4.

138 Liu DY, Clarke GN, Baker HW. Inhibition of human sperm-zona pellucida and sperm-oolemma binding by antisperm antibodies. Fertil Steril 1991; 55: 440-2.

139 Naz RK. Effects of antisperm antibodies on early cleavage of fertilized ova. Biol Reprod 1992; 46: 130-9.

140 Munuce MJ, Berta CL, Pauluzzi F, Caille AM. Relationship between antisperm antibodies, sperm movement, and semen quality. Urol Int 2000; 65: 200-3.

141 Blasco L. Clinical tests of sperm fertilization ability. Fertil Steril 1984; 41: 177-92.

142 Ford WC, Williams KM, McLaughin EA, Harrison S, Ray B, Hull MG. The indirect immunobead test for seminal antisperm antibodies and fertilization rates at in vitro fertilization. Hum Reprod 1996; 11: 1418-22.

143 Patrizio P, Silber SJ, Ord T, Moretti Rojas I, Asch RH. Relationship of epididymal sperm antibodies to their in vitro fertilization capacity in men with congenital absence of the vas deferens. Fertil Steril 1992; 58: 1006-10.

144 Lenzi A, Lombardo F, Sgr+P, Salacone P, Caponnecchia L, Dondero F. Use of carnitine therapy in selected cases of male factor infertility: a double blind cross over trial. Fertil Steril+2003; 79: 292-300.

145 Lenzi A, Sgr+P, Salacone P, Paoli D, Gilio B, Lombardo F, et al. A placebo-controlled double-blind randomized trial of the use of combined l-carnitine and l-acetyl-carnitine treatment in men with asthenozoospermia. Fertil Steril 2004; 81: 1578-84.

146 Cavallini G, Ferraretti AP, Gianaroli L, Biagiotti G, Vitali G. Cinnoxicam and L-carnitine/acetyl-L-carnitine treatment for idiopathic and varicocele-associated oligoasthenospermia. J Androl 2004; 25: 761-70.

147 Weidner W, Frause W, Ludwig M. Relevance of male of accessory gland infection for subsequent fertility with special focus on prostatitis. Hum Reprod Update 1999; 5: 421-32.

148 Keck C, Gerber-Schafer C, Clad A, Wilhelm C, Breckwoldt M. Seminal tract infections: impact on male fertility and treatment options. Hum Reprod Update 1998; 4: 891-903.

149 Vicari E. Effectiveness and limits of antimicrobial treatment on seminal leukocyte concentration and related reactive oxygen species production in patients with male accessory gland infection. Hum Reprod 2000; 15: 2536-44.

150 Zorn B, Virant-Klun I, Meden-Vrtovec H. Semen granulocyte elastase: its relevance for the diagnosis and prognosis of silent genital tract inflammation. Hum Reprod 2000; 15: 1978-84.

151 Eggert-Kruse W, Boit R, Rohr G, Aufenanger J, Hund M, Strowitzki T. Relationship of seminal plasma interleukin (IL)-8 and IL-6 with semen quality. Hum Reprod 2001; 16: 517-8.

152 Foresta C, Bettella A, Merico M, Garolla A, Ferlin A, Rossato M. Use of recombinant human follicle-stimulating hormone in the treatment of male infertility. Fertil Steril 2002; 77: 238-44.

153 Baccetti P, Piomboni P, Bruni E, Capitani S, Gambera L, Moretti E, et al. Effect of follicle-stimulating hormone on sperm quality and pregnancy rate. Asian J Androl 2004; 6: 133-7.

154 Kamische A, Behre HM, Bergman M, Simoni M, Schafer T, Nieschlag E. Recombinant human follicle stimulating hormone for treatment of male idiopathic infertility: a randomized, double-blind, placebo-controlled, clinical trial. Hum Reprod 1998; 13: 596-603.

155 Sallam H, El-Garem Y, Agameya AF. FSH therapy for oligoasthenospermia - a meta-analysis of randomized trial. Hum Reprod 2004; 19 (Suppl 1) i23. Proecedings of the 20th Annual Meeting of thed European Society of Human Reproduction and Embriology. Berlin, Germany, 27th-30th June 2004.

156 Jansen RP. Relative infertility: modeling clinical paradoxes. Fertil Steril 1993; 59: 1041-5.

157 Adamopoulos DA, Pappa A, Billa E, Nicopoulou S, Koukkou E, Michopoulos J. Effectiveness of combined tamoxifen citrate and testosterone undecanoate treatment in men with idiopathic oligozoospermia. Fertil Steril 2003; 80: 914-20.

158 Comhaire F. Clinical andrology: from evidence-base to ethics. The E-quintet in clinical andrology. Hum Reprod 2000; 15: 2067-71.

159 Vandekerckove P, Lilford R, Vail A, Hughes E.. Clomiphene or tamoxifen for idiopathic oligo/asthenospermia. Cochrane Database Syst Rev 2000; (2): CD000151.

160 Adamopoulos DA. Medical treatment of idiopathic oligozoospermia and male factor subfertility. Asian J Androl 2000; 2: 25-32.

161 Adamopoulos DA, Nicopoulou S, Kapolla N, Karamertzanis M, Andreou E. The combination of testosterone undecanoate with tamoxifen citrate enhances the effects of each agent given independently on seminal parameters in men with idiopathic oligozoospermia. Fertil Steril 1997; 67: 756-62.

162 Vermeulen A, Comhaire F. Hormonal effects of an antiestrogen, tamoxifen, in normal and oligospermic men. Fertil Steril. 1978; 29: 320-7.

163 Willis KJ, London DR, Bevis MA, Butt WR, Lynch SS, Holder G. Hormonal effects of tamoxifen in oligospermic men. J Endocrinol 1977; 73: 171-8.

164 Vermeulen A, Comhaire F. Hormonal effects of an antiestrogen, tamoxifen, in normal and oligospermic men. Fertil Steril 1978; 29: 320-7.

165 Smals AG, Pieters GF, Drayer JI, Boers GH, Benraad TJ, Kloppenborg PW. Tamoxifen suppresses gonadotropin-induced 17 alpha-hydroxyprogesterone accumulation in normal men. J Clin Endocrinol Metab 1980; 52: 1026-9.

166 Wong WY, Merkus HM, Thomas CM, Menkveld R, Zielhuis GA, Steegers-Theunissen RP. Effects of folic acid and zinc sulfate on male factor subfertility: a double-blind, randomized, placebo-controlled trial. Fertil Steril 2002; 77: 491-8.

167 Gulmez I, Tatlisen A, Karacagil M, Kesekci S.. Seminal parameters of ejaculates collected successively with sixty minute interval in infertile men: effect of combination of prazosin and terbutaline on these parameters. Andrologia 1991; 23: 167-9.

168 Yamamoto M, Hibi H, Miyake K. Comparison of the effectiveness of placebo and alpha-blocker therapy for the treatment of idiopathic oligozoospermia. Fertil Steril 1995; 63: 396-400.

169 Gregoriou O, Vitoratos N, Papadias C, Gargaropoulos A, Konidaris S, Giannopoulos V, et al. Treatment of idiopathic oligozoospermia with an alpha-blocker: a placebo-controlled double-blind trial. Int J Fertil Womens Med 1997; 42: 301-5.

170 Apa DD, Cayan S, Polat A, Akaby E. Mast cells and fibrosis on testicular biopsies in male infertility. Arch Androl 2002; 48: 337-44.

171 Yamamoto M, Hibi H, Miyake K. New treatment of idiopathic severe oligozoospermia with mast cell blocker: results of a single-blind study. Fertil Steril 1995: 64: 1221-3.

172 Hibi H, Kato K, Mitsui K, Taki T, Yamada Y, Honda N, et al. The treatment with tranilast, a mast cell blocker, for idiopathic oligozoospermia. Arch Androl 2001; 47: 107-11.

173 Hibi H, Kato K, Mitsui K, Taki T, Yamada Y, Honda N, et al. Treatment of oligoasthenozoospermia with tranilast, a mast cell blocker, after long-term administration. Arch Androl 2002; 48: 451-9.

174 Cayan S, Apa DD, Akbay E. Effect of fexofenadine, a mast cell blocker, in infertile men with significantly increased testicular mast cells. Asian J Androl 2002; 4: 291-4.

175 Vicari E, Calogero AE. Effects of treatment with carnitines in infertile patients with prostato-vesiculo-epididymitis. Hum Reprod 2001; 16: 2338-42.

176 Cavallini G, Biagiotti G, Ferraretti AP, Gianaroli L, Vitali G. Medical therapy of oligoasthenospermia associated to left varicocele: an option. BJU Int 2003; 91: 513-8.

177 Comhaire FH, Mahmoud AM. [Editorial commentary on: Cavallini G, Ferraretti AP, Gianaroli L, Biagiotti G, Vitali G. Cinnoxicam and L-carnitine/acetyl-L-carnitine treatment for idiopathic and varicocele-associated oligoasthenospermia. J Androl 2004; 25: 761-70]. J Androl 2004; 25: 771-2.

178 Loescher W, Littengau H, Schlegel W. Pharmacokinetics of non steroidal anti-inflammatory drugs in male rabbits after acute and chronic administration and effect of chronic treatment on seminal plasma. J Reprod Fertil 1988; 82: 353-64.

179 Cipolla MJ, Nnicoloff A, Rebello T, Amato A, Porter JM. Propionyl-L-carnitine dilates human subcutaneous arteries through an endothelium-dependent mechanism. J Vasc Surg 1999; 29: 1097-103.

180 Fuse H, Minagawa H, Ito H, Shimazaki J. The effects of prostaglandin synthetase inhibitor on male infertility. Hinyokika Kiyo 1984; 30: 1439-45.

181 Bendvold E, Gottlieb C, Svanborg K, Bygdeman M, Eneroth P. Concentration of prostaglandins in seminal fluid of fertile men. Int J Androl 1987; 10: 463-9.

182 Comhaire FH, Christophe AB, Zalata AA, Dhooge WS, Mahmoud AM, Depuydt CE. The effects of combined conventional treatment, oral antioxidants and essential fatty acids on sperm biology in subfertile men. Prostaglandins Leukot Essent Fatty Acids 2000; 6: 159-65.

183 Kesker-Ammar L, Feki Chacroun N, Rebai T, Sahnoun Z, Ghozzi H, Hammami S, et al. Sperm oxidative stress and the effect of an oral vitamin E and selenium supplement on semen quality in infertile men. Arch Androl 2003; 49: 83-94.

184 Rolf C, Cooper TG, Yeung CH, Nieschlag E. Antioxidant treatment of patients with asthenozoospermia or moderate oligoasthenozoospermia with high-dose vitamin C and vitamin E: a randomized, placebo-controlled, double-blind study. Hum Reprod 1999; 14: 1028-33.

185 Gyllenborg J, Skakkebaek K, Nielsen NC. Secular and seasonal changes in semen quality among young Danish men: a statistical analysis of semen samples from 1927 donor candidates during 1977-1995. Int J Androl 1999; 22: 28-36.

186 Jorgensen N, Andersen AG, Eustache F, Irvine DS, Suominen J, Petersen JH, et al. Regional differences in semen quality in Europe. Hum Reprod 2001; 16: 1012-9.

187 von Eckardstein S, Syska A, Gromoll J, Kamischke A, Simoni M, Nieschlag E. Inverse correlation between sperm concentration and number of androgen receptor CAG repeats in normal men. J Endocrinol Clin Metab 2001; 86: 2585-90.

กก