Journal List > World J Mens Health > v.33(3) > 1088765

Harlev, Agarwal, Gunes, Shetty, and du Plessis: Smoking and Male Infertility: An Evidence-Based Review

Abstract

Many studies have reported that the contents of cigarette smoke negatively affect sperm parameters, seminal plasma, and various other fertility factors. Nevertheless, the actual effect of smoking on male fertility is not clear. The effect of smoking on semen parameters is based on the well-established biological finding that smoking increases the presence of reactive oxygen species, thereby resulting in oxidative stress (OS). OS has devastating effects on sperm parameters, such as viability and morphology, and impairs sperm function, hence reducing male fertility. However, not all studies have come to the same conclusions. This review sheds light upon the arguable association between smoking and male fertility and also assesses the impact of non-smoking routes of tobacco consumption on male infertility. It also highlights the evidence that links smoking with male infertility, including newly emerging genetic and epigenetic data, and discusses the clinical implications thereof.

INTRODUCTION

Despite the well-known detrimental impacts of smoking on health in general and on the male reproductive system in particular [1], smoking remains a worldwide phenomenon. According to the World Health Organization (WHO), 30% of all 15 years and older men, smoke [2]. Men of reproductive age (20~39 years of age) make up approximately 46% of smokers [3]. Although the prevalence of male smokers, based on nationally representative sources from 187 countries, decreased from 41.2% in 1980 to 31.1% in 2012, the actual number of everyday smokers increased from 721 million in 1980 to 967 million in 2012 [4].
The WHO reports that approximately 8% of couples worldwide and 10% to 15% of those in industrialized nations experience infertility [5], and that male factor infertility is involved in 30% to 35% of all cases [6]. In order to identify possible associations between smoking and male infertility, numerous studies have been conducted, with some reporting contradictory results. While many studies have reported a negative impact of smoking on semen analysis parameters and male infertility [78910], others have found no such effects, and in some cases, have even found positive effects on sperm motility [811] and the extent of nuclear DNA damage in sperm [12]. Other inconsistent and conflicting data regarding the influence of smoking on male infertility have been reported [121314]. Moreover, even studies reporting an effect of smoking on semen parameters have not clearly demonstrated any effect of smoking on male fertility [151617].
These opposing and confusing results are not surprising. Three possible explanations for the emergence of contradictory results can be hypothesized. First, several metrics are used to assess the influence of smoking on male fertility, such as semen parameters, spermatozoa function, histologic alterations, and others. The use of different assessments could result in inconsistent findings among studies. Second, the mechanisms of how smoking may influence male fertility have not been securely established. Third, it is challenging to directly compare data among the studies due to the difficulty of adjusting for confounders such as exposure to alcohol use, medical illnesses, toxins, and socioeconomic status.
In this review, we discuss the content of cigarettes and detail the effect of various forms of tobacco exposure on male infertility. Next, we characterize the effect of smoking on different aspects of the male reproductive system, focusing on both the physiological and pathological impacts of smoking. Subsequently, this study summarizes the possible mechanisms through which smoking causes genetic and epigenetic alternations in male reproductive function, a topic that has not been thoroughly reviewed previously. Finally, we discuss how this area of research should advance to meet the clinical needs of infertile smokers.

SMOKING: AN OVERVIEW

1. Content of cigarette smoke

Cigarette smoke is composed of gases, vaporized liquid, and particles. Nearly 4,000 compounds are released through the chemical processes of hydrogenation, pyrolysis, oxidation, decarboxylation, and dehydration. The smoke release is biphasic (gaseous and particulate). In the gaseous phase, carbon monoxide is released [18], and in the particulate phase, nicotine and tar are released. Cigarette smoke contains several toxic chemicals, mutagenic substances, and carcinogens, including nicotine and its metabolites, cotinine, radioactive polonium, benzopyrene, dimethylbenzanthracene, naphthalene, methylnaphthalene, polycyclic aromatic hydrocarbons (PAHs), and cadmium [1920].
The primary psychoactive component of tobacco is nicotine. Nicotine is the active element responsible for tobacco addiction. The majority of nicotine in humans is metabolized to cotinine, and cotinine is further metabolized to trans-3'-hydroxycotinine (3HC) [21]. In cigarette smokers, seminal and serum levels of cotinine and 3HC appear to be similar, whereas seminal nicotine levels are generally higher than serum levels [22]. While total sperm motility is negatively correlated with seminal cotinine and 3HC levels, forward sperm motility is correlated with seminal cotinine levels [22].
The major active components of smoke that influence semen parameters are the heavy metals cadmium and lead. The negative effect of cadmium on sperm parameters has been previously shown in animal studies [23]. Elevated seminal cadmium in smokers has been observed if >20 cigarettes/day are consumed, and cadmium levels in the blood have been found show a statistically significant positive correlation with cigarette-years and a statistically significant negative correlation with sperm density [24]. Additionally, lead levels in seminal plasma have been shown to be higher in infertile smokers than in fertile men and infertile non-smokers [25]. Moreover, negative associations between seminal lead and cadmium concentrations and sperm concentration, motility, and morphological abnormalities in abnormal spermatozoa have been observed [26].

2. Means of smoking and routes of nicotine exposure

Several types of smoke are produced when a cigarette is smoked (Fig. 1). Mainstream smoke is the actual mouth-to-cigarette inhalation of cigarette smoke into the smoker's lungs. Exhaled mainstream smoke is the smoke that escapes into the environment after it is drawn through the cigarette, filtered by the smoker's lungs, and exhaled. Sidestream smoke arises from the burning end of the cigarette and enters directly the environment directly [7]. Firsthand or active smoking includes inhaled and exhaled mainstream smoke, as well as sidestream smoke. This type of smoking may affect sperm quality, most notably sperm concentration, motility, and morphology [27282930].
Passive or secondhand smoking mainly refers to the involuntary inhalation of tobacco smoke present in the air. Most of this smoke comes from the cigarette's burning end, while about 10% originates from exhaled mainstream smoke [31]. Approximately 50% of non-smokers are passive smokers. Serum cotinine, a biomarker of tobacco smoke exposure, can be detected in approximately 50% of the non-smoking USA population. Secondhand smoke contains thousands of compounds, many of which are known to be harmful to humans [32]. The negative effects of sidestream and passive smoke are fairly well understood, and these types of smoke can negatively affect reproduction in non-smokers with excessive exposure to cigarette smoke [17].
Comparing the effect of mainstream and sidestream smoke on mouse male germ cells, Polyzos et al [33] showed that sidestream smoke reduced sperm motility, whereas mainstream smoke increased sperm DNA fragmentation. Both mainstream and sidestream smoke adversely affected sperm chromatin integrity and reduced fertilization rates. They concluded that male exposure to secondhand smoke prior to fertilization is likely to have negative results on the reproduction process.
The findings of epidemiological studies of people exposed to secondhand smoke can be difficult to evaluate due to the difficulties in determining the amount of secondhand smoke exposure, confounding exposures (the occasional cigarette or environmental exposure), and the lack of parallel but fully unexposed control groups [34]. For instance, maternal cigarette smoke exposure is often a confounding variable that makes analyzing the effects of paternal secondhand smoke exposure on fertilization and early embryonic development particularly challenging. However, a number of toxicants are at higher levels in sidestream smoke than in mainstream smoke and have been shown to effect sperm motility, including reactive oxygen species (ROS) such as superoxide and hydrogen peroxide [35363738], as well as cadmium [3738].
Electronic cigarettes have a liquid-filled container that holds concentrated flavors, humectant (i.e., propylene glycol, vegetable glycerin, and/or polyethylene glycol 400), and variable concentrations of nicotine [39]. Several analyses have detected toxicants and carcinogens similar to those found in cigarette smoke, although usually at much lower levels [4041]. The safety of electronic cigarettes has not yet been well established [42]. A recent systematic literature review [43] concluded that the current data do not warrant health concerns, at least according to the standards used to guarantee workplace safety. However, electronic cigarette use exposes users to aerosol that contains toxicants, making further research necessary.
Chewing tobacco is one of the most common methods of smokeless tobacco intake, with approximately 6 million to 22 million users of smokeless tobacco products in the USA alone [44]. Sales of smokeless tobacco have increased over the past decade [4546]. The nicotine distribution features of smokeless tobacco make it both addictive and a feasible substitute for cigarette smoking for many consumers, since it delivers a higher nicotine dose than other tobacco cessation products [47]. Chewing tobacco is markedly less harmful than smoking, but it is not harmless. Its nicotine absorbance is equal to that from cigarettes, but with a less toxic effect [48]. Chewing tobacco increases the risk of multiple oral premalignant lesions [4950], respiratory tract cancers [51], and cardiovascular disease [48]. Moreover, chewing tobacco affects semen parameters in a dose-dependent manner. Sperm concentration, percentage of motility, morphology, and viability have been found to be significantly reduced in tobacco chewers [5253]. Structural defects in the sperm head and cytoplasmic residues were correlated with the intensive use of chewing tobacco, but anomalies in the midpiece and tail were not observed [53].
Nicotine patches and nicotine gum are commonly used as nicotine replacement therapy for those who want to stop smoking. Since orally ingested nicotine is metabolized first in the liver, the bioavailability of swallowed nicotine medication is reduced. Skin transdermal patches and nicotine intake through the oral mucosa (chewing gum, sublingual tablets, etc.) avoid this reduction in bioavailability [54]. Although nicotine is absorbed transdermally, no major side effects or complications have been reported with these products [555657]. We found no well-conducted studies dealing with the influence of nicotine patches or gum on fertility.

EFFECTS AND POSSIBLE MECHANISM OF SMOKING ON MALE INFERTILITY

In studies investigating the influence of cigarette smoke on semen quality, most researchers have studied semen parameters based on the basic WHO recommendations for semen analysis, which include volume, count, concentration, motility, and morphology.

