Next Article in Journal
The Anti-Microbial Peptide Citrocin Controls Pseudomonas aeruginosa Biofilms by Breaking Down Extracellular Polysaccharide
Previous Article in Journal
The Golgi Apparatus: A Key Player in Innate Immunity
Previous Article in Special Issue
Exploring the Mitogenomes of Mantodea: New Insights from Structural Diversity and Higher-Level Phylogenomic Analyses
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

Elucidating the Role of OXPHOS Variants in Asthenozoospermia: Insights from Whole Genome Sequencing and an In Silico Analysis

by
Maria-Anna Kyrgiafini
1,
Themistoklis Giannoulis
2,
Alexia Chatziparasidou
1,3 and
Zissis Mamuris
1,*
1
Laboratory of Genetics, Comparative and Evolutionary Biology, Department of Biochemistry and Biotechnology, University of Thessaly, Viopolis, Mezourlo, 41500 Larissa, Greece
2
Laboratory of Biology, Genetics and Bioinformatics, Department of Animal Sciences, University of Thessaly, Gaiopolis, 41336 Larissa, Greece
3
Embryolab IVF Unit, St. 173-175 Ethnikis Antistaseos, Kalamaria, 55134 Thessaloniki, Greece
*
Author to whom correspondence should be addressed.
Int. J. Mol. Sci. 2024, 25(7), 4121; https://doi.org/10.3390/ijms25074121
Submission received: 6 March 2024 / Revised: 31 March 2024 / Accepted: 5 April 2024 / Published: 8 April 2024
(This article belongs to the Special Issue Mitochondrial DNA and RNA 2.0)

Abstract

:
Infertility is a global health challenge that affects an estimated 72.4 million people worldwide. Between 30 and 50% of these cases involve male factors, showcasing the complex nature of male infertility, which can be attributed to both environmental and genetic determinants. Asthenozoospermia, a condition characterized by reduced sperm motility, stands out as a significant contributor to male infertility. This study explores the involvement of the mitochondrial oxidative phosphorylation (OXPHOS) system, crucial for ATP production and sperm motility, in asthenozoospermia. Through whole-genome sequencing and in silico analysis, our aim was to identify and characterize OXPHOS gene variants specific to individuals with asthenozoospermia. Our analysis identified 680,099 unique variants, with 309 located within OXPHOS genes. Nine of these variants were prioritized due to their significant implications, such as potential associations with diseases, effects on gene expression, protein function, etc. Interestingly, none of these variants had been previously associated with male infertility, opening up new avenues for research. Thus, through our comprehensive approach, we provide valuable insights into the genetic factors that influence sperm motility, laying the foundation for future research in the field of male infertility.

1. Introduction

Infertility is a significant global health concern, estimated to impact 72.4 million individuals worldwide [1]. Male factors are implicated in 30–50% of infertility cases among couples of reproductive age [2]. Male infertility is a multifaceted condition influenced by a combination of environmental and genetic factors [3], and it encompasses a wide range of subcategories, each involving various qualitative and quantitative sperm defects [4]. One prevalent cause of male infertility is asthenozoospermia, defined by the fifth edition of the World Health Organization (WHO) guidelines (https://apps.who.int/iris/handle/10665/44261, (accessed on 5 March 2024)) as having reduced sperm motility (<40%) or progressive motility below 32%. In this condition, although sperm are present in the semen, they lack the ability to move or progress sufficiently for the journey from the vagina to the fallopian tube. Consequently, fertilization becomes unattainable.
In the context of asthenozoospermia, the motility of sperm hinges on the availability of energy. Thus, recent investigations into sperm physiology have cast the spotlight on the mitochondrion, acknowledged as the cellular powerhouse. The mitochondrion plays a pivotal role in sperm function, impacting not only motility but also other critical aspects of fertilization, including capacitation, hyperactivation, and the acrosome reaction, thereby influencing overall male fertility [5]. Furthermore, current research highlights two primary metabolic pathways that contribute to ATP production for optimal sperm function: oxidative phosphorylation (OXPHOS) and glycolysis [6,7,8,9]. OXPHOS, the predominant metabolic pathway occurring within the mitochondria, is essential for proper sperm function, and disruptions in mitochondrial oxidative phosphorylation are posited to compromise normal sperm activity and especially sperm motility [7].
The oxidative phosphorylation system (OXPHOS) on the inner mitochondrial membrane consists of five enzymes organized into complexes I–V: NADH: ubiquinone reductase (Complex I), succinate dehydrogenase (Complex II), quinol-cytochrome c reductase (Complex III), cytochrome c oxidase (Complex IV), and H+-transporting two-sector ATPase or FoF1-ATPase (Complex V), which synthesizes ATP. In humans, these enzymes are mostly multimeric, with subunits encoded in both the mitochondrial genome (mtDNA) and the nuclear genome (nDNA), excluding Complex II [10].
However, despite the recent extensive research in the field, the molecular mechanisms underlying asthenozoospermia remain not fully understood in the majority of cases. Additionally, there is limited literature on specific mutations in OXPHOS genes that may cause asthenozoospermia.
Therefore, this study aimed to conduct whole-genome sequencing (WGS) in individuals with asthenozoospermia and those with normal sperm motility (normozoospermic), with the primary goal of identifying and characterizing variants within OXPHOS genes (both nuclear- and mtDNA-encoded) exclusive to asthenozoospermic men. By doing so, we sought to elucidate genetic factors contributing to the pathogenic phenotype associated with reduced sperm motility. It should be noted that the overarching objective of our research was to provide a comprehensive reference for future investigations into asthenozoospermia, offering insights into variants that potentially influence the functionality of the OXPHOS machinery and impact sperm motility. This endeavor involved an integrated approach that combines whole-genome sequencing with bioinformatics and in silico tools.

2. Results

2.1. WGS Results—Variant Calling and Annotation

After whole-genome sequencing, data analysis was conducted. Specifically, a comparison was performed between normozoospermic and asthenozoospermic individuals to identify unique variants present exclusively in one of the two groups. A total of 680,099 variants were observed exclusively in asthenozoospermic individuals, while 2,329,803 variants were found only in normozoospermic men. These variants were then mapped to 30,362 and 26,019 genes in normozoospermic and asthenozoospermic males, respectively.
For the purpose of this study, only the variants identified in asthenozoospermic individuals were selected for further analysis. This decision was made because the objective was to identify and investigate variants in OXPHOS genes that could potentially contribute to the reduced sperm motility observed in asthenozoospermic men.

2.2. Unique OXPHOS Variants in Asthenozoospermic Men

Out of the 680,099 unique variants found in men with asthenozoospermia, 309 were identified within OXPHOS genes, as shown in Table S1. Table 1 displays the distribution of these variants among the mitochondrial respiratory complexes and their associated genes, with no variants detected in Complex V.
Furthermore, as shown in Table 2, the majority of the unique variants were found in nuclear-encoded OXPHOS genes (94.5%) rather than in mitochondrial-encoded OXPHOS genes (5.5%).
It is worth noting that out of the 309 variants mapped on OXPHOS genes in men with asthenozoospermia, 22 (7.1%) were novel variants.

2.3. Unique OXPHOS Variants in Asthenozoospermic Men—Genomic Consequences and Missense Variants

The unique OXPHOS variants identified in asthenozoospermic men were also categorized based on their genomic consequences. As shown in Figure 1, the majority of variants were intronic (89%), followed by synonymous variants (3.8%), missense variants (3.1%), variants in 3′ untranslated (UTR) regions (3.1%), and variants in 5′ UTR regions (0.9%).
Regarding missense variants, as shown in Table 3, we evaluated them using SIFT [11] and Polyphen2 [12] scores to determine their impact on protein functionality. We found that only one variant, rs35462421, is deemed to have a damaging effect according to both SIFT [11] and Polyphen2 [12]. Table 3 also presents the allele frequencies for the European population, as this study focused on Europeans.

2.4. Unique OXPHOS Variants in Asthenozoospermic Men—Variants with Potential Functional Effect

To comprehensively evaluate the functional significance of the unique OXPHOS variants identified in asthenozoospermic men, we utilized two different databases: RegulomeDB [13] and 3DSNP 2.0 [14]. As presented in Table 4, we identified a total of twenty-eight variants that are most likely to have a functional impact, as they have a RegulomeDB rank ranging from 1a to 3b and a 3DSNP score greater than 10.

2.5. Unique OXPHOS Variants in Asthenozoospermic Men—Expression Quantitative Trait Loci (eQTL) and Splicing Quantitative Trait Loci (sQTL)

Expression Quantitative Trait Loci (eQTLs) and Splicing Quantitative Trait Loci (sQTLs) are two types of genetic loci that influence gene expression and RNA splicing, respectively, at a quantitative level. Both eQTLs and sQTLs are fundamental in genomics and molecular biology because they offer insights into how genetic variation affects phenotypic variation [15,16,17]. Therefore, we investigated the unique variants identified in OXPHOS genes in asthenozoospermic men using the GTex portal [18]. The identified variants are presented in Table 5, with only the variants associated with eQTLs and sQTLs in testis and prostate tissues being selected.

2.6. Unique OXPHOS Variants in Asthenozoospermic Men—Association with Diseases

SNPnexus [19] was utilized to identify the association between unique OXPHOS variants in asthenozoospermic men and diseases, as reported in previous studies. As shown in Table 6, five variants were identified as being associated with Leigh syndrome or/and Mitochondrial complex I deficiency.

2.7. Unique OXPHOS Variants in Asthenozoospermic Men—Interactions with miRNAs

MicroRNAs (miRNAs) play a crucial role in gene regulation by targeting messenger RNAs (mRNAs) for degradation or translational repression. They achieve this by binding to complementary sequences within the mRNA [20,21]. Therefore, studying variants that affect miRNA-mRNA interactions is essential because these variations can disrupt the delicate balance of gene expression, ultimately leading to abnormal protein production [22].
To determine if the unique variants in OXPHOS genes have an impact on mRNA-miRNA interactions, miRNASNP v3 [23] was utilized. Table 7 presents the findings, showing that seven variants were identified to affect the binding sites of miRNAs, resulting in either miRNA loss or/and gain. All of these variants were located in the 3’ UTR of OXPHOS genes.

3. Discussion

Asthenozoospermia, characterized by reduced sperm motility, stands out as a key contributor to male infertility. The etiology of this condition is closely linked to energy metabolism, as optimal energy production is essential for sperm motility [8]. This underscores the critical role of research into oxidative phosphorylation (OXPHOS) genes, which are fundamental for cellular energy production, in understanding the molecular basis of asthenozoospermia.
More specifically, in the past years, emerging studies have focused on the interplay between OXPHOS, mitochondrial function, and asthenozoospermia. Notably, Barbagallo et al. (2020) [24] observed diminished activities of mitochondrial respiratory complexes I, II, and IV in asthenozoospermic patients compared to fertile men, suggesting a direct link between OXPHOS dysfunction and reduced sperm motility. Furthermore, mutations in mitochondrial DNA (mtDNA) and their association with male infertility, particularly asthenozoospermia, have garnered significant scientific interest [25,26]. Studies also show that key features of non-motile sperm include mitochondrial membrane integrity disruption and compromised sheath function, with mitochondria contributing significantly to movement energy [9,27]. Alterations in mitochondrial chain enzyme activities can impinge on sperm motility too. Research indicates a correlation between mitochondrial enzyme activity, sperm motility, and idiopathic asthenozoospermia suggesting that mitochondrial impairment may be a causative factor [24].
Therefore, pinpointing specific genetic variants within OXPHOS genes linked to decreased sperm motility may unveil new aspects of asthenozoospermia pathophysiology and open avenues for targeted therapeutic interventions.
In the present study, we performed whole genome sequencing on blood samples obtained from asthenozoospermic and normozoospermic men. Our objective was to identify specific genetic variants that were exclusive to either group, as these variants could potentially contribute to the underlying pathology or serve as potential biomarkers. We specifically focused on variants within the OXPHOS pathway, which were found exclusively in asthenozoospermic men. Through the use of a comprehensive set of analytical tools, we identified variants within the OXPHOS genes that are most likely to impact the asthenozoospermic phenotype. More specifically, out of the 680,099 unique variants detected in individuals with asthenozoospermia, 309 were located within OXPHOS genes. Notably, as indicated in Table 8, nine of these variants were deemed high-priority for further investigation due to their significant effects, such as associations with diseases, expression quantitative trait loci (eQTLs), etc., as determined by our in silico analyses.
It should be emphasized that no previous studies have linked the identified prioritized OXPHOS variants with male infertility. According to SNPnexus [19], many of these variants are associated with conditions such as Leigh syndrome or mitochondrial complex I deficiency. Leigh Syndrome is a severe neurological disorder that typically manifests in infancy or early childhood and mitochondrial complex I deficiency is one of the most common biochemical defects observed [28]. Despite these associations, the potential implications of these variants in the context of reproduction, particularly male infertility, remain unexplored.
Regarding the genes on which the prioritized variants were mapped, NDUFA10 was identified as deregulated in the spermatozoa of first (F1) and second (F2) generation male mice following gestational bisphenol A (BPA) exposure, which was associated with decreased sperm count, motility, and intracellular ATP levels [29]. Similarly, another mouse study demonstrated that benzo[a]pyrene exposure inhibits testosterone through NDUFA10-mediated mitochondrial compromise in Leydig cells [30]. Additionally, NDUFA5 was also found to be downregulated in mouse testes after benzo[a]pyrene exposure [30]. However, BPA exposure significantly increased NDUFV2 in mouse testes, according to another study [31].
Furthermore, another interesting study in mice revealed a significant difference in the abundance of Ndufv3 between heavy (orthodox) and light (condensed) mitochondria in mouse testis, with condensed mitochondria originating from orthodox ones during meiosis and being essential for acrosomal matrix formation [32]. Lie et al. (2022) [33] reported also that reduced COX6C expression led to impaired COX enzyme activity, affecting mitochondrial ATP production and thus sperm motility in buffalo. Moreover, oxidative phosphorylation was found to be enriched in all undifferentiated spermatogonia subtypes studied, with significant differences in the relative abundances of Ndufv1 and Cox6c transcripts between control and E4f1-depleted spermatogonia, the latter condition leading to a progressive loss of undifferentiated spermatogonial cells [34]. Other studies highlighted the differential expression of COX6C, NDUFA5, NDUFS7, and NDUFV2 between progenitor and differentiating spermatogonia, too [35].
All the above findings indicate that OXPHOS genes play a pivotal role in the regulation of sperm motility, and these insights underline the importance of mitochondrial integrity in sperm development. Therefore, the study of the prioritized reported above could serve as a roadmap for future research.
Furthermore, seven variants were identified to create or disrupt miRNA binding sites, according to miRNASNP v3 [23]. MiRNAs typically bind to complementary sequences within the 3′ untranslated regions (3’ UTRs) of target messenger RNAs (mRNAs), leading to mRNA degradation or inhibition of translation and, thus, downregulation of gene expression. This interaction is highly sequence-specific; therefore, an SNP within the miRNA binding site can significantly alter miRNA binding affinity, resulting in disrupted gene regulation [36]. Although no studies have directly linked the variants reported in Table 7 with gene regulation through miRNAs in asthenozoospermia, several miRNAs presented in this table have been previously associated with male infertility.
Notably, miR-7-1-3p is upregulated in patients with idiopathic azoospermia [37], and a significant negative correlation between its expression levels and sperm concentration has been reported [38]. Similarly, miR-122 has been linked to infertility and identified as a potential sperm quality biomarker [39], with various studies supporting its crucial role in male infertility [38,40,41,42,43]. Furthermore, miR-21 has been shown to regulate the self-renewal of mouse spermatogonial stem cells [44], with potential implications for spermatogenesis also reported in other animal studies [45,46]. MiR-495, recognized also for its role in reproduction through various animal studies [47,48,49], is expressed in the testis [47] and has been associated with the progression of human pregnancy [50], as well as mitochondrial metabolism [51]. Additionally, miR-329 is expressed in rat Leydig cells and plays a role during development from the progenitor to the adult stage [52]. Finally, several miRNAs have been indicated to play a role in female fertility, such as miR-299 [53], the miR-548 family expressed in the female reproductive tract with various regulatory roles [54], miR-589 [55], and miR-1266 and miR-340 associated with pregnancy progression [50].
All the above indicate that miRNAs play a crucial role in male infertility and reproduction in general, and further studies are needed to decipher their specific interactions. It is also paramount for future studies to perform functional experiments to validate the interactions reported here as well as the impact of the reported SNPs in these interactions and to further elucidate the role of the reported miRNAs in conditions such as asthenozoospermia.
In discussing the limitations of our study, it is important to note that the research was conducted on a relatively small cohort. This cohort consisted of five individuals with asthenozoospermia and ten with normozoospermia. We acknowledge that the small sample size may limit the statistical power of our analyses and the generalizability of our findings. This is a critical aspect to consider when interpreting our results, as the conclusions drawn from a limited dataset may not fully represent broader populations. Therefore, we strongly encourage future research to involve larger and more diverse cohorts, which would help replicate and broaden our observations, thereby enhancing the reliability and applicability of our findings. However, it is worth noting that, despite the limited sample size, our study provides valuable preliminary insights into the distinct genomic profile between asthenozoospermia and normozoospermia. This is particularly significant considering the scarcity of studies that specifically examine these specific conditions using next-generation sequencing in this research area. Furthermore, much of the existing research on male infertility also involves relatively small cohorts, similar to ours [56,57,58]. This commonality underscores the broader challenge within the field and emphasizes the urgent need for larger-scale studies. From this perspective, our findings contribute meaningfully to the existing body of literature and serve as a starting point for more extensive future research. Furthermore, we primarily used bioinformatics approaches and in silico analyses to investigate the significance of SNPs within OXPHOS genes in relation to male infertility. Similarly, we employed computational algorithms to predict the interactions between mRNAs and miRNAs (miRNASNP v3 [23]). As these interactions were not validated through experimental procedures, it is possible that some of the miRNA interactions proposed in this study may not be influenced by SNPs in OXPHOS genes, as suggested. However, to mitigate these limitations, we employed whole genome sequencing, which, due to its comprehensive nature, facilitates the examination of the entire genome and provides a thorough overview of the genetic landscape. More importantly, the number of studies that utilize whole genome sequencing to investigate male infertility is limited. Additionally, we leveraged a broad spectrum of databases and varied analytical tools to bolster the robustness of our findings, including SNPnexus [19], RegulomeDB [13], and the GTEx portal [18], among others. We also adhered to stringent selection criteria, particularly with regard to RegulomeDB and 3DSNP scores, to enhance the reliability of our results. Finally, in several instances, we further validated our findings by concurrently utilizing pairs of databases, such as SIFT [11] and Polyphen2 [12], as well as RegulomeDB [13] and 3DSNP 2.0 [14], to ensure the accuracy and consistency of our conclusions.
Thus, regarding future directions, studies involving larger, more diverse cohorts are essential to validate our findings. Genome-wide association studies (GWAS) could also play a crucial role in determining whether the variants identified in this study are associated with asthenozoospermia. Additionally, it is of utmost importance to conduct functional experiments, potentially utilizing knockout models, in order to verify the impact of the reported variants on protein functionality and to comprehend their specific contributions to the asthenozoospermic phenotype. Further investigations are also required to elucidate the involvement of OXPHOS SNPs in male infertility, including an assessment of whether the mutations we have reported exhibit dominant or recessive inheritance patterns. Lastly, it is essential to experimentally validate the predicted interactions, such as those between miRNAs and mRNAs, in order to confirm their biological significance. It would also be interesting to investigate the role of other metabolic pathways, such as glycolysis, in future studies. These pathways are involved in energy production, and thus their disruption can also potentially affect sperm motility. It should also be noted that incorporating Computer-Assisted Sperm Analysis (CASA) and flow cytometry into future genetic studies on male infertility offers a promising avenue that could enhance our understanding of the complex mechanisms underlying this condition. CASA, with its advanced quantitative analysis capabilities, provides a comprehensive evaluation of sperm motility and kinematics. Meanwhile, flow cytometry introduces high-throughput analysis at the cellular level, enabling rapid assessment of sperm cell populations, detection of cellular heterogeneity, and detailed examination of phenotypic characteristics. These advanced techniques hold significant potential for genetic studies, where the complex interplay between genotype and phenotype demands a nuanced understanding of sperm function. They are especially particularly effective when analyzing large datasets, providing a level of detail and efficiency that traditional methods cannot match. However, we should keep in mind that these techniques require specific standardization protocols and extensive training to ensure reliable results.

4. Materials and Methods

4.1. Patient Recruitment

Human blood and sperm samples were collected from willing volunteers in partnership with the “Embryolab IVF Unit” in Thessaloniki, Greece, as part of the Spermogene research program. Ethical approval was granted by the University of Thessaly Ethics Committee in Volos, Greece, and all participants provided written informed consent. To gather comprehensive information, volunteers completed a questionnaire regarding their health, medical history, and other relevant details.
Exclusion criteria encompassed patients with varicocele, reproductive tract infections, testicular injuries or pathologies, a history of cryptorchidism, orchitis, epididymitis, as well as those with certain systemic diseases, Y microdeletions, or other chromosomal abnormalities. Additionally, all participants self-reported having Greek ethnicity.
All volunteers underwent an andrological examination, and semen analysis was conducted. Sperm samples were collected through masturbation following a minimum abstinence period of two to three days and these were allowed to liquefy at 37 °C for 30 min before analysis. Semen analyses were performed according to the fifth edition of the World Health Organization (WHO) manual for the examination and processing of human semen (available at: https://apps.who.int/iris/handle/10665/44261 (accessed on 5 March 2024)). More specifically, the main analysis (seminogram) was performed using the cell vision counting slides (Tek-Event) for cell counting and observation on Nikon Eclipse TS100, Nikon Eclipse E200, and Nikon Eclipse Ts2 microscopes (Nikon Instruments, Tokyo, Japan). Furthermore, all semen analyses were performed by the same laboratory, undergoing regular internal and external quality controls. The reference values from this edition were used to categorize individuals into normozoospermic and asthenozoospermic phenotypes.

4.2. DNA Extraction and Sample Preparation

Genomic DNA was isolated from whole EDTA blood samples using the PureLink Genomic DNA Mini Kit (Invitrogen, Waltham, MA, USA—Catalog number: K182002) following the manufacturer’s instructions. DNA concentration was determined using the Qubit 2.0 fluorometer with the Qubit dsDNA BR Assay Kit (Invitrogen, Waltham, MA, USA—Catalog number: Q32850). The DNA quality was also accessed through agarose gel electrophoresis.
For sample preparation for WGS, three sequencing pools were created. Specifically, DNA obtained from ten normozoospermic individuals was divided into two pools, each comprising DNA from five individuals. Additionally, a third pool was generated using DNA from five asthenozoospermic individuals. The DNAs in each pool were mixed equimolar, resulting in a final concentration of 100 ng/uL and a total quantity of 2 mg.

4.3. Whole Genome Sequencing (WGS)

Following the sample preparation, whole genome sequencing was performed by Novogene (Cambridge, UK). Genomic DNA prepared as explained above was utilized to construct libraries for WGS. After normalization and stringent quality control, the qualified libraries were sequenced on an Illumina HiSeq 3000 platform (Illumina Inc., San Diego, CA, USA) as 100-bp paired-end reads. The average sequencing coverage achieved was 30×.
Standard bioinformatics analysis was conducted on the sequencing data. Initially, the quality of the reads was assessed using FASTQC (available online at: http://www.bioinformatics.babraham.ac.uk/projects/fastqc/ (accessed on 5 March 2024)), followed by the removal of low-quality reads (with a minimum PHRED score of 30) and adapter sequences using Trimmomatic (v0.39) [59]. The remaining reads were aligned to the human reference genome (GRCh37/hg19) obtained from the Ensembl database (accessed on 5 March 2024) [60], employing the Burrows-Wheeler aligner (BWA) [61]. Duplicate reads resulting from polymerase chain reaction (PCR) were eliminated using Picard tools, and subsequently, SAM files were converted to BAM files with SAMtools (v1.19.2) [62]. At this point, individual BAM files for normozoospermic pools were merged into a single file. Following this, variant calling was performed using freeBayes (v1.3.7) [63], and the results were stored in variant call format (VCF). BCFtools (v1.17) [62] was utilized to compare VCF files from normozoospermic and asthenozoospermic individuals to identify unique variants specific to each group. Finally, the unique variants were annotated using the VEP tool (available at: https://www.ensembl.org/Tools/VEP (accessed on 5 March 2024)) to gather biological information and predict their effects on protein functionality and pathogenicity.

4.4. Investigation of Unique Mutations in OXPHOS Genes—Bioinformatics Approach and Tools

Among the unique variants exclusively found in asthenozoospermic individuals after WGS, those mapped to OXPHOS genes were selected and categorized by type (nuclear or mitochondrial encoded) and OXPHOS complexes (I–V).
Evaluation of these mutations extended to understanding their impact on OXPHOS function and their potential role in asthenozoospermia using various databases. Specifically, RegulomeDB (accessed on 5 March 2024) [13] and 3DSNP 2.0 [14] databases were utilized to assess potential functional effects. RegulomeDB [13] categorizes SNPs based on the presence or absence of functional elements, encompassing protein binding sites, chromatin structure, eQTL (expression quantitative trait loci), histone modifications, and more. Each SNP receives a ranking on a scale from 1 to 7, with lower values signifying a higher likelihood of having a regulatory function. Similarly, the 3DSNP 2.0 [14] is a comprehensive database that consolidates data about 3D-interacting genes, enhancer states, transcription factor binding sites, etc. This information is used to compute a functional score for each SNP, with higher scores signaling a greater probability of SNP functionality. Additionally, information on population genetics was gathered from the 1000 Genomes Project [64], gnomAD [65], and NCBI ALFA (Allele Frequency Aggregator) [66]. SNPnexus [19] provided details about the consequences of unique variants on OXPHOS genes (e.g., intergenic, synonymous, missense, etc.) and their previous associations with other diseases. Additionally, for assessing the impact on protein functionality, Polymorphism Phenotyping v2 (PolyPhen2) [12] and Sorting Intolerant From Tolerant (SIFT) [11] tools were employed. Information on expression quantitative trait loci (eQTL) and splicing quantitative trait loci (sQTL) were obtained through the Genotype-Tissue Expression Project (GTEx) [18]. Finally, miRNASNP v3 [23] was utilized to determine whether the identified variants influenced the binding of noncoding RNAs, potentially affecting the regulation of genes crucial for the OXPHOS machinery.

5. Conclusions

In summary, this investigation stands as the first comprehensive study to utilize whole genome sequencing for examining OXPHOS genes and their variants in the context of asthenozoospermia. By identifying 309 OXPHOS gene variants exclusively in asthenozoospermic men, our research offers a pioneering roadmap for future studies on the genetic basis of male infertility. More specifically, the extensive dataset of variants provides invaluable insights into the genetic landscape of asthenozoospermia and the significance of these variants by using different tools and databases. Finally, it not only enhances our understanding of the condition but also sets the foundations for advancements in assisted reproductive technologies (ART) and the development of diagnostic and prognostic tools, as the diverse range of previously unexplored variants identified here presents a rich resource for further study, potentially serving as biomarkers for the diagnosis of male infertility.

Supplementary Materials

The following supporting information can be downloaded at: https://www.mdpi.com/article/10.3390/ijms25074121/s1.

Author Contributions

Conceptualization, Z.M. and M.-A.K.; methodology, Z.M. and M.-A.K., formal analysis, M.-A.K. and T.G.; data interpretation, M.-A.K. and Z.M.; data acquisition, A.C.; writing—original draft preparation, M.-A.K.; writing—review and editing, Z.M.; visualization, M.-A.K., supervision, Z.M.; project administration, Z.M.; funding acquisition, Z.M. All authors have read and agreed to the published version of the manuscript.

Funding

This research was funded by the Spermogene (Fertilaid) project which is co-financed by the European Regional Development Fund of the European Union and Greek national funds through the Operational Program Competitiveness, Entrepreneurship, and Innovation, under the call RESEARCH–CREATE–INNOVATE (Grant number T1EΔK-02787).

Institutional Review Board Statement

The study was conducted in accordance with the Declaration of Helsinki and approved by the ethics committee of the Medical Faculty of the University of Thessaly (protocol code 20.04/2016 and date of approval 20 April 2016).

Informed Consent Statement

Informed consent was obtained from all subjects involved in this study.

Data Availability Statement

Whole-genome sequencing data of normozoospermic men used in this study are available through SRA (BioProject ID PRJNA875412, http://www.ncbi.nlm.nih.gov/bioproject/875412 (accessed on 5 March 2024)).

Conflicts of Interest

The authors declare no conflicts of interest.

References

  1. Fainberg, J.; Kashanian, J.A. Recent Advances in Understanding and Managing Male Infertility. F1000Research 2019, 8, 670. [Google Scholar] [CrossRef] [PubMed]
  2. Eisenberg, M.L.; Esteves, S.C.; Lamb, D.J.; Hotaling, J.M.; Giwercman, A.; Hwang, K.; Cheng, Y.-S. Male Infertility. Nat. Rev. Dis. Prim. 2023, 9, 49. [Google Scholar] [CrossRef] [PubMed]
  3. Kothandaraman, N.; Agarwal, A.; Abu-Elmagd, M.; Al-Qahtani, M.H. Pathogenic Landscape of Idiopathic Male Infertility: New Insight towards Its Regulatory Networks. NPJ Genom. Med. 2016, 1, 16023. [Google Scholar] [CrossRef] [PubMed]
  4. Sudhakar, D.V.S.; Shah, R.; Gajbhiye, R.K. Genetics of Male Infertility—Present and Future: A Narrative Review. J. Hum. Reprod. Sci. 2021, 14, 217–227. [Google Scholar] [CrossRef] [PubMed]
  5. Moraes, C.R.; Meyers, S. The Sperm Mitochondrion: Organelle of Many Functions. Anim. Reprod. Sci. 2018, 194, 71–80. [Google Scholar] [CrossRef] [PubMed]
  6. Ford, W.C.L. Glycolysis and Sperm Motility: Does a Spoonful of Sugar Help the Flagellum Go Round? Hum. Reprod. Update 2006, 12, 269–274. [Google Scholar] [CrossRef] [PubMed]
  7. Ferramosca, A.; Zara, V. Bioenergetics of Mammalian Sperm Capacitation. BioMed Res. Int. 2014, 2014, 902953. [Google Scholar] [CrossRef] [PubMed]
  8. Maurya, S.; Kesari, K.K.; Roychoudhury, S.; Kolleboyina, J.; Jha, N.K.; Jha, S.K.; Sharma, A.; Kumar, A.; Rathi, B.; Kumar, D. Metabolic Dysregulation and Sperm Motility in Male Infertility. Adv. Exp. Med. Biol. 2022, 1358, 257–273. [Google Scholar] [CrossRef] [PubMed]
  9. Du Plessis, S.S.; Agarwal, A.; Mohanty, G.; Van Der Linde, M. Oxidative Phosphorylation versus Glycolysis: What Fuel Do Spermatozoa Use? Asian J. Androl. 2015, 17, 230–235. [Google Scholar] [CrossRef] [PubMed]
  10. Signes, A.; Fernandez-Vizarra, E. Assembly of Mammalian Oxidative Phosphorylation Complexes I–V and Supercomplexes. Essays Biochem. 2018, 62, 255–270. [Google Scholar] [PubMed]
  11. Vaser, R.; Adusumalli, S.; Ngak Leng, S.; Sikic, M.; Ng, P.C. SIFT Missense Predictions for Genomes. Nat. Protoc. 2015, 11, 1–9. [Google Scholar] [CrossRef] [PubMed]
  12. Adzhubei, I.A.; Schmidt, S.; Peshkin, L.; Ramensky, V.E.; Gerasimova, A.; Bork, P.; Kondrashov, A.S.; Sunyaev, S.R. A Method and Server for Predicting Damaging Missense Mutations. Nat. Methods 2010, 7, 248–249. [Google Scholar] [CrossRef] [PubMed]
  13. Boyle, A.P.; Hong, E.L.; Hariharan, M.; Cheng, Y.; Schaub, M.A.; Kasowski, M.; Karczewski, K.J.; Park, J.; Hitz, B.C.; Weng, S.; et al. Annotation of Functional Variation in Personal Genomes Using RegulomeDB. Genome Res. 2012, 22, 1790–1797. [Google Scholar] [CrossRef] [PubMed]
  14. Lu, Y.; Quan, C.; Chen, H.; Bo, X.; Zhang, C. 3DSNP: A Database for Linking Human Noncoding SNPs to Their Three-Dimensional Interacting Genes. Nucleic Acids Res. 2017, 45, D643–D649. [Google Scholar] [CrossRef] [PubMed]
  15. Westra, H.J.; Peters, M.J.; Esko, T.; Yaghootkar, H.; Schurmann, C.; Kettunen, J.; Christiansen, M.W.; Fairfax, B.P.; Schramm, K.; Powell, J.E.; et al. Systematic Identification of Trans EQTLs as Putative Drivers of Known Disease Associations. Nat. Genet. 2013, 45, 1238–1243. [Google Scholar] [CrossRef] [PubMed]
  16. Ardlie, K.G.; DeLuca, D.S.; Segrè, A.V.; Sullivan, T.J.; Young, T.R.; Gelfand, E.T.; Trowbridge, C.A.; Maller, J.B.; Tukiainen, T.; Lek, M.; et al. Human Genomics. The Genotype-Tissue Expression (GTEx) Pilot Analysis: Multitissue Gene Regulation in Humans. Science 2015, 348, 648–660. [Google Scholar] [CrossRef]
  17. Yamaguchi, K.; Ishigaki, K.; Suzuki, A.; Tsuchida, Y.; Tsuchiya, H.; Sumitomo, S.; Nagafuchi, Y.; Miya, F.; Tsunoda, T.; Shoda, H.; et al. Splicing QTL Analysis Focusing on Coding Sequences Reveals Mechanisms for Disease Susceptibility Loci. Nat. Commun. 2022, 13, 4659. [Google Scholar] [CrossRef] [PubMed]
  18. Lonsdale, J.; Thomas, J.; Salvatore, M.; Phillips, R.; Lo, E.; Shad, S.; Hasz, R.; Walters, G.; Garcia, F.; Young, N.; et al. The Genotype-Tissue Expression (GTEx) Project. Nat. Genet. 2013, 45, 580. [Google Scholar] [CrossRef] [PubMed]
  19. Oscanoa, J.; Sivapalan, L.; Gadaleta, E.; Dayem Ullah, A.Z.; Lemoine, N.R.; Chelala, C. SNPnexus: A Web Server for Functional Annotation of Human Genome Sequence Variation (2020 Update). Nucleic Acids Res. 2020, 48, W185–W192. [Google Scholar] [CrossRef]
  20. Naeli, P.; Winter, T.; Hackett, A.P.; Alboushi, L.; Jafarnejad, S.M. The Intricate Balance between MicroRNA-Induced MRNA Decay and Translational Repression. FEBS J. 2023, 290, 2508–2524. [Google Scholar] [CrossRef] [PubMed]
  21. O’Brien, J.; Hayder, H.; Zayed, Y.; Peng, C. Overview of MicroRNA Biogenesis, Mechanisms of Actions, and Circulation. Front. Endocrinol. 2018, 9, 402. [Google Scholar] [CrossRef] [PubMed]
  22. Xi, E.; Bai, J.; Zhang, K.; Yu, H.; Guo, Y. Genomic Variants Disrupt MiRNA-MRNA Regulation. Chem. Biodivers. 2022, 19, e202200623. [Google Scholar] [CrossRef] [PubMed]
  23. Liu, C.J.; Fu, X.; Xia, M.; Zhang, Q.; Gu, Z.; Guo, A.Y. MiRNASNP-v3: A Comprehensive Database for SNPs and Disease-Related Variations in MiRNAs and MiRNA Targets. Nucleic Acids Res. 2021, 49, D1276–D1281. [Google Scholar] [CrossRef] [PubMed]
  24. Barbagallo, F.; La Vignera, S.; Cannarella, R.; Aversa, A.; Calogero, A.E.; Condorelli, R.A. Evaluation of Sperm Mitochondrial Function: A Key Organelle for Sperm Motility. J. Clin. Med. 2020, 9, 363. [Google Scholar] [CrossRef] [PubMed]
  25. Holyoake, A.J.; McHugh, P.; Wu, M.; O’Carroll, S.; Benny, P.; Sin, I.L.; Sin, F.Y.T. High Incidence of Single Nucleotide Substitutions in the Mitochondrial Genome Is Associated with Poor Semen Parameters in Men. Int. J. Androl. 2001, 24, 175–182. [Google Scholar] [CrossRef] [PubMed]
  26. Shamsi, M.B.; Kumar, R.; Bhatt, A.; Bamezai, R.N.K.; Kumar, R.; Gupta, N.P.; Das, T.K.; Dada, R. Mitochondrial DNA Mutations in Etiopathogenesis of Male Infertility. Indian J. Urol. 2008, 24, 150–154. [Google Scholar] [PubMed]
  27. Du Plessis, S.S.; Agarwal, A.; Halabi, J.; Tvrda, E. Contemporary Evidence on the Physiological Role of Reactive Oxygen Species in Human Sperm Function. J. Assist. Reprod. Genet. 2015, 32, 509–520. [Google Scholar] [CrossRef] [PubMed]
  28. Lake, N.J.; Bird, M.J.; Isohanni, P.; Paetau, A. Leigh Syndrome: Neuropathology and Pathogenesis. J. Neuropathol. Exp. Neurol. 2015, 74, 482–492. [Google Scholar] [CrossRef] [PubMed]
  29. Rahman, M.S.; Kwon, W.S.; Karmakar, P.C.; Yoon, S.J.; Ryu, B.Y.; Pang, M.G. Gestational Exposure to Bisphenol A Affects the Function and Proteome Profile of F1 Spermatozoa in Adult Mice. Environ. Health Perspect. 2017, 125, 238–245. [Google Scholar] [CrossRef] [PubMed]
  30. Yang, W.; Cui, H.; Chai, Z.; Zou, P.; Shi, F.; Yang, B.; Zhang, G.; Yang, H.; Chen, Q.; Liu, J.; et al. Benzo[a]Pyrene Inhibits Testosterone Biosynthesis via NDUFA10-Mediated Mitochondrial Compromise in Mouse Leydig Cells: Integrating Experimental and in Silico Toxicological Approaches. Ecotoxicol. Environ. Saf. 2022, 244, 114075. [Google Scholar] [CrossRef]
  31. Ryu, D.Y.; Pang, W.K.; Adegoke, E.O.; Rahman, M.S.; Park, Y.J.; Pang, M.G. Bisphenol-A Disturbs Hormonal Levels and Testis Mitochondrial Activity, Reducing Male Fertility. Hum. Reprod. Open 2023, 2023, hoad044. [Google Scholar] [CrossRef] [PubMed]
  32. Ren, M.; Xu, Y.; Phoon, C.K.L.; Erdjument-Bromage, H.; Neubert, T.A.; Rajan, S.; Hussain, M.M.; Schlame, M. Condensed Mitochondria Assemble Into the Acrosomal Matrix During Spermiogenesis. Front. Cell Dev. Biol. 2022, 10, 867175. [Google Scholar] [CrossRef]
  33. Liu, R.; Huang, X.; Sun, Q.; Hou, Z.; Yang, W.; Zhang, J.; Zhang, P.; Huang, L.; Lu, Y.; Fu, Q. Comparative Proteomic Analyses of Poorly Motile Swamp Buffalo Spermatozoa Reveal Low Energy Metabolism and Deficiencies in Motility-Related Proteins. Animals 2022, 12, 1706. [Google Scholar] [CrossRef] [PubMed]
  34. Yan, R.-G.; He, Z.; Wang, F.-C.; Li, S.; Shang, Q.-B.; Yang, Q.-E. Transcription Factor E4F1 Dictates Spermatogonial Stem Cell Fate Decisions by Regulating Mitochondrial Functions and Cell Cycle Progression. Cell Biosci. 2023, 13, 177. [Google Scholar] [CrossRef] [PubMed]
  35. Lord, T.; Nixon, B. Metabolic Changes Accompanying Spermatogonial Stem Cell Differentiation. Dev. Cell 2020, 52, 399–411. [Google Scholar] [CrossRef] [PubMed]
  36. Moszyńska, A.; Gebert, M.; Collawn, J.F.; Bartoszewski, R. SNPs in MicroRNA Target Sites and Their Potential Role in Human Disease. Open Biol. 2017, 7, 170019. [Google Scholar] [CrossRef] [PubMed]
  37. Wu, W.; Qin, Y.; Li, Z.; Dong, J.; Dai, J.; Lu, C.; Guo, X.; Zhao, Y.; Zhu, Y.; Zhang, W.; et al. Genome-Wide MicroRNA Expression Profiling in Idiopathic Non-Obstructive Azoospermia: Significant up-Regulation of MiR-141, MiR-429 and MiR-7-1-3p. Hum. Reprod. 2013, 28, 1827–1836. [Google Scholar] [CrossRef] [PubMed]
  38. Mokánszki, A.; Molnár, Z.; Varga Tóthné, E.; Bodnár, B.; Jakab, A.; Bálint, B.L.; Balogh, I. Altered MicroRNAs Expression Levels of Sperm and Seminal Plasma in Patients with Infertile Ejaculates Compared with Normozoospermic Males. Hum. Fertil. 2020, 23, 246–255. [Google Scholar] [CrossRef] [PubMed]
  39. Joshi, M.; Andrabi, S.W.; Yadav, R.K.; Sankhwar, S.N.; Gupta, G.; Rajender, S. Qualitative and Quantitative Assessment of Sperm MiRNAs Identifies Hsa-MiR-9-3p, Hsa-MiR-30b-5p and Hsa-MiR-122-5p as Potential Biomarkers of Male Infertility and Sperm Quality. Reprod. Biol. Endocrinol. 2022, 20, 122. [Google Scholar] [CrossRef] [PubMed]
  40. Rahbar, S.; Pashaiasl, M.; Ezzati, M.; Ahmadi AsrBadr, Y.; Mohammadi-Dehcheshmeh, M.; Mohammadi, S.A.; Ghaffari Novin, M. MicroRNA-Based Regulatory Circuit Involved in Sperm Infertility. Andrologia 2020, 52, e13453. [Google Scholar] [CrossRef] [PubMed]
  41. Abu-Halima, M.; Hammadeh, M.; Schmitt, J.; Leidinger, P.; Keller, A.; Meese, E.; Backes, C. Altered MicroRNA Expression Profiles of Human Spermatozoa in Patients with Different Spermatogenic Impairments. Fertil. Steril. 2013, 99, P1249–P1255. [Google Scholar] [CrossRef] [PubMed]
  42. Liu, T.; Huang, Y.; Liu, J.; Zhao, Y.; Jiang, L.; Huang, Q.; Cheng, W.; Guo, L. MicroRNA-122 Influences the Development of Sperm Abnormalities from Human Induced Pluripotent Stem Cells by Regulating TNP2 Expression. Stem Cells Dev. 2013, 22, 1839–1850. [Google Scholar] [CrossRef] [PubMed]
  43. Abu-Halima, M.; Hammadeh, M.; Backes, C.; Fischer, U.; Leidinger, P.; Lubbad, A.M.; Keller, A.; Meese, E. Panel of Five MicroRNAs as Potential Biomarkers for the Diagnosis and Assessment of Male Infertility. Fertil. Steril. 2014, 102, 989–997.e1. [Google Scholar] [CrossRef] [PubMed]
  44. Niu, Z.; Goodyear, S.M.; Rao, S.; Wu, X.; Tobias, J.W.; Avarbock, M.R.; Brinster, R.L. MicroRNA-21 Regulates the Self-Renewal of Mouse Spermatogonial Stem Cells. Proc. Natl. Acad. Sci. USA 2011, 108, 12740–12745. [Google Scholar] [CrossRef] [PubMed]
  45. Xu, Z.; Xie, Y.; Zhou, C.; Hu, Q.; Gu, T.; Yang, J.; Zheng, E.; Huang, S.; Xu, Z.; Cai, G.; et al. Expression Pattern of Seminal Plasma Extracellular Vesicle Small RNAs in Boar Semen. Front. Vet. Sci. 2020, 7, 585276. [Google Scholar] [CrossRef] [PubMed]
  46. Wu, J.; Liao, M.Z.; Zhu, H.J.; Kang, K.; Mu, H.L.; Song, W.C.; Niu, Z.W.; He, X.; Bai, C.L.; Li, G.P.; et al. CD49f-Positive Testicular Cells in Saanen Dairy Goat Were Identified as Spermatogonia-Like Cells by MiRNA Profiling Analysis. J. Cell. Biochem. 2014, 115, 1712–1723. [Google Scholar] [CrossRef] [PubMed]
  47. Salas-Huetos, A.; Ribas-Maynou, J.; Mateo-Otero, Y.; Tamargo, C.; Llavanera, M.; Yeste, M. Expression of MiR-138 in Cryopreserved Bovine Sperm Is Related to Their Fertility Potential. J. Anim. Sci. Biotechnol. 2023, 14, 129. [Google Scholar] [CrossRef]
  48. Zhang, Z.B.; Tang, J.S.; Di, R.; Liu, Q.Y.; Wang, X.Y.; Gan, S.Q.; Zhang, X.S.; Zhang, J.L.; Chu, M.X.; Hu, W.P. Integrated Hypothalamic Transcriptome Profiling Reveals the Reproductive Roles of MRNAs and MiRNAs in Sheep. Front. Genet. 2020, 10, 1296. [Google Scholar] [CrossRef]
  49. Gebremedhn, S.; Salilew-Wondim, D.; Hoelker, M.; Held-Hoelker, E.; Neuhoff, C.; Tholen, E.; Schellander, K.; Tesfaye, D. Exploring Maternal Serum MicroRNAs during Early Pregnancy in Cattle. Theriogenology 2018, 121, 196–203. [Google Scholar] [CrossRef]
  50. Vashukova, E.S.; Kozyulina, P.Y.; Illarionov, R.A.; Yurkina, N.O.; Pachuliia, O.V.; Butenko, M.G.; Postnikova, T.B.; Ivanova, L.A.; Eremeeva, D.R.; Zainulina, M.S.; et al. High-Throughput Sequencing of Circulating MicroRNAs in Plasma and Serum during Pregnancy Progression. Life 2021, 11, 1055. [Google Scholar] [CrossRef]
  51. Miguel, V.; Ramos, R.; García-Bermejo, L.; Rodríguez-Puyol, D.; Lamas, S. The Program of Renal Fibrogenesis Is Controlled by MicroRNAs Regulating Oxidative Metabolism. Redox Biol. 2021, 40, 101851. [Google Scholar] [CrossRef] [PubMed]
  52. Chen, H.; Guo, X.; Xiao, X.; Ye, L.; Huang, Y.; Lu, C.; Su, Z. Identification and Functional Characterization of MicroRNAs in Rat Leydig Cells during Development from the Progenitor to the Adult Stage. Mol. Cell. Endocrinol. 2019, 493, 110453. [Google Scholar] [CrossRef] [PubMed]
  53. Xu, Y.W.; Wang, B.; Ding, C.H.; Li, T.; Gu, F.; Zhou, C. Differentially Expressed MicoRNAs in Human Oocytes. J. Assist. Reprod. Genet. 2011, 28, 559–566. [Google Scholar] [CrossRef] [PubMed]
  54. Rooda, I.; Kaselt, B.; Liivrand, M.; Smolander, O.P.; Salumets, A.; Velthut-Meikas, A. Hsa-Mir-548 Family Expression in Human Reproductive Tissues. BMC Genom. Data 2021, 22, 40. [Google Scholar] [CrossRef] [PubMed]
  55. Li, D.; Xu, W.; Wang, X.; Dang, Y.; Xu, L.; Lu, G.; Chan, W.Y.; Leung, P.C.K.; Zhao, S.; Qin, Y. LncRNA DDGC Participates in Premature Ovarian Insufficiency through Regulating RAD51 and WT1. Mol. Ther. Nucleic Acids 2021, 26, 1092–1106. [Google Scholar] [CrossRef] [PubMed]
  56. Sun, T.C.; Zhang, Y.; Yu, K.; Li, Y.; Yu, H.; Zhou, S.J.; Wang, Y.P.; Deng, S.L.; Tian, L. LncRNAs Induce Oxidative Stress and Spermatogenesis by Regulating Endoplasmic Reticulum Genes and Pathways. Aging 2021, 13, 13764–13787. [Google Scholar] [CrossRef]
  57. Lü, M.; Tian, H.; Cao, Y.X.; He, X.; Chen, L.; Song, X.; Ping, P.; Huang, H.; Sun, F. Downregulation of MiR-320a/383-Sponge-like Long Non-Coding RNA NLC1-C (Narcolepsy Candidate-Region 1 Genes) Is Associated with Male Infertility and Promotes Testicular Embryonal Carcinoma Cell Proliferation. Cell Death Dis. 2015, 6, E1960. [Google Scholar] [CrossRef] [PubMed]
  58. Kyrgiafini, M.A.; Giannoulis, T.; Chatziparasidou, A.; Christoforidis, N.; Mamuris, Z. Unveiling the Genetic Complexity of Teratozoospermia: Integrated Genomic Analysis Reveals Novel Insights into LncRNAs’ Role in Male Infertility. Int. J. Mol. Sci. 2023, 24, 15002. [Google Scholar] [CrossRef] [PubMed]
  59. Bolger, A.M.; Lohse, M.; Usadel, B. Trimmomatic: A Flexible Trimmer for Illumina Sequence Data. Bioinformatics 2014, 30, 2114–2120. [Google Scholar] [CrossRef] [PubMed]
  60. Martin, F.J.; Amode, M.R.; Aneja, A.; Austine-Orimoloye, O.; Azov, A.G.; Barnes, I.; Becker, A.; Bennett, R.; Berry, A.; Bhai, J.; et al. Ensembl 2023. Nucleic Acids Res. 2023, 51, D933–D941. [Google Scholar] [CrossRef] [PubMed]
  61. Li, H.; Durbin, R. Fast and Accurate Short Read Alignment with Burrows-Wheeler Transform. Bioinformatics 2009, 25, 1754–1760. [Google Scholar] [CrossRef] [PubMed]
  62. Danecek, P.; Bonfield, J.K.; Liddle, J.; Marshall, J.; Ohan, V.; Pollard, M.O.; Whitwham, A.; Keane, T.; McCarthy, S.A.; Davies, R.M.; et al. Twelve Years of SAMtools and BCFtools. Gigascience 2021, 10, 1–4. [Google Scholar] [CrossRef] [PubMed]
  63. Garrison, E.; Marth, G. Haplotype-Based Variant Detection from Short-Read Sequencing. arXiv 2012, arXiv:1207.3907. [Google Scholar]
  64. Auton, A.; Abecasis, G.R.; Altshuler, D.M.; Durbin, R.M.; Bentley, D.R.; Chakravarti, A.; Clark, A.G.; Donnelly, P.; Eichler, E.E.; Flicek, P.; et al. A Global Reference for Human Genetic Variation. Nature 2015, 526, 68–74. [Google Scholar] [CrossRef] [PubMed]
  65. Chen, S.; Francioli, L.C.; Goodrich, J.K.; Collins, R.L.; Wang, Q.; Alföldi, J.; Watts, N.A.; Vittal, C.; Gauthier, L.D.; Poterba, T.; et al. A Genome-Wide Mutational Constraint Map Quantified from Variation in 76,156 Human Genomes. bioRxiv 2022. [Google Scholar] [CrossRef]
  66. Phan, L.; Jin, Y.; Zhang, H.; Qiang, W.; Shekhtman, E.; Shao, D.; Revoe, D.; Villamarin, R.; Ivanchenko, E.; Kimura, M.; et al. ALFA: Allele Frequency Aggregator. Available online: www.ncbi.nlm.nih.gov/snp/docs/gsr/alfa/ (accessed on 5 March 2024).
Figure 1. Unique OXPHOS variants in asthenozoospermic men and their genomic consequences.
Figure 1. Unique OXPHOS variants in asthenozoospermic men and their genomic consequences.
Ijms 25 04121 g001
Table 1. Analysis of unique OXPHOS variants in asthenozoospermic men: Distribution across mitochondrial respiratory complexes and associated genes, gene length to variant ratio, and proportion of OXPHOS variants to total variants.
Table 1. Analysis of unique OXPHOS variants in asthenozoospermic men: Distribution across mitochondrial respiratory complexes and associated genes, gene length to variant ratio, and proportion of OXPHOS variants to total variants.
GenesVariant
Number
Length of Gene/
Variant Number (%)
OXPHOS Variants/Total Variants
in Asthenozoospermic (%)
Mitochondrial Respiratory Complex I
NDUFS130.00670.0004
NDUFS230.01700.0004
NDUFS310.00520.0001
NDUFS770.05800.0010
NDUFV110.01450.0001
NDUFV250.05800.0007
MT-ND210.09610.0001
MT-ND5100.55220.0001
MT-ND610.19080.0001
NDUFAB130.01960.0004
NDUFA530.01430.0004
NDUFA830.01910.0004
NDUFA980.01770.0012
NDUFA10420.31510.0062
NDUFA1110.00790.0001
NDUFA12300.02770.0044
NDUFA1320.01500.0003
NDUFB150.08630.0007
NDUFB240.01250.0006
NDUFB340.02790.0006
NDUFB420.03240.0003
NDUFB520.00870.0003
NDUFB630.01490.0004
NDUFB820.03190.0003
NDUFB9100.02410.0015
NDUFB1010.04100.0001
NDUFC120.00560.0003
NDUFC220.01730.0003
NDUFS4110.00900.0016
NDUFS550.06010.0007
NDUFS610.00680.0001
NDUFV350.01490.0007
Mitochondrial Respiratory Complex II
SDHA30.00770.0004
SDHB80.02260.0012
SDHC110.02250.0016
SDHD480.14460.0071
Mitochondrial Respiratory Complex III
UQCRC240.01320.0006
MT-CYB20.17540.0003
Mitochondrial Respiratory Complex IV
COX5A10.00550.0001
COX6B180.07650.0012
COX6C40.01950.0006
COX7B2340.01950.0050
MT-CO220.29280.0003
MT-CO310.12770.0001
Table 2. Distribution of unique variants in nuclear- and mitochondrial-encoded OXPHOS genes.
Table 2. Distribution of unique variants in nuclear- and mitochondrial-encoded OXPHOS genes.
Mitochondrial Respiratory Complex I
Variants in mitochondrial-encoded genes12
Variants in nuclear-encoded genes171
Mitochondrial Respiratory Complex II
Variants in mitochondrial-encoded genes0
Variants in nuclear-encoded genes70
Mitochondrial Respiratory Complex III
Variants in mitochondrial-encoded genes2
Variants in nuclear-encoded genes4
Mitochondrial Respiratory Complex IV
Variants in mitochondrial-encoded genes3
Variants in nuclear-encoded genes47
Table 3. SIFT [11] and Polyphen2 [12] scores of missense OXPHOS variants identified in asthenozoospermic men.
Table 3. SIFT [11] and Polyphen2 [12] scores of missense OXPHOS variants identified in asthenozoospermic men.
Genomic
Coordinates
AlleleAllele
Frequency
(Europeans)
VariantGeneSIFT ScorePolyphen2 Score
MT:12406-12406A0.2%rs28617389MT-ND50.45 (tolerated)0 (benign)
MT:13708-13708A11.6%rs28359178MT-ND50.26 (tolerated)0 (benign)
MT:13780-13780G2.9%rs41358152MT-ND50.01 (deleterious)0.003 (benign)
MT:13928-13928C0.2%rs28359184MT-ND51 (tolerated)0.18 (benign)
MT:14178-14178C0.2%rs28357671MT-ND60.4 (tolerated)0.023 (benign)
MT:14793-14793G3.7%rs2853504MT-CYB0.04 (deleterious)0.003 (benign)
MT:9477-9477A8.5%rs2853825MT-CO30.1 (tolerated)0 (benign)
2:240951071-240951071T1.1%rs35462421NDUFA100.01 (deleterious)0.995 (probably damaging)
16:21976762-21976762A4.3%rs4850UQCRC20.04 (deleterious)0.003 (benign)
5:52942083-52942083C96%rs31304NDUFS4-0 (unknown)
Table 4. Unique OXPHOS variants in asthenozoospermic men with potential functional significance according to RegulomeDB [13] and 3DSNP 2.0 [14] databases.
Table 4. Unique OXPHOS variants in asthenozoospermic men with potential functional significance according to RegulomeDB [13] and 3DSNP 2.0 [14] databases.
Genomic
Coordinates
AlleleAllele
Frequency
(Europeans)
VariantGeneGenomic
Consequences
RegulomeDB Rank3DSNP Score
19:1394865-1394865C2.1%rs73515054NDUFS73′ UTR variant, intron variant2b13.72
9:124897110-124897110T8.7%rs11998959NDUFA8Intron variant1f36.76
9:124897088-124897088T8.3%rs11998958NDUFA8Intron variant1f36.36
7:123197559-123197559C8.6%rs17146099NDUFA55′ UTR variant, intron variant1f146.4
2:240897460-240897460C3.5%rs7588974NDUFA103′ UTR variant, intron variant2b10.56
16:2011653-2011667CCCCCA0.03%rs774819361NDUFB10Intron variant2a103.27
8:125551858-125551858G3.5%rs72713101NDUFB9Intron variant1f108.59
8:125554452-125554452T3.3%rs111795428NDUFB9Intron variant1f11.6
8:125552526-125552527-3.3%rs112295879NDUFB9Intron variant1b116.14
11:77790158-77790158AAAAA0.1%rs752264424NDUFC2Intron variant2b104.37
1:161175652-161175652A1.8%rs145629160NDUFS2Intron variant1f13.25
1:161171736-161171736G1.8%rs115518404NDUFS2Intron variant1b146.84
21:44313221-44313221C20.2%rs35197797NDUFV3Intron variant1a211.4
8:100903890-100903890G14.1%rs12544943COX6CIntron variant1f66.33
11:67374581-67374581C38.2%rs1871043NDUFV1Intron variant1f208.1
18:9119489-9119489T9.1%rs41274300NDUFV2Synonymous variant1f28.68
14:92586558-92586558A16.3%rs79507139NDUFB1Intron variant1f16.69
12:95376507-95376507T9.2%rs4923659NDUFA12Intron variant1b16.37
12:95371804-95371806-9.2%rs113060515NDUFA12Intron variant1f13.75
12:95374449-95374449C9.2%rs76835653NDUFA12Intron variant1b59.71
12:95397275-95397275T10.1%rs17321986NDUFA12Intron variant1b201
11:112044398-112044398C22.9%rs12420476SDHDIntron variant1f11.55
11:112034062-112034063AA22%rs5744230SDHDIntron variant1d33.1
11:112037730-112037730A10.8%rs72992972SDHDIntron variant1d14.26
11:112047061-112047061A12.2%rs10431036SDHDIntron variant1f20.65
11:112043614-112043614A12.2%rs11214108SDHDIntron variant1f12.25
11:112048051-112048051Τ22.7%rs7121554SDHDIntron variant1f12.03
11:111991866-111991868-0.3%rs1453244355SDHDIntron variant2b11.47
Table 5. Unique OXPHOS variants in asthenozoospermic identified as eQTL and sQTL according to the GTex portal [18].
Table 5. Unique OXPHOS variants in asthenozoospermic identified as eQTL and sQTL according to the GTex portal [18].
Genomic
Coordinates
AlleleAllele
Frequency (Europeans)
VariantGeneGenomic
Consequences
Functionp-Value
7:123197559-123197559C8.6%rs17146099NDUFA55′ UTR variant, intron varianteQTL (Testis)0.000089
7:123197559-123197559C8.6%rs17146099NDUFA55′ UTR variant, intron variantsQTL (Testis)9.3 × 10−8
7:123190928-123190928T8.6%rs34225533NDUFA5Intron varianteQTL (Testis)0.000036
7:123190928-123190928T8.6%rs34225533NDUFA5Intron variantsQTL (Testis)9.8 × 10−7
2:240872465-240872465A14.7%rs11684384NDUFA10Intron varianteQTL (Testis)8.4 × 10−10
2:240872465-240872465A14.7%rs11684384NDUFA10Intron varianteQTL (Prostate)2.1 × 10−15
21:44325525-44325525T20.2%rs8134542NDUFV3Intron varianteQTL (Prostate)3.9 × 10−12
21:44328278-44328278A20.2%rs35893787NDUFV3Intron varianteQTL (Prostate)7.8 × 10−13
21:44313221-44313221C20.2%rs35197797NDUFV3Intron varianteQTL (Prostate)9.5 × 10−13
8:100894978-100894986AAAC18.2%rs71274941COX6CIntron variantsQTL (Testis)1.1 × 10−59
8:100894978-100894986AAAC18.2%rs71274941COX6CIntron variantsQTL (Prostate)1.4 × 10−28
8:100903890-100903890G14.1%rs12544943COX6CIntron variantsQTL (Testis)9.1 × 10−36
8:100903890-100903890G14.1%rs12544943COX6CIntron variantsQTL (Prostate)1.8 × 10−16
11:67374581-67374581C38.2%rs1871043NDUFV1Intron varianteQTL (Prostate)7.6 × 10−9
4:46775623-46775623G4.7%rs78130313COX7B2Intron varianteQTL (Testis)0.000032
4:46908004-46908004A5.4%rs371114117COX7B2Intron varianteQTL (Testis)0.00010
4:46908004-46908004A5.4%rs371114117COX7B2Intron variantsQTL (Testis)3.9 × 10−7
12:95387542-95387542Τ44.4%rs4923660NDUFA12Intron varianteQTL (Testis)0.000015
12:95387542-95387542Τ44.4%rs4923660NDUFA12Intron variantsQTL (Testis)0.0000037
Table 6. Association of unique OXPHOS variants in asthenozoospermic with diseases according to SNPnexus [19].
Table 6. Association of unique OXPHOS variants in asthenozoospermic with diseases according to SNPnexus [19].
Genomic
Coordinates
AlleleAllele
Frequency
(Europeans)
VariantGeneGenomic
Consequence
Association with
Diseases
19:1391059-1391059T1.9%rs2074896NDUFS7intron variantLeigh syndrome, Mitochondrial complex I deficiency (Benign/Likely benign)
2:240897460-240897460C3.5%rs7588974NDUFA103′ UTR variant, intron variantLeigh syndrome, Mitochondrial complex I deficiency
2:240951071-240951071T1.1%rs35462421NDUFA10Missense variantLeigh syndrome (Benign/Likely benign)
5:52942083-52942083C96%rs31304NDUFS4Synonymous variantLeigh syndrome, Mitochondrial complex I deficiency (Benign)
18:9119489-9119489T9.1%rs41274300NDUFV2Synonymous variantMitochondrial complex I deficiency (Benign/Likely benign)
Table 7. Unique OXPHOS variants in asthenozoospermic men leading to miRNA binding site gain and/or loss according to miRNASNP v3 [23].
Table 7. Unique OXPHOS variants in asthenozoospermic men leading to miRNA binding site gain and/or loss according to miRNASNP v3 [23].
Genomic
Coordinates
AlleleAllele
Frequency
(Europeans)
VariantGenemiRNA LossmiRNA Gain
3:120320652-120320652C0.7%rs190013694NDUFB4hsa-miR-1273h-3p, hsa-miR-1245b-3p, hsa-miR-5700, hsa-miR-3678-3phsa-miR-1193, hsa-miR-105-3p, hsa-miR-4754, hsa-miR-6850-5p
19:1394865-1394865C2.1%rs73515054NDUFS7hsa-miR-495-3p, hsa-miR-5688, hsa-miR-7-2-3p, hsa-miR-589-3p, hsa-miR-7-1-3p, hsa-miR-4773hsa-miR-2278, hsa-miR-548p, hsa-miR-6501-3p
7:123180937-123180942GCG0.6%rs201784621NDUFA5hsa-miR-4536-3p, hsa-miR-4787-3phsa-miR-8064, hsa-miR-6821-5p, hsa-miR-4783-5p
2:240897460-240897460C3.5%rs7588974NDUFA10hsa-miR-3155b, hsa-miR-3155a, hsa-miR-4518, hsa-miR-1266-5p, hsa-miR-484, hsa-miR-3664-3phsa-miR-6829-3p, hsa-miR-6741-3p, hsa-miR-6778-3p, hsa-miR-6791-3p
12:4798415-4798415Τ0.1%rs181096156NDUFA9hsa-miR-4712-3p, hsa-miR-580-3p, hsa-miR-539-5phsa-miR-577
4:46736853-46736853Τ13.4%rs11736008COX7B2-hsa-miR-12135, hsa-miR-4748, hsa-miR-299-5p, hsa-miR-548m, hsa-miR-4464, hsa-miR-548at-5p, hsa-miR-561-3p, hsa-miR-329-5p
11:111966122-111966122G0.7%rs184654032SDHDhsa-miR-3120-5p, hsa-miR-200a-3p, hsa-miR-1208, hsa-miR-6757-3p, hsa-miR-141-3p, hsa-miR-6760-3phsa-miR-340-3p, hsa-miR-122b-3p, hsa-miR-6827-3p, hsa-miR-21-3p
Table 8. Prioritized OXPHOS variants for further investigation.
Table 8. Prioritized OXPHOS variants for further investigation.
VariantGeneAllele
Frequency (Europeans)
Missense
Variant
Functional SignificanceAssociation with
Diseases
eQTLs/sQTLsmiRNA Interactions
rs35462421NDUFA101.1%Damaging according to both databases---
rs31304NDUFS496%Unknown impact---
rs73515054NDUFS72.1%---
rs17146099NDUFA58.6%---
rs7588974NDUFA103.5%--
rs35197797NDUFV320.2%---
rs12544943COX6C14.1%---
rs1871043NDUFV138.2%---
rs41274300NDUFV29.1%---
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Kyrgiafini, M.-A.; Giannoulis, T.; Chatziparasidou, A.; Mamuris, Z. Elucidating the Role of OXPHOS Variants in Asthenozoospermia: Insights from Whole Genome Sequencing and an In Silico Analysis. Int. J. Mol. Sci. 2024, 25, 4121. https://doi.org/10.3390/ijms25074121

AMA Style

Kyrgiafini M-A, Giannoulis T, Chatziparasidou A, Mamuris Z. Elucidating the Role of OXPHOS Variants in Asthenozoospermia: Insights from Whole Genome Sequencing and an In Silico Analysis. International Journal of Molecular Sciences. 2024; 25(7):4121. https://doi.org/10.3390/ijms25074121

Chicago/Turabian Style

Kyrgiafini, Maria-Anna, Themistoklis Giannoulis, Alexia Chatziparasidou, and Zissis Mamuris. 2024. "Elucidating the Role of OXPHOS Variants in Asthenozoospermia: Insights from Whole Genome Sequencing and an In Silico Analysis" International Journal of Molecular Sciences 25, no. 7: 4121. https://doi.org/10.3390/ijms25074121

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop