Skip to main content

Seminal cell-free nucleic acids as possible biomarker in male infertility: a mini-review article

Abstract

Background

Male infertility is a major problem for many couples in the world. Many factors could cause male infertility such as environmental and genetic factors, life style, aging, inflammation, endocrinological etiologies, and antisperm antibodies.

Main body

Circulating cell-free nucleic acids (cfNAs) may play a key role in male infertility. cfNAs are obtained from different body fluids such as blood plasma, cerebrospinal fluid, amniotic fluid, urine, bronchoalveolar lavage fluid, and seminal plasma. The different types of cfNAs present in human semen include cell-free DNAs, cell free RNAs and cell-free mitochondrial DNAs and they are differentially higher than those in other body fluids. Few evidence have been done regarding the direct relationship between cfNAs and male infertility in serum and seminal plasma of infertile men compared to the fertile men.

Conclusions

This document aimed to compile data about the main causes influencing male infertility focusing on seminal cfNA/cfDNA and its possible role as differential biomarker to diagnosis the main source of spermatogenesis abnormalities and male infertility.

1 Introduction

The incapability to conceive after 1 year orderly unprotected sex is called infertility. Infertility has been a concern for couples in reproductive age and also a serious clinical problem today. It affects about 15% of couples who are at the age of procreate worldwide [1]. According to the estimation of WHO, 60–80 million couples in the world suffer from infertility [2]. Females and males are equally responsible for the reason of infertility. 40% of infertility is related to women, 40% to men, and 20% to both sexes [3]. Three major reasons including tubal-peritoneal disease, male factor, and ovulatory dysfunction are the main causes of infertility in couples [4]. The most common causes of infertility in women are lack of ovulation, menstrual cycle problems, impossibility of embryo implantation and infection [5]. Male factors are also factors affecting fertility, explained in the following [6]. Nowadays, male infertility affects many couples in the world, but the cause and molecular mechanism of idiopathic infertility in men are ambiguous. However, with the advancement of new molecular and genetic methods, new realizations have shown that cell-free DNA fragments (Cf-DNA) may be a valuable molecular tool to easily determine the causes of infertility and to choose the best assisted reproductive technology (ART) programs for infertile couples [7]. The existence of cell-free DNA (cfDNA) in human plasma and its relationship with diseases was first presented in 1948 by Mendel and Matais [8]. However, this pivotal discovery had received little attention until Tan and his colleagues discovered an increase in cfDNA in the blood of patients with systemic lupus erythematosus compared to healthy individuals [9]. Today, the relationship between cfDNA and various diseases such as leukemia, rheumatoid arthritis and malignant tumors has also been determined [10]. Increased cfDNA may reflect physiological and non-malignant pathological processes such as inflammation, diabetes, tissue trauma, sepsis and infarction [11]. cfDNA can be obtained from different body fluids, such as blood plasma, cerebrospinal fluid, amniotic fluid, urine, bronchoalveolar lavage fluid, and seminal plasma [12]. The mechanism for the release of cfDNA is not fully understood, but there are two main mechanisms, including apoptosis/ necrosis and release of intact cells into blood and then lysis [13]. Therefore, it can be a non-invasive and/or prognostic biomarker for some cancers and other severe pathologies [14]. The isolated cfDNA is applied to evaluate DNA integrity, loss of heterozygosity, polymorphisms, microsatellite instability, mutations, and DNA methylation [15]. The increase in the amount of cfDNA in different tissues is related to the increase in reactive oxygen species (ROS) and the increase in oxidative stress, as well as inflammation and infection [16]. Given that very few studies have been conducted on the relationship between semen and serum cfDNA with male infertility, this review article examines the recent findings on cfDNA in male infertility and its potential use as a biomarker of male infertility.

2 Method

Studies related to the relationship between cell free DNA and men infertility until the end of 2023 by searching for articles and studies in scientific databases such as ISI Web of Science, Medline/Pubmed, ScienceDirect, Embase, Scopus, Biological Abstract, Chemical Abstract and Google Scholar was collected. To select the desired articles, the titles and abstracts of these articles were reviewed and the eligible articles were selected and summarized. The search was performed with the keywords cfDNA, men infertility, seminal CfDNA, and the relevant articles were collected, classified and summarized after evaluation. The summary of the studies conducted in this field is presented in Table 1.

Table 1 The studies regarding the relationship between serum and seminal cfDNA with male infertility

3 Male infertility

Decreased fertility in men can be due to genetic abnormalities (single or multiple gene mutations, chromosomal aberrations, polymorphisms), acquired and congenital urogenital abnormalities, varicocele, mitochondrial dysfunction, infections of the accessory glands, and immunological factors [17]. When there is no casual factor and there are abnormalities in semen including no detectable spermatozoa (azoospermia), decreased motility (asthenozoospermia), abnormal morphology (teratozoospermia), and decreased number of spermatozoa (oligozoospermia) male infertility is defined as idiopathic case [18]. The occurrence of the last three simultaneous abnormalities is demonstrated as moderate or severe oligoasthenoteratozoospermia. In ART and clinical andrology, male infertility is diagnosed by endocrine and genetic evaluation, clinical examination, semen analysis, and testicular biopsy or puncture [19]. But these tests are mostly unsuccessful in elucidating the critical reason for infertility in all men [20]. Furthermore, these tests are usually costly, intensive (especially endocrinology and genetic tests), and invasive (biopsy). Studies have shown that free sperm mtDNA copy number is correlated with semen parameters and may serve as a new diagnostic marker of semen quality [21]. According to the evidence, the increased levels of seminal cfDNA are associated with the defect of sperm morphology and motility, indicating that cfDNA could be a biomarker of sperm quality [22].

4 Main factors influencing male infertility

4.1 Environmental factors

Various environmental factors influence fertility via epi/genetic pathways. The epigenome is related to the environment and genome and propagates epigenetic tags during generations [23]. For example, occupational exposure to toxic chemical and physical agents is associated with poor semen quality, a reduced count of motile sperm, and an increased risk of male infertility,as well. Exposure to high temperatures, including metallurgical industries, and bakeries, prolonged sitting, and high stress also affect fertility. In addition, the workload, physical and sexual psychological symptoms are associated with early andropause [24]. Other factors are radiation factors via laptops, tight-fitting underwear, mobile phones, and endocrine-disrupting chemicals such as phthalates, bisphenol A, pesticide residue, and dioxins. The relation between mobile phone exposure and decreased sperm motility and viability is also shown [25].

4.2 Lifestyle

Lifestyle includes all behavioral factors influencing diet, health, exercise, and the use of tobacco and alcohol. Obesity induced by diet affects fertility in men via changing sexual behavior, sleep, semen parameters, hormonal profiles, and scrotal temperature [26]. The risk of azoospermia is also higher in overweight and underweight men than in those with normal weight. The decreased level of sex-hormone-binding globulin is seen in obese males, leading to hyperinsulinemia and an enhanced total level of estradiol. In addition, weight loss is associated with decreased cellular DNA damage, improved semen morphology, and enhanced total motile sperm count [27]. The change in lifestyle, especially the quality of food is useful to treat poor semen quality. Among the factors related to lifestyle, two factors of high consumption of caffeine and exogenous androgen are also mentioned. However, there exists no definitive evidence to suggest that caffeine has any impact on one’s ability to conceive [28]. Studies provide insufficient evidence that there is no increased risk of infertility associated with low, moderate, or high caffeine consumption. Nevertheless, it is imperative to approach this conclusion with caution. It has been shown that exogenous testosterone impedes spermatogenesis by eliminating the feedback response to low testosterone at the hypothalamus and pituitary. This leads to a decrease in the synthesis and secretion of gonadotropins, which are vital for promoting endogenous testosterone production and facilitating spermatogenesis [29].

4.3 Aging

Aging progressively impairs the function of cells and facilitates vulnerability to illness. Aging is associated with reproductive endocrine disorders which cause late-onset hypogonadism in men [30]. But the molecular mechanism affecting semen quality and routine tests are poorly known. Andropause (age-related hypogonadism) is associated with the risk of complications and spontaneous abortions in infancy, including autism, genetic diseases, schizophrenia, lower birth weight, and male infertility [31]. In addition, andropause also suppresses DNA repair machinery and the antioxidant defense system, enhancing the production of ROS, and causing genomic instability [32]. However, the effect of andropause on sperm DNA damage remains controversial. Aging is also associated with various cumulative cellular and molecular events such as sperm telomere shortening, and DNA damage leading to apoptosis [33].

4.4 Inflammation

Inflammation is a process in which the human body reacts to traumatic, chemical, and infectious insults, leading to an influx of activated leukocytes, and different supporting cells and extracellular proteins [34]. Although chronic inflammation usually progresses after an acute symptomatic insult, it may happen in tissues without a history of injury or insult. In fact, most men with genitourinary tract inflammation have no inflammation symptoms. It is the latter insidious process in the male reproductive tract that has caused concern [35]. The assessment of testicular tissue specimens from asymptomatic infertile men shows leukocytic infiltration in more than 50% of men. Various inflammatory factors including the type of pathogen and the chronic and acute nature of the disease can affect male fertility. In addition, noninfectious inflammatory reactions may influence the reproductive system in men [36]. Pro-inflammatory cytokines, including interleukin (IL)-1a, and IL-1b, as well as tumor necrosis factor-alpha are the most important inflammatory response in the reproductive systems of men. The IL-6, IL-8, and IL-10 cytokines are released to inflammation status and are found in the semen of men with diverse seminal defects, indicating semen cytokines may be used to detect inflammation in the reproductive tract of men [37].

4.5 Genetic factor

Infertility in men is a multifactorial pathological situation in which genetic factors are involved. The genetic perspective of infertility in men is highly complex and at least 2000 genes involve in spermatogenesis [38]. Genetic screening is relevant for diagnosis, decision-making, and genetic counseling. Autosome-linked gene mutations play a main role in monomorphic teratozoospermia, central hypogonadism, asthenozoospermia, and familial cases of spermatogenic disturbances. In addition, the study of the whole genome proposes a marginal role for routine variants as causative factors; but some of these variants are important for pharmacogenetic purposes [39]. The most genetic factor which affects male infertility is related to azoospermia (25%) [38]. Researchers reported azoospermic individuals with microscopic deletions of distal euchromatic part of long arm of the Y-chromosome and on the basis of these findings in azoospermic men, they proposed existence of a spermatogenesis gene complex called “azoospermia factor” (AZF) on Yq [40]. According to global estimate about 10% cases of idiopathic azoospermia and oligozoospermia occur due to be deletions in AZF region. Therefore AZF deletions are among most common causes of spermatogenic failure in man identifiable by molecular genetics tool [41]. However, the genetic abnormalities which are recognized in other patients with different sperm abnormalities are increasing, indicating the important role of genetic factors in male infertility.

4.6 Endocrinological aspects

The pituitary failure for secreting follicle-stimulating hormone (FSH) and luteinizing hormone (LH) leads to disrupt fertility, and testicular function. But gonadotropin deficiency contains less than 0.5% of the causative factors in male infertility [42]. The elevated level of LH and low level of testosterone is seen in approximately 30% of men with severe degrees of testicular damage. The measurement of prolactin is mainly associated with impotency compared to infertility. Taken together, sexual hormone therapy, such as FSH, LH, and prolactin seems to be a beneficial management of infertility in men [43].

4.7 Antisperm antibodies

In addition to the customary analysis of semen, there exists the possibility of conducting other extended examinations, one of which involves the identification of antisperm antibodies (ASA). Male immune infertility is characterized as infertility that is brought about by ASA. The production of ASA is because of an unusual exposure of germ cells to the immune system. Also, ASA is present in 70 to 100% men after vasectomy and its association with post-vasectomy obstructive azoospermia is obviously declared [44]. Exogenous antigens, including bacteria, viruses, fungi, and allergens can create ASA during cross-immune reactions [45]. For example, the probability of ASA in patients with chronic prostatitis is three times higher than in patients without this disease. Its mechanism is not known; however, it may be associated with inflammatory damage to genital glands in men, and local immune dysregulation. The laboratory manual from the World Health Organization (WHO) states that “the mere existence of sperm antibodies is inadequate for diagnosing sperm autoimmunity and so, it is necessary to establish that the antibodies severely disrupt sperm function” [46]. They can be detected in 16% of infertile men and approximately 2% of fertile men. ASA can lead to the sperm cells clumping even though it may occur due to other factors, such as the presence of E.coli in the semen [47, 48]. However, the extent to which ASA alone affects fertility outcomes (both natural and assisted) remains to be clearly elucidated.

5 Cell-free nucleic acid

Circulating cell-free nucleic acids (cfNAs) have a main role in the physiology of human and male infertility. Mandel et al. in 1948 recognized cfNAs, however in early 1990s, its importance was identified as a candidate biomarker [49]. cfNAs are divided into cell-free DNA (cfDNA), and cell-free RNA, which contains messenger RNAs (mRNAs) and small non-coding RNAs (microRNAs, and piwi-interacting RNAs, as well as small interfering RNAs (siRNAs) [50].

5.1 Cell-free DNA

DNA circulating in the blood plasma or serum is called cell-free DNA (cfDNA). It is a double-stranded DNA with lower molecular weight compared to genomic DNA. cfDNA presents in healthy individuals with low concentration (< 50 ng/ml) which is discovered in the 1950s. The level of cfDNA in semen is significantly higher than other body fluids. it was found that cfDNA can be used as a biomarker of sperm quality, and promising diagnostic or prognostic biomarker [51, 52]. Seminal cfDNA contains information such as epigenetic modification of the male genital tract. Few studies have been done regarding the relationship between cfDNA and male infertility summarized in Table 1.