1. Smoking and sperm parameters: concentration, motility, and morphology

Table 1 [5859606162636465666768697071727374757677787980] summarizes the results of studies evaluating the effect of smoking on semen parameters. Although many studies did observe a negative influence of smoking on semen quality, many other studies did not show a significant effect on one or more basic sperm parameters.
Künzle et al [10] prospectively studied 2,105 participants, of whom 839 persons (39.9%) were smokers and 1,266 persons (60.1%) were non-smokers. Sperm concentration, motility, and morphology were significantly affected in smokers. Nevertheless, sperm vitality, ejaculate volume, and progressive motility concentrations were slightly but not significantly reduced among smokers. In a large cross-sectional study involving the general healthy population, Ramlau-Hansen et al [80] observed a significant reduction in sperm concentration, total sperm count, and motility in smokers. In a meta-analysis, Vine et al [81] reported that the sperm concentration in smokers was approximately 13% lower than in non-smokers on average. Among normal healthy males, a 24% lower sperm concentration was observed in smokers in comparison to non-smokers [81].
In order to investigate the effects of smoking on sperm parameters, we searched the literature for studies that compared differences between smokers and non-smokers, since simply comparing data from smokers with WHO reference values cannot provide a true reflection of the changes caused by smoking. A total of 38 relevant studies were found, five of which were excluded based on methodological concerns and sample size. Twenty-eight of the remaining 33 studies were prospective and five were retrospective studies (Table 1).
When stratified according to study population, 13 studies were performed on a healthy population and 20 were performed on infertile men. Due to the diversity of the studies and of the study populations, it was challenging to extrapolate the data to draw definitive conclusions. When all studies comparing smokers to non-smokers were considered, 36.4% reported a significant reduction in motility and concentration, and 41.4% showed a decrease in normal morphology. When the studies were stratified based on the studied population, it was observed that motility, concentration, and morphology were affected in 23.1%, 38.5%, and 44.4% of the healthy population and in 40%, 30%, and 42.1% of the infertile population studies, respectively, when smokers were compared to non-smokers (Fig. 2).
A number of studies in our analysis reported adverse effects of smoking on sperm parameters. However, a large number of studies showed no significant effect, resulting in a lack of conclusive evidence. To date, only a single meta-analysis has been performed on the effects of smoking on semen parameters, but that study mainly focused on sperm concentration. The authors showed that exposure to more than 10 cigarettes per day reduced sperm concentration by 13% to 17% [81]. The latest American Society for Reproductive Medicine Committee opinion [82] states that smoking can affect basic semen parameters in a dose-dependent manner. However, the values still mostly remain within the normal reference ranges. Therefore, future studies should investigate the effect of cigarette smoking on sperm function at a molecular level.

2. Smoking and male infertility: dose-response correlations

Despite the large number of studies performed using basic semen parameters, the available data do not conclusively indicate the impact of smoking on male fertility [17]. However, two conclusions may be drawn. First, a more significant association between smoking and basic sperm parameters may be observed in studies that investigated the general population than in studies involving infertile men [83]. Second, a dose-dependent relationship between the amount of cigarette consumption and negative effects on semen parameters was observed [808485]. In a large cross-sectional study, Ramlau-Hansen et al [80] reported an inverse dose-based association between smoking and semen volume, total sperm count, and the percentage of motile spermatozoa. The sperm concentration of heavy smokers was 19% lower than that of non-smokers. Therefore, smoking cessation, or at least a reduction in cigarette use, may be advised as a way of reducing toxin exposure [86].

3. Smoking and spermatozoa: biological function

The axoneme, a basic structural organ of motile cilia and flagella, is composed of microtubules. Variations in both the quantity and positioning of axonemal microtubules in smokers has been described [87]. In one study, ultrastructural assessment of the axoneme revealed aberrations in 99% of smokers and in 26% of non-smokers. These smoke-induced variations in the normal axoneme structure could impair flagellar movement, causing the sperm motility pathologies seen in smokers [88].
Another important biological activity is sperm motility. Creatine kinase is an enzyme expressed by cells, such as spermatozoa, that require large amounts of energy. It plays a major role in adenosine triphosphate (ATP) and adenosine diphosphate metabolism [89], and also provides an ATP buffering system. Ghaffari and Rostami [90] reported that creatine kinase activity in sperm was reduced in smokers, affecting sperm motility and overall fertility.
Acrosin is a proteolytic enzyme that is released by the sperm when it comes into contact with an ovum, degrading the zona pellucida of the oocyte and allowing the sperm to penetrate it [91]. Smokers have been found to display lower acrosin activity than non-smokers, even in the presence of normal semen parameters [9192].
The effect of smoking on sperm function and ability to fertilize have been investigated in vivo, and a negative correlation was observed [93]. Furthermore, sperm penetration assays in cigarette smokers have shown a correlation with poor sperm function [2894].
The functional impairment of sperm highlights the possibility of male subfertility in the presence of normal semen parameters, and also calls into question the ability of semen-based parameters to detect subfertility in infertile males who smoke.

4. Oxidative stress and leukocytospermia

Oxidative stress (OS) develops from an imbalance between ROS and natural antioxidant defenses. Such an imbalance may be caused by an elevation in ROS, a decrease in total antioxidant capacity (TAC), or both.
Cigarette smoking has been correlated with increased seminal OS markers, as shown by a prominent increase in ROS levels [95] and a decrease in ROS-TAC measures [96]. Spermatozoa are particularly prone to damage caused by excessive ROS due to the large amount of polyunsaturated fatty acids in the plasma membrane that are substrates for ROS [97] and the low concentrations of scavenging enzymes in their cytoplasm [98].
Elevated seminal ROS levels in smokers could stem from either endogenous or exogenous sources. While the exogenic sources of ROS consist of environmental factors such as smoking [99], alcohol use [100], and air pollutants [101], leukocytes (neutrophils and macrophages) are the main endogenic source of ROS [102], although immature spermatozoa contribute as well [103]. A correlation between cigarette smoking and leukocytospermia has been well established [294]. As a major producer of ROS in seminal fluid, elevated leukocyte levels hinder fertility by exposing spermatozoa to OS [104], which in turn damages sperm DNA and the bilayer lipid membrane [105]. In addition, tobacco metabolites can generate an inflammatory response, triggering the activation and infiltration of leukocytes into the seminal plasma [106]. Sequentially, activated leukocytes elevate levels of ROS in the seminal fluid, which may overwhelm antioxidant defenses, resulting in OS [107]. An additional source of ROS in smokers is cigarette smoke itself, which contains high levels of ROS such as superoxide anion, hydrogen peroxide, and hydroxyl radicals [108].
Intracellular antioxidant enzymes function to reduce ROS levels. When these enzymes are not capable of protecting the plasma membrane, cell function is altered [109]. The seminal plasma supports and protects spermatozoa from pathological levels of ROS through free radical scavengers such as ascorbate, alpha-tocopherol, uric acid, and ROS-metabolizing enzymes such as superoxide dismutase (against superoxide), catalase (against hydrogen peroxide), and glutathione peroxidase (against glutathione peroxide) [110]. Ascorbic acid is an essential antioxidant in seminal fluid. Human seminal plasma contains up to 10 mg/dL of ascorbic acid, which is more than nine times the blood plasma concentration [111]. For instance, male heavy smokers have been found to have a 20% to 40% reduction in serum ascorbic acid levels, but when they are given as supplements the sperm quality has been shown to improve [111112]. Likewise, Zhang et al [71] compared seminal zinc, copper, and superoxide dismutase levels in medium and heavy long-term smokers and non-smokers, and found negative correlations between the amount and duration of cigarette smoking and semen quality as reflected by antioxidant levels.

DIRECT AND INDIRECT EFFECTS OF SMOKING ON THE MALE REPRODUCTIVE SYSTEM

1. Epididymis

The epididymis is the site of spermatozoa maturation where sperm develop their tails. However, the adequate maturation of sperm is known to be inhibited by various factors, including smoking [113]. Detached ciliary tufts have been detected in the semen of smokers [114]. It is thought that detached ciliary tufts are derived from the epithelium of the epididymal lining. Detached ciliary tufts may indicate a testicular pathology with an epididymal involvement as a consequence of smoking [114].

2. Varicocele

Varicocele is the abnormal dilation of the pampiniform plexus of the veins near the upper and lateral parts of the scrotum. This dilation is similar to that of varicose veins in the legs, which occurs due to valve insufficiency that prevents blood backflow. As a consequence, blood pools in the pampiniform plexus of veins, dilating the blood vessels and causing an elevated temperature in the groin. Hyperthermia has been proven to be harmful to the normal development of spermatozoa and may result in reduced male fertility [115116].
The combination of smoking and varicocele has been found to be correlated with a 10 times greater incidence of oligozoospermia than observed in non-smoking men with varicocele and a five times greater than the incidence in men who smoked but did not have varicocele [117]. This impairment may be attributed to the increased catecholamine secretion from the adrenal medulla that results from smoking [118]. The secreted catecholamines reach the testes by flowing down through the internal spermatic vein. The significant increase of OS due to smoking, varicocele, and hyperthermia of the scrotal region is a major contributor to dysfunction in the male reproductive system [79].

3. Erectile dysfunction

Both firsthand and secondhand smoking have been conclusively proven to be modifiable risk factors for erectile dysfunction [119120121]. In a systematic review that included four prospective cohort studies and four case-control studies, Cao et al [121] reported that smoking significantly increased the risk of erectile dysfunction. Smoking cessation significantly improved both physiological and sexual health in male smokers, regardless of their baseline level of erectile dysfunction [122123].

4. Accessory glands and seminal plasma

The accessory sex glands of the male reproductive tract include the prostate gland, seminal vesicles, and bulbourethral glands. The role of these accessory glands is to secrete a fluid that helps maintain the normal homeostasis of spermatozoa once they leave the epididymis, where they have matured and gained motility. Seminal fluid contains enzymatic and non-enzymatic antioxidants as well as physiological ROS [124].
The function of the accessory glands in smokers has been studied through the evaluation of a number of glandular markers in the ejaculate, including N-acetyl amino sugar, total phosphate (a marker of the functionality seminal vesicles), zinc, acid phosphatase (a marker of prostate gland functionality), and alpha-1,4-glucosidase (a marker of epididymal function). It was found that smoking reduced both vesicular and prostatic parameters in smokers [125]. Exposing spermatozoa from non-smokers to the seminal plasma of smokers significantly reduced sperm motility and the acrosome reaction, as well as elevating malondialdehyde levels [126]. Conversely, exposing spermatozoa from smokers to non-smokers' seminal plasma led to a nonsignificant improvement in the functional parameters of the sperm. Hence, removing a smoker's seminal plasma and subsequently reconstituting the sample with physiological media could be of clinical significance in the various assisted reproductive technology programs utilized for smokers [70127].

5. Hypothalamic-pituitary-gonadal axis

Nicotine can alter the hypothalamic-pituitary axis by stimulating the release of growth hormone, cortisol, vasopressin, and oxytocin, which in turn inhibit luteinizing hormone (LH) and prolactin [128]. In a study assessing the effects of tobacco smoking on hormone levels, Ochedalski et al [129] reported that mean estradiol levels were higher and mean levels of LH, follicle-stimulating hormone (FSH), and prolactin were lower in smokers than in non-smokers, whereas the mean levels of testosterone and dehydroepiandrosterone did not differ. Similar results were reported in other studies investigating plasma or seminal fluid hormone levels [130]. Ramlau-Hansen et al [80] observed a positive dose-response relationship between smoking and testosterone, LH, and the ratio of LH to free testosterone. Moreover, FSH and inhibin B levels were also found to be increased in smokers. With regard to the physiology of the hypothalamic-pituitary-gonadal (HPG) axis, when FSH and LH levels increase, a resulting elevation of testosterone and inhibin B is anticipated, which in turn decreases FSH and LH levels by negative feedback. The results of this study led to the proposal that elements of tobacco smoke might interrupt the regular functioning of the HPG system, leading to Leydig cell failure in smokers [80].
However, in a study evaluating hormone levels in 889 fertile men divided into mild, moderate, and heavy smokers, no significant differences in FSH, LH, or serum total testosterone levels were observed [79].

6. Testicular endocrine and spermatogenetic function

Direct toxic testicular damage in smokers may interrupt testicular endocrine and spermatogenetic function. The two major roles of the testis are spermatogenesis and the hormonal secretion of androgens. Contradictory results have been found regarding the influence of smoking on the final products of the HPG axis, mainly androgens. Some evidence indicates that smoking influences the secretory function of Sertoli and Leydig cells in the testis, thereby causing the impaired sperm quality seen in smokers.
The vascular blood supply in the spermatic cord is relatively insufficient, and smoking is known to compromise oxygen delivery [131]. The process of spermatogenesis has a high metabolic demand. However, the testis is physiologically vulnerable to hypoxia. Oxygen insufficiency, which can be caused by smoking, is therefore suspected to negatively affect testicular function.

SMOKING-RELATED GENETIC, EPIGENETIC AND MOLECULAR ALTERATIONS: RELATIONSHIP WITH MALE INFERTILITY

1. Smoking-induced DNA damage in sperm

Tobacco smoke contains harmful compounds and generates reactive intermediates, such as ROS and reactive nitrogen species, which can induce multiple genetic and epigenetic changes. Through the interaction of these intermediates, exposure to tobacco smoke can directly or indirectly cause the formation of DNA and protein adducts, mutations, chromosomal abnormalities, micronucleus formation, sister chromatid exchange (SCE), and promoter methylation (Fig. 3). Interestingly, even tissues that are not directly exposed to tobacco smoke show elevated levels of DNA adducts due to indirect systemic exposure. Furthermore, tobacco smoke and its contents lead to oxidative DNA damage [132133].
PAHs are formed during the combustion of tobacco and target endothelial cells. Well-known examples of PAHs include benzo[a]pyrene and benzo[a]pyrene diol epoxide. These PAHs cause the formation of DNA adducts at the N2 position of guanine and can be detected in both somatic cells and spermatozoa [133134135]. Some studies have found that higher levels of DNA adduct formation and DNA damage were associated with sperm parameters. In a small subfertile group, DNA adducts were found to be inversely related, to a statistically nonsignificant extent, with both sperm concentration and motility [136137]. Another study showed that these adducts were transmitted to the zygote and that they were not always repaired by the ovum [132].
No consensus exists about whether smoking affects spermatozoa DNA. Gallagher et al [138] found no significant variation in DNA adduct levels in sperm from heavy smokers (≥20 cigarettes/d), light smokers (<20 cigarettes/d), and non-smokers. However, a more recent study found significantly higher levels of DNA adducts in the spermatozoa of smokers [134]. 8-oxo-2'-deoxyguanosine, a mutagenic base modification of DNA that occurs upon exposure to carcinogens such as tobacco smoke, was also shown to be present at a significantly higher level in the DNA of spermatozoa from smokers than in the DNA of spermatozoa from age-matched non-smokers (Fig. 3) [74].
As summarized by Begum [135], several studies have reported phenotypic and molecular abnormalities in cell lines treated with cigarette smoke condensate, including lagging chromosomes, sticky chromosomes during anaphase and telophase, micronucleus formation, and an increased frequency of SCEs [135]. A micronucleus is the formation of a small nucleus as a result of lagging whole chromosome or chromosomal fragments during anaphase in both meiosis and mitosis. DNA damage, as well as spindle and kinetochore defects, may lead to micronucleus formation in the germ line. It is also widely recognized that a higher incidence of chromosomal instability, as assessed using micronuclei as a marker, is present in the infertile population [139]. Lähdetie [140] analyzed micronuclei in Golgi-phase or cap-phase spermatids found in semen samples from 62 subfertile men. Smokers had a 1.7-fold increase in micronuclei formation compared to non-smokers, although the difference was not statistically significant and was not related to the number of cigarettes smoked daily. These findings are supported by those of another study that did not find an association between smoking habits and the frequency of micronuclei in immature seminal germ cells [141].
SCE is the exchange of genetic material between two parental sister chromatids during replication. During these exchanges, which are caused by DNA damage, both DNA strands break, allowing whole DNA duplexes to cross between the chromatids. Two studies have reported an association between infertility and SCE [142143]. Since SCE can be caused by cigarette smoke, it is therefore possible that cigarette smoke is responsible for genetic causes of infertility via this mechanism.
It is apparent that all smokers are not infertile, which suggests that genetic variations or polymorphisms in DNA repair, apoptosis, and xenobiotic metabolism genes among smokers may increase susceptibility to infertility. Recently published studies have reported a link between genetic variants of xenobiotic metabolism genes and infertility in smokers. Glutathione S-transferases (GSTs) are isoenzymes involved in the detoxification of both endogenous and exogenous compounds. Yarosh et al [144] found a statistically significant relationship between the GSTM1+/GSTT1 del genotypes and the GST gene GSTP1 105IV/GSTT1 polymorphism and smoking in infertile men. The GSTM1, GSTT1, and GSTP1 genes are collectively involved in the development of idiopathic male infertility, and their phenotypic effects on the risk of disease are potentiated by cigarette smoking [144]. N-acetyltransferase (NAT2) is a xenobiotic metabolism enzyme that plays a role in the detoxification of toxicants of tobacco, such as hydrazines, aromatic amines, and arylamines. Hypoacetylated genotypes of NAT2 have been shown to be associated with a link between cigarette smoking and infertility, presumably by increasing ROS production and OS (odds ratio [OR]=1.71, 95% confidence interval [CI]=1.02~2.87, p=0.042) [145].
The polymorphism 590G>A of NAT2 can therefore be used as a novel genetic marker for susceptibility to idiopathic male infertility, but the risk is potentiated by exposure to various environmental oxidants, including cigarette smoke.
Cytochrome P450, a family 1, subfamily A polypeptide 1 (CYP1A1) is another xenobiotic metabolizing enzyme that participates in the metabolism of PAHs. In another study by Yarosh et al. [146], an increased risk of idiopathic male infertility was reported in male smokers who were carriers of the 462Ile/Val genotype of the CYP1A1 gene (OR=1.91; 95% CI=1.01~3.59), while non-smokers did not show an increased risk of infertility [146].
In another study, amniocytes grown in nicotine-containing media showed numerical and structural abnormalities in their autosomal chromosomes, including chromosomes 8, 15, 20, 21, and 22 [147]. In addition, the accumulation of such smoking-induced molecular alterations may increase aneuploidy levels in spermatozoa. Numerical chromosomal alterations in both paternal and maternal gametes are a direct cause of infertility and/or spontaneous abortions. Nondisjunction and anaphase lagging in paternal meiosis are also responsible for about 5% to 10% of autosomal aneuploidies, approximately half of Klinefelter syndrome cases (47,XXY), nearly 70% of Turner syndrome cases (45,X), and all cases of 47,XYY syndrome [148]. A recent study in a small cohort showed a significant increase in disomy frequency of chromosomes 3 and XY in the spermatozoa of smokers. Interestingly, these findings were independent of the number of cigarettes smoked [149]. These studies make it quite evident that the male meiotic process is affected by genotoxic damage from tobacco use.

