Next Article in Journal
Association of miRNA-17-92 Cluster with Muscle Invasion in Bladder Cancer
Previous Article in Journal
Antimicrobial Photodynamic Activity of the Zn(II) Phthalocyanine RLP068/Cl Versus Antimicrobial-Resistant Priority Pathogens
Previous Article in Special Issue
Revisiting the Injury Mechanism of Goat Sperm Caused by the Cryopreservation Process from a Perspective of Sperm Metabolite Profiles
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Exploring Human Sperm Metabolism and Male Infertility: A Systematic Review of Genomics, Proteomics, Metabolomics, and Imaging Techniques

by
Achraf Zakaria
1,2,
Idrissa Diawara
1,3,4,
Amal Bouziyane
1,5,6,* and
Noureddine Louanjli
2,7
1
Research Laboratory of Microbiology, Infectious Diseases, Allergology and Pathogen Surveillance (LARMIAS), Mohammed VI Faculty of Medicine, Mohammed VI University of Sciences and Health (UM6SS), Casablanca 82403, Morocco
2
LABOMAC IVF Centers and Clinical Laboratory Medicine, Casablanca 20080, Morocco
3
Mohammed VI Higher Institute of Biosciences and Biotechnologies, Mohammed VI University of Sciences and Health (UM6SS), Casablanca 82403, Morocco
4
Research Unit, Mohammed VI Center for Research and Innovation, Rabat 10112, Morocco
5
Cheikh Khalifa International University Hospital, Casablanca 82403, Morocco
6
Mohammed VI International University Hospital, Bouskoura 27182, Morocco
7
IVF Center IRIFIV, Iris Clinic, Casablanca 20330, Morocco
*
Author to whom correspondence should be addressed.
Int. J. Mol. Sci. 2025, 26(15), 7544; https://doi.org/10.3390/ijms26157544
Submission received: 9 May 2025 / Revised: 7 June 2025 / Accepted: 20 June 2025 / Published: 5 August 2025

Abstract

Male infertility is a multifactorial condition often associated with disruptions in sperm metabolism and mitochondrial function, yet traditional semen analysis provides limited insight into these molecular mechanisms. Understanding sperm bioenergetics and metabolic dysfunctions is crucial for improving the diagnosis and treatment of conditions such as asthenozoospermia and azoospermia. This systematic review synthesizes recent literature, focusing on advanced tools and techniques—including omics technologies, advanced imaging, spectroscopy, and functional assays—that enable comprehensive molecular assessment of sperm metabolism and development. The reviewed studies highlight the effectiveness of metabolomics, proteomics, and transcriptomics in identifying metabolic biomarkers linked to male infertility. Non-invasive imaging modalities such as Raman and magnetic resonance spectroscopy offer real-time metabolic profiling, while the seminal microbiome is increasingly recognized for its role in modulating sperm metabolic health. Despite these advances, challenges remain in clinical validation and implementation of these techniques in routine infertility diagnostics. Integrating molecular metabolic assessments with conventional semen analysis promises enhanced diagnostic precision and personalized therapeutic approaches, ultimately improving reproductive outcomes. Continued research is needed to standardize biomarkers and validate clinical utility. Furthermore, these metabolic tools hold significant potential to elucidate the underlying causes of previously misunderstood and unexplained infertility cases, offering new avenues for diagnosis and treatment.

1. Introduction

Human spermatozoa are highly specialized cells responsible for delivering the paternal genome to the oocyte, a process requiring substantial energy, primarily in the form of adenosine triphosphate (ATP), to maintain sperm integrity and motility [1]. ATP is generated mainly through glycolysis and oxidative phosphorylation (OXPHOS) pathways localized in distinct sperm compartments [2,3].These metabolic pathways are dynamically regulated during sperm development, maturation, capacitation, and transit through the male and female reproductive tracts [4]. Despite its critical role, sperm metabolism and bioenergetics remain underexplored, limiting advances in diagnosing and treating male infertility conditions such as asthenozoospermia and azoospermia [5].
Infertility affects approximately 8–12% of couples worldwide, with male factors contributing to nearly half of these cases [6,7].
Male infertility can result from reproductive tract blockages, hormonal imbalances, testicular dysfunction, and abnormal sperm quality or motility. Lifestyle factors such as smoking, excessive alcohol use, and obesity also negatively impact fertility. Additionally, exposure to environmental toxins can reduce sperm count and quality. These diverse factors collectively contribute to impaired male reproductive function [6].
Among male infertility disorders, sperm motility defects have been increasingly linked to mitochondrial dysfunction and impaired energy metabolism [8].
Emerging evidence also implicates the seminal microbiome in modulating sperm DNA integrity and metabolism, suggesting novel therapeutic targets [9]. Advances in omics technologies—including genomics, proteomics, and metabolomics combined with advanced analytical tools—now enable comprehensive profiling of sperm metabolic pathways and identification of molecular biomarkers associated with fertility outcomes [10,11,12].
This systematic review synthesizes recent advances in tools and techniques for evaluating human sperm metabolism and development. The objective is to critically assess methodologies that elucidate molecular mechanisms underlying infertility and improve the prediction of sperm retrieval success, particularly in azoospermia and metabolic impairments. We also aim to highlight the roles of mitochondria, oxidative stress, and the seminal microbiome, providing a comprehensive framework to inform ongoing clinical strategies and support the interpretation of emerging findings in male fertility diagnostics and therapeutics.
Energy metabolism in sperm cells is critical for their motility and fertilization capacity, relying primarily on glycolysis, oxidative phosphorylation (OXPHOS), and fatty acid oxidation pathways. Glycolysis occurs mainly in the sperm head and principal piece, producing ATP and metabolic intermediates, while OXPHOS and fatty acid oxidation take place in the mitochondria-rich midpiece, generating the majority of cellular ATP through the electron transport chain. However, these metabolic processes also produce reactive oxygen species (ROS), which can induce oxidative stress and impair sperm function if not adequately neutralized by antioxidant defenses. A schematic overview of these interconnected pathways and their localization within sperm cells is presented in Figure 1, providing a comprehensive framework for understanding sperm bioenergetics and oxidative balance.

2. Methods

2.1. Search Strategy

A systematic search was conducted independently by the authors using PubMed, Scopus, and Web of Science databases, covering all records published up to 15 February 2025. Search strings combined Medical Subject Headings (MeSH) and keywords linked by Boolean operators (AND, OR) to capture literature on sperm metabolism and development. The main search terms included (“sperm metabolism” OR “sperm bioenergetics”) AND (“glycolysis” OR “oxidative phosphorylation” OR “mitochondrial function” OR “mitochondrial membrane potential”) AND (“sperm motility” OR “asthenozoospermia” OR “azoospermia” OR “male infertility”) AND (“metabolomics” OR “proteomics” OR “transcriptomics” OR “antioxidants” OR “sperm capacitation” OR “microbiome”). The full search strategy is provided in Supplementary File S1.
No protocol registration (e.g., PROSPERO) was performed for this review.

2.2. Inclusion and Exclusion Criteria

Studies were included if they were original research articles (observational studies, clinical trials, or technical evaluations) involving human males with infertility of various etiologies, including idiopathic and metabolic-related dysfunctions affecting sperm quality.
Exclusion criteria comprised review articles, animal studies, and male infertility cases with well-established causes (e.g., varicocele, cryptorchidism, infections, genetic abnormalities, systemic diseases). The rationale for excluding animal studies was to focus on clinical relevance to human infertility.

2.3. Data Extraction and Synthesis

Data on study design, sample size, diagnostic tools, and outcomes were extracted independently by two authors. Studies were grouped by analytical technique (proteomics, metabolomics, imaging, etc.) and infertility condition to facilitate synthesis. A PRISMA flowchart detailing the study selection process is provided in Figure 2, and the PRISMA checklist is included in the Supplementary File S1. The included studies are summarized and grouped by technique in Table 1A (Genomics), Table 1B (Proteomics), Table 1C (Metabolomics), and Table 1D (Imaging, Functional Assays, and Others).

