Skip to main content
Translational Andrology and Urology logoLink to Translational Andrology and Urology
. 2025 Oct 28;14(10):3387–3401. doi: 10.21037/tau-2025-440

Antidiabetic agents and male fertility: unraveling the impact of glucose-lowering therapies on sperm quality in diabetic males—a narrative review

Zuoxi Luo 1, Li Wang 1, Jinli Luo 1, Li Shi 1, Lina Wang 1, Guangmei Xie 1,
PMCID: PMC12603833  PMID: 41230150

Abstract

Background and Objective

Diabetes mellitus imposes a substantial burden on male fertility, adversely affecting sperm quality through structural damage in the reproductive tract, endocrine disruption, and dysregulation of cellular metabolism. While glucose-lowering agents help achieve glycemic control, they may also ameliorate diabetes-induced impairment of sperm quality. However, these drugs exert heterogeneous effects on male reproduction via divergent mechanisms, with outcomes often varying between in vivo and in vitro settings. This review aims to systematically evaluate the impacts of different antihyperglycemic therapies on semen quality and elucidate their mechanistic bases, to guide therapy selection for improving fertility in diabetic men.

Methods

A comprehensive search of the PubMed and Embase databases was conducted, covering the period from their inception to 2025.

Key Content and Findings

Insulin enhances sperm DNA integrity via Hsp70-2/TP-1/TP-2 upregulation and acts as a cryoprotectant. Metformin activates AMPK-mediated autophagy and synergizes with zinc to preserve fertility. SGLT-2 inhibitors suppress testicular apoptosis via Nrf2/HO-1 antioxidant pathways. Critically, sulfonylureas improve in vivo sperm quality but exert spermicidal effects in vitro by disrupting calcium homeostasis.

Conclusions

Current evidence supports the potential of certain antihyperglycemic agents in mitigating diabetes-related male infertility, though drug-specific and context-dependent effects must be considered. Further phenotype-targeted studies are needed to optimize clinical treatment strategies.

Keywords: Diabetes mellitus, male infertility, antidiabetic drugs, sperm quality, personalized medicine

Introduction

Infertility is increasingly recognized as a critical public-health challenge that substantially impedes demographic expansion; its global prevalence currently approximates 10–15%. Concurrently, a recent modelling study forecasts that the age-standardized worldwide prevalence of male infertility will escalate at a markedly accelerated rate compared with that of female infertility during the period 2022–2040 (1). Among these, the population of infertile men concomitantly afflicted with diabetes mellitus is exhibiting an escalating trend (2), and a substantial body of investigations has already delineated the deleterious impact of diabetes on human spermatozoa (3-6). The blood-testis barrier (BTB), one of the most impermeable vascular barriers, exerts stringent control entry of molecules into the seminiferous epithelium. Diabetic insults compromise male fertility via oxidative stress-mediated structural disruption of the BTB, thereby precipitating a perturbed testicular immune milieu. Supporting cells not only constitute the physical framework of the BTB but also deliver indispensable nutritional support for spermatogenic progression; consequently, any diabetes-induced metabolic derangement within these BTB-forming cells can disturb spermatogenesis and ultimately impair sperm quality (7,8). In addition, diabetes mellitus also affects spermatogenesis and development by altering the secretory activity of the hypothalamic-pituitary-testicular (HPT) axis (9,10). In the testicular stage, this impairment is mediated through reduced testosterone secretion (11); in the hypothalamic stage, low insulin levels lead to reduced levels of leptin, and consequently, gonadotropin-releasing hormone (GnRH) secretion, and subsequently, reduced luteinizing hormone (LH) and follicle-stimulating hormone (FSH) signaling to the testis, which in turn causes secondary hypogonadism (12-14). There are particular effects on sperm viability, DNA fragmentation, and chromatin quality (15,16).

In a clinical investigation, both lean and obese diabetic individuals exhibited markedly attenuated conventional semen parameters alongside significantly reduced circulating total testosterone and sex hormone-binding globulin concentrations relative to normoglycemic, normal-weight controls; concurrently, serum leptin concentrations were elevated, whereas serum insulin levels displayed inverse correlations with sperm concentration, viability and morphology (17). In a subsequent case-control study, patients with type 1 diabetes mellitus demonstrated a higher prevalence of infertility and erectile dysfunction, accompanied by impaired conventional semen indices—most notably diminished semen volume, total sperm count and progressive sperm motility. Additionally, type 1 diabetic men exhibited an increased incidence of positive anti-sperm antibodies in seminal plasma (18).

Antihyperglycemic agents, while executing both direct and indirect glycemic control at the systemic level, simultaneously exert precise regulatory effects on sperm viability, morphological integrity, and DNA fragmentation indices. These pharmacotherapeutic entities not only enhance intrinsic semen quality in vivo, but also serve as exogenous “sperm agonists” under in vitro conditions, thereby imparting measurable protective advantages to cryopreserved-thawed spermatozoa. The mechanistic repertoire underpinning these actions encompasses the modulation of testicular redox equilibrium, the preservation of acrosomal membrane structural integrity, and the fine-tuning of mitochondrial bioenergetics within spermatozoa; these phenomena have been comprehensively delineated across both experimental animal paradigms and human clinical studies. As shown in Figure 1, there are mechanisms of the effect of diabetes mellitus on sperm quality in males. We present this article in accordance with the Narrative Review reporting checklist (available at https://tau.amegroups.com/article/view/10.21037/tau-2025-440/rc).

Figure 1.

Figure 1

Mechanisms of the effect of diabetes mellitus on sperm quality in males. BTB, blood-testis barrier; FSH, follicle-stimulating hormone; GnRH, gonadotropin-releasing hormone; HPT, hypothalamic-pituitary-testicular; LH, luteinizing hormone; NO, nitric oxide.