2. Smoking-induced epigenetic alterations in infertile men

Epigenetic changes alter gene expression without changing the DNA sequence, and can be transmitted through both mitotic and meiotic cell divisions [150]. Several epigenetic mechanisms have been defined, including DNA methylation, histone modifications, and chromatin remodeling. However, the activity of noncoding RNA and miRNA are not considered to be an epigenetic process [151].
Epigenetic and DNA methylation studies have indicated that smoking induces methylation in the genome in the majority of loci studied [152153154155]. A recent genome-wide DNA methylation study reported alterations in the methylation profile of 95 sites in smokers, and furthermore suggested a possible association between methylation status and infertility [156].
In somatic cells and oocytes, DNA is packaged by proteins known as histones. Protamines are proteins smaller than histones that enable the further condensation of DNA in spermatozoa. During the advanced stages of spermatogenesis, histones are replaced by protamines. Protamination is an epigenetic regulatory process specific to spermatozoa [75]. The protamination of sperm chromatin facilitates compaction of the nucleus, protecting the paternal genome during its passage and delivery to the oocyte. Moreover, the stability of this nuclear structure protects the sperm genome from oxidation and other detrimental changes in the female reproductive tract [157].
Yu et al [75] reported that the histone abnormality rate in a Chinese population was lowest in normozoospermic non-smoking men and highest in heavy smokers with oligospermia. Similarly, a European population-based study showed a significant decrease in protamine 2 (P2) concentrations in smokers, which subsequently caused an abnormal elevation of the P1:P2 protein ratio [158]. Furthermore, OS and smoking markers such as ROS, malondialdehyde, 8-hydroxyguanosine (8-OHdG), and cotinine were significantly higher (p<0.010) in smokers than in non-smokers and correlated significantly (p<0.050) with P1:P2 ratios. ROS, 8-OHdG, and cotinine also correlated negatively with P2 levels.

1) miRNA/noncoding RNA

MicroRNAs (miRNAs) are non-coding short RNAs that regulate translation. A recent study showed that over 100 miRNAs can be found in spermatozoa. In addition, 28 miRNAs were found to be differentially expressed between smokers and non-smokers, and the expression of four of them (has-miR-146b-5p, has-miR-509-5p, has-miR-146d, and has-miR-652) was altered in infertile men. Moreover, the authors reported that these four miRNAs played a role in pathways involved in cell proliferation, differentiation, and apoptosis in spermatozoa as well as early embryogenesis [159]. Additionally, Yu et al [160] investigated the association between IκBα rs696 polymorphism and defective spermatogenesis in humans. Their study suggested that cigarette smoking-associated ROS may affect spermatogenesis via nuclear factor-κB signaling and involve mechanisms such as interference with inflammatory signaling pathways in the reproductive system. These findings allow us to better understand the genetic mechanisms that impact male infertility.
Although the epigenetic mechanisms by which benzo[a]pyrene and nicotine-induced alterations can be vertically transmitted has not been resolved, smoking-induced abnormal protamination may cause alterations in the methylation pattern of CpG in the promoter regions of DNA in the offspring of smokers [161].

THE EFFECT OF SMOKING CESSATION ON MALE INFERTILITY

Much like the relatively inconclusive effects of smoking on male infertility, definitive information on the effects of smoking cessation on male reproductive parameters is rather scanty as well. In an animal study, Oyeyipo et al [162] showed that exposure to a high dose of nicotine negatively impacted semen concentration, motility, and morphology, as well as the libido of rats, in a dose-dependent manner. Interestingly, an increase in morphologically normal spermatozoa and libido scores were observed in the nicotine recovery group. The authors concluded that nicotine cessation improved fertility in male rats [162].
A recent retrospective cross-sectional study of infertile couples compared 229 never-smokers, 56 past smokers, and 109 current smokers [163]. Lower testosterone levels were observed in both never-smokers and previous smokers than in current smokers. Other researchers have also found decreased testosterone levels in previous smokers [3164]. In a study of the effect of smoking cessation on erectile dysfunction, Harte and Meston [165] examined physiological and subjective sexual arousal indices during and after a smoking cessation program. The authors reported that smoking cessation considerably improved both physiological and self-reported indices of sexual health, irrespective of baseline erectile impairment.
In conclusion, no conclusive data exist on the actual influence of smoking cessation on male infertility in human subjects beyond the finding that smoking cessation is likely to improve erectile dysfunction.

CONCLUSIONS

This review discussed the possible effects of smoking on male fertility. Although no unambiguous and clear-cut conclusions can be drawn, a few inferences can be made. First, smoking has a stronger measurable effect on semen quality and function in fertile men than in subfertile men. This may be attributed to impaired semen parameters and spermatozoa function at the primary level in the infertile population. Critically evaluating the current data on the effect of smoking on semen parameters did not yield an unambiguous result, but fertility can be affected even in the presence of normal semen parameters. Secondly, OS and the resulting genetic and epigenetic changes that result from smoking may correlate directly with reduced sperm function and reduced fertility, thereby providing a possible mechanism for the effect of smoking on male fertility. This correlation should be further investigated. Third, dose-dependent correlations exist between smoking and semen quality and sperm function. Smoking cessation should certainly be advised to any male smoker, especially if he is trying to conceive with his partner. Healthcare providers should facilitate smoking cessation by education, monitoring, and constant support. The data on smoking and male fertility reinforce the preferred preventive approach of discouraging smoking and eliminating exposure to tobacco smoke among both males and females in general, and in particular, while trying to conceive.

ACKNOWLEDGEMENTS

This study was supported by funds from the American Center for Reproductive Medicine at Cleveland Clinic, Cleveland, OH, USA.

Notes

CONFLICT OF INTEREST: No potential conflict of interest relevant to this article was reported.

References

1. Zhang ZH, Zhu HB, Li LL, Yu Y, Zhang HG, Liu RZ. Decline of semen quality and increase of leukocytes with cigarette smoking in infertile men. Iran J Reprod Med. 2013; 11:589–596. PMID: 24639795.
2. Saleh RA, Agarwal A, Sharma RK, Nelson DR, Thomas AJ Jr. Effect of cigarette smoking on levels of seminal oxidative stress in infertile men: a prospective study. Fertil Steril. 2002; 78:491–499. PMID: 12215323.
crossref
3. Trummer H, Habermann H, Haas J, Pummer K. The impact of cigarette smoking on human semen parameters and hormones. Hum Reprod. 2002; 17:1554–1559. PMID: 12042277.
crossref
4. Ng M, Freeman MK, Fleming TD, Robinson M, Dwyer-Lindgren L, Thomson B, et al. Smoking prevalence and cigarette consumption in 187 countries, 1980-2012. JAMA. 2014; 311:183–192. PMID: 24399557.
crossref
5. World Health Organization. Infections, pregnancies, and infertility: perspectives on prevention. Fertil Steril. 1987; 47:964–968. PMID: 3595902.
6. Odisho AY, Nangia AK, Katz PP, Smith JF. Temporal and geospatial trends in male factor infertility with assisted reproductive technology in the United States from 1999-2010. Fertil Steril. 2014; 102:469–475. PMID: 24931206.
crossref
7. Zinaman MJ, Brown CC, Selevan SG, Clegg ED. Semen quality and human fertility: a prospective study with healthy couples. J Androl. 2000; 21:145–153. PMID: 10670528.
8. Lewin A, Gonen O, Orvieto R, Schenker JG. Effect of smoking on concentration, motility and zona-free hamster test on human sperm. Arch Androl. 1991; 27:51–54. PMID: 1772308.
crossref
9. Chia SE, Lim ST, Tay SK, Lim ST. Factors associated with male infertility: a case-control study of 218 infertile and 240 fertile men. BJOG. 2000; 107:55–61. PMID: 10645862.
crossref
10. Künzle R, Mueller MD, Hänggi W, Birkhäuser MH, Drescher H, Bersinger NA. Semen quality of male smokers and nonsmokers in infertile couples. Fertil Steril. 2003; 79:287–291. PMID: 12568836.
crossref
11. Adelusi B, al-Twaijiri MH, al-Meshari A, Kangave D, al-Nuaim LA, Younnus B. Correlation of smoking and coffee drinking with sperm progressive motility in infertile males. Afr J Med Med Sci. 1998; 27:47–50. PMID: 10456129.
12. Sergerie M, Ouhilal S, Bissonnette F, Brodeur J, Bleau G. Lack of association between smoking and DNA fragmentation in the spermatozoa of normal men. Hum Reprod. 2000; 15:1314–1321. PMID: 10831562.
crossref
13. Hoidas S, Williams AE, Tocher JL, Hargreave TB. Scoring sperm morphology from fertile and infertile cigarette smokers using the scanning electron microscope and image analysis. Fertil Steril. 1985; 43:595–598. PMID: 3987928.
crossref
14. Vogt HJ, Heller WD, Borelli S. Sperm quality of healthy smokers, ex-smokers, and never-smokers. Fertil Steril. 1986; 45:106–110. PMID: 3943641.
crossref
15. Brugo-Olmedo S, Chillik C, Kopelman S. Definition and causes of infertility. Reprod Biomed Online. 2001; 2:41–53. PMID: 12537824.
crossref
16. Marinelli D, Gaspari L, Pedotti P, Taioli E. Mini-review of studies on the effect of smoking and drinking habits on semen parameters. Int J Hyg Environ Health. 2004; 207:185–192. PMID: 15330386.
crossref
17. Practice Committee of American Society for Reproductive Medicine. Smoking and infertility. Fertil Steril. 2008; 90:S254–S259. PMID: 19007641.
18. Hammond D, Fong GT, Cummings KM, O'Connor RJ, Giovino GA, McNeill A. Cigarette yields and human exposure: a comparison of alternative testing regimens. Cancer Epidemiol Biomarkers Prev. 2006; 15:1495–1501. PMID: 16896039.
crossref
19. Colagar AH, Jorsaraee GA, Marzony ET. Cigarette smoking and the risk of male infertility. Pak J Biol Sci. 2007; 10:3870–3874. PMID: 19090244.
20. Richthoff J, Elzanaty S, Rylander L, Hagmar L, Giwercman A. Association between tobacco exposure and reproductive parameters in adolescent males. Int J Androl. 2008; 31:31–39. PMID: 17376217.
crossref
21. Zhu AZ, Zhou Q, Cox LS, Ahluwalia JS, Benowitz NL, Tyndale RF. Variation in trans-3'-hydroxycotinine glucuronidation does not alter the nicotine metabolite ratio or nicotine intake. PLoS One. 2013; 8:e70938. PMID: 23936477.
crossref
22. Pacifici R, Altieri I, Gandini L, Lenzi A, Pichini S, Rosa M, et al. Nicotine, cotinine, and trans-3-hydroxycotinine levels in seminal plasma of smokers: effects on sperm parameters. Ther Drug Monit. 1993; 15:358–363. PMID: 8249041.
23. Oliveira H, Spanò M, Santos C, Pereira Mde L. Adverse effects of cadmium exposure on mouse sperm. Reprod Toxicol. 2009; 28:550–555. PMID: 19695322.
crossref
24. Oldereid NB, Thomassen Y, Purvis K. Seminal plasma lead, cadmium and zinc in relation to tobacco consumption. Int J Androl. 1994; 17:24–28. PMID: 8005705.
crossref
25. Chia SE, Xu B, Ong CN, Tsakok FM, Lee ST. Effect of cadmium and cigarette smoking on human semen quality. Int J Fertil Menopausal Stud. 1994; 39:292–298. PMID: 7820163.
26. Pant N, Kumar G, Upadhyay AD, Gupta YK, Chaturvedi PK. Correlation between lead and cadmium concentration and semen quality. Andrologia. 2015; 47:887–891. PMID: 25228328.
crossref
27. Stillman RJ, Rosenberg MJ, Sachs BP. Smoking and reproduction. Fertil Steril. 1986; 46:545–566. PMID: 3530822.
crossref
28. Sofikitis N, Miyagawa I, Dimitriadis D, Zavos P, Sikka S, Hellstrom W. Effects of smoking on testicular function, semen quality and sperm fertilizing capacity. J Urol. 1995; 154:1030–1034. PMID: 7637048.
crossref
29. Vine MF, Tse CK, Hu P, Truong KY. Cigarette smoking and semen quality. Fertil Steril. 1996; 65:835–842. PMID: 8654648.
crossref
30. Merino G, Lira SC, Martínez-Chéquer JC. Effects of cigarette smoking on semen characteristics of a population in Mexico. Arch Androl. 1998; 41:11–15. PMID: 9642454.
crossref
31. Meeker JD, Benedict MD. Infertility, pregnancy loss and adverse birth outcomes in relation to maternal secondhand tobacco smoke exposure. Curr Womens Health Rev. 2013; 9:41–49. PMID: 23888128.
crossref
32. Pirkle JL, Bernert JT, Caudill SP, Sosnoff CS, Pechacek TF. Trends in the exposure of nonsmokers in the U.S. population to secondhand smoke: 1988-2002. Environ Health Perspect. 2006; 114:853–858. PMID: 16759984.
crossref
33. Polyzos A, Schmid TE, Piña-Guzmán B, Quintanilla-Vega B, Marchetti F. Differential sensitivity of male germ cells to mainstream and sidestream tobacco smoke in the mouse. Toxicol Appl Pharmacol. 2009; 237:298–305. PMID: 19345701.
crossref
34. Office on Smoking and Health (US). The health consequences of involuntary exposure to tobacco smoke: a report of the surgeon general. Atlanta (GA): Centers for Disease Control and Prevention (US);2006.
35. Tremellen K. Oxidative stress and male infertility: a clinical perspective. Hum Reprod Update. 2008; 14:243–258. PMID: 18281241.
crossref
36. Kao SH, Chao HT, Chen HW, Hwang TI, Liao TL, Wei YH. Increase of oxidative stress in human sperm with lower motility. Fertil Steril. 2008; 89:1183–1190. PMID: 17669405.
crossref
37. Xu LC, Wang SY, Yang XF, Wang XR. Effects of cadmium on rat sperm motility evaluated with computer assisted sperm analysis. Biomed Environ Sci. 2001; 14:312–317. PMID: 11862611.
38. Kumosani TA, Elshal MF, Al-Jonaid AA, Abduljabar HS. The influence of smoking on semen quality, seminal microelements and Ca2+-ATPase activity among infertile and fertile men. Clin Biochem. 2008; 41:1199–1203. PMID: 18708043.
crossref
39. Besaratinia A, Tommasi S. Electronic cigarettes: the road ahead. Prev Med. 2014; 66:65–67. PMID: 24952095.
crossref
40. Goniewicz ML, Knysak J, Gawron M, Kosmider L, Sobczak A, Kurek J, et al. Levels of selected carcinogens and toxicants in vapour from electronic cigarettes. Tob Control. 2014; 23:133–139. PMID: 23467656.
crossref
41. Kim HJ, Shin HS. Determination of tobacco-specific nitrosamines in replacement liquids of electronic cigarettes by liquid chromatography-tandem mass spectrometry. J Chromatogr A. 2013; 1291:48–55. PMID: 23602640.
crossref
42. Harrell PT, Simmons VN, Correa JB, Padhya TA, Brandon TH. Electronic nicotine delivery systems ("e-cigarettes"): review of safety and smoking cessation efficacy. Otolaryngol Head Neck Surg. 2014; 151:381–393. PMID: 24898072.
43. Burstyn I. Peering through the mist: systematic review of what the chemistry of contaminants in electronic cigarettes tells us about health risks. BMC Public Health. 2014; 14:18. PMID: 24406205.
crossref
44. Nair U, Bartsch H, Nair J. Alert for an epidemic of oral cancer due to use of the betel quid substitutes gutkha and pan masala: a review of agents and causative mechanisms. Mutagenesis. 2004; 19:251–262. PMID: 15215323.
crossref
45. Burkey MD, Feirman S, Wang H, Choudhury SR, Grover S, Johnston FM. The association between smokeless tobacco use and pancreatic adenocarcinoma: a systematic review. Cancer Epidemiol. 2014; 38:647–653. PMID: 25262376.
crossref
46. Connolly GN, Alpert HR. Trends in the use of cigarettes and other tobacco products, 2000-2007. JAMA. 2008; 299:2629–2630. PMID: 18544722.
crossref
47. Bates C, Fagerström K, Jarvis MJ, Kunze M, McNeill A, Ramström L. European Union policy on smokeless tobacco: a statement in favour of evidence based regulation for public health. Tob Control. 2003; 12:360–367. PMID: 14660767.
crossref
48. Benowitz NL, Porchet H, Sheiner L, Jacob P 3rd. Nicotine absorption and cardiovascular effects with smokeless tobacco use: comparison with cigarettes and nicotine gum. Clin Pharmacol Ther. 1988; 44:23–28. PMID: 3391001.
crossref
49. Thomas G, Hashibe M, Jacob BJ, Ramadas K, Mathew B, Sankaranarayanan R, et al. Risk factors for multiple oral premalignant lesions. Int J Cancer. 2003; 107:285–291. PMID: 12949809.
crossref
50. Amagasa T, Yamashiro M, Uzawa N. Oral premalignant lesions: from a clinical perspective. Int J Clin Oncol. 2011; 16:5–14. PMID: 21225307.
crossref
51. Rodu B, Cole P. Smokeless tobacco use and cancer of the upper respiratory tract. Oral Surg Oral Med Oral Pathol Oral Radiol Endod. 2002; 93:511–515. PMID: 12075196.
crossref
52. Said TM, Ranga G, Agarwal A. Relationship between semen quality and tobacco chewing in men undergoing infertility evaluation. Fertil Steril. 2005; 84:649–653. PMID: 16169398.
crossref
53. Sunanda P, Panda B, Dash C, Ray PK, Padhy RN, Routray P. Prevalence of abnormal spermatozoa in tobacco chewing sub-fertile males. J Hum Reprod Sci. 2014; 7:136–142. PMID: 25191028.
crossref
54. Stead LF, Perera R, Bullen C, Mant D, Hartmann-Boyce J, Cahill K, et al. Nicotine replacement therapy for smoking cessation. Cochrane Database Syst Rev. 2012; 11:CD000146. PMID: 23152200.
crossref
55. Joseph AM, Fu SS. Safety issues in pharmacotherapy for smoking in patients with cardiovascular disease. Prog Cardiovasc Dis. 2003; 45:429–441. PMID: 12800126.
crossref
56. Greenland S, Satterfield MH, Lanes SF. A meta-analysis to assess the incidence of adverse effects associated with the transdermal nicotine patch. Drug Saf. 1998; 18:297–308. PMID: 9565740.
crossref
57. Cooper S, Taggar J, Lewis S, Marlow N, Dickinson A, Whitemore R, et al. Effect of nicotine patches in pregnancy on infant and maternal outcomes at 2 years: follow-up from the randomised, double-blind, placebo-controlled SNAP trial. Lancet Respir Med. 2014; 2:728–737. PMID: 25127405.
crossref
58. Osser S, Beckman-Ramirez A, Liedholm P. Semen quality of smoking and non-smoking men in infertile couples in a Swedish population. Acta Obstet Gynecol Scand. 1992; 71:215–218. PMID: 1317645.
crossref
59. Chia SE, Tay SK, Lim ST. What constitutes a normal seminal analysis? Semen parameters of 243 fertile men. Hum Reprod. 1998; 13:3394–3398. PMID: 9886521.
crossref
60. Dikshit RK, Buch JG, Mansuri SM. Effect of tobacco consumption on semen quality of a population of hypofertile males. Fertil Steril. 1987; 48:334–336. PMID: 3609347.
61. Dunphy BC, Barratt CL, von Tongelen BP, Cooke ID. Male cigarette smoking and fecundity in couples attending an infertility clinic. Andrologia. 1991; 23:223–225. PMID: 1741486.
crossref
62. Gerhard I, Lenhard K, Eggert-Kruse W, Runnebaum B. Clinical data which influence semen parameters in infertile men. Hum Reprod. 1992; 7:830–837. PMID: 1323572.
crossref
63. Hassa H, Yildirim A, Can C, Turgut M, Tanir HM, Senses T, et al. Effect of smoking on semen parameters of men attending an infertility clinic. Clin Exp Obstet Gynecol. 2006; 33:19–22. PMID: 16761532.
64. Jeng HA, Chen YL, Kantaria KN. Association of cigarette smoking with reproductive hormone levels and semen quality in healthy adult men in Taiwan. J Environ Sci Health A Tox Hazard Subst Environ Eng. 2014; 49:262–268. PMID: 24279617.
crossref
65. Liu RZ, Gao JC, Zhang HG, Wang RX, Zhang ZH, Liu XY. Seminal plasma zinc level may be associated with the effect of cigarette smoking on sperm parameters. J Int Med Res. 2010; 38:923–928. PMID: 20819428.
crossref
66. Mak V, Jarvi K, Buckspan M, Freeman M, Hechter S, Zini A. Smoking is associated with the retention of cytoplasm by human spermatozoa. Urology. 2000; 56:463–466. PMID: 10962316.
crossref
67. Ochedalski T, Lachowicz-Ochedalska A, Dec W, Czechowski B. Evaluating the effect of smoking tobacco on some semen parameters in men of reproductive age. Ginekol Pol. 1994; 65:80–86. PMID: 8070715.
68. Oldereid NB, Rui H, Clausen OP, Purvis K. Cigarette smoking and human sperm quality assessed by laser-Doppler spectroscopy and DNA flow cytometry. J Reprod Fertil. 1989; 86:731–736. PMID: 2760898.
crossref
69. Sepaniak S, Forges T, Gerard H, Foliguet B, Bene MC, Monnier-Barbarino P. The influence of cigarette smoking on human sperm quality and DNA fragmentation. Toxicology. 2006; 223:54–60. PMID: 16621218.
crossref
70. Zavos PM, Correa JR, Antypas S, Zarmakoupis-Zavos PN, Zarmakoupis CN. Effects of seminal plasma from cigarette smokers on sperm viability and longevity. Fertil Steril. 1998; 69:425–429. PMID: 9531871.
71. Zhang JP, Meng QY, Wang Q, Zhang LJ, Mao YL, Sun ZX. Effect of smoking on semen quality of infertile men in Shandong, China. Asian J Androl. 2000; 2:143–146. PMID: 11232793.
72. Kiziler AR, Aydemir B, Onaran I, Alici B, Ozkara H, Gulyasar T, et al. High levels of cadmium and lead in seminal fluid and blood of smoking men are associated with high oxidative stress and damage in infertile subjects. Biol Trace Elem Res. 2007; 120:82–91. PMID: 17916958.
crossref
73. Shaarawy M, Mahmoud KZ. Endocrine profile and semen characteristics in male smokers. Fertil Steril. 1982; 38:255–257. PMID: 6809501.
crossref
74. Shen HM, Chia SE, Ni ZY, New AL, Lee BL, Ong CN. Detection of oxidative DNA damage in human sperm and the association with cigarette smoking. Reprod Toxicol. 1997; 11:675–680. PMID: 9311575.
crossref
75. Yu B, Qi Y, Liu D, Gao X, Chen H, Bai C, et al. Cigarette smoking is associated with abnormal histone-to-protamine transition in human sperm. Fertil Steril. 2014; 101:51–57. PMID: 24112532.
crossref
76. Taha EA, Ez-Aldin AM, Sayed SK, Ghandour NM, Mostafa T. Effect of smoking on sperm vitality, DNA integrity, seminal oxidative stress, zinc in fertile men. Urology. 2012; 80:822–825. PMID: 23021663.
crossref
77. Martini AC, Molina RI, Estofán D, Senestrari D, Fiol de Cuneo M, Ruiz RD. Effects of alcohol and cigarette consumption on human seminal quality. Fertil Steril. 2004; 82:374–377. PMID: 15302286.
crossref
78. Meri ZB, Irshid IB, Migdadi M, Irshid AB, Mhanna SA. Does cigarette smoking affect seminal fluid parameters? A comparative study. Oman Med J. 2013; 28:12–15. PMID: 23386938.
crossref
79. Pasqualotto FF, Sobreiro BP, Hallak J, Pasqualotto EB, Lucon AM. Cigarette smoking is related to a decrease in semen volume in a population of fertile men. BJU Int. 2006; 97:324–326. PMID: 16430638.
crossref
80. Ramlau-Hansen CH, Thulstrup AM, Aggerholm AS, Jensen MS, Toft G, Bonde JP. Is smoking a risk factor for decreased semen quality? A cross-sectional analysis. Hum Reprod. 2007; 22:188–196. PMID: 16966350.
crossref
81. Vine MF, Margolin BH, Morrison HI, Hulka BS. Cigarette smoking and sperm density: a meta-analysis. Fertil Steril. 1994; 61:35–43. PMID: 8293842.
82. Practice Committee of the American Society for Reproductive Medicine. Smoking and infertility: a committee opinion. Fertil Steril. 2012; 98:1400–1406. PMID: 22959451.
83. Vine MF. Smoking and male reproduction: a review. Int J Androl. 1996; 19:323–337. PMID: 9051418.
crossref
84. Gaur DS, Talekar M, Pathak VP. Effect of cigarette smoking on semen quality of infertile men. Singapore Med J. 2007; 48:119–123. PMID: 17304390.
85. Hassan MA, Killick SR. Negative lifestyle is associated with a significant reduction in fecundity. Fertil Steril. 2004; 81:384–392. PMID: 14967378.
crossref
86. Benowitz NL, Jacob P 3rd, Kozlowski LT, Yu L. Influence of smoking fewer cigarettes on exposure to tar, nicotine, and carbon monoxide. N Engl J Med. 1986; 315:1310–1313. PMID: 3773954.
crossref
87. Zavos PM, Correa JR, Karagounis CS, Ahparaki A, Phoroglou C, Hicks CL, et al. An electron microscope study of the axonemal ultrastructure in human spermatozoa from male smokers and nonsmokers. Fertil Steril. 1998; 69:430–434. PMID: 9531872.
88. Yeung CH, Tüttelmann F, Bergmann M, Nordhoff V, Vorona E, Cooper TG. Coiled sperm from infertile patients: characteristics, associated factors and biological implication. Hum Reprod. 2009; 24:1288–1295. PMID: 19221095.
crossref
89. Miyaji K, Kaneko S, Ishikawa H, Aoyagi T, Hayakawa K, Hata M, et al. Creatine kinase isoforms in the seminal plasma and the purified human sperm. Arch Androl. 2001; 46:127–134. PMID: 11297066.
crossref
90. Ghaffari MA, Rostami M. The effect of cigarette smoking on human sperm creatine kinase activity: as an ATP buffering system in sperm. Int J Fertil Steril. 2013; 6:258–265. PMID: 24520449.
91. Gerhard I, Fröhlich E, Eggert-Kruse W, Klinga K, Runnebaum B. Relationship of sperm acrosin activity to semen and clinical parameters in infertile patients. Andrologia. 1989; 21:146–154. PMID: 2712369.
92. El Mulla KF, Köhn FM, El Beheiry AH, Schill WB. The effect of smoking and varicocele on human sperm acrosin activity and acrosome reaction. Hum Reprod. 1995; 10:3190–3194. PMID: 8822441.
93. Sofikitis N, Takenaka M, Kanakas N, Papadopoulos H, Yamamoto Y, Drakakis P, et al. Effects of cotinine on sperm motility, membrane function, and fertilizing capacity in vitro. Urol Res. 2000; 28:370–375. PMID: 11221915.
crossref
94. Close CE, Roberts PL, Berger RE. Cigarettes, alcohol and marijuana are related to pyospermia in infertile men. J Urol. 1990; 144:900–903. PMID: 2398564.
crossref
95. La Maestra S, De Flora S, Micale RT. Effect of cigarette smoke on DNA damage, oxidative stress, and morphological alterations in mouse testis and spermatozoa. Int J Hyg Environ Health. 2015; 218:117–122. PMID: 25260855.
crossref
96. Sharma RK, Pasqualotto FF, Nelson DR, Thomas AJ Jr, Agarwal A. The reactive oxygen species-total antioxidant capacity score is a new measure of oxidative stress to predict male infertility. Hum Reprod. 1999; 14:2801–2807. PMID: 10548626.
crossref
97. Alvarez JG, Storey BT. Differential incorporation of fatty acids into and peroxidative loss of fatty acids from phospholipids of human spermatozoa. Mol Reprod Dev. 1995; 42:334–346. PMID: 8579848.
crossref
98. de Lamirande E, Gagnon C. Impact of reactive oxygen species on spermatozoa: a balancing act between beneficial and detrimental effects. Hum Reprod. 1995; 10(Suppl 1):15–21. PMID: 8592032.
crossref
99. Murphy MP, Holmgren A, Larsson NG, Halliwell B, Chang CJ, Kalyanaraman B, et al. Unraveling the biological roles of reactive oxygen species. Cell Metab. 2011; 13:361–366. PMID: 21459321.
crossref
100. Logan-Garbisch T, Bortolazzo A, Luu P, Ford A, Do D, Khodabakhshi P, et al. Developmental ethanol exposure leads to dysregulation of lipid metabolism and oxidative stress in Drosophila. G3 (Bethesda). 2014; 5:49–59. PMID: 25387828.
crossref
101. Cho DY, Le W, Bravo DT, Hwang PH, Illek B, Fischer H, et al. Air pollutants cause release of hydrogen peroxide and interleukin-8 in a human primary nasal tissue culture model. Int Forum Allergy Rhinol. 2014; 4:966–971. PMID: 25400124.
crossref
102. Agarwal A, Prabakaran SA. Mechanism, measurement, and prevention of oxidative stress in male reproductive physiology. Indian J Exp Biol. 2005; 43:963–974. PMID: 16315393.
103. Griveau JF, Le Lannou D. Reactive oxygen species and human spermatozoa: physiology and pathology. Int J Androl. 1997; 20:61–69. PMID: 9292315.
crossref
104. Shen HM, Chia SE, Ong CN. Evaluation of oxidative DNA damage in human sperm and its association with male infertility. J Androl. 1999; 20:718–723. PMID: 10591610.
105. Kim JG, Parthasarathy S. Oxidation and the spermatozoa. Semin Reprod Endocrinol. 1998; 16:235–239. PMID: 10101805.
crossref
106. 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; 5:393–398. PMID: 10582779.
crossref
107. Aitken RJ, Buckingham DW, Brindle J, Gomez E, Baker HW, Irvine DS. Analysis of sperm movement in relation to the oxidative stress created by leukocytes in washed sperm preparations and seminal plasma. Hum Reprod. 1995; 10:2061–2071. PMID: 8567843.
108. Stone KK, Bermúdez E, Pryor WA. Aqueous extracts of cigarette tar containing the tar free radical cause DNA nicks in mammalian cells. Environ Health Perspect. 1994; 102(Suppl 10):173–178. PMID: 7705294.
crossref
109. Iwasaki A, Gagnon C. Formation of reactive oxygen species in spermatozoa of infertile patients. Fertil Steril. 1992; 57:409–416. PMID: 1735495.
110. Alvarez JG, Storey BT. Role of glutathione peroxidase in protecting mammalian spermatozoa from loss of motility caused by spontaneous lipid peroxidation. Gamete Res. 1989; 23:77–90. PMID: 2545584.
crossref
111. Dawson EB, Harris WA, Teter MC, Powell LC. Effect of ascorbic acid supplementation on the sperm quality of smokers. Fertil Steril. 1992; 58:1034–1039. PMID: 1426355.
112. Smith JL, Hodges RE. Serum levels of vitamin C in relation to dietary and supplemental intake of vitamin C in smokers and nonsmokers. Ann N Y Acad Sci. 1987; 498:144–152. PMID: 3475996.
crossref
113. Dacheux JL, Dacheux F. New insights into epididymal function in relation to sperm maturation. Reproduction. 2013; 147:R27–R42. PMID: 24218627.
crossref
114. Bornman MS, Kok EL, du Plessis DJ, Otto BS. Clinical features of patients with detached ciliary tufts in semen. Andrologia. 1989; 21:18–22. PMID: 2712358.
115. Jarow JP. Effects of varicocele on male fertility. Hum Reprod Update. 2001; 7:59–64. PMID: 11212076.
crossref
116. Masson P, Brannigan RE. The varicocele. Urol Clin North Am. 2014; 41:129–144. PMID: 24286772.
crossref
117. Klaiber EL, Broverman DM, Pokoly TB, Albert AJ, Howard PJ Jr, Sherer JF Jr. Interrelationships of cigarette smoking, testicular varicoceles, and seminal fluid indexes. Fertil Steril. 1987; 47:481–486. PMID: 3556626.
118. Hiremagalur B, Sabban EL. Nicotine elicits changes in expression of adrenal catecholamine biosynthetic enzymes, neuropeptide Y and immediate early genes by injection but not continuous administration. Brain Res Mol Brain Res. 1995; 32:109–115. PMID: 7494448.
crossref
119. Polsky JY, Aronson KJ, Heaton JP, Adams MA. Smoking and other lifestyle factors in relation to erectile dysfunction. BJU Int. 2005; 96:1355–1359. PMID: 16287457.
crossref
120. Kupelian V, Link CL, McKinlay JB. Association between smoking, passive smoking, and erectile dysfunction: results from the Boston Area Community Health (BACH) Survey. Eur Urol. 2007; 52:416–422. PMID: 17383811.
crossref
121. Cao S, Yin X, Wang Y, Zhou H, Song F, Lu Z. Smoking and risk of erectile dysfunction: systematic review of observational studies with meta-analysis. PLoS One. 2013; 8:e60443. PMID: 23573257.
crossref
122. Maiorino MI, Bellastella G, Esposito K. Lifestyle modifications and erectile dysfunction: what can be expected? Asian J Androl. 2015; 17:5–10. PMID: 25248655.
crossref
123. Pourmand G, Alidaee MR, Rasuli S, Maleki A, Mehrsai A. Do cigarette smokers with erectile dysfunction benefit from stopping?: a prospective study. BJU Int. 2004; 94:1310–1313. PMID: 15610111.
crossref
124. O WS, Chen H, Chow PH. Male genital tract antioxidant enzymes--their ability to preserve sperm DNA integrity. Mol Cell Endocrinol. 2006; 250:80–83. PMID: 16442705.
crossref
125. Pakrashi A, Chatterjee S. Effect of tobacco consumption on the function of male accessory sex glands. Int J Androl. 1995; 18:232–236. PMID: 8567092.
crossref
126. Arabi M, Moshtaghi H. Influence of cigarette smoking on spermatozoa via seminal plasma. Andrologia. 2005; 37:119–124. PMID: 16164428.
crossref
127. Mehran A. The toxic effect of seminal plasma from smokers on sperm function in non-smokers. Zhonghua Nan Ke Xue. 2005; 11:647–651. PMID: 16209200.
128. Weisberg E. Smoking and reproductive health. Clin Reprod Fertil. 1985; 3:175–186. PMID: 3910212.
129. Ochedalski T, Lachowicz-Ochedalska A, Dec W, Czechowski B. Examining the effects of tobacco smoking on levels of certain hormones in serum of young men. Ginekol Pol. 1994; 65:87–93. PMID: 8070716.
130. Jurasović J, Cvitković P, Pizent A, Colak B, Telisman S. Semen quality and reproductive endocrine function with regard to blood cadmium in Croatian male subjects. Biometals. 2004; 17:735–743. PMID: 15689116.
crossref
131. Jensen JA, Goodson WH, Hopf HW, Hunt TK. Cigarette smoking decreases tissue oxygen. Arch Surg. 1991; 126:1131–1134. PMID: 1929845.
crossref
132. Perrin J, Tassistro V, Mandon M, Grillo JM, Botta A, Sari-Minodier I. Tobacco consumption and benzo(a)pyrene-diol-epoxide-DNA adducts in spermatozoa: in smokers, swim-up procedure selects spermatozoa with decreased DNA damage. Fertil Steril. 2011; 95:2013–2017. PMID: 21406302.
crossref
133. Phillips DH, Venitt S. DNA and protein adducts in human tissues resulting from exposure to tobacco smoke. Int J Cancer. 2012; 131:2733–2753. PMID: 22961407.
crossref
134. Zenzes MT, Puy LA, Bielecki R, Reed TE. Detection of benzo[a]pyrene diol epoxide-DNA adducts in embryos from smoking couples: evidence for transmission by spermatozoa. Mol Hum Reprod. 1999; 5:125–131. PMID: 10065867.
crossref
135. Begum S. Molecular changes in smoking-related lung cancer. Expert Rev Mol Diagn. 2012; 12:93–106. PMID: 22133122.
crossref
136. Horak S, Polanska J, Widlak P. Bulky DNA adducts in human sperm: relationship with fertility, semen quality, smoking, and environmental factors. Mutat Res. 2003; 537:53–65. PMID: 12742507.
crossref
137. Gaspari L, Chang SS, Santella RM, Garte S, Pedotti P, Taioli E. Polycyclic aromatic hydrocarbon-DNA adducts in human sperm as a marker of DNA damage and infertility. Mutat Res. 2003; 535:155–160. PMID: 12581533.
crossref
138. Gallagher J, Mumford J, Li X, Shank T, Manchester D, Lewtas J. DNA adduct profiles and levels in placenta, blood and lung in relation to cigarette smoking and smoky coal emissions. IARC Sci Publ. 1993; (124):283–292. PMID: 8225497.
139. Trková M, Kapras J, Bobková K, Stanková J, Mejsnarová B. Increased micronuclei frequencies in couples with reproductive failure. Reprod Toxicol. 2000; 14:331–335. PMID: 10908836.
crossref
140. Lähdetie J. Micronucleated spermatids in the seminal fluid of smokers and nonsmokers. Mutat Res. 1986; 172:255–263. PMID: 3785273.
crossref
141. Fenech M. Micronuclei and their association with sperm abnormalities, infertility, pregnancy loss, pre-eclampsia and intra-uterine growth restriction in humans. Mutagenesis. 2011; 26:63–67. PMID: 21164184.
crossref
142. Papachristou F, Simopoulou M, Touloupidis S, Tsalikidis C, Sofikitis N, Lialiaris T. DNA damage and chromosomal aberrations in various types of male factor infertility. Fertil Steril. 2008; 90:1774–1781. PMID: 18082736.
crossref
143. El Ghamrasni S, Cardoso R, Halaby MJ, Zeegers D, Harding S, Kumareswaran R, et al. Cooperation of Blm and Mus81 in development, fertility, genomic integrity and cancer suppression. Oncogene. 2015; 34:1780–1789. PMID: 24858046.
crossref
144. Yarosh SL, Kokhtenko EV, Churnosov MI, Solodilova MA, Polonikov AV. Joint effect of glutathione S-transferase genotypes and cigarette smoking on idiopathic male infertility. Andrologia. 2015; 47:980–986. PMID: 25348056.
crossref
145. Yarosh SL, Kokhtenko EV, Churnosov MI, Ataman AV, Solodilova MA, Polonikov AV. Synergism between the N-acetyltransferase 2 gene and oxidant exposure increases the risk of idiopathic male infertility. Reprod Biomed Online. 2014; 29:362–369. PMID: 24928356.
crossref
146. Yarosh SL, Kokhtenko EV, Starodubova NI, Churnosov MI, Polonikov AV. Smoking status modifies the relation between CYP1A1*2C gene polymorphism and idiopathic male infertility: the importance of gene-environment interaction analysis for genetic studies of the disease. Reprod Sci. 2013; 20:1302–1307. PMID: 23548414.
147. Demirhan O, Demir C, Tunç E, nandıklıoğlu N, Sütcü E, Sadıkoğlu N, et al. The genotoxic effect of nicotine on chromosomes of human fetal cells: the first report described as an important study. Inhal Toxicol. 2011; 23:829–834. PMID: 22035122.
crossref
148. Hassold T, Hunt P. To err (meiotically) is human: the genesis of human aneuploidy. Nat Rev Genet. 2001; 2:280–291. PMID: 11283700.
crossref
149. Pereira CS, Juchniuk de Vozzi MS, Dos Santos SA, Vasconcelos MA, de Paz CC, Squire JA, et al. Smoking-induced chromosomal segregation anomalies identified by FISH analysis of sperm. Mol Cytogenet. 2014; 7:58. PMID: 25264457.
crossref
150. Boissonnas CC, Jouannet P, Jammes H. Epigenetic disorders and male subfertility. Fertil Steril. 2013; 99:624–631. PMID: 23714437.
crossref
151. Kotaja N. MicroRNAs and spermatogenesis. Fertil Steril. 2014; 101:1552–1562. PMID: 24882619.
crossref
152. Shenker NS, Ueland PM, Polidoro S, van Veldhoven K, Ricceri F, Brown R, et al. DNA methylation as a long-term biomarker of exposure to tobacco smoke. Epidemiology. 2013; 24:712–716. PMID: 23867811.
crossref
153. Joubert BR, Håberg SE, Nilsen RM, Wang X, Vollset SE, Murphy SK, et al. 450K epigenome-wide scan identifies differential DNA methylation in newborns related to maternal smoking during pregnancy. Environ Health Perspect. 2012; 120:1425–1431. PMID: 22851337.
crossref
154. Monick MM, Beach SR, Plume J, Sears R, Gerrard M, Brody GH, et al. Coordinated changes in AHRR methylation in lymphoblasts and pulmonary macrophages from smokers. Am J Med Genet B Neuropsychiatr Genet. 2012; 159B:141–151. PMID: 22232023.
crossref
155. Philibert RA, Beach SR, Brody GH. Demethylation of the aryl hydrocarbon receptor repressor as a biomarker for nascent smokers. Epigenetics. 2012; 7:1331–1338. PMID: 23070629.
crossref
156. Besingi W, Johansson A. Smoke-related DNA methylation changes in the etiology of human disease. Hum Mol Genet. 2014; 23:2290–2297. PMID: 24334605.
crossref
157. Fortes MR, Satake N, Corbet DH, Corbet NJ, Burns BM, Moore SS, et al. Sperm protamine deficiency correlates with sperm DNA damage in Bos indicus bulls. Andrology. 2014; 2:370–378. PMID: 24634207.
158. Hammadeh ME, Hamad MF, Montenarh M, Fischer-Hammadeh C. Protamine contents and P1/P2 ratio in human spermatozoa from smokers and non-smokers. Hum Reprod. 2010; 25:2708–2720. PMID: 20823111.
crossref
159. Marczylo EL, Amoako AA, Konje JC, Gant TW, Marczylo TH. Smoking induces differential miRNA expression in human spermatozoa: a potential transgenerational epigenetic concern? Epigenetics. 2012; 7:432–439. PMID: 22441141.
crossref
160. Yu B, Ding Q, Zheng T, Jiang L, Li Q, Sun X, et al. Smoking attenuated the association between IκBα rs696 polymorphism and defective spermatogenesis in humans. Andrologia. 2015; 47:987–994. PMID: 25352423.
161. Aston KI, Punj V, Liu L, Carrell DT. Genome-wide sperm deoxyribonucleic acid methylation is altered in some men with abnormal chromatin packaging or poor in vitro fertilization embryogenesis. Fertil Steril. 2012; 97:285–292. PMID: 22154369.
162. Oyeyipo IP, Raji Y, Emikpe BO, Bolarinwa AF. Effects of nicotine on sperm characteristics and fertility profile in adult male rats: a possible role of cessation. J Reprod Infertil. 2011; 12:201–207. PMID: 23926503.
163. Lotti F, Corona G, Vitale P, Maseroli E, Rossi M, Fino MG, et al. Current smoking is associated with lower seminal vesicles and ejaculate volume, despite higher testosterone levels, in male subjects of infertile couples. Hum Reprod. 2015; 30:590–602. PMID: 25567620.
crossref
164. Camacho EM, Huhtaniemi IT, O'Neill TW, Finn JD, Pye SR, Lee DM, et al. Age-associated changes in hypothalamic-pituitary-testicular function in middle-aged and older men are modified by weight change and lifestyle factors: longitudinal results from the European Male Ageing Study. Eur J Endocrinol. 2013; 168:445–455. PMID: 23425925.
crossref
165. Harte CB, Meston CM. Association between smoking cessation and sexual health in men. BJU Int. 2012; 109:888–896. PMID: 21883852.
crossref
Fig. 1

Types of smoke produced when a cigarette is smoked.

wjmh-33-143-g001
Fig. 2

Percentage of studies reporting a negative effect of smoking on semen parameters.

wjmh-33-143-g002
Fig. 3

Likely mechanisms through which smoking affects sperm parameters and quality, including genetic and epigenetic alternations as well as oxidative stress formation. ROS: reactive oxygen species, 8-OHdG: 8-hydroxyguanosine.

wjmh-33-143-g003
Table 1

Studies reporting a negative effect of smoking on semen parameters

wjmh-33-143-i001
Study No. Author Year Study type No. of participant No. of smoker No. of non-smoker Study group Association between semen parameters and smoking
Motility Concentration Morphology
1 Osser et al [58] 1992 Prospective 350 186 164 Infertile smokers vs. infertile non-smokers No No No
2 Künzle et al [10] 2003 Prospective 1,786 655 1,131 Infertile smokers vs. infertile non-smokers Yes Yes Yes
3 Chia et al [59] 1998 Prospective 243 152 91 Healthy population No No No
4 Dikshit et al [60] 1987 Prospective 626 219 288 Infertile smokers vs. infertile non-smokers No No No
5 Dunphy et al [61] 1991 Prospective 330 135 195 Infertile smokers vs. infertile non-smokers No No No
6 Gerhard et al [62] 1992 Prospective 225 136 89 Infertile smokers vs. infertile non-smokers No No No
7 Hassa et al [63] 2006 Prospective 223 126 97 Infertile smokers vs. infertile non-smokers No No No
8 Vogt et al [14] 1986 Prospective 333 150 183 Healthy population No No No
9 Colagar et al [19] 2007 Prospective 101 53 48 Infertile smokers vs. infertile non-smokers and fertile smokers compared to infertile smokers Yes Yes Yes
10 Jeng et al [64] 2014 Prospective 192 103 89 Healthy population No No Yes
11 Lewin et al [8] 1991 Prospective 675 293 382 Healthy population No Yes NA
12 Liu et al [65] 2010 Prospective 147 68 79 Infertile smokers vs. infertile non-smokers Yes Yes Yes
13 Mak et al [66] 2000 Prospective 87 18 69 Infertile smokers vs. infertile non-smokers vs. fertile non-smokers No No No
14 Merino et al [30] 1998 Prospective 358 197 161 Infertile smokers vs. infertile non-smokers Yes Yes Yes
15 Ochedalski et al [67] 1994 Prospective 70 40 30 Infertile smokers vs. infertile non-smokers Yes Yes No
16 Oldereid et al [68] 1989 Prospective 350 147 203 Infertile smokers vs. infertile non-smokers No No No
17 Richthoff et al [20] 2008 Prospective 302 217 85 Healthy population No No NA
18 Saleh et al [2] 2002 Prospective 65 20 32 Infertile smokers vs. infertile non-smokers No No No
19 Sepaniak et al [69] 2006 Prospective 108 51 57 Infertile smokers vs. infertile non-smokers No No No
20 Sergerie et al [12] 2000 Prospective 97 69 28 Healthy population No No No
21 Trummer et al [3] 2002 Prospective 1,104 478 517 Infertile smokers vs. infertile non-smokers No No No
22 Zavos et al [70] 1998 Prospective 40 20 20 Healthy population Yes Yes Yes
23 Zhang et al [71] 2000 Prospective 362 191 110 Infertile smokers vs. infertile non-smokers vs. fertile non-smokers Yes Yes Yes
24 Kiziler et al [72] 2007 Prospective 71 26 22 Infertile smokers vs. infertile non-smokers vs. fertile non-smokers Yes Yes Yes
25 Shaarawy et al [73] 1982 Prospective 40 20 20 Healthy population No Yes Yes
26 Shen et al [74] 1997 Prospective 60 28 32 Healthy population No No No
27 Yu et al [75] 2014 Prospective 322 147 175 Healthy population No No NA
28 Taha et al [76] 2012 Prospective 160 80 80 Healthy population Yes Yes Yes
29 Martini et al [77] 2004 Retrospective 3,546 372 3,174 Infertile smokers vs. infertile non-smokers No No No
30 Meri et al [78] 2013 Retrospective 960 396 564 Infertile smokers vs. infertile non-smokers Yes No Yes
31 Pasqualotto et al [79] 2006 Retrospective 889 367 522 Healthy population No No No
32 Ramlau-Hansen et al [80] 2007 Retrospective 2,542 1,052 1,490 Healthy population Yes Yes NA
33 Zhang et al [1] 2013 Retrospective 1,512 737 775 Infertile smokers vs. infertile non-smokers Yes No Yes

All studies evaluated semen parameters according to the World Health Organization guidelines at the time the study was conducted.

NA: not applicable.

TOOLS
Similar articles