3. Results

A total of 2775 records were identified, with 2433 remaining after duplicate removal. Screening titles and abstracts excluded 977 records, leaving 1456 for full-text review. After further exclusion of 1150 studies based on eligibility criteria, 306 texts were assessed, resulting in 43 studies being included in this review (Figure 2). Figure 2 follows the PRISMA (Preferred Reporting Items for Systematic Reviews and Meta-Analyses) guidelines.
This review synthesizes the most relevant and recent tools and techniques used to evaluate human sperm metabolism and development, focusing on their application in male infertility diagnostics and research. The included studies provide a comprehensive overview of methodologies assessing metabolic pathways, mitochondrial function, and molecular biomarkers associated with various infertility causes.
Before discussing the findings on these techniques and their clinical relevance, subsequent sections summarize current knowledge on sperm metabolism and its critical role in male fertility, providing the foundation for future research and clinical applications.

4. Energy Metabolism Pathways in Spermatozoa

Energy metabolism in spermatozoa depends on a series of tightly regulated biochemical reactions culminating in the production of adenosine triphosphate (ATP), the cell’s primary energy currency [51]. Two principal pathways generate ATP: glycolysis and oxidative phosphorylation (OXPHOS). Glycolysis occurs mainly in the sperm’s principal piece and head, providing a rapid but limited ATP supply without requiring oxygen [2]. This pathway yields only two net ATP molecules per glucose molecule.
OXPHOS, in contrast, takes place in the mitochondria densely packed within the sperm midpiece. This oxygen-dependent pathway is more efficient and produces the bulk of ATP needed to meet the high energy demands of sperm motility [3]. ATP generated in the midpiece must be transported along the flagellum to power movement. While diffusion has been proposed as a mechanism, its efficiency is limited by the narrow space within the flagellum, suggesting specialized transport systems might be involved, though these remain to be fully elucidated in humans and other mammals [52,53].

5. Metabolic Substrates and Adaptations

5.1. Metabolic Substrates and Transport

Sperm utilize both endogenous and exogenous carbohydrates to satisfy their metabolic needs. Seminal plasma from various mammals, including humans, contains fructose as the main metabolic substrate [54,55], along with glucose and, in some species, mannose [56]. These sugars are internalized through glucose transporters (GLUTs) with varying substrate specificities [57,58]. Additionally, sugars such as sorbitol serve as energy substrates in species including humans, rams, and mice [59,60].
Mitochondrial substrates include lactate and pyruvate, metabolized via lactate dehydrogenase (LDH) to regenerate NAD+ or enter the Krebs cycle for OXPHOS [61]. Pyruvate and lactate may originate from glycolysis or be taken up externally via monocarboxylate transporters [62,63]. Human sperm express specific LDH isoforms, enhancing mitochondrial utilization of lactate, permitting efficient respiratory substrate use [64,65]. Fatty acid β-oxidation also contributes to ATP production. Boar and bull sperm metabolize long-chain fatty acids such as oleic, palmitic, and myristic acids via mitochondrial β-oxidation [66,67]. Human sperm possess enzymatic machinery for both saturated and unsaturated fatty acid β-oxidation; inhibition of this pathway impairs motility [68]. Glycogen reserves, once thought absent in mature sperm, have been found in some species, such as dogs, where sperm can synthesize and utilize glycogen as an energy source [53,69,70].

5.2. Metabolic Adaptations During Maturation and Transit

Many studies use epididymal sperm to assess basal metabolic activity; however, epididymal sperm lack exposure to seminal plasma, which provides a fluid rich in energetic substrates and metabolic modulators [71]. Differences in metabolic activity between epididymal and ejaculated sperm have been observed. For example, ejaculated bull sperm rely heavily on OXPHOS for motility, whereas epididymal sperm maintain ATP levels primarily through glycolysis [71,72,73]. In mice, epididymal sperm depend mainly on glucose, but ejaculated sperm utilize a broader range of substrates, including citrate and pyruvate, through nonglycolytic pathways [74].
Sperm acquire fertilizing capacity through capacitation, a process involving extensive biochemical and physiological changes such as plasma membrane remodeling, ion fluxes, and activation of signaling cascades including cAMP-dependent pathways. Metabolic activity increases substantially during capacitation, reflecting heightened energy demands [75]. During transit through the female reproductive tract, sperm encounter variable environments with differing oxygen and substrate availability, necessitating metabolic adaptability to maintain motility and viability until fertilization [74,76,77].

6. Molecular Biomarkers and Oxidative Stress

Disruptions in sperm metabolism are frequently characterized by mitochondrial dysfunction and heightened oxidative stress, both pivotal in male infertility pathophysiology. Mitochondria, the powerhouse of sperm cells, are essential for ATP production necessary for motility and overall sperm function. However, excessive generation of reactive oxygen species (ROS)—a byproduct of impaired mitochondrial electron transport chains—leads to oxidative damage targeting mitochondrial membranes, proteins, and mitochondrial DNA [42]. This oxidative insult diminishes sperm motility and compromises nuclear DNA integrity, increasing the risk of mutations and fragmentation that can adversely affect fertilization and embryo development [4].
Advances in high-throughput omics technologies, including proteomics, metabolomics, and transcriptomics, have enabled the identification of specific molecular biomarkers indicative of these metabolic disturbances. Proteomic analyses reveal aberrant expression of key mitochondrial proteins and antioxidant enzymes, while metabolomic profiling uncovers altered concentrations of metabolites involved in energy metabolism and redox balance. Transcriptomic studies highlight dysregulation in gene expression pathways related to oxidative stress response and mitochondrial function [11]. Together, these molecular signatures provide valuable insight into sperm dysfunction’s biochemical underpinnings and hold promise for developing diagnostic tools and targeted therapies aimed at mitigating oxidative damage in infertile men.

7. Influence of the Seminal Microbiome

Recent research elucidates the seminal microbiome’s significant role—the diverse microbial communities residing within seminal fluid and associated with spermatozoa—in modulating sperm metabolic health and fertility outcomes. These microbial populations influence seminal plasma’s biochemical milieu, impacting pH, nutrient availability, and oxidative stress levels. Certain bacterial species have been implicated in either protective or detrimental effects on sperm function, with some microbes producing metabolites that exacerbate oxidative stress or trigger inflammatory responses, compromising sperm DNA integrity and metabolic efficiency [9]. The interaction between the seminal microbiome and sperm cells represents a complex, dynamic system potentially contributing to idiopathic male infertility. In healthy semen samples, the microbiota is predominantly composed of a core group of bacterial genera. The most commonly identified genera include Lactobacillus, Streptococcus, Corynebacterium, Prevotella, and Staphylococcus. These bacteria form the primary microbial communities in normal seminal fluid and are thought to contribute to maintaining reproductive tract health and sperm function [9]. Understanding how microbial dysbiosis alters sperm metabolism and function opens new avenues for therapeutic intervention, such as probiotics, antibiotics, or microbiome modulation strategies, to restore a healthy seminal environment. This emerging field holds promise for enhancing fertility treatments by addressing both host cellular dysfunction and microbial factors influencing reproductive health.