Methods

We conducted a literature review of the most relevant articles on the overview topic within the PubMed and Embase databases, limiting the search scope to the period from database inception to May 2025 to ensure capture of the latest relevant research. Search keywords included “diabetes”, “sperm”, “antidiabetic drugs”, “biguanides”, “sulfonylureas”, “glucagon-like peptide-1 (GLP-1) receptor agonists”, “SGLT-2 inhibitors”, “thiazolidinediones”, and “insulin”. To ensure comprehensive coverage, we manually reviewed reference lists of articles to include additional papers relevant to the topic. Our screening process comprised two stages: title/abstract screening and full-text screening. During the title/abstract screening phase, three independent researchers conducted an initial review of each document’s title and abstract to exclude studies irrelevant to the research topic. In the full-text screening phase, three researchers independently assessed the full text of each article and screened them according to predetermined inclusion and exclusion criteria. The summary of the search strategy is shown in Table 1 (all included studies are compiled in Table S2).

Table 1. The search strategy summary.

Items Specification
Date of search June 5, 2025
Databases PubMed and Embase
Search terms used “diabetes”, “sperm”, “antidiabetic drugs”, “biguanides”, “sulfonylureas”, “glucagon-like peptide-1 (GLP-1) receptor agonists”, “SGLT-2 inhibitors”, “thiazolidinediones”, and “insulin” (for details on the PubMed database search strategy, please refer to Table S1)
Timeframe Database inception–May 2025
Inclusion and exclusion criteria (I) Including randomized controlled trials, clinical trials, animal studies, case reports, and systematic reviews concerning antidiabetic drugs and male sperm quality. (II) Duplicate studies and studies without English versions were excluded
Selection process During the title/abstract screening phase, Z.L., L.S., Lina Wang conducted an initial review of each document’s title and abstract to exclude studies irrelevant to the research topic. In the full-text screening phase, J.L., L.S., Lina Wang independently assessed the full text of each article and screened them according to predetermined inclusion and exclusion criteria

Insulin

Insulin is a protein hormone secreted by pancreatic β-cells in the pancreatic organ of the body, which dominates the blood glucose regulation of the body. In type I diabetes mellitus, autoimmune- and genetically-mediated destruction of pancreatic β-cells results in an absolute deficiency of insulin secretion that is insufficient to maintain euglycemia (19); conversely, in type II diabetes mellitus, the dysregulation of blood glucose arises from a state of relative insulin secretory insufficiency coupled with peripheral insulin resistance (20). Persistent hyperglycemia constitutes a detrimental prognostic indicator for male seminal quality, whereas exogenous insulin replacement therapy has been demonstrated to elicit favorable and clinically meaningful improvements in sperm parameters.

In an in vivo study, Guo et al. found that insulin can up-regulate the expression of lipocalin (an insulin sensitizing hormone, which can be secreted locally by the testis to regulate testicular function through the autocrine/paracrine pathway) and its receptor and down-regulate oxidative stress and inflammatory cytokine levels, and that lipocalin binds to its receptor and activates and phosphorylates the AMPK, thus regulating the activity of steroidogenic enzymes (6). This regulates the activity of steroidogenic enzymes, which controls the synthesis of steroid hormones and regulates sperm production and reproductive system function, as well as inhibiting the inflammatory response and decreasing inflammatory cytokines in order to increase testosterone secretion by Leydig cells. In addition, it was found that insulin may promote NO release and regulate testicular microcirculation by modulating the Akt/eNOS/NO signaling pathway, exerting a protective effect on the testes of diabetic rats, improving the diabetes-induced low levels of sperm counts, viability, serum and testicular testosterone, as well as lipocalin and its receptor, and decreasing the level of reactive oxygen species (ROS) in the testes of rats (21,22). Meanwhile, it has been shown that insulin intervention in combination with the micronutrients zinc, Omega-3 and moderate-intensity exercise showed better synergistic effects on sperm quality than insulin alone, mainly in terms of sperm morphology, counts, viability, DNA integrity and testosterone production (23-25). When utilizing insulin in combination with other therapies to modulate sperm quality, careful consideration must be given to exposure dose and duration. For instance, low zinc levels are associated with impaired spermatogenesis and male infertility, whereas elevated zinc levels can induce oxidative stress and other deleterious changes contributing to infertility (26). Similarly, moderate-intensity exercise training protects testicular Leydig cells from diabetes-induced apoptosis, thereby maintaining testicular cellular integrity and promoting spermatogenesis. However, when combining exercise with insulin therapy, the antioxidant effects of exercise on sperm quality must be accounted for. The above study indicates that while this exercise-mediated mechanism independently improves spermatogenesis, primarily through enhanced testosterone production, it more significantly potentiates the beneficial effects of insulin on sperm parameters (24).

In in vitro studies, the addition of insulin and leptin to washed human sperm increased sperm total motility, forward motility, and NO production, thereby increasing in vitro sperm fertilization; However, it is worth exploring the fact that, although leptin is involved in the regulation of reproduction-related hormones, its direct effects on sperm motility and sperm morphology are controversial. Lampiao et al. demonstrates that the synergistic effect of leptin and insulin on enhancing sperm fertilization potential primarily manifests through modulation of sperm signaling and function during capacitation (27), but it is not clear whether this positive effect is an effect on the process of capacitation or a stimulation of the acrosome reaction itself (28). Insulin can also be used as a sperm agonist to improve the quality of frozen and thawed spermatozoa. Diogo et al. found that spermatozoa from diabetic males exhibited a significant reduction in sperm viability, plasma membrane activity, and DNA integrity (15). After cryopreservation of spermatozoa, it exhibits no elongation of telomere length; nevertheless, the procedure triggers oxidative stress via both physical and chemical pathways, resulting in an elevated proportion of oxidized DNA fragmentation, concomitant with reductions in sperm viability and the percentage of morphologically normal cells (29,30). Shokri et al. found that the addition of different doses of insulin to frozen spermatozoa reduced the level of ROS in sperm mitochondria, and in particular, the 1,000 ng/mL dose of insulin significantly reduced DNA fragmentation rate and increased sperm viability, implying that insulin needs in high levels to induce protection against sperm DNA fragmentation. However, the above studies are, after all, for animal experiments, in human spermatozoa has not been explored, and the lack of mechanistic studies on the effect of insulin on spermatozoa in vitro, it remains to be further clarified the function of insulin on human semen in vitro (31).

The molecular protective mechanism of insulin in hyperglycemic sperm DNA is mainly realized by enhancing the antioxidant capacity of spermatozoa, and it has been demonstrated that insulin treatment improves the expression of Hsp70-2a, TP-1 and TP-2, and enhances the role of Hsp90 in the DNA repair process, and TP-1 and TP-2 are mainly involved in the chromatin composition and precision changes of haploid spermatozoa, whereas the Hsp70-2a and Hsp90 of the heat shock protein family are involved in epigenetic processes related to nuclear chromatin condensation, and all of the above pathways contribute to the maintenance of sperm DNA integrity (32-34). At the gene expression level, it has been suggested that type I diabetes inhibits B-cell lymphoma-2 (Bcl-2) and increases BAX expression, initiating intrinsic apoptosis signaling through caspase-3 overexpression, and that insulin inhibits type I-triggered intrinsic apoptosis in the testis by maintaining preexisting Bcl-2 protein and inhibiting BAX overexpression and thus caspase-3 expression; The Physiological levels of cell cycle protein D1 interact with its kinase Cdk-4 to help accelerate the cell cycle transition from G1 to S phase, whereas its overexpression or under-expression promotes apoptosis, and insulin improves testicular cell proliferation by restoring diabetes-induced deficiencies in cell cycle protein D1; Moreover, diabetic conditions upregulate p53—a DNA damage checkpoint protein in the G1 phase that promotes apoptosis upon DNA damage. Conversely, insulin therapy suppresses p53-dependent pathways, inhibiting G1 cell cycle arrest and/or apoptotic onset (35).

Biguanides

Biguanide antihyperglycemic agents represent the first-line pharmacotherapy for type II diabetes mellitus, and their principal glucose-lowering mechanisms encompass: (I) enhancement of anaerobic glycolysis within skeletal muscle; (II) inhibition of hepatic gluconeogenesis; (III) augmentation of peripheral glucose uptake and glycolytic flux (36); and (IV) amplification of systemic insulin sensitivity (37). Type II diabetes mellitus constitutes the predominant form of diabetes among infertile males, and biguanides have accordingly been extensively prescribed in this population. Over recent years, a growing body of clinical investigations and animal studies has begun to examine the impact of biguanide-based hypoglycemic therapy on human and experimental spermatozoa. However, the majority of these works have concentrated on seminal quality in obesity-related male infertility, whereas investigations specifically targeting sperm parameters within diabetic males remain comparatively scarce (38,39).

In in vivo studies, Naghibi et al. found metformin significantly improves sperm quality, increases testosterone levels, and reduces levels of ROS, glucose, and BAX to Bcl-2 ratios in blood and testicular tissues in diabetic rats. The levels of BAX to Bcl-2 ratios correlate with apoptosis, and a decrease in their levels inhibited apoptosis in testicular cells, which promoted the preservation of male fertility. The combination of metformin and mauve forskolin especially had significant effects on testosterone levels and antioxidant capacity, but for other routine sperm parameters there was little change compared to the two alone (40). Metformin augments testicular antioxidant capacity primarily via AMPK activation, whereby oxidative stress within the testis is attenuated through both AMPK-dependent and AMPK-independent signaling cascades. In parallel, accumulating evidence demonstrates that metformin further mitigates testicular oxidative injury by reducing the levels of membrane lipid-peroxidation product malondialdehyde (MDA) and apoptotic marker caspase-3 (41), concomitant with a decrease in the BAX/Bcl-2 ratio, thereby inhibiting germ-cell apoptosis and preserving testicular cytoarchitecture. Forskolin likewise attenuates oxidative stress while promoting testosterone biosynthesis and spermatogenesis; nevertheless, it does not confer a significant synergistic advantage when combined with metformin, possibly attributable to disparities in exposure duration or dosage, or to an as-yet-unidentified antagonistic interaction that warrants further investigation (40). A study by Zhao et al. found a significant increase in sperm motility, sperm membrane functional integrity and acrosome integrity with the addition of 150 mg/kg dose of metformin to the diet of male goats. In contrast, the addition of metformin at a dose of 300 mg/kg to the diet of male goats revealed a significant increase in catalase (CAT), glutathione peroxidase (GSH-px) and superoxide dismutase (SOD) activities, as well as AMPK and p-AMPK protein expression. It is through the activation of AMPK, Belicin-1 and LC3II/I protein expression that metformin maintains sperm energetic homeostasis and regulates autophagy and oxidative stress, decreasing ROS levels and improving sperm quality and plasma membrane integrity. Furthermore, the present study revealed that serum testosterone concentrations exhibited a progressive decline as the metformin dose escalated, implying that an optimal therapeutic window, rather than simple dose escalation, must be identified to maximize patient benefit during in vivo metformin therapy. Concomitantly, the protein expression levels of key autophagy markers beclin-1 and LC3-II/I were up-regulated in a dose-dependent manner, indicating that metformin alleviates oxidative stress by facilitating the autophagic clearance of senescent or aberrant cells, thereby attenuating oxidative injury within the testicular microenvironment (42). The combination of metformin and the trace element zinc promotes a significant increase in the expression of blood-epididymis barrier (BEB)-related molecules (including ZO-1, β-catenin, and N-cadherin) as well as water channel proteins (AQP3, AQP9, and AQP11) in epididymal tissues, which ameliorates diabetes-induced imbalance of epididymal zinc homeostasis, BEB, and uptake dysfunction, and consequently increases spermatozoa. There are studies found that metformin combined with zinc could correct diabetes-induced zinc homeostasis imbalance and improve sperm quality by activating the PI3K/AKT/mTOR pathway and improving the expression of related proteins, such as PI3K, p-AKT/AKT and p-mTOR/mTOR (43,44). Meanwhile, it was found that PI3K, p-AKT/AKT, and p-mTOR/mTOR are key signaling pathway proteins in the regulation of autophagy, and their increased expression contributes to the elimination of testicular senescent and abnormal cells and the reduction of oxidative stress induced by cellular metabolites (45).