5.2 Cell-free RNA

The investigation has revealed that mRNA and miRNA profiles in testis samples are highly expressed in individuals with non-obstructive azoospermia rather than men with obstructive azoospermia or normozoospermia [53]. Similarly, cell-free seminal mRNAs have been investigated as novel noninvasive biomarker for the diagnosis of male infertility. According to these findings, cfs-mRNA DDX4 may be applied to assess the type of azoospermia [54, 55]. In addition, miRNAs play a main role in cell differentiation, and metabolism as well as apoptosis. They are non-coding and small RNAs modulating the expression of genes, post-transcriptionally via targeting particular mRNAs. Also, miRNAs can mediate the gene expression involved in idiopathic male infertility [56]. Piwi-interacting RNAs (piRNAs) are another cell-free RNAs that have been expressed in germ cells, particularly pachytene spermatocytes and spermatids in human testes and seminal plasma. piRNAs, including piR-31925, piR-31068, piR-43771, piR-43773, and piR-30198 can distinguish fertile patients from infertile men, indicating these are specific non-invasive biomarkers of male infertility [57].

5.3 Cell-free mitochondrial DNA

Mitochondria are the major source of ATP in the electron transport chain in sperm. ATP needs for sperm hyperactivation and motility, proposing that mitochondria is key factor for sperm fertilizing capacity and flagellar movement. Additionally, mitochondria regulate apoptosis through releasing some apoptosis-inducing factors and cytochrome C. Since mitochondria play an important role in sperm function and spermatogenesis, very few reports have been done regarding seminal cell-free mtDNA (CFMD) [58]. Chen et al. [59]., revealed a decreased copy number of CFMD in patients with oligoasthenozoospermia and asthenozoospermia. There is a positive correlation between seminal CFMD copy number and semen parameters, including morphology, motility, and velocity. Therefore, the content of seminal CFMD could be a potential diagnostic marker for evaluating the semen quality. Additionally, seminal ROS is negatively correlated with semen parameters [60]. Thus, a negative relationship was seen between the copy number of CFMD in semen and ROS level. There is a low feasible explanation to support this data. The first explanation is that the decreased copy numbers of CFMD in seminal plasma indicates more content of mitochondria in spermatozoa of infertile men (the increased number of mitochondria may be related to excessive ROS). The second is that the enhanced copy number of mtDNA in poor-quality sperm may be due to retarded elimination process and excessive oxidative stress levels during spermatogenesis [61].

6 Clinical collection and analysis methods for cfNAs

The isolation and analysis of cfDNA in semen involve various approaches, including the use of commercial kits based on selective binding and elution technologies, magnetic-bead methods, and organic solvent extractions. Quantitative real-time PCR has demonstrated that cfDNA levels in semen can differentiate between men with sperm abnormalities and those with normal sperm, with significantly higher cfDNA levels observed in patients with azoospermia and teratozoospermia [1]. Proper sample collection is critical for cfDNA analysis. Standardization ensures consistency, with semen donors needing to follow specific guidelines and undergo routine analyses [62]. The separation of cells from plasma or fluid is typically achieved through centrifugation. High-speed centrifugation is preferred for its efficiency in preparing plasma for cfDNA isolation [63].

Several methods are employed for cfDNA isolation and purification:

Silica-Membrane Technology: This method uses selective binding and elution on silica membranes. It is fast and easy but may lose small DNA fragments [64]. Magnetic Bead Technology: This method involves magnetic beads coated to bind DNA. It is efficient and suitable for high-throughput applications, but requires specialized equipment [65]. Organic Solvent Extraction: Methods like phenol–chloroform extraction recover more cfDNA, including smaller fragments. These methods are time-consuming but flexible for protocol adjustments [66,67,68,69].

Analytical Methods Key techniques for cfDNA quantification and analysis include: Real-Time qPCR: Targets specific genes for cfDNA quantification and size distribution [63]. Digital and Droplet Digital PCR: Highly sensitive and precise for detecting low-abundance cfDNA [62]. Next-Generation Sequencing (NGS): Provides comprehensive data on cfDNA [70].

7 Biological mechanisms of cfNA levels and male fertility

The increased concentration and the DNA ladder pattern of cfDNA fragments in men with azoospermia suggest that the apoptosis of germ cells might be a contributing mechanism to the secretion of cfDNA [70].

Oxidative stress (OS) has been suggested as an additional source of cfDNA. An in vitro study demonstrated that exposing semen to paraquat, a toxic compound that induces oxidative stress by generating superoxide anions, led to increased levels of double-stranded cfDNA. This exposure also resulted in decreased sperm viability, motility, and normal morphology. Consequently, the concentration of cfDNA was proposed as a marker for OS in semen [22].

Subsequent research aimed to identify the primary origin of cfDNA secretion. To this end, researchers compared cfDNA concentrations between normozoospermic and vasectomized men. The findings revealed that cfDNA levels in normal semen were four times higher than in vasectomized men. Since vasectomized men lack testis and epididymis ejaculations, the lower cfDNA levels in their semen suggest that these organs are the main sources of cfDNA. Consequently, cfDNA could provide extensive information about the DNA status of these critical reproductive organs, particularly the testis, which directly influences male fertility. This led to further studies focusing on the practical applications of cfDNA, particularly as biomarkers in reproductive medicine [71].

Several factors may explain the high cfDNA concentration in semen: (1) DNA from dying cells through apoptosis, necrosis, and netosis, particularly during spermatogenesis; (2) DNA secreted by glandular cells, as secretions from seminal vesicles, prostate, and bulbourethral glands form about 90% of semen; (3) Chemicals in seminal plasma protect cfDNA from degradation and influence DNase activity, with cations like Ca2 + , Mg2 + , and Zn2 + playing a role; (4) Pathological conditions such as inflammation, cancer, or trauma may also contribute to elevated cfDNA levels [72,73,74,75,76,77,78,79,80].

As previously noted, male infertility can be attributed to obesity and being overweight. Evaluating the potential effects of overweight and obesity on DNA integrity is essential, as increased sperm DNA damage has been associated with lower pregnancy success rates and higher miscarriage rates [81]. Kort et al. [82] identified an increase in sperm DNA damage, assessed via the Sperm Chromatin Structure Assay (SCSA), in overweight and obese patients. Chavarro et al. [83] and Ferreiro et al. [84], using the Comet assay, and La Vignera et al. [85], using the TUNEL assay along with flow cytometry, noted higher sperm DNA damage in obese men, but not in those who were merely overweight. On the other hand, other studies did not find a significant association between BMI and the health of sperm DNA when utilizing the TUNEL and SCSA techniques [86,87,88].

Through the utilization of the TUNEL test on a more extensive sample size, another study revealed a heightened susceptibility to sperm DNA damage in obese males, although no such correlation was detected in overweight men. These results support and validate earlier research findings. This increased risk remained present even after accounting for age and smoking, which are characteristics that are frequently overlooked in previous research despite their recognized influence on sperm DNA fragmentation [81].

Additionally, since varicocele is recognized as a contributing factor to male infertility, A meta-analysis study analyzed studies including seven on sperm DNA damage in varicocele patients and six on the effectiveness of varicocele repair, with one study covering both aspects. In the DNA damage studies, involving 240 patients and 176 controls, varicocele patients showed significantly higher sperm DNA damage, with a mean difference of 9.84% (95% CI 9.19 to 10.49; P < 0.00001). In the repair studies, 177 patients had surgery, resulting in a notable improvement in sperm DNA integrity, with a mean difference of − 3.37% (95% CI − 4.09 to − 2.65; P < 0.00001). As mentioned before Oxidative stress and oxidative DNA damage were associated with impaired spermatogenesis in varicocele patients. High OS levels in these patients indicate its role in sperm DNA damage. The link between varicoceles and OS might be due to increased nitric oxide and related enzymes in the dilated veins. Additionally, elevated intratesticular temperature in varicocele patients may impair testicular function and directly damage nuclear DNA in seminiferous tubules [89]. Also The underlying mechanisms of DNA damage in varicocele patients can include apoptosis, and abnormal chromatin packaging [90, 91].

8 Cost-effectiveness

Traditional semen analysis continues to be a fundamental part of the first assessment of male fertility. This standard method evaluates crucial factors like sperm count, motility, morphology, and viability. Although standard semen analysis is commonly used, it has limitations and often fails to discover the root reasons of infertility in situations when sperm parameters appear to be normal [92].

Several research have examined the cost-effectiveness of using cfDNA testing for managing male infertility, while the majority of these studies have mostly focused on perinatal situations. An important study to take into account is the investigation conducted by Mbaye et al. [93]. This study emphasizes that cfDNA is a very efficient diagnostic tool for detecting several biomarkers associated with infertility. Therefore, it has the potential to be a cost-effective complement to conventional diagnostic approaches. However, it does not provide a thorough and direct comparison of cost-effectiveness between the new approach and established procedures for male infertility. Boissière et al. [94] examined the role of cfDNA in male infertility and demonstrated that this method could serve as a biomarker for diagnosing male infertility. Due to its non-invasive nature and ease of detection, this method could be a suitable alternative to invasive and traditional methods.

An extensive examination of cfDNA in many medical scenarios reveals its capacity for reducing costs and enhancing diagnostic precision, indicating that its use in male infertility may exhibit comparable cost-effectiveness trends.

9 Future of seminal cfNA as biomarker in male infertility

While the current evidence highlights the potential of circulating cfNAs as biomarkers for male infertility, there are several limitations in the existing research. Variations in the handling of samples, differences in methodology and technical techniques among laboratories are typically not consistent, which might result in conflicting outcomes. Furthermore, as recently explained by Dong et al. [57], there may be deceptive elements that might affect the measurement of seminal cf-RNA levels. For example, being exposed to heat or abstaining from sexual activity for a lengthy period of time might lead to an increase in amounts of cf-RNA in semen.

Additional investigation is required to rectify these deficiencies. Conducting extensive research involving several centers and following defined methods for collecting samples and analyzing cfNAs is crucial to confirm the clinical usefulness of cfNAs as biomarkers. Furthermore, investigating the molecular mechanisms that connect cfNAs to infertility might offer a more profound comprehension of their function in male reproductive health. It is essential to develop sophisticated analytical methods to improve the accuracy and precision of cfNA detection in order to advance this field.

Although current evidence suggests that cfDNA may be used as a biomarker for male infertility, there are several limitations and uncertainties in the existing studies that need careful consideration. One of these limitations is the variation in demographic characteristics of the participants across different studies. Such variations can include age, race, general health status, and lifestyle habits like diet and smoking. These differences may lead to inconsistent results and reduce the generalizability of the findings [95].

Additionally, technical differences in experimental methods and laboratory analyses can also result in variable outcomes. For example, different methods of cfDNA isolation and purification may yield varying amounts of cfDNA, and diverse analytical techniques such as PCR, qPCR, and NGS may have different sensitivities and accuracies. Therefore, future studies should aim to standardize sample collection methods, cfDNA isolation procedures, and laboratory analyses to minimize these discrepancies [1].

Ultimately, conducting extensive research with collaboration across multiple research centers and following standardized protocols for sample collection and cfDNA analysis can help validate the clinical utility of cfDNA as a biomarker for male infertility. Furthermore, deeper investigation into the molecular mechanisms linking cfDNA to infertility may provide a better understanding of their role in male reproductive health.

10 Conclusion

The data given in this concise analysis emphasize the potential significance of circulating cfNAs, specifically cfDNA, as promising indicators for the diagnosis of male infertility. In order to properly evaluate their therapeutic value, it is necessary to overcome various limitations and inconsistencies that exist in the existing study, notwithstanding their promise. Differences in the way samples are handled, variations in methodology, and technical disparities across laboratories might result in inconsistent results, which reduce the reliability of cfNA measurements. In addition, extrinsic variables such as exposure to high temperatures or extended periods of sexual abstinence might impact the quantities of cfNA (cell-free nucleic acid) in semen, thereby providing further variability.

In order to make progress in the area, it is crucial to conduct thorough multi-center research that adheres to established methods for both sample collection and cfNA analysis. Studying the molecular pathways that connect cfNAs to infertility can offer a more profound understanding of their impact on male reproductive well-being. It will be essential to create advanced analytical techniques to improve the accuracy and precision of cfNA detection. Although existing data indicate that cfDNA can be a reliable and non-invasive biomarker for male infertility, it is crucial to overcome the limitations and establish standardized research techniques in order to demonstrate its clinical significance and enhance diagnostic tools for male infertility.

Data availability

Data sharing is not applicable as no new data were generated or analyzed during this study.

References

  1. Draškovič T (2017) Isolation of cell-free DNA from seminal fluid. J Pharm Pharmacol 5:554–560

    Google Scholar 

  2. Cox C, Thoma M, Tchangalova N, Mburu G, Bornstein M, Johnson C, Kiarie J (2022) Infertility prevalence and the methods of estimation from 1990 to 2021: a systematic review and meta-analysis. Hum Reprod Open 2022(4):hoac051

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  3. Akbarinejad V, Fathi R, Shahverdi A, Esmaeili V, Rezagholizadeh A, Ghaleno LR (2020) The relationship of mitochondrial membrane potential, reactive oxygen species, adenosine triphosphate content, sperm plasma membrane integrity, and kinematic properties in warmblood stallions. J Equine Vet 94:103267

    Article  Google Scholar 

  4. La Marca A, Mastellari E (2020) Infertility: epidemiology and etiology. In: Petraglia Felice, Fauser Bart C (eds) Female reproductive dysfunction. Springer International Publishing, Cham, pp 211–33

    Chapter  Google Scholar 

  5. Pallotti F, Barbonetti A, Rastrelli G, Santi D, Corona G, Lombardo F (2022) The impact of male factors and their correct and early diagnosis in the infertile couple’s pathway: 2021 perspectives. J Endocrinol Invest 45(10):1807–1822

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  6. Babakhanzadeh E, Nazari M, Ghasemifar S, Khodadadian A (2020) Some of the factors involved in male infertility: a prospective review. Intl J Gen Med. https://doi.org/10.2147/IJGM.S241099

    Article  Google Scholar 

  7. Qasemi M, Mahdian R, Amidi F (2021) Cell-free DNA discoveries in human reproductive medicine: providing a new tool for biomarker and genetic assays in ART. J Assist Reprod Genet 38:277–288

    Article  PubMed  PubMed Central  Google Scholar 

  8. Gahan PB, Schwarzenbach H, Anker P (2022) The history and future of basic and translational cell-free DNA research at a glance. Diagnostics 12(5):1192

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  9. Liu J, Chen X, Wang J, Zhou S, Wang C, Ye M et al (2019) Biological background of the genomic variations of cf-DNA in healthy individuals. Ann Oncol 30(3):464–470

    Article  PubMed  CAS  Google Scholar 

  10. Duvvuri B, Lood C (2019) Cell-free DNA as a biomarker in autoimmune rheumatic diseases. Front Immunol 10:502

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  11. Spector BL, Harrell L, Sante D, Wyckoff GJ, Willig L (2023) The methylome and cell-free DNA: current applications in medicine and pediatric disease. Pediatr Res 94(1):89–95

    Article  PubMed  Google Scholar 

  12. Yang SR, Mooney KL, Libiran P, Jones CD, Joshi R, Lau HD et al (2020) Targeted deep sequencing of cell-free DNA in serous body cavity fluids with malignant, suspicious, and benign cytology. Cancer Cytopathol 128(1):43–56

    Article  PubMed  CAS  Google Scholar 

  13. Grabuschnig S, Bronkhorst AJ, Holdenrieder S, Rosales Rodriguez I, Schliep KP, Schwendenwein D et al (2020) Putative origins of cell-free DNA in humans: a review of active and passive nucleic acid release mechanisms. Int J Mol Sci 21(21):8062

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  14. de Miranda FS, Barauna VG, Dos Santos L, Costa G, Vassallo PF, Campos LCG (2021) Properties and application of cell-free DNA as a clinical biomarker. Int J Mol Sci 22(17):9110

    Article  PubMed  PubMed Central  Google Scholar 

  15. Gielis E, Ledeganck K, Winter D, Del Favero J, Bosmans JL, Claas F et al (2015) Cell-free DNA: an upcoming biomarker in transplantation. Am J Transpl 15(10):2541–2551

    Article  CAS  Google Scholar 

  16. Casteleiro Alves MM, Oliani L, Almeida M, Cardoso HJ, Oliani AH, Breitenfeld L, Ramalhinho AC (2023) Cell-free DNA as a new biomarker of IVF success, independent of any infertility factor, including endometriosis. Diagnostics 13(2):208

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  17. Skakkebæk NE, Lindahl-Jacobsen R, Levine H, Andersson AM, Jørgensen N, Main KM et al (2022) Environmental factors in declining human fertility. Nat Rev Endocrinol 18(3):139–157

    Article  PubMed  Google Scholar 

  18. Boeri L, Fallara G, Pozzi E, Belladelli F, Corsini C, Raffo M et al (2022) The impact of different WHO reference criteria for semen analysis in clinical practice: who will benefit from the new 2021 thresholds for normal semen parameters? Andrology 10(6):1134–1142

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  19. GhoshRoy D, Alvi P, Santosh K (2023) AI tools for assessing human fertility using risk factors: a state-of-the-art review. J Med Syst 47(1):91

    Article  PubMed  Google Scholar 

  20. Montoya JM, Bernal A, Borrero C (2002) Diagnostics in assisted human reproduction. Reprod Biomed Online 5(2):198–210

    Article  PubMed  Google Scholar 

  21. Venkatesh S, Deecaraman M, Kumar R, Shamsi M, Dada R (2009) Role of reactive oxygen species in the pathogenesis of mitochondrial DNA (mtDNA) mutations in male infertility. Indian J Med Res 129(2):127–137

    PubMed  CAS  Google Scholar 

  22. Costa F, Barbisan F, Assmann C, Araújo N, de Oliveira A, Signori J et al (2017) Seminal cell-free DNA levels measured by PicoGreen fluorochrome are associated with sperm fertility criteria. Zygote 25(2):111–119

    Article  PubMed  CAS  Google Scholar 

  23. Kumar N, Singh AK (2022) Impact of environmental factors on human semen quality and male fertility: a narrative review. Environ Sci Eur 34:1–13

    Article  Google Scholar 

  24. Rodprasert W, Toppari J, Virtanen HE (2023) Environmental toxicants and male fertility. Best Pract Res Clin Obstet Gynaecol 86:102298

    Article  PubMed  Google Scholar 

  25. Martínez MÁ, Marquès M, Salas-Huetos A, Babio N, Domingo JL, Salas-Salvadó J (2023) Lack of association between endocrine disrupting chemicals and male fertility: a systematic review and meta-analysis. Environ Res 217:114942

    Article  PubMed  Google Scholar 

  26. Girela JL, Gil D, Johnsson M, Gomez-Torres MJ, De Juan J (2013) Semen parameters can be predicted from environmental factors and lifestyle using artificial intelligence methods. Biol Reprod 88(4):99

    Article  PubMed  Google Scholar 

  27. Jurewicz J, Radwan M, Sobala W, Ligocka D, Radwan P, Bochenek M, Hanke W (2014) Lifestyle and semen quality: role of modifiable risk factors. Syst Biol Reprod Med 60(1):43–51

    Article  PubMed  Google Scholar 

  28. Jensen TK, Swan SH, Skakkebæk NE, Rasmussen S, Jørgensen N (2010) Caffeine intake and semen quality in a population of 2,554 young Danish men. Am J Epidemiol 171(8):883–891

    Article  PubMed  Google Scholar 

  29. Fusco F, Verze P, Capece M, Napolitano L (2021) Suppression of spermatogenesis by exogenous testosterone. Curr Pharm Des 27(24):2750–2753

    Article  PubMed  CAS  Google Scholar 

  30. van den Beld AW, Kaufman J-M, Zillikens MC, Lamberts SW, Egan JM, van der Lely AJ (2018) The physiology of endocrine systems with ageing. Lancet Diabet Endocrinol 6(8):647–658

    Article  Google Scholar 

  31. Katib AA, Al–Hawsawi K, Motair W, Bawa AM (2014) Secondary infertility and the aging male, overview. Cent Eur J Urol 67(2):184

    Article  Google Scholar 

  32. Ali M, Parekh N (2020) Male age and andropause. In: Parekattil SJ, Esteves SC, Agarwal A (eds) Male infertility: contemporary clinical approaches, andrology, ART and antioxidants. Springer International Publishing, Springer Cham, pp 469–77

    Chapter  Google Scholar 

  33. Pohl E, Gromoll J, Wistuba J, Laurentino S (2021) Healthy ageing and spermatogenesis. Reproduction 161(4):R89–R101

    Article  PubMed  CAS  Google Scholar 

  34. Libby P (2007) Inflammatory mechanisms: the molecular basis of inflammation and disease. Nutr Rev 65(suppl_3):S140–S6

    Article  Google Scholar 

  35. Schaeffer AJ (2002) Infection and inflammation of the genitourinary tract. J Urol 167(4):1926–1935

    Article  Google Scholar 

  36. Christoffersen M, Troedsson M (2017) Inflammation and fertility in the mare. Reprod Domest Anim 52:14–20

    Article  PubMed  Google Scholar 

  37. Martínez-Prado E, Camejo Bermúdez MI (2010) Expression of IL-6, IL-8, TNF-α, IL-10, HSP-60, Anti-HSP-60 antibodies, and anti-sperm antibodies, in semen of men with leukocytes and/or bacteria. Am J Reprod Immunol 63(3):233–243

    Article  PubMed  Google Scholar 

  38. Huynh T, Mollard R, Trounson A (2002) Selected genetic factors associated with male infertility. Hum Reprod Update 8(2):183–198

    Article  PubMed  Google Scholar 

  39. Krausz C, Cioppi F, Riera-Escamilla A (2018) Testing for genetic contributions to infertility: potential clinical impact. Expert Rev Mol Diagn 18(4):331–346

    Article  PubMed  CAS  Google Scholar 

  40. Colaco S, Modi D (2018) Genetics of the human Y chromosome and its association with male infertility. Reprod Biol Endocrinol 16:1–24

    Article  Google Scholar 

  41. Navarro-Costa P, Gonçalves J, Plancha CE (2010) The AZFc region of the Y chromosome: at the crossroads between genetic diversity and male infertility. Hum Reprod Updat 16(5):525–542

    Article  CAS  Google Scholar 

  42. Keskin MZ, Budak S, Zeyrek T, Çelik O, Mertoglu O, Yoldas M, Ilbey YÖ (2015) The relationship between serum hormone levels (follicle-stimulating hormone, luteinizing hormone, total testosterone) and semen parameters. Archivio Italiano Di Urologia E Andrologia 87(3):194–197

    Article  PubMed  CAS  Google Scholar 

  43. Messing EM, Manola J, Sarosdy M, Wilding G, Crawford ED, Trump D (1999) Immediate hormonal therapy compared with observation after radical prostatectomy and pelvic lymphadenectomy in men with node-positive prostate cancer. N Engl J Med 341(24):1781–1788

    Article  PubMed  CAS  Google Scholar 

  44. Silva AF, Ramalho-Santos J, Amaral S (2021) The impact of antisperm antibodies on human male reproductive function: an update. Reproduction 162(4):R55–R71

    Article  PubMed  CAS  Google Scholar 

  45. Bohring C, Krause W (2005) The role of antisperm antibodies during fertilization and for immunological infertility. Immun Gametes Embryo Implant 88:15–26

    Article  CAS  Google Scholar 

  46. Bohring C, Krause W (2003) Immune infertility: towards a better understanding of sperm (auto)-immunity: the value of proteomic analysis. Hum Reprod 18(5):915–924

    Article  PubMed  CAS  Google Scholar 

  47. Nagaria T, Patra P, Sahu JP (2011) Evaluation of serum antisperm antibodies in infertility. J Obstet Gynecol India 61:307–316

    Article  CAS  Google Scholar 

  48. Wdowiak A, Gujski M, Bojar I, Raczkiewicz D, Bartosińska J, Wdowiak-Filip A, Filip R (2021) Chronic inflammation impairs male fertility—a case-control study in ulcerative colitis patients. J Clin Med 10(7):1460

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  49. Pös O, Biró O, Szemes T, Nagy B (2018) Circulating cell-free nucleic acids: characteristics and applications. Eur J Hum Genet 26(7):937–945

    Article  PubMed  PubMed Central  Google Scholar 

  50. Szilágyi M, Pös O, Márton É, Buglyó G, Soltész B, Keserű J et al (2020) Circulating cell-free nucleic acids: main characteristics and clinical application. Int J Mol Sci 21(18):6827

    Article  PubMed  PubMed Central  Google Scholar 

  51. Kustanovich A, Schwartz R, Peretz T, Grinshpun A (2019) Life and death of circulating cell-free DNA. Cancer Biol Ther 20(8):1057–1067

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  52. Norton ME, Jacobsson B, Swamy GK, Laurent LC, Ranzini AC, Brar H et al (2015) Cell-free DNA analysis for noninvasive examination of trisomy. N Engl J Med 372(17):1589–1597

    Article  PubMed  CAS  Google Scholar 

  53. Dai Y, Kong X, Yao C, Xiong C, Li Z, Li H (2022) Multi-stage screening cell-free seminal mRNAs to diagnose completion of meiosis and predict testicular sperm retrieval in men with non-obstructive azoospermia. Andrology 10(4):749–757

    Article  PubMed  CAS  Google Scholar 

  54. Yu Q, Gu X, Shang X, Li H, Xiong C (2016) Discrimination and characterization of Sertoli cell-only syndrome in non-obstructive azoospermia using cell-free seminal DDX4. Reprod Biomed Online 33(2):189–196

    Article  PubMed  CAS  Google Scholar 

  55. Tzimagiorgis G, Michailidou EZ, Kritis A, Markopoulos AK, Kouidou S (2011) Recovering circulating extracellular or cell-free RNA from bodily fluids. Cancer Epidemiol 35(6):580–589

    Article  PubMed  CAS  Google Scholar 

  56. Timofeeva AV, Chagovets VV, Drapkina YS, Makarova NP, Kalinina EA, Sukhikh GT (2019) Cell-free, embryo-specific sncRNA as a molecular biological bridge between patient fertility and IVF efficiency. Int J Mol Sci 20(12):2912

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  57. Dong T, Yu Q, Qing X, Ma X, Dong W, Shi J, Li H (2016) Potential confounding factors in measurement of specific cell-free seminal mRNA s and micro RNA s derived from human reproductive organs. Andrology 4(6):1010–1019

    Article  PubMed  CAS  Google Scholar 

  58. Aitken RJ, Whiting S, Connaughton H, Curry B, Reinheimer T, van Duin M (2020) A novel pathway for the induction of DNA damage in human spermatozoa involving extracellular cell-free DNA. Mutat Res Fundam Mol Mech Mutagen 821:111722

    Article  CAS  Google Scholar 

  59. Chen Y, Liao T, Zhu L, Lin X, Wu R, Jin L (2018) Seminal plasma cell-free mitochondrial DNA copy number is associated with human semen quality. Eur J Obstet Gynecol Reprod Biol 231:164–168

    Article  PubMed  CAS  Google Scholar 

  60. Srivastava A, Srivastava P, Mathur S, Mishra S, Abbas S, Gupta A et al (2022) Analysis of cellular and cell free mitochondrial DNA content and reactive oxygen species levels in maternal blood during normal pregnancy: a pilot study. BMC Pregnancy Childbirth 22(1):845

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  61. Bisserier M, Shanmughapriya S, Rai AK, Gonzalez C, Brojakowska A, Garikipati VNS et al (2021) Cell-Free mitochondrial DNA as a potential biomarker for astronauts’ health. J Am Heart Assoc 10(21):e022055

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  62. Jung K, Fleischhacker M, Rabien A (2010) Cell-free DNA in the blood as a solid tumor biomarker—a critical appraisal of the literature. Clin Chim Acta 411(21–22):1611–1624

    Article  PubMed  CAS  Google Scholar 

  63. Chou JS, Jacobson JD, Patton WC, King A, Chan PJ (2004) Modified isocratic capillary electrophoresis detection of cell-free DNA in semen. J Assist Reprod Genet 21:397–400

    Article  PubMed  PubMed Central  Google Scholar 

  64. Devonshire AS, Whale AS, Gutteridge A, Jones G, Cowen S, Foy CA, Huggett JF (2014) Towards standardisation of cell-free DNA measurement in plasma: controls for extraction efficiency, fragment size bias and quantification. Anal Bioanal Chem 406:6499–6512

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  65. Stormer M, Kleesiek K, Dreier J (2007) High-volume extraction of nucleic acids by magnetic bead technology for ultrasensitive detection of bacteria in blood components. Clin Chem 53(1):104–110

    Article  PubMed  Google Scholar 

  66. Fong SL, Zhang JT, Lim CK, Eu KW, Liu Y (2009) Comparison of 7 methods for extracting cell-free DNA from serum samples of colorectal cancer patients. Clin Chem 55(3):587–589

    Article  PubMed  CAS  Google Scholar 

  67. Schmidt B, Weickmann S, Witt C, Fleischhacker M (2005) Improved method for isolating cell-free DNA. Clin Chem 51(8):1561–1563

    Article  PubMed  CAS  Google Scholar 

  68. Yuan H, Zhu ZZ, Lu Y, Liu F, Zhang W, Huang G et al (2012) A modified extraction method of circulating free DNA for epidermal growth factor receptor mutation analysis. Yonsei Med J 53(1):132–137

    Article  PubMed  CAS  Google Scholar 

  69. Xue X, Teare MD, Holen I, Zhu YM, Woll PJ (2009) Optimizing the yield and utility of circulating cell-free DNA from plasma and serum. Clin Chim Acta 404(2):100–104

    Article  PubMed  CAS  Google Scholar 

  70. Li HG, Huang SY, Zhou H, Liao AH, Xiong CL (2009) Quick recovery and characterization of cell-free DNA in seminal plasma of normozoospermia and azoospermia: implications for non-invasive genetic utilities. Asian J Androl 11(6):703

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  71. Wu C, Ding X, Li H, Zhu C, Xiong C (2013) Genome-wide promoter methylation profile of human testis and epididymis: identified from cell-free seminal DNA. BMC Genomics 14:1–10

    Article  CAS  Google Scholar 

  72. Li HG, Huang SY, Zhou H, Liao AH, Xiong CL (2009) Quick recovery and characterization of cell-free DNA in seminal plasma of normozoospermia and azoospermia: implications for non-invasive genetic utilities. Asian J Androl 11(6):703–709

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  73. Fleischhacker M, Schmidt B (2007) Circulating nucleic acids (CNAs) and cancer—a survey. Biochimica Et Biophysica Acta BBA Rev Cancer 1775(1):181–232

    Article  CAS  Google Scholar 

  74. Moreira VG, García BP, de la Cera MT, Menéndez FVÁ (2009) Elevated transrenal DNA (cell-free urine DNA) in patients with urinary tract infection compared to healthy controls. Clin Biochem 42(7–8):729–731

    Article  Google Scholar 

  75. Su Y-H, Wang M, Brenner DE, Ng A, Melkonyan H, Umansky S et al (2004) Human urine contains small, 150 to 250 nucleotide-sized, soluble DNA derived from the circulation and may be useful in the detection of colorectal cancer. J Mol Diagn 6(2):101–107

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  76. Lapaire O, Bianchi DW, Peter I, O’BrienStrohCowan BHJM et al (2007) Cell-free fetal DNA in amniotic fluid unique fragmentation signatures in euploid and aneuploid fetuses. Clin Chem 53(3):405–11

    Article  PubMed  CAS  Google Scholar 

  77. Sadeghian-Nodoushan F, Aflatoonian R, Borzouie Z, Akyash F, Fesahat F, Soleimani M, Aghajanpour S, Moore HD, Aflatoonian B (2016) Pluripotency and differentiation of cells from human testicular sperm extraction: An investigation of cell stemness. Mol Reprod Dev 83(4):312–323

    Article  PubMed  CAS  Google Scholar 

  78. Blanco-Rodriguez J (1998) A matter of death and life: the significance of germ cell death during spermatogenesis. Int J Androl 21(5):236–248

    Article  PubMed  CAS  Google Scholar 

  79. Quinn P (1968) Deoxyribonuclease activity in semen. Reproduction 17(1):35–39

    Article  CAS  Google Scholar 

  80. Hashida T, Tanaka Y, Matsunami N, Yoshihara K, Kamiya T, Tanigawa Y, Koide SS (1982) Purification and properties of bull seminal plasma Ca2+, Mg2+-dependent endonuclease. J Biol Chem 257(21):13114–13119

    Article  PubMed  CAS  Google Scholar 

  81. Dupont C, Faure C, Sermondade N, Boubaya M, Eustache F, Clément P et al (2013) Obesity leads to higher risk of sperm DNA damage in infertile patients. Asian J Androl 15(5):622

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  82. Kort HI, Massey JB, Elsner CW, Mitchell-Leef D, Shapiro DB, Witt MA, Roudebush WE (2006) Impact of body mass index values on sperm quantity and quality. J Androl 27(3):450–452

    Article  PubMed  Google Scholar 

  83. Chavarro JE, Toth TL, Wright DL, Meeker JD, Hauser R (2010) Body mass index in relation to semen quality, sperm DNA integrity, and serum reproductive hormone levels among men attending an infertility clinic. Fertil Steril 93(7):2222–2231

    Article  PubMed  CAS  Google Scholar 

  84. Fariello RM, Pariz JR, Spaine DM, Cedenho AP, Bertolla RP, Fraietta R (2012) Association between obesity and alteration of sperm DNA integrity and mitochondrial activity. BJU Int 110(6):863–867

    Article  PubMed  CAS  Google Scholar 

  85. La Vignera S, Condorelli RA, Vicari E, Calogero AE (2012) Negative effect of increased body weight on sperm conventional and nonconventional flow cytometric sperm parameters. J Androl 33(1):53–58

    Article  PubMed  Google Scholar 

  86. Tunc O, Bakos H, Tremellen K (2011) Impact of body mass index on seminal oxidative stress. Andrologia 43(2):121–128

    Article  PubMed  CAS  Google Scholar 

  87. Rybar R, Kopecka V, Prinosilova P, Markova P, Rubes J (2011) Male obesity and age in relationship to semen parameters and sperm chromatin integrity. Andrologia 43(4):286–291

    Article  PubMed  CAS  Google Scholar 

  88. Hammiche F, Laven J, Boxmeer J, Dohle G, Steegers E, Steegers-Theunissen R (2011) Sperm quality decline among men below 60 years of age undergoing IVF or ICSI treatment. J Androl 32(1):70–76

    Article  PubMed  CAS  Google Scholar 

  89. Wang YJ, Zhang RQ, Lin YJ, Zhang RG, Zhang WL (2012) Relationship between varicocele and sperm DNA damage and the effect of varicocele repair: a meta-analysis. Reprod Biomed Online 25(3):307–314

    Article  PubMed  CAS  Google Scholar 

  90. Wood GJA, Cardoso JPG, Paluello DV, Nunes TF, Cocuzza M (2021) Varicocele-associated infertility and the role of oxidative stress on sperm DNA fragmentation. Front Reprod Health 3:695992

    Article  PubMed  PubMed Central  Google Scholar 

  91. Esfahani MHN, Tavalaee M (2012) Origin and role of DNA damage in varicocele. Int J Fertil Steril 6(3):141

    Google Scholar 

  92. Barbăroșie C, Agarwal A, Henkel R (2021) Diagnostic value of advanced semen analysis in evaluation of male infertility. Andrologia 53(2):e13625

    Article  PubMed  Google Scholar 

  93. Mbaye MM, Khalfi BE, Louanjli N, Saadani B, Kaarouch I, Madkour A, Soukri A (2019) Seminal cell-free DNA test for the management of male infertility. Ann Res Rev Biol 30(5):1–10

    Google Scholar 

  94. Boissière A, Gala A, Ferrières-Hoa A, Mullet T, Baillet S, Petiton A et al (2017) Cell-free and intracellular nucleic acids: new non-invasive biomarkers to explore male infertility. Basic Clin Androl 27:1–8

    Article  Google Scholar 

  95. Llavanera M, Delgado-Bermudez A, Ribas-Maynou J, Salas-Huetos A, Yeste M (2022) A systematic review identifying fertility biomarkers in semen: a clinical approach through omics to diagnose male infertility. Fertil Steril 118(2):291–313

    Article  PubMed  CAS  Google Scholar 

  96. Mbaye MM, Zekhnini H, Khalfi BE, Louanjli N, Zakaria M, Elmellouli F, Soukri A (2021) Determination of free DNA (cfDNA) by RT-qPCR in individuals in sperm alterations. Ann Res Rev Biol 36(5):1–6

    Article  Google Scholar 

  97. Di Pizio P, Celton N, Menoud PA, Belloc S, Cohen Bacrie M, Belhadri-Mansouri N et al (2021) Seminal cell-free DNA and sperm characteristic’s: an added biomarker for male infertility investigation. Andrologia 53(1):e13822

    Article  PubMed  Google Scholar 

  98. Ponti G, Maccaferri M, Mandrioli M, Manfredini M, Micali S, Cotugno M et al (2018) Seminal cell-free DNA assessment as a novel prostate cancer biomarker. Pathol Oncol Res 24:941–945

    Article  PubMed  CAS  Google Scholar 

  99. Hazout A, Montjean D, Cassuto N, Belloc S, Dalleac A, Tesarik J, Benkhalifa M (2018) Free circulating nucleic acids and infertility. J Gynecol Women Health 11:1–11

    Google Scholar 

  100. Wu C, Ding X, Tan H, Li H, Xiong C (2016) Alterations of testis-specific promoter methylation in cell-free seminal deoxyribonucleic acid of idiopathic nonobstructive azoospermic men with different testicular phenotypes. Fertil Steril 106(6):1331–1337

    Article  PubMed  CAS  Google Scholar 

  101. Spindler KLG, Pallisgaard N, Vogelius I, Jakobsen A (2012) Quantitative cell-free DNA, KRAS, and BRAF mutations in plasma from patients with metastatic colorectal cancer during treatment with cetuximab and irinotecan. Clin Cancer Res 18(4):1177–85

    Article  PubMed  CAS  Google Scholar 

  102. Stroun M, Maurice P, Vasioukhin V, Lyautey J, Lederrey C, Lefort F et al (2000) The origin and mechanism of circulating DNA. Ann N Y Acad Sci 906(1):161–168

    Article  PubMed  CAS  Google Scholar 

  103. Tournaye H, Popovic-Todorovic B, Hamamah S, Nilsson B, Aasted H, Klein B, Arce J (2018) Cell-free DNA is elevated in men with abnormal semen. Fertil Steril 110(4):e298

    Article  Google Scholar 

Download references

Acknowledgements

The authors thank all contributors to this article

Funding

This research did not receive any specific funding.

Author information

Authors and Affiliations

Authors

Contributions

All authors contributed to the conception and design. ART and AS were responsible for overall supervision. DJ, FF and FH Drafted the manuscript. All authors contributed to conception and design. All authors have read and approved the manuscript.

Corresponding authors

Correspondence to Ali Reza Talebi or Abdolhossein Shahverdi.

Ethics declarations

Ethics approval and consent to participate

Not applicable.

Consent for publication

Not applicable.

Competing interests

The authors declare no conflicts of interest.

Additional information

Publisher's Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Rights and permissions

Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by/4.0/.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Javidmehr, D., Fesahat, F., Hassani, F. et al. Seminal cell-free nucleic acids as possible biomarker in male infertility: a mini-review article. Afr J Urol 30, 54 (2024). https://doi.org/10.1186/s12301-024-00450-1

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s12301-024-00450-1

Keywords