8. Advanced Techniques and Tools for Evaluating Sperm Metabolism

The evaluation of sperm metabolism and its role in male infertility has advanced considerably through diverse methodologies encompassing proteomics, metabolomics, transcriptomics, imaging, and functional assays. Agarwal et al. conducted proteomic analyses on seminal plasma from infertile men, identifying a 35-protein pathway linked to sperm dysfunction, including overexpression of membrane metallo-endopeptidase (MME) [20]. Amaral et al. characterized the sperm tail proteome, identifying 1049 proteins with prominent lipid metabolism enzymes and active mitochondrial and peroxisomal pathways, highlighting metabolic complexity essential for motility [8].
Metabolomic studies have revealed biochemical alterations in infertile populations. Boguenet et al. applied targeted metabolomics to seminal plasma from severe oligoasthenospermia patients, documenting decreased metabolites associated with membrane deterioration and energy defects [31]. Li et al. identified nine metabolites differing significantly in asthenozoospermic men, with nicotinamide levels correlating positively with sperm count and motility [78]. Zhao et al. detected disturbed metabolites implicating disrupted energy and amino acid metabolism in idiopathic asthenozoospermia [39]. Mitochondrial dysfunction emerges as a central theme linking molecular changes to impaired sperm function. Cassina et al. demonstrated mitochondrial respiratory deficits and nitro-oxidative protein modifications correlating with reduced motility [42]. Paoli et al. showed a positive correlation between mitochondrial membrane potential and sperm motility, reinforcing mitochondrial health as a fertility marker [45].
Transcriptomic and epigenetic analyses further enrich biomarker discovery. Larriba et al. profiled small RNAs in seminal extracellular vesicles from azoospermic men, identifying microRNAs discriminating azoospermia origin [16]. Li et al. reported differentially methylated regions in asthenozoospermic spermatozoa affecting genes essential for spermatogenesis and motility, suggesting epigenetic regulation contributes to metabolic dysfunction [78]. Imaging and spectroscopic techniques provide complementary functional insights. Raman microspectroscopy detects sperm DNA damage and is suitable for analyzing live sperm [46]. Magnetic resonance spectroscopy reveals motile sperm preferentially metabolize glucose, fructose, and pyruvate into lactate, highlighting increased glycolytic activity [32,38].
Oxidative stress remains critical in sperm dysfunction. Aziz et al. and Irigoyen et al. quantified ROS production, demonstrating correlations between elevated ROS and reduced mitochondrial respiratory control ratios and motility [43,48]. Proteomic analyses identified proteins differentially expressed in spermatozoa with elevated ROS, mainly related to energy metabolism and antioxidant defense [11]. Emerging research implicates the seminal microbiome in sperm health; combined 16S rRNA sequencing and metabolomics revealed Lactobacillus iners enrichment in men with high sperm DNA fragmentation, associating microbial metabolites with DNA damage and metabolic disruptions [9].
Integration of multi-dimensional data sets has been facilitated by bioinformatics approaches. Milardi et al. and Sharma et al. utilized pathway enrichment and network modeling to identify disrupted metabolic nodes in infertile men, accelerating biomarker discovery and therapeutic targeting{Citation}.Such systems biology approaches are crucial for translating complex molecular insights into clinical practice.
Collectively, these studies underscore sperm metabolism and its perturbations involving mitochondrial dysfunction, oxidative stress, epigenetic alterations, and microbial influences as pivotal in male infertility. The high sensitivity and specificity of these advanced methodologies promise improved diagnostic precision and personalized treatment strategies. Table 1A–D summarizes key methods and their contributions, illustrating progress toward a comprehensive assessment of sperm metabolism beyond traditional semen analysis.

9. Clinical Implications: Metabolic Dysfunction in Male Infertility

9.1. Diagnostic Implications

Metabolic biomarkers identified through omics and imaging techniques offer enhanced diagnostic precision beyond conventional semen analysis. These biomarkers can detect subtle metabolic dysfunctions contributing to infertility, enabling earlier and more accurate diagnosis.

9.2. Prognostic Potential

Proteomic and microRNA profiles provide prognostic information predictive of spermatogenic status and sperm retrieval success, guiding clinical decisions and personalized treatment strategies.

9.3. Therapeutic Strategies

Emerging interventions targeting metabolic dysfunction and seminal microbiome modulation hold promise for improving sperm function and fertility outcomes.

9.4. Integration with Clinical Workflows

Challenges remain in standardizing assays and integrating multi-omics data into routine clinical practice. Future research should focus on validating biomarkers in larger cohorts and developing accessible platforms for widespread clinical application.

9.5. Disadvantages of Current Methods and Tools to Evaluate Sperm Metabolism

Despite significant advancements, several limitations hinder the clinical translation of sperm metabolism assessment techniques. Omics approaches, while comprehensive, are often costly, time-consuming, and require specialized equipment and expertise, limiting their accessibility in routine clinical settings. Imaging techniques such as fluorescence microscopy and metabolic flux analysis provide valuable spatial and functional insights but may involve invasive sample preparation or rely on indirect metabolic indicators, potentially affecting sperm viability. Additionally, variability in sample handling, assay protocols, and data interpretation poses challenges for reproducibility and standardization across laboratories. These limitations underscore the need for developing rapid, cost-effective, and standardized tools that can reliably assess metabolism to facilitate their integration into routine infertility diagnostics.

9.6. Complementary Strengths of Metabolic Assessment Techniques

Different techniques offer unique strengths for studying sperm metabolism: proteomics excels in analyzing protein dynamics, metabolomics captures metabolic pathway changes, and transcriptomics reveals gene regulation. Imaging and spectroscopy provide real-time, non-invasive metabolic insights, while functional assays assess mitochondrial health and oxidative stress. Combining these complementary methods into integrated platforms will enhance biomarker discovery, improve diagnostics, and guide personalized treatments in male infertility.

10. Conclusions

The exploration of human sperm metabolism has greatly benefited from a variety of analytical methods and tools, ranging from traditional biochemical assays to advanced molecular and imaging technologies. This systematic review highlights the diversity and evolution of these approaches, underscoring their critical role in deepening our understanding of sperm bioenergetics and metabolic regulation. However, despite technological advances, significant challenges remain in translating these tools into standardized, clinically applicable protocols. Many methods, while powerful in research settings, lack validation for routine diagnostic use, and variability in sample preparation, assay conditions, and data interpretation limits comparability across studies.
Furthermore, the majority of current tools focus on isolated aspects of metabolism, such as glycolysis or oxidative phosphorylation, often overlooking the integrated and dynamic nature of sperm metabolic networks. There is a pressing need for comprehensive, multi-parametric platforms that can simultaneously assess multiple metabolic pathways with high sensitivity and specificity. Additionally, the accessibility and cost-effectiveness of these technologies remain barriers to widespread clinical adoption, particularly in low-resource settings.
Future research should prioritize the development and validation of standardized, user-friendly tools that can reliably assess sperm metabolism in clinical laboratories. Integration of metabolic assessments with conventional semen analysis and emerging omics data will be key to advancing personalized diagnostics and treatment strategies in male infertility. Moreover, longitudinal studies are necessary to establish the prognostic value of metabolic biomarkers and to understand how metabolic profiles evolve in response to therapeutic interventions.
Key questions for future exploration include the following: Which combinations of metabolic parameters provide the most clinically relevant information? How can emerging technologies be optimized for routine use without compromising accuracy? What are the best practices for harmonizing protocols across laboratories to ensure reproducibility? Addressing these will be essential for bridging the gap between research innovations and practical clinical applications.
In summary, while significant progress has been made in the methodological exploration of human sperm metabolism, translating these advances into clinical practice requires concerted efforts in validation, standardization, and integration. By overcoming these hurdles, we can unlock the full potential of metabolic diagnostics to improve the understanding and management of male infertility through precision medicine.

Supplementary Materials

The following supporting information can be downloaded at https://www.mdpi.com/article/10.3390/ijms26157544/s1.

Author Contributions

Conceptualization, A.Z. and I.D.; methodology, A.Z. and I.D.; software, A.Z.; validation, A.Z., I.D., A.B. and N.L.; formal analysis, A.Z.; investigation, A.B.; resources, I.D.; data curation, I.D., A.B. and N.L.; writing—original draft preparation, A.Z.; writing—review and editing, A.Z., I.D., A.B. and N.L.; visualization, A.Z., I.D., A.B. and N.L.; supervision, I.D., A.B. and N.L.; project administration, A.B. All authors have read and agreed to the published version of the manuscript.

Funding

This study was conducted without external funding.

Conflicts of Interest

The authors declare no conflicts of interest.

References

  1. Bohnensack, R.; Halangk, W. Control of respiration and of motility in ejaculated bull spermatozoa. Biochim. Et Biophys. Acta (BBA)-Bioenerg. 1986, 850, 72–79. [Google Scholar] [CrossRef]
  2. 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]
  3. Visconti, P. Sperm Bioenergetics in a Nutshell1. Biol. Reprod. 2012, 87, 72. [Google Scholar] [CrossRef]
  4. Sciorio, R.; Fleming, S.D. Intracytoplasmic sperm injection vs. in-vitro fertilization in couples in whom the male partners had a semen analysis within normal reference ranges: An open debate. Andrology 2023, 12, 20–29. [Google Scholar] [CrossRef] [PubMed]
  5. Ombelet, W.; Van Blerkom, J.; Boshoff, G.; Huyser, C.; Lopes, F.; Nargund, G.; Sallam, H.; Vanmechelen, K.; Campo, R. Now is the time to introduce new innovative assisted reproduction methods to implement accessible, affordable, and demonstrably successful advanced infertility services in resource-poor countries. Hum. Reprod. Open 2025, 2025, hoaf001. [Google Scholar] [CrossRef] [PubMed]
  6. World Health Organization. Infertility. Available online: https://www.who.int/news-room/fact-sheets/detail/infertility (accessed on 17 April 2025).
  7. Agarwal, A.; Mulgund, A.; Hamada, A.; Chyatte, M.R. A unique view on male infertility around the globe. Reprod. Biol. Endocrinol. 2015, 13, 37. [Google Scholar] [CrossRef]
  8. Amaral, A.; Castillo, J.; Estanyol, J.M.; Ballescà, J.L.; Ramalho-Santos, J.; Oliva, R. Human Sperm Tail Proteome Suggests New Endogenous Metabolic Pathways. Mol. Cell Proteom. 2013, 12, 330–342. [Google Scholar] [CrossRef]
  9. He, J.; Ma, M.; Xu, Z.; Guo, J.; Chen, H.; Yang, X.; Chen, P.; Liu, G. Association between semen microbiome disorder and sperm DNA damage. Microbiol. Spectr. 2024, 12, e00759-24. [Google Scholar] [CrossRef]
  10. Milardi, D.; Grande, G.; Vincenzoni, F.; Giampietro, A.; Messana, I.; Castagnola, M.; Marana, R.; De Marinis, L.; Pontecorvi, A. Novel Biomarkers of Androgen Deficiency From Seminal Plasma Profiling Using High-Resolution Mass Spectrometry. J. Clin. Endocrinol. Metab. 2014, 99, 2813–2820. [Google Scholar] [CrossRef]
  11. Sharma, R.; Agarwal, A.; Mohanty, G.; Jesudasan, R.; Gopalan, B.; Willard, B.; Yadav, S.P.; Sabanegh, E. Functional proteomic analysis of seminal plasma proteins in men with various semen parameters. Reprod. Biol. Endocrinol. 2013, 11, 38. [Google Scholar] [CrossRef]
  12. Sharma, R.; Agarwal, A.; Mohanty, G.; Hamada, A.J.; Gopalan, B.; Willard, B.; Yadav, S.; du Plessis, S. Proteomic analysis of human spermatozoa proteins with oxidative stress. Reprod. Biol. Endocrinol. 2013, 11, 48. [Google Scholar] [CrossRef] [PubMed]
  13. Abruzzese, G.A.; Sanchez-Rodriguez, A.; Roldan, E.R. Sperm Metabolism. Mol. Reprod. Dev. 2024, 91, e23772. [Google Scholar] [CrossRef] [PubMed]
  14. Barceló, M.; Mata, A.; Bassas, L.; Larriba, S. Exosomal microRNAs in seminal plasma are markers of the origin of azoospermia and can predict the presence of sperm in testicular tissue. Hum. Reprod. 2018, 33, 1087–1098. [Google Scholar] [CrossRef] [PubMed]
  15. 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]
  16. Larriba, S.; Sánchez-Herrero, J.F.; Pluvinet, R.; López-Rodrigo, O.; Bassas, L.; Sumoy, L. Seminal extracellular vesicle sncRNA sequencing reveals altered miRNA/isomiR profiles as sperm retrieval biomarkers for azoospermia. Andrology 2023, 12, 137–156. [Google Scholar] [CrossRef]
  17. Li, J.; Xu, J.; Yang, T.; Chen, J.; Larriba Li, F.; Shen, B.; Fan, C. Genome-Wide Methylation Analyses of Human Sperm Unravel Novel Differentially Methylated Regions in Asthenozoospermia. Epigenomics 2022, 14, 951–964. [Google Scholar] [CrossRef]
  18. Wainstein, A.; Hassan, S.; Barda, S.; Lehavi, O.; Azem, F.; Ben-Dov, I.Z.; Hauser, R.; Kleiman, S.E. MicroRNAs expression in semen and testis of azoospermic men. Andrology 2023, 11, 687–697. [Google Scholar] [CrossRef]
  19. Wang, C.; Yang, C.; Chen, X.; Yao, B.; Yang, C.; Zhu, C.; Li, L.; Wang, J.; Li, X.; Shao, Y.; et al. Altered Profile of Seminal Plasma MicroRNAs in the Molecular Diagnosis of Male Infertility. Clin. Chem. 2011, 57, 1722–1731. [Google Scholar] [CrossRef]
  20. Agarwal, A.; Ayaz, A.; Samanta, L.; Sharma, R.; Assidi, M.; Abuzenadah, A.M.; Sabanegh, E. Comparative proteomic network signatures in seminal plasma of infertile men as a function of reactive oxygen species. Clin. Proteom. 2015, 12, 23. [Google Scholar] [CrossRef]
  21. Bragado, M.; Garcia-Marin, L.; Calle-Guisado, V.; de Llera, A.; Martin-Hidalgo, D.; Mijares, J.; Gil, M.; Alvarez, I. AMP-activated kinase in human spermatozoa: Identification, intracellular localization, and key function in the regulation of sperm motility. Asian J. Androl. 2017, 19, 707–714. [Google Scholar] [CrossRef]
  22. Deng, T.; Wang, W.; Fu, Z.; Xie, Y.; Zhou, Y.; Pu, J.; Chen, K.; Yao, B.; Li, X.; Yao, J. Lipidomics random forest algorithm of seminal plasma is a promising method for enhancing the diagnosis of necrozoospermia. Metabolomics 2024, 20, 57. [Google Scholar] [CrossRef]
  23. Fietz, D.; Sgaier, R.; O’donnell, L.; Stanton, P.G.; Dagley, L.F.; Webb, A.I.; Schuppe, H.-C.; Diemer, T.; Pilatz, A. Proteomic biomarkers in seminal plasma as predictors of reproductive potential in azoospermic men. Front. Endocrinol. 2024, 15, 1327800. [Google Scholar] [CrossRef] [PubMed]
  24. Hashemitabar, M.; Sabbagh, S.; Orazizadeh, M.; Ghadiri, A.; Bahmanzadeh, M. A proteomic analysis on human sperm tail: Comparison between normozoospermia and asthenozoospermia. J. Assist. Reprod. Genet. 2015, 32, 853–863. [Google Scholar] [CrossRef] [PubMed]
  25. Liang, J.; Zheng, Y.; Zeng, W.; Chen, L.; Yang, S.; Du, P.; Wang, Y.; Yu, X.; Zhang, X. Proteomic Profile of Sperm in Infertile Males Reveals Changes in Metabolic Pathways. Protein J. 2021, 40, 929–939. [Google Scholar] [CrossRef] [PubMed]
  26. Torra-Massana, M.; Jodar, M.; Barragán, M.; Soler-Ventura, A.; Delgado-Dueñas, D.; Rodríguez, A.; Oliva, R.; Vassena, R. Altered mitochondrial function in spermatozoa from patients with repetitive fertilization failure after ICSI revealed by proteomics. Andrology 2021, 9, 1192–1204. [Google Scholar] [CrossRef]
  27. Martins, A.D.; Selvam, M.K.P.; Agarwal, A.; Alves, M.G.; Baskaran, S. Alterations in seminal plasma proteomic profile in men with primary and secondary infertility. Sci. Rep. 2020, 10, 7539. [Google Scholar] [CrossRef]
  28. Soler, L.; Labas, V.; Thélie, A.; Grasseau, I.; Teixeira-Gomes, A.-P.; Blesbois, E. Intact Cell MALDI-TOF MS on Sperm: A Molecular Test For Male Fertility Diagnosis. Mol. Cell Proteom. 2016, 15, 1998–2010. [Google Scholar] [CrossRef]
  29. Sulc, A.; Czétány, P.; Máté, G.; Balló, A.; Semjén, D.; Szántó, Á.; Márk, L. MALDI Imaging Mass Spectrometry Reveals Lipid Alterations in Physiological and Sertoli Cell-Only Syndrome Human Testicular Tissue Sections. Int. J. Mol. Sci. 2024, 25, 8358. [Google Scholar] [CrossRef]
  30. Xu, Y.; Fan, Y.; Fan, W.; Jing, J.; Xue, K.; Zhang, X.; Ye, B.; Ji, Y.; Liu, Y.; Ding, Z. RNASET2 impairs the sperm motility via PKA/PI3K/calcium signal pathways. Reproduction 2018, 155, 383–392. [Google Scholar] [CrossRef]
  31. Boguenet, M.; Bocca, C.; Bouet, P.; Serri, O.; Chupin, S.; Tessier, L.; Blanchet, O.; El Hachem, H.; de la Barca, J.M.C.; Reynier, P.; et al. Metabolomic signature of the seminal plasma in men with severe oligoasthenospermia. Andrology 2020, 8, 1859–1866. [Google Scholar] [CrossRef]
  32. Calvert, S.J.; Reynolds, S.; Paley, M.N.; Walters, S.J.; Pacey, A.A. Probing human Kyrgiafini using 13C-magnetic resonance spectroscopy. Mol. Hum. Reprod. 2018, 25, 30–41. [Google Scholar] [CrossRef] [PubMed]
  33. Jayaraman, V.; Ghosh, S.; Sengupta, A.; Srivastava, S.; Sonawat, H.M.; Narayan, P.K. Identification of biochemical differences between different forms of male infertility by nuclear magnetic resonance (NMR) spectroscopy. J. Assist. Reprod. Genet. 2014, 31, 1195–1204. [Google Scholar] [CrossRef] [PubMed]
  34. Olesti, E.; Boccard, J.; Rahban, R.; Girel, S.; Moskaleva, N.E.; Zufferey, F.; Rossier, M.F.; Nef, S.; Rudaz, S.; González-Ruiz, V. Low-polarity untargeted metabolomic profiling as a tool to gain insight into seminal fluid. Metabolomics 2023, 19, 53. [Google Scholar] [CrossRef] [PubMed]
  35. Olesti, E.; Garcia, A.; Rahban, R.; Rossier, M.F.; Boccard, J.; Nef, S.; González-Ruiz, V.; Rudaz, S. Steroid profile analysis by LC-HRMS in human seminal fluid. J. Chromatogr. B 2020, 1136, 121929. [Google Scholar] [CrossRef]
  36. Paiva, C.; Amaral, A.; Rodriguez, M.; Canyellas, N.; Correig, X.; Ballescà, J.L.; Ramalho-Santos, J.; Oliva, R. Identification of endogenous metabolites in human sperm cells using proton nuclear magnetic resonance (1H-NMR) spectroscopy and gas chromatography-mass spectrometry (GC-MS). Andrology 2015, 3, 496–505. [Google Scholar] [CrossRef]
  37. Qiao, S.; Wu, W.; Chen, M.; Tang, Q.; Xia, Y.; Jia, W.; Wang, X. Seminal plasma metabolomics approach for the diagnosis of unexplained male infertility. PLoS ONE 2017, 12, e0181115. [Google Scholar] [CrossRef]
  38. Reynolds, S.; Calvert, S.J.; Paley, M.N.; Pacey, A.A. 1H Magnetic Resonance Spectroscopy of live human sperm. Mol. Hum. Reprod. 2017, 23, 441–451. [Google Scholar] [CrossRef]
  39. Zhao, K.; Zhang, J.; Xu, Z.; Xu, Y.; Xu, A.; Chen, W.; Miao, C.; Liu, S.; Wang, Z.; Jia, R. Metabolomic Profiling of Human Spermatozoa in Idiopathic Asthenozoospermia Patients Using Gas Chromatography-Mass Spectrometry. BioMed Res. Int. 2018, 2018, 8327506. [Google Scholar] [CrossRef]
  40. Zhao, K.; Zhang, Q.; Cong, R.; Xu, Z.; Xu, Y.; Han, J. Metabolomic profiling of human semen in patients with oligospermia using high performance liquid chromatography-tandem mass spectrometry. Sci. Rep. 2024, 14, 23739. [Google Scholar] [CrossRef]
  41. Almeida, C.; Cunha, M.; Ferraz, L.; Silva, J.; Barros, A.; Sousa, M. Caspase-3 detection in human testicular spermatozoa from azoospermic and non-azoospermic patients. Int. J. Androl. 2011, 34, e407–e414. [Google Scholar] [CrossRef]
  42. Cassina, A.; Silveira, P.; Cantu, L.; Montes, J.M.; Radi, R.; Sapiro, R. Defective Human Sperm Cells Are Associated with Mitochondrial Dysfunction and Oxidant Production1. Biol. Reprod. 2015, 93, 119. [Google Scholar] [CrossRef]
  43. Irigoyen, P.; Pintos-Polasky, P.; Rosa-Villagran, L.; Skowronek, M.F.; Cassina, A.; Sapiro, R. Mitochondrial metabolism determines the functional status of human sperm and correlates with semen parameters. Front. Cell Dev. Biol. 2022, 10, 926684. [Google Scholar] [CrossRef]
  44. Fukuda, T.; Miyake, H.; Enatsu, N.; Matsushita, K.; Fujisawa, M. Seminal level of clusterin in infertile men as a significant biomarker reflecting spermatogenesis. Andrologia 2016, 48, 1188–1194. [Google Scholar] [CrossRef] [PubMed]
  45. Paoli, D.; Gallo, M.; Rizzo, F.; Baldi, E.; Francavilla, S.; Lenzi, A.; Lombardo, F.; Gandini, L. Mitochondrial membrane potential profile and its correlation with increasing sperm motility. Fertil. Steril. 2011, 95, 2315–2319. [Google Scholar] [CrossRef] [PubMed]
  46. Da Costa, R.; Amaral, S.; Redmann, K.; Kliesch, S.; Schlatt, S. Spectral features of nuclear DNA in human sperm assessed by Raman Microspectroscopy: Effects of UV-irradiation and hydration. PLoS ONE 2018, 13, e0207786. [Google Scholar] [CrossRef] [PubMed]
  47. Liu, Y.; Zhu, Y.; Di, L.; Osterberg, E.C.; Liu, F.; He, L.; Hu, H.; Huang, Y.; Li, P.S.; Li, Z. Raman spectroscopy as an ex vivo noninvasive approach to distinguish complete and incomplete spermatogenesis within human seminiferous tubules. Fertil. Steril. 2014, 102, 54–60.e2. [Google Scholar] [CrossRef]
  48. Aziz, N.; Novotny, J.; Oborna, I.; Fingerova, H.; Brezinova, J.; Svobodova, M. Comparison of chemiluminescence and flow cytometry in the estimation of reactive oxygen and nitrogen species in human semen. Fertil. Steril. 2010, 94, 2604–2608. [Google Scholar] [CrossRef]
  49. Torrezan-Nitao, E.; Brown, S.G.; Mata-Martínez, E.; Treviño, C.L.; Barratt, C.; Publicover, S. [Ca2+]i oscillations in human sperm are triggered in the flagellum by membrane potential-sensitive activity of CatSper. Hum. Reprod. 2020, 36, 293–304. [Google Scholar] [CrossRef]
  50. Vashisht, A.; Ahluwalia, P.K.; Gahlay, G.K. A Comparative Analysis of the Altered Levels of Human Seminal Plasma Constituents as Contributing Factors in Different Types of Male Infertility. Curr. Issues Mol. Biol. 2021, 43, 1307–1324. [Google Scholar] [CrossRef]
  51. Quijano, C.; Trujillo, M.; Castro, L.; Trostchansky, A. Interplay between oxidant species and energy metabolism. Redox Biol. 2016, 8, 28–42. [Google Scholar] [CrossRef]
  52. Tombes, R.M.; Shapiro, B.M. Metabolite channeling: A phosphorylcreatine shuttle to mediate high energy phosphate transport between sperm mitochondrion and tail. Cell 1985, 41, 325–334. [Google Scholar] [CrossRef] [PubMed]
  53. Ford, W. Glycolysis and sperm motility: Does a spoonful of sugar help the flagellum go round? Hum. Reprod. Update 2006, 12, 269–274. [Google Scholar] [CrossRef]
  54. Mann, T. Fructose, a Constituent of Semen. Nature 1946, 157, 79. [Google Scholar] [CrossRef] [PubMed]
  55. Frenette, G.; Lessard, C.; Madore, E.; Fortier, M.A.; Sullivan, R. Aldose Reductase and Macrophage Migration Inhibitory Factor Are Associated with Epididymosomes and Spermatozoa in the Bovine Epididymis1. Biol. Reprod. 2003, 69, 1586–1592. [Google Scholar] [CrossRef]
  56. Rigau, T.; Rivera, M.; Palomo, M.; Fernandez-Novell, J.; Mogas, T.; Ballester, J.; Pena, A.; Otaegui, P.; Guinovart, J.; Rodriguez-Gil, J. Differential effects of glucose and fructose on hexose metabolism in dog spermatozoa. Reproduction 2002, 123, 579–591. [Google Scholar] [CrossRef]
  57. Burant, C.; Takeda, J.; Brot-Laroche, E.; Bell, G.; Davidson, N. Fructose transporter in human spermatozoa and small intestine is GLUT5. J. Biol. Chem. 1992, 267, 14523–14526. [Google Scholar] [CrossRef] [PubMed]
  58. Bucci, D.; Rodriguez-Gil, J.E.; Vallorani, C.; Spinaci, M.; Galeati, G.; Tamanini, C. GLUTs and Mammalian Sperm Metabolism. J. Androl. 2010, 32, 348–355. [Google Scholar] [CrossRef]
  59. King, T.E.; Mann, T.R.R. Sorbitol metabolism in spermatozoa. Proc. R. Soc. Lond. Ser. B Biol. Sci. 1959, 151, 226–243. [Google Scholar] [CrossRef]
  60. Cao, W.; Aghajanian, H.K.; Haig-Ladewig, L.A.; Gerton, G.L. Sorbitol Can Fuel Mouse Sperm Motility and Protein Tyrosine Phosphorylation via Sorbitol Dehydrogenase1. Biol. Reprod. 2009, 80, 124–133. [Google Scholar] [CrossRef]
  61. Darr, C.R.; Varner, D.D.; Teague, S.; Cortopassi, G.A.; Datta, S.; Meyers, S.A. Lactate and Pyruvate Are Major Sources of Energy for Stallion Sperm with Dose Effects on Mitochondrial Function, Motility, and ROS Production. Biol. Reprod. 2016, 95, 34. [Google Scholar] [CrossRef]
  62. Halestrap, A.P.; Price, N.T. The proton-linked monocarboxylate transporter (MCT) family: Structure, function and regulation. Biochem. J. 1999, 343, 281–299. [Google Scholar] [CrossRef] [PubMed]
  63. Lee, S.; Lee, J.; Lee, D.R. The genetic variation in Monocarboxylic acid transporter 2 (MCT2) has functional and clinical relevance with male infertility. Asian J. Androl. 2014, 16, 694–697. [Google Scholar] [CrossRef] [PubMed]
  64. Blanco, A.; Zinkham, W.H. Lactate Dehydrogenases in Human Testes. Science 1963, 139, 601–602. [Google Scholar] [CrossRef]
  65. Swegen, A.; Curry, B.J.; Gibb, Z.; Lambourne, S.R.; Smith, N.D.; Aitken, R.J. Investigation of the stallion sperm proteome by mass spectrometry. Reproduction 2015, 149, 235–244. [Google Scholar] [CrossRef]
  66. Zhu, Z.; Li, R.; Feng, C.; Liu, R.; Zheng, Y.; Hoque, S.A.M.; Wu, D.; Lu, H.; Zhang, T.; Zeng, W. Exogenous Oleic Acid and Palmitic Acid Improve Boar Sperm Motility via Enhancing Mitochondrial Β-Oxidation for ATP Generation. Animals 2020, 10, 591. [Google Scholar] [CrossRef] [PubMed]
  67. Islam, M.; Umehara, T.; Tsujita, N.; Shimada, M. Saturated fatty acids accelerate linear motility through mitochondrial ATP production in bull sperm. Reprod. Med. Biol. 2021, 20, 289–298. [Google Scholar] [CrossRef]
  68. Amaral, A.; Lourenço, B.; Marques, M.; Ramalho-Santos, J. Mitochondria functionality and sperm quality. Reproduction 2013, 146, R163–R174. [Google Scholar] [CrossRef]
  69. Ballester, J.; Rutllant, J.; Mogas, T.; Rigau, T.; Guinovart, J.J.; Fernández-Novell, J.M.; García-Rocha, M.; Palomo, M.J.; Peña, A.; Rodríguez-Gil, J.E. Evidence for a functional glycogen metabolism in mature mammalian spermatozoa. Mol. Reprod. Dev. 2000, 56, 207–219. [Google Scholar] [CrossRef]
  70. Palomo, M.J.; Fernández-Novell, J.M.; Peña, A.; Guinovart, J.J.; Rigau, T.; Rodríguez-Gil, J.E. Glucose- and fructose-induced dog-sperm glycogen synthesis shows specific changes in the location of the sperm glycogen deposition. Mol. Reprod. Dev. 2003, 64, 349–359. [Google Scholar] [CrossRef]
  71. Losano, J.; Angrimani, D.; Dalmazzo, A.; Rui, B.; Brito, M.; Mendes, C.; Kawai, G.; Vannucchi, C.; Assumpção, M.; Barnabe, V.; et al. Effect of mitochondrial uncoupling and glycolysis inhibition on ram sperm functionality. Reprod. Domest. Anim. 2017, 52, 289–297. [Google Scholar] [CrossRef]
  72. Moraes, C.R.; Moraes, L.E.; Blawut, B.; Benej, M.; Papandreou, I.; Denko, N.C.; da Silva, M.C. Effect of glucose concentration and cryopreservation on mitochondrial functions of bull spermatozoa and relationship with sire conception rate. Anim. Reprod. Sci. 2021, 230, 106779. [Google Scholar] [CrossRef] [PubMed]
  73. Bulkeley, E.A.; Foutouhi, A.; Wigney, K.; Santistevan, A.C.; Collins, C.; McNabb, B.; Meyers, S. Effects from disruption of mitochondrial electron transport chain function on bull sperm motility. Theriogenology 2021, 176, 63–72. [Google Scholar] [CrossRef]
  74. Balbach, M.; Ghanem, L.; Violante, S.; Kyaw, A.; Romarowski, A.; Cross, J.R.; Visconti, P.E.; Levin, L.R.; Buck, J. Capacitation induces changes in metabolic pathways supporting motility of epididymal and ejaculated sperm. Front. Cell Dev. Biol. 2023, 11, 1160154. [Google Scholar] [CrossRef] [PubMed]
  75. Jin, S.-K.; Yang, W.-X. Factors and pathways involved in capacitation: How are they regulated? Oncotarget 2016, 8, 3600–3627. [Google Scholar] [CrossRef] [PubMed]
  76. Tourmente, M.; Sansegundo, E.; Roldan, E.R.S. Capacitation promotes a shift in energy metabolism in murine sperm. Front. Cell Dev. Biol. 2022, 10, 950979. [Google Scholar] [CrossRef]
  77. Mateo-Otero, Y.; Madrid-Gambin, F.; Llavanera, M.; Gomez-Gomez, A.; Haro, N.; Pozo, O.J.; Yeste, M. Sperm physiology and in vitro fertilising ability rely on basal metabolic activity: Insights from the pig model. Commun. Biol. 2023, 6, 344. [Google Scholar] [CrossRef]
  78. Li, L.; Hao, X.; Chen, H.; Wang, L.; Chen, A.; Song, X.; Hu, Z.; Su, Y.; Lin, H.; Fan, P. Metabolomic characterization of semen from asthenozoospermic patients using ultra-high-performance liquid chromatography–tandem quadrupole time-of-flight mass spectrometry. Biomed. Chromatogr. 2020, 34, e4897. [Google Scholar] [CrossRef]
Figure 1. Adapted and modified from [13].
Figure 1. Adapted and modified from [13].
Ijms 26 07544 g001
Figure 2. PRISMA flowchart illustrating the selection process of studies included in the systematic review.
Figure 2. PRISMA flowchart illustrating the selection process of studies included in the systematic review.
Ijms 26 07544 g002
Table 1. (A) Genomics studies based on techniques used in sperm metabolism and male infertility; (B) proteomics studies based on techniques used in sperm metabolism and male infertility; (C) metabolomics studies based on techniques used in sperm metabolism and male infertility; (D) imaging, functional assays, and other techniques used in sperm metabolism and male infertility.
Table 1. (A) Genomics studies based on techniques used in sperm metabolism and male infertility; (B) proteomics studies based on techniques used in sperm metabolism and male infertility; (C) metabolomics studies based on techniques used in sperm metabolism and male infertility; (D) imaging, functional assays, and other techniques used in sperm metabolism and male infertility.
(A) Genomics Studies Based on Techniques Used in Sperm Metabolism and Male Infertility
Methods/ToolsStudy AimResultsSample SizeStudy TypeReferences
  • MiRNA quantitative PCR panels
  • Exosome isolation
Evaluate seminal plasma exosomal MiRNAs as markers for Azoospermia origin and sperm presenceMir-31-5p as biomarker for azoospermia origin combined with FSH improves prediction39Case-control prospective studyBarceló et al., 2018 [14]
  • Whole-genome sequencing
  • RNA expression analysis
Identify lncRNA mutations and expression in teratozoospermia1166 unique mutations in Differentially Expressed lncRNAs
Variants affect structure/function and MiRNA interactions
N/A *Observational studyKyrgiafini et al., 2023 [15]
  • High-throughput small RNA profiling
  • RT-qPCR validation

Small RNA profiling in seminal extracellular vesicles for azoospermia classificationCanonical and isomiR microRNAs discriminate azoospermia origin
tRNA and piRNAs less discriminatory
17Observational case-control studyLarriba et al., 2024 [16]
  • Whole-genome bisulfite sequencing
Investigate DNA methylation patterns in asthenozoospermia238 differentially methylated regions annotated to 114 genes related to spermatogenesis and motility12Comparative observational studyLi et al., 2022 [17]
  • Small RNA deep sequencing
  • RT-qPCR validation

Study microRNA profiles in semen and testicular tissue of azoospermic menMir-202-3p reduced in azoospermic semen
mir-370-3p elevated in azoospermia without sperm in testis
54Observational comparative studyWainstein et al., 2023 [18]
  • SOLEXA sequencing
  • RT-qPCR

Seminal plasma miRNAs in infertile men and diagnostic valueSeven miRNAs altered
miRNAs better diagnostic markers than routine parameters
457Observational case-control studyWang et al., 2011 [19]
(B) Proteomics studies based on techniques used in sperm metabolism and male infertility
Methods/ToolsStudy AimResultsSample sizeStudy TypeReferences
  • Chemiluminescence assay
  • Proteomic analysis via 1D gel electrophoresis
  • In-gel digestion
  • LC/MS-MS



Identify seminal plasma proteins involved in ROS-mediated male infertilityMembrane Metallo-Endopeptidase (MME) overexpressed; Proteins modulated in infertile groups
35-protein pathway linked to sperm dysfunction
59Comparative observational studyAgarwal et al., 2015 [20]
  • LC-MS/MS
  • Sonication and sucrose-gradient ultracentrifugation for tail fractions

Characterize human sperm tail proteome, focusing on metabolism-related proteins1049 proteins identified; lipid metabolism enzymes prominent
Mitochondrial and peroxisomal pathways active
N/A *Experimental proteomic studyAmaral et al., 2013 [8]
  • Western blotting
  • Indirect immunofluorescence

Identify and localize AMP- activated protein kinase (AMPK) in human sperm and evaluate its role in sperm motilityAMPK localized in acrosome, midpiece, tail
Active AMPK higher in high motility sperm; inhibition reduces motility
N/A *Experimental studyCalle-Guisado et al., 2017 [21]
  • LC-MS lipidomic analysis
Profile seminal plasma lipid composition in necrozoospermia and evaluate lipid biomarkersIdentified 1267 lipids; 20 lipids predictive for necrozoospermia
Lipidomics improves diagnosis
56Comparative observational studyDeng et al., 2024 [22]
  • Label-free LC-MS/MS proteomic analysis
  • Western blot validation

Discover seminal plasma biomarkers for non-invasive differential diagnosis of Obstructive Azoospermia (OA) vs. Non-Obstructive Azoospermia (NOA)42 proteins downregulated in NOA SCO patients
Testis-specific proteins as biomarkers
30Prospective observational proteomic studyFietz et al., 2024 [23]
  • 2D GEL ELECTROPHORESIS
  • MALDI-TOF-TOF MS

Identify novel biomarkers for asthenozoospermia via sperm tail proteomic analysis14 proteins altered in asthenozoospermia including tubulin beta 2B, glutathione S-transferase Mu 3, clusterinN/A *Experimental proteomic studyHashemitabar et al., 2015 [24]
  • Isobaric tag labeling; LC-MS/MS
  • Immunofluorescence western blot

Proteomic profiling of sperm in severe oligoasthenoteratozoospermia938 proteins differentially expressed
Metabolic pathways enriched
YBX1 upregulated
AK1 and ACO2 downregulated
N/A *Experimental proteomic studyLiang et al., 2021 [25]
  • LC-MS/MS
Proteomic profile changes in spermatozoa with elevated ROS15 proteins differentially expressed
Energy metabolism and oxidative stress pathways affected
52Prospective observational proteomic studySharma et al., 2013 [12]
  • 2D-LC-MS/MS
  • Western blot
  • Flow cytometry
  • Fluorometric assays



Proteomic analysis of sperm in fertilization failure after ICSIAltered mitochondrial and proteasomal proteins in fertilization failure17Observational proteomic studyMassana et al., 2021 [26]
  • LTQ ORBITRAP mass spectrometry
  • Protein interaction network

Identify seminal biomarkers for secondary male hypogonadism (HH)33 proteins absent in hypogonadic patients
14 recovered after testosterone therapy
30Observational proteomic studyMilardi et al., 2014 [10]
  • LC-MS/MS PROTEOMIC ANALYSIS
Identify seminal plasma proteins as biomarkers of sperm quality20 proteins differentially expressed
Biological regulation affected
N/A *Observational proteomic studySharma et al., 2013 [11]
  • Proteomic shotgun analysis
  • Western blot

Characterize seminal plasma proteome in primary and secondary infertilityDysregulated proteins linked to secretion and immune response59Pilot observational proteomic studyMartins et al., 2020 [27]
  • MALDI-TOF MS
  • TOP-DOWN PROTEIN IDENTIFICATION

Fertility-predictive model profiles of spermatozoaHigh diagnostic accuracy
Proteins involved in energy metabolism and sperm structure
N/A *Experimental studySoler et al., 2016 [28]
  • MALDI imaging mass spectrometry
Phospholipid expression in Sertoli cell-only syndrome (SCOS) testisPhospholipid levels correlated with spermatogenesis
Potential microTESE diagnostic tool
N/A *Experimental studySulc et al., 2024 [29]
  • Protein quantification
  • Incubation experiments
  • Immunoprecipitation
  • Mass spectrometry



Ribonuclease
(RNASET2) levels in sperm and relation to motility
RNASET2 elevated in asthenozoospermia
Inversely correlated with motility
Interacts with AKAP4
205Experimental observational studyXu et al., 2018 [30]
(C) Metabolomics studies based on techniques used in sperm metabolism and male infertility
Methods/ToolsStudy AimResultsSample sizeStudy TypeReferences
  • Targeted quantitative metabolomics
  • HPLC-TANDEM MS

Assess metabolomic signatures of seminal plasma in severe OligoasthenospermiaDecreased metabolites linked to sperm membrane deterioration and energy defects40Comparative observational studyBoguenet et al., 2020 [31]
  • Magnetic Resonance Spectroscopy (MRS)
Investigate sperm metabolismSperm metabolize glucose, fructose, pyruvate to lactate
Motile sperm show higher glycolytic activity
97Experimental observational studyCalvert et al., 2019 [32]
  • 16S rRNA SEQUENCING
  • UNTARGETED METABOLOMICS

Investigate seminal microbiome and metabolome role in high sperm DNA fragmentation index (HDFI)Lactobacillus iners enriched in HDFI
Microbial profiles linked to sperm DNA fragmentation
Butanoate fermentation implicated
102Observational case-control studyHe et al., 2024 [9]
  • 1H NMR spectroscopy (700 mhz)
Analyze seminal plasma metabolic profiles in idiopathic/male factor infertilityDistinct metabolic profiles in idiopathic infertility: Altered lysine, arginine, tyrosine, citrate, fructose103Observational comparative studyJayaraman et al., 2014 [33]
  • Untargeted metabolomics
Correlate metabolomic profiles with semen quality in young menMetabolites including acyl-carnitines and steroids distinguish low vs. high sperm count2700Observational cohort studyOlesti et al., 2023 [34]
  • LC-HRMS
  • Extraction optimization

Develop steroidomics strategy for human seminal fluidDetected 41 steroids including androgens; steroid profile stable7Method development and validation studyOlesti et al., 2020 [35]
  • 1H NMR AND GC-MS
  • 2D NMR SPECTROSCOPY

Comprehensive metabolomic characterization of human sperm cellIdentified 69 metabolites
Pathways linked to sperm physiology and dysfunction
N/A *Methodological studyPaiva et al., 2015 [36]
  • GC-MS
Metabolic profiling of unexplained male infertility (UMI)Identified 44 differential metabolites
Metabolic pathways altered
N/A *Two-stage population observational studyQiao et al., 2017 [37]
  • 1H MRS scanning of sperm
Examine sperm moleculesDensity gradient concentration (DGC) with two washes minimized contamination
Metabolite peaks differed between sperm populations
20Experimental observational studyReynolds et al., 2017 [38]
  • Untargeted GC-MS metabolomics
Metabolic profiling of idiopathic asthenozoospermia sperm cells33 metabolites identified
Disturbed energy and amino acid metabolism
213Experimental observational studyZhao et al., 2018 [39]
  • HPLC-MS/MS metabolomics
Semen metabolic profiling in oligospermia patients72–89 metabolites as potential markers
Altered amino acid and ketone body metabolism
40Comparative observational studyZhao et al., 2024 [40]
(D) Imaging, functional assays, and other techniques used in sperm metabolism and male infertility
Methods/ToolsStudy AimResultsSample sizeStudy TypeReferences
  • Fluorescence microscopy
Quantify testicular sperm apoptosis via active caspase-3 in normal and impaired spermatogenesisHigher active caspase-3 rates in obstructive azoospermia (OA) and hypospermatogenesis
Distinct localization patterns
24Observational comparative studyAlmeida et al., 2011 [41]
  • High-resolution respirometry
  • Oxidative/nitrative protein detection

Analyze mitochondrial function and oxidative stress in human sperm affecting fertilityMitochondrial dysfunction correlated with reduced motility with nitro-oxidative modifications in midpiece and headN/A *Experimental observational studyCassina et al., 2015 [42]
  • High-resolution respirometry
  • H2O2 measurement
  • Antioxidant capacity assays


Assess sperm mitochondrial metabolism and ROS production as tools to complement semen analysisMitochondrial respiratory control ratio correlates with motility; H2O2 inversely correlatedN/A *Experimental observational studyIrigoyen et al., 2022 [43]
  • ELISA
  • Immunohistochemistry

Assess impact of seminal clusterin level on spermatogenesis and sperm retrieval in infertile menSeminal clusterin lower in NOA and oligozoospermia
Correlation with testicular expression
FSH and clusterin predict retrieval
89Observational clinical studyFukuda et al., 2016 [44]
  • JC-1 fluorometric staining
Correlate sperm mitochondrial integrity with motilityPositive correlation between mitochondrial membrane potential and sperm motility213Experimental studyPaoli et al., 2011 [45]
  • Raman microspectroscopy with principal component analysis
Assessement of sperm DNA damage and biochemical featuresDNA damage detected only in dry conditions
Technique applicable to live sperm analysis
N/A *Experimental methodological studyCosta et al., 2018 [46]
  • Raman spectroscopy
  • Immunohistochemistry
  • Metabolomic analysis


Identify spermatogenesis in testicular tissueRaman spectra distinguished NOA from OA with ~90% sensitivity and 86% specificity52Observational diagnostic studyLiu et al., 2014 [47]
  • Chemiluminescence
  • Flow cytometry with fluorescent probes

Determine cell type contributions to intracellular H2O2 and peroxynitrite production in spermCell-type specific ROS production
H2O2 and peroxynitrite correlated
197Prospective observational studyAziz et al., 2010 [48]
  • Fluorescence imaging;
  • Whole-cell current clamp
  • Pharmacological inhibitors


Progesterone-induced Ca2+ oscillations in human spermCa2+ oscillations generated by CatSper channels
Membrane potential modulates oscillations
20Laboratory experimental studyNitao et al., 2021 [49]
  • Assays for 21 analytes in seminal plasma
  • ROC analysis

Evaluate seminal plasma biochemical and immunological markers in male infertility15 markers significantly altered
Monocytes/lymphocytes in NOA
Platelets in asthenozoospermia
100Observational case-control studyVashisht et al., 2021 [50]
* N/A: The information was not available in the original study.
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

Zakaria, A.; Diawara, I.; Bouziyane, A.; Louanjli, N. Exploring Human Sperm Metabolism and Male Infertility: A Systematic Review of Genomics, Proteomics, Metabolomics, and Imaging Techniques. Int. J. Mol. Sci. 2025, 26, 7544. https://doi.org/10.3390/ijms26157544

AMA Style

Zakaria A, Diawara I, Bouziyane A, Louanjli N. Exploring Human Sperm Metabolism and Male Infertility: A Systematic Review of Genomics, Proteomics, Metabolomics, and Imaging Techniques. International Journal of Molecular Sciences. 2025; 26(15):7544. https://doi.org/10.3390/ijms26157544

Chicago/Turabian Style

Zakaria, Achraf, Idrissa Diawara, Amal Bouziyane, and Noureddine Louanjli. 2025. "Exploring Human Sperm Metabolism and Male Infertility: A Systematic Review of Genomics, Proteomics, Metabolomics, and Imaging Techniques" International Journal of Molecular Sciences 26, no. 15: 7544. https://doi.org/10.3390/ijms26157544

APA Style

Zakaria, A., Diawara, I., Bouziyane, A., & Louanjli, N. (2025). Exploring Human Sperm Metabolism and Male Infertility: A Systematic Review of Genomics, Proteomics, Metabolomics, and Imaging Techniques. International Journal of Molecular Sciences, 26(15), 7544. https://doi.org/10.3390/ijms26157544

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