In in vitro study, Rindone et al. found that metformin interfered with the metabolism and tight composition of testicular supporting cells during BTB formation, did not impair BTB permeability, and did not affect spermatogenesis (46). Metformin also plays a positive role in the improvement of sperm quality; the viability of freeze-thawed spermatozoa mainly depends on the amount of sperm adenosine triphosphate (ATP) stored before activation. During sperm freezing and thawing, the mitochondria will be impaired due to a significant decrease in high membrane potential affecting the production of ATP. Thus, maintaining sufficient ATP content during storage will be beneficial to thawed spermatozoa viability enhancement, similar to the in vivo mechanism of action. Metformin can improve sperm viability and fertilization ability while prolonging the storage capacity of spermatozoa in vivo through activation of the AMPK pathway. Moreover, similar to its in vivo mechanism, metformin maintains sperm ATP content by activating AMPK expression and enhancing glucose uptake, thereby increasing sperm viability and fertilization potential and further extending the storage capacity of spermatozoa in vitro (47). Low concentrations of metformin exert no deleterious effects on fresh spermatozoa; however, exposure to high concentrations significantly compromises fresh sperm viability. Importantly, supplementation of cryopreservation medium with metformin not only enhances post-thaw sperm motility and fertilization competence, but also diminishes the incidence of abnormal zygotes and reduces lipid peroxidation following in vitro fertilization. Lipid peroxidation of the sperm plasma membrane precipitates impaired motility and viability, and concurrently disrupts the spermatozoon’s ability to bind to the zona pellucida. The observed improvement in embryonic quality conferred by metformin may be attributable to enhanced genomic stability of the male gamete. Certainly, the concentration of metformin tolerated by spermatozoa varies between species, and the optimal concentration for human spermatozoa needs to be further investigated (48,49). Meanwhile, metformin has been found to limit mitochondrial respiration and reduce oxidative stress in spermatozoa in vitro; 50 μM metformin improves sperm viability and increases spermatozoa mitochondrial activity and nicotinamide adenine dinucleotide+ (NAD+) content, which is positively correlated with the functional state of the mitochondria, and the NAD+ content decreases in case of mitochondrial damage or dysfunction (50).

However, the use of metformin in diabetic men is controversial, with Wensink et al. noting in a cohort study that metformin takers were at increased risk of birth defects in their offspring, with a particular increased risk of developing genital defects (51). Controversially, Rotem et al. in a cohort study noted the safety of its use and found that metformin monotherapy did not increase the risk of birth defects in newborns (52). Obesity and diabetes often go hand in hand, McPherson et al. found that short-term metformin treatment in men who are obese could be a potential intervention for the treatment of subfertility and increase fetal weights and lengths (53). Further validation of the safety of metformin on birth defects in offspring is still needed from a large number of clinical studies and evidence-based medicine. As shown in Figure 2, there are mechanisms by which metformin and insulin improve sperm quality in diabetes males.

Figure 2.

Figure 2

Mechanisms by which metformin and insulin improve sperm quality in diabetic males. BEB, blood-epididymal barrier.

Sulfonylureas

Sulfonylurea hypoglycemic agents belong to insulinotropic agents, which stimulate insulin secretion to lower blood glucose by blocking ATP-sensitive K+ channels and cAMP-dependent signaling proteins on β-cell membranes (54,55), and common medications include glibenclamide, glipizide, gliclazide, gliquidone, and glimepiride.

In an in vivo study, Shokri et al. demonstrated that glipalamide significantly enhanced fundamental sperm parameters, reduced testicular MDA concentrations, and elevated systemic antioxidant capacity in diabetic rats. The drug also mitigated diabetes-induced pathological alterations, as evidenced by decreased testicular varicocele diameter, restoration of spermatogenic epithelial height, and overall improvement in testicular histological architecture. However, the mechanistic relationship between glipalamide-mediated suppression of ROS and the reversal of testicular histological lesions was not investigated in the study (56). Animal experiments revealed that the sulfonylurea antidiabetic drugs glimepiride and glibenclamide could increase the normal sperm morphology rate and enhance the antioxidant capacity of the testis, and glibenclamide could not only reduce the lipid peroxidation of sperm plasma membrane but also increase the levels of the antioxidant enzymes SOD, CAT, and glutathione-s-transferase (GST). Rabbani et al. suggested that glibenclamide might reduce xanthine oxidase reaction by inhibiting the ATP-sensitive potassium channel, and then decrease the production of ROS, which in turn reduces the abnormal oxidative stress caused by the abnormal sperm morphology. ATP-sensitive potassium channels reduce the xanthine oxidase reaction and the production of ROS, thereby reducing the oxidative stress that causes spermatozoa morphological abnormalities (57,58).

Glibenclamide has strong systemic antioxidant activity outside of the reproductive system and can reverse the diabetic-induced decrease in CAT and SOD activity in the liver, thereby reducing systemic ROS production (59,60). However, it is noteworthy that the combination of pioglitazone and glimepiride did not improve the diabetes-induced sperm morphology abnormalities and oxidative stress state, probably due to the lack of free radical clarity, which might be related to the dose and duration of exposure to the combination of the two (61). Öztaş et al. reported that glipizide elevated sperm count and the proportion of morphologically normal spermatozoa while ameliorating testicular architecture in diabetic rats, yet exerted no discernible influence on the HPT axis. Notably, testosterone concentrations remained persistently low in glipalamide-treated diabetic animals, implying that glipalamide may confer protection to the GnRH-LH signaling cascade of the HPT axis against diabetes-induced damage, thereby preserving physiological testosterone levels. Consequently, the observed improvements in spermatozoa quality were primarily ascribed to glipalamide-mediated enlargement of testicular seminiferous tubule diameter and restoration of inter-Sertoli-cell tight-junction integrity (62).

Glibenclamide can increase GSH, CAT and SOD activities and reduce ROS levels, and carrying glibenclamide through nanocarriers can increase the drug dissolution rate, which can significantly reduce ROS levels in type 2 diabetes compared with glibenclamide alone. In addition, nanocarriers carrying glibenclamide can increase the transcriptional levels of Nrf2, HO-1, and GPx4 through Nrf2/HO-1 and GPx4 signaling pathways, and protect sperm cell membranes from oxidative stress damage through adenylate-rich uridine, which is important for spermatogenesis and motility. GPx4 transcript levels, resist oxidative stress and protect sperm cell membranes from oxidative stress damage, Nrf2 plays an important role in spermatogenesis and motility, and stimulates antioxidant enzymes to reduce ROS levels through adenylate-uridylic acid-rich elements (63,64). Glibenclamide nano-carrying shows significant clinical benefits and provides direction for new drug development and fertility protection in diabetic men.

However, the in vitro effects of sulfonylurea hypoglycemic agents on spermatozoa diverge markedly from those of insulin and bisphosphonates. Spermatozoa function within the female reproductive tract is critically dependent on calcium-channel modulation. When sulfonylureas such as glipalamide or gliclazide are added to semen in vitro, they exert a dual, opposing action: first, they block ATP-sensitive K+ channels, provoking membrane depolarization and subsequent opening of voltage-gated calcium channels, which promotes calcium influx; second, they simultaneously inhibit the Na+-Ca2+ exchanger, thereby preventing Ca2+ efflux from spermatozoa. The net consequence is a sustained elevation of intracellular Ca2+ concentration and disruption of calcium homeostasis, ultimately compromising sperm viability and, in extreme cases, inducing spermatozoa death (65,66). The in vitro properties of this class of drugs can mostly be utilized in male contraception and in vitro spermicide.

GLP-1 receptor agonists

GLP-1 is a multifunctional peptide hormone that regulates blood glucose, and GLP-1 receptor agonists commonly used in clinical practice, including somatostatin, liraglutide, dulaglutide, etc. The hypoglycemic mechanisms of GLP-1 receptor agonists include promotion of insulin secretion and inhibition of glucagon secretion, and they can inhibit gastrointestinal motility and reduce appetite (67,68).

Pourheydar et al. demonstrated that, under physiological conditions, liraglutide may exert deleterious effects on the normal metabolic machinery of germ cells, thereby eliciting excessive generation of ROS and compromising sperm DNA integrity. This adverse outcome is presumably attributable to the route-dependent administration of liraglutide, which inhibits the endogenous redox system and subsequently impairs the scavenging of ROS derived from aberrant cellular metabolism. In diabetic mice, neither 1.20 nor 1.8 mg/kg liraglutide significantly increased sperm survival or viability; nevertheless, both doses effectively ameliorated diabetes-induced impairments in sperm DNA integrity and chromatin condensation. These findings imply that, despite its insulin-mimetic properties, liraglutide fails to attenuate diabetes-evoked, mitochondria-dependent apoptosis, thereby conferring no substantial improvement on overall spermatogenesis or seminal quality (69). However, a recent animal study found that liraglutide failed to affect sperm concentration and viability, both in vivo and in vitro, and that sperm concentration was instead increased in mice if GLP-1 receptor was absent, further validating the negative effects of liraglutide on germ cell metabolism under physiological conditions (70). The improvement of male sperm quality by liraglutide was mostly found in animal studies in obesity models (71,72). Although in a randomized open clinical study, somatostatin was found to significantly improve sperm quality in patients with obesity combined with type 2 diabetes mellitus, which not only increased the normal sperm morphology, concentration and number of subjects, but also increased total testosterone and libido (73). However, this study does not directly prove whether the improvement in sperm quality acts on obesity-induced sperm damage mechanisms, diabetes-induced sperm damage mechanisms, or both interventions, and further studies are needed.

A meta-analysis showed that GLP-1 receptor agonists do not act directly to intervene in testicular function (74); meanwhile, in a randomized, double-blind, placebo-controlled crossover study, it was found that the GLP-1 receptor agonist, dulaglutide, did not significantly intervene in the secretion of hormones of the Hypothalamic-Pituitary-Ovarian (HPO) axis (75). It has been hypothesized that the effect of GLP-1 receptor agonists on sperm quality may be either direct by triggering sperm GLP-1 receptor expression or indirect through paracrine bursts of mesenchymal cells acting on the GLP-1 receptor present in supporting cells (76,77). However, evidence from relevant studies is still lacking to support that the mechanism of action of GLP-1 receptor agonists on spermatozoa needs to be further explored.

SGLT-2 inhibitors

Sodium-glucose cotransporter proteins are active transport proteins that carry out isotropic transport of glucose relying on the difference in sodium ion concentration gradient on both sides of the cell membrane (78). SGLT-2 inhibitors lower blood glucose mainly by blocking glucose reabsorption in the kidney. It has also been demonstrated that SGLT-2 inhibitors have a positive effect on male spermatozoa as well. The common clinical drugs include dapagliflozin and empagliflozin.

In an animal study, it was found that empagliflozin improved oxidative stress, sperm quality and pathological changes in testicular tissues, and indirectly modulated serum sex hormone, insulin, leptin levels and expression of kisspeptin (a hypothalamic neuropeptide hormone involved in the regulation of energy metabolism, GnRH, and reproductive function) in testicular tissues in diabetic rats, which enhances fertility (79). Kiani et al. found that empagliflozin improved the antioxidant capacity of spermatozoa by increasing the viability of SOD, as well as significantly lowering the values of single-stranded DNA (AO+), immature nucleated spermatozoa (AB+), and the BAX/Bcl-2 ratio to improve sperm quality and reduce apoptosis. The improvement of sperm viability by Empagliflozin is mainly due to the reduction of oxidative stress, the increase of ATP content required by spermatozoa, and the modulation of calcium levels. It is worth noting that empagliflozin, while improving the morphological abnormalities of the head of the spermatozoa, increases the morphological abnormalities of the tail of spermatozoa, which may be related to the lowering of testicular cholesterol levels, and a high proportion of the spermatozoa tails are more susceptible to damage after lowering of cholesterol (80). In addition, dapagliflozin inhibited the testicular apoptotic process by up-regulating the expression of the anti-apoptotic proteins Bcl-2 and X-linked inhibitor of apoptosis (XIAP) and suppressed testicular apoptotic process by enhancing the total antioxidant capacity, total SOD activity, and GPx activity as well as by decreasing the level of 4-hydroxynonenal (4-HNE) oxidative stress (81).

In addition to the mechanism of action at the level of oxidative stress, at the chromosomal and genetic levels, diabetes increases the incidence of diploid nondisjunction or chromosomal defects in male spermatozoa, as well as disruption of genes such as Ogg1, Parp1, and P53, and dapagliflozin repairs diabetes-induced DNA damage thereby decreasing the oxidative stress response and reducing the rate of diabetes-induced spermatozoa aneuploidy (82). At the level of intestinal flora metabolism, dapagliflozin modulates microbial composition and cascade regulates the metabolism of adenosine, cAMP, and 2'-deoxyinosine and 2'-deoxyhypoxanthine in serum and testis, and decreases apoptosis and ROS production in spermatogenic cells. High levels of adenosine in the cecum, cAMP in plasma, and 2'-deoxyinosine in testis induces ROS production and apoptosis, in which cAMP is involved in oxidative stress and apoptosis through the cAMP-PKA signaling pathway (83). Based on the above studies, it can be found that SGLT-2 inhibitors protect male fertility and improve sperm quality mainly by responding to antioxidant stress, decreasing apoptosis and promoting DNA repair.

Meanwhile, the combination of SGLT-2 inhibitors and other hypoglycemic agents also showed positive improvements in sperm quality in diabetic males. Ribeiro et al. found that the combination of the GLP-1 agonist, exenatide, and dapagliflozin had a cumulative effect on the production of lactic acid and alanine in the testicular supportive cells and on glucose depletion, which was favorable for improving spermatogenesis and development. Alshamrani et al. found that the combination of dipeptidyl peptidase-4 (DPP-4) inhibitor saxagliptin and dapagliflozin reduced testicular ROS levels and increased glutathione levels to restore the gonadal redox imbalance in diabetic mice, but unlike dapagliflozin, saxagliptin exacerbated the decline in spermatozoa viability and number in diabetic mice. in diabetic mice, so caution should be exercised when combining the two (84,85). A randomized controlled trial exploring the effects of dapagliflozin on sperm quality in pre-diabetic individuals is currently underway, and further validation of the effects of SGLT-2 inhibitors on human fertility is awaited (86).

Thiazolidinediones

Thiazolidinediones hypoglycemic agents are insulin sensitizers that lower blood glucose by increasing insulin sensitivity and do not directly promote insulin secretion (87,88). Common clinical thiazolidinediones hypoglycemic agents include rosiglitazone and pioglitazone, and also act as peroxisome proliferator-activated receptor γ (PPARγ) agonists, which can affect spermatozoa quality by directly acting on spermatozoa PPARγ receptors (89).

In in vivo studies, Rabbani et al. found that higher dose (10 mg/kg) of pioglitazone significantly ameliorated the oxidative stress status of male spermatozoa and increased the normal morphology ratio of spermatozoa as well as sperm counts by decreasing the levels of serum lipid peroxide (LPO), GSH, and GPx in diabetic rats (90). Some studies have shown that pioglitazone can also reduce early and late sperm cell apoptosis by improving sperm mitochondrial activity and increase sperm viability and linear velocity, and pioglitazone has a time- and concentration-dependent effect in vivo, with lower concentrations of pioglitazone instead showing higher mitochondrial activity. At the same time, pioglitazone can act directly on damaged mitochondria, altering glycolytic metabolism and fuel Substrate-specific changes, increasing mitochondrial activity and reducing apoptosis (91,92). It is noteworthy that, in leptin-receptor-deficient obese mice, the PPARγ agonist pioglitazone failed to reverse diabetes-induced degeneration of the spermatogenic epithelium or to restore testicular architecture; instead, it diminished epididymal and testicular weight. Moreover, pioglitazone did not attenuate testicular lipid peroxidation or oxidative DNA damage in diabetic mice and further suppressed the base excision repair (BER) machinery within diabetic testes, as evidenced by down-regulation of the BER proteins 8-oxoguanine DNA glycosylase (OGG1/2), X-ray repair cross-complementary protein-1 (XRCC1), and DNA polymerase δ. Elevated levels of OGG1/2 and DNA polymerase δ suggest that the BER mechanism attempts to remove damaged bases and generate depurination/pyrimidineless sites and initiate single-strand breaks, exhibiting a negative effect on diabetic male spermatozoa. However, PPAR-γ stimulation in leptin receptor-intact lean mice reduced testicular ROS levels, suggesting that the reduction of testicular ROS by pioglitazone may be mediated through the leptin and its receptor-associated pathway, and that when leptin and its receptor-associated pathway are deficient, PPAR-γ stimulation instead exacerbates the production of ROS and exacerbates apoptosis (93,94). Thus, the in vivo effects of pioglitazone on diabetic male spermatozoa are controversial, and the effects on human male fertility require further clinical validation.

In in vitro studies, thiazolidinediones showed mostly positive effects on spermatozoa in vitro. The addition of rosiglitazone at a concentration of 60 µM to the frozen-thawed spermatozoa of rams formed a protective layer on the outside of the sperm membrane, protected the sperm membrane from lipid peroxidation caused by oxidative stress during freezing and thawing, and significantly improved sperm viability, sperm acrosomes and membrane integrity. It was mainly due to the regulation of the energy metabolism of spermatozoa and the protection of the mitochondrial membrane potential, while rosiglitazone reduced the level of ROS, mainly due to the activation of the AMPK-dependent mechanism to inhibit NAD(P)H oxidase. The reduction of ROS levels by rosiglitazone is mainly due to its activation of AMPK, an AMPK-dependent mechanism that inhibits NAD(P)H oxidase, and the hydroxylation of the phenyl and pyridine rings in the chemical structure of rosiglitazone, which may be able to remove hydroxyl radicals (95,96). Ortiz-Rodriguez et al. found that rosiglitazone reduces oxidative stress induced by the freezing process and regulates sperm mitochondrial function by increasing Akt phosphorylation and decreasing caspase 3 activation (97). Meanwhile, Rabbani et al. found that pioglitazone in combination with metformin improved the state of oxidative stress and increased the percentage of normal morphology due to diabetes (61). Further evidence of synergistic effects between hypoglycemic agents provides options for individualized diagnosis and treatment.

It is important to note that advanced age is a significant confounding factor in this context, as type II diabetes predominantly affects older men who are already experiencing age-related decline in fertility (98-100). This factor has been accounted for in Table S2.

Conclusions

To date, animal experiments, clinical studies, and meta-analyses focusing on the relationship between glucose-lowering drugs and male sperm quality have yielded different conclusions. Firstly, in in vivo studies, insulin, biguanide hypoglycemic agents, sulfonylureas, and SGLT-2 inhibitors have shown positive effects on improving sperm quality. However, the effects of GLP-1 receptor agonists and thiazolidinediones on sperm quality in vivo are controversial and further meta-analyses are needed to clarify their significance. Secondly, in in vitro studies, insulin, biguanide hypoglycemic agents and thiazolidinediones showed positive effects on in vitro sperm quality, however, sulfonylurea hypoglycemic agents showed negative effects on in vitro sperm quality.

Glucose-lowering drugs have different mechanisms of action in vivo and in vitro, and antidiabetic drugs play a dual role in male fertility. Insulin improves sperm quality by enhancing antioxidant capacity, upregulating DNA repair proteins (e.g., Hsp70-2, TP-1/2), and acting as a “sperm booster” in cryopreservation. Metformin maintains fertility through AMPK-mediated autophagy, reduction of oxidative stress, and synergistic effects with zinc through PI3K/AKT/mTOR signaling, and SGLT-2 inhibitors (e.g., empagliflozin, dapagliflozin) show promise in combating testicular apoptosis and ROS through metabolic reprogramming and antioxidant upregulation. In contrast, sulfonylureas (e.g., glibenclamide) exhibit spermicidal effects in vitro due to disruption of calcium homeostasis, despite their antioxidant effects in vivo. GLP-1 receptor agonists show contradictory results, potentially improving DNA integrity in diabetic models, but exhibit neutral or unfavorable effects on sperm metabolism in a physiological setting thiazolidinedione (e.g., pioglitazone) enhances sperm mitochondrial function in vitro but exhibits inconsistent efficacy in vivo.

The literature on this line of research is mostly animal experiments, rarely supported by clinical evidence, and the only relevant clinical studies are still in progress. The mechanisms and results explored in the current animal studies may provide support and direction for clinical studies, but the effects on sperm quality in human males need to be further explored in clinical studies.

In conclusion, antidiabetic drugs have great potential as adjuncts to fertility management in diabetic male. Currently, there are few or several studies aimed at exploring the effects of synergistic use of glucose-lowering drugs on sperm quality, such as the combination of metformin with zinc and insulin omega-3, which provides a new opportunity for exploring nanocarrier-delivered sulphonylureas to improve efficacy while mitigating off-target spermicidal effects. Also, the tissue-specific expression of GLP-1 receptor in testis and spermatozoa provides new directions to study the epigenetic modifications induced by antidiabetic drugs in germ cells. Clarifying the effects of different types of hypoglycemic agents on sperm quality in diabetic males will not only help to rationally avoid reproductive toxicity drugs, but also help to provide individualized treatment for infertile men.

Supplementary

The article’s supplementary files as

tau-14-10-3387-rc.pdf (94.9KB, pdf)
DOI: 10.21037/tau-2025-440
tau-14-10-3387-coif.pdf (225.7KB, pdf)
DOI: 10.21037/tau-2025-440
DOI: 10.21037/tau-2025-440

Acknowledgments

We appreciate the contributions made to this study by every member of the staff.

Ethical Statement: The authors are accountable for all aspects of the work in ensuring that questions related to the accuracy or integrity of any part of the work are appropriately investigated and resolved.

Footnotes

Reporting Checklist: The authors have completed the Narrative Review reporting checklist. Available at https://tau.amegroups.com/article/view/10.21037/tau-2025-440/rc

Funding: This research was supported by the Major Scientific Research Project for Technological Innovation in the Health Sector of Gansu Province (No. GSWSZD2025-18).

Conflicts of Interest: All authors have completed the ICMJE uniform disclosure form (available at https://tau.amegroups.com/article/view/10.21037/tau-2025-440/coif). The authors have no conflicts of interest to declare.

References

  • 1.Liang Y, Huang J, Zhao Q, et al. Global, regional, and national prevalence and trends of infertility among individuals of reproductive age (15-49 years) from 1990 to 2021, with projections to 2040. Hum Reprod 2025;40:529-44. 10.1093/humrep/deae292 [DOI] [PubMed] [Google Scholar]
  • 2.Graziani A, Scafa R, Grande G, et al. Diabetes and male fertility disorders. Mol Aspects Med 2024;99:101303. 10.1016/j.mam.2024.101303 [DOI] [PubMed] [Google Scholar]
  • 3.Delbarba A, Anelli V, Bambini F, et al. Type 1 diabetes mellitus and sperm quality: A case-control study. Andrology 2025;13:208-16. 10.1111/andr.13681 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4.Biasi A, Ambruosi MR, Romano MZ, et al. Impact of Type 1 Diabetes on Testicular Microtubule Dynamics, Sperm Physiology, and Male Reproductive Health in Rat. Int J Mol Sci 2025;26:4579. 10.3390/ijms26104579 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5.Carvalho RPR, Guimarães-Ervilha LO, Viana AGA, et al. A systematic review and meta-analysis of the effects of diabetes on human and murine epididymis. Andrology 2025. [Epub ahead of print]. doi: . 10.1111/andr.70026 [DOI] [PubMed] [Google Scholar]
  • 6.Guo Z, Yan X, Wang L, et al. Effect of Telmisartan or Insulin on the Expression of Adiponectin and its Receptors in the Testis of Streptozotocin-Induced Diabetic Rats. Horm Metab Res 2016;48:404-12. 10.1055/s-0042-101549 [DOI] [PubMed] [Google Scholar]
  • 7.He Z, Yin G, Li QQ, et al. Diabetes Mellitus Causes Male Reproductive Dysfunction: A Review of the Evidence and Mechanisms. In Vivo 2021;35:2503-11. 10.21873/invivo.12531 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.Lee JE, Lee CW, Han AR, et al. Human embryonic stem cell-derived Sertoli cells as an immune modulator of cell transplantation therapy in a diabetic mice model. Stem Cell Res Ther 2025;16:403. 10.1186/s13287-025-04532-x [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.Venditti M, Romano MZ, Boccella S, et al. Type 1 diabetes impairs the activity of rat testicular somatic and germ cells through NRF2/NLRP3 pathway-mediated oxidative stress. Front Endocrinol (Lausanne) 2024;15:1399256. 10.3389/fendo.2024.1399256 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.He Z, Liu T, He X, et al. Molecular mechanisms of libido influencing semen quality in geese through the hypothalamic-pituitary-testicular-external genitalia axis. Poult Sci 2025;104:104756. 10.1016/j.psj.2024.104756 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.Cignarelli A, Genchi VA, D'Oria R, et al. Role of Glucose-Lowering Medications in Erectile Dysfunction. J Clin Med 2021;10:2501. 10.3390/jcm10112501 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.Genchi VA, Rossi E, Lauriola C, et al. Adipose Tissue Dysfunction and Obesity-Related Male Hypogonadism. Int J Mol Sci 2022;23:8194. 10.3390/ijms23158194 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.George JT, Millar RP, Anderson RA. Hypothesis: kisspeptin mediates male hypogonadism in obesity and type 2 diabetes. Neuroendocrinology 2010;91:302-7. 10.1159/000299767 [DOI] [PubMed] [Google Scholar]
  • 14.Badejogbin OC, Akano OP, Julius OEB, et al. Potential Role of CREM in Diabetes-Associated Testicular Dysfunction: Current Evidence and Future Perspectives. Reprod Med Biol 2025;24:e12661. 10.1002/rmb2.12661 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Diogo P, Martins G, Simão M, et al. Type I Diabetes in Zebrafish Reduces Sperm Quality and Increases Insulin and Glucose Transporter Transcripts. Int J Mol Sci 2023;24:7035. 10.3390/ijms24087035 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Agbaje IM, McVicar CM, Schock BC, et al. Increased concentrations of the oxidative DNA adduct 7,8-dihydro-8-oxo-2-deoxyguanosine in the germ-line of men with type 1 diabetes. Reprod Biomed Online 2008;16:401-9. 10.1016/s1472-6483(10)60602-5 [DOI] [PubMed] [Google Scholar]
  • 17.AbbasiHormozi S , Kouhkan A, Shahverdi A, et al. How much obesity and diabetes do impair male fertility? Reprod Biol Endocrinol 2023;21:48. 10.1186/s12958-022-01034-w [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Longo M, Caruso P, Varro C, et al. Semen quality and metabolic profile in people with type 1 diabetes with and without erectile dysfunction: a cross-sectional study. J Endocrinol Invest 2024;47:1787-95. 10.1007/s40618-023-02285-z [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.Atkinson MA, Eisenbarth GS, Michels AW. Type 1 diabetes. Lancet 2014;383:69-82. 10.1016/S0140-6736(13)60591-7 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Patil PD, Mahajan UB, Patil KR, et al. Past and current perspective on new therapeutic targets for Type-II diabetes. Drug Des Devel Ther 2017;11:1567-83. 10.2147/DDDT.S133453 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Otčenášková T, Macíčková E, Vondráková J, et al. Proteomic analysis of the mouse sperm acrosome - towards an understanding of an organelle with diverse functionality. Eur J Cell Biol 2023;102:151296. 10.1016/j.ejcb.2023.151296 [DOI] [PubMed] [Google Scholar]
  • 22.Xu Q, Qiu L, Gu Q, et al. P407 hydrogel loaded with nitric oxide microbubbles promotes angiogenesis and functional improvement in testicular transplantation. Biomater Sci 2024;12:1004-15. 10.1039/d3bm01521a [DOI] [PubMed] [Google Scholar]
  • 23.Ujah G, Emmanuel IB, Ansa F, et al. Insulin and Zinc Co-Administration Ameliorate Diabetes Mellitus-Induced Reproductive Dysfunction in Male Rats. Niger J Physiol Sci 2022;37:49-58. 10.54548/njps.v37i1.7 [DOI] [PubMed] [Google Scholar]
  • 24.Samadian Z, Tofighi A, Razi M, et al. Moderate-intensity exercise training ameliorates the diabetes-suppressed spermatogenesis and improves sperm parameters: Insole and simultaneous with insulin. Andrologia 2019;51:e13457. 10.1111/and.13457 [DOI] [PubMed] [Google Scholar]
  • 25.Aminzadeh E, Haghpanah T, Ganjalikhan Hakemi S, et al. Omega-3 with and without insulin replacement alleviates detrimental effects of type II diabetes on reproductive system of male C57BL/6 mice. Gen Comp Endocrinol 2023;344:114370. 10.1016/j.ygcen.2023.114370 [DOI] [PubMed] [Google Scholar]
  • 26.Zečević N, Veselinović A, Perović M, et al. Association Between Zinc Levels and the Impact of Its Deficiency on Idiopathic Male Infertility: An Up-to-Date Review. Antioxidants (Basel) 2025;14:165. 10.3390/antiox14020165 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Lampiao F, du Plessis SS. Insulin and leptin enhance human sperm motility, acrosome reaction and nitric oxide production. Asian J Androl 2008;10:799-807. 10.1111/j.1745-7262.2008.00421.x [DOI] [PubMed] [Google Scholar]
  • 28.Chang B, Song C, Gao H, et al. Leptin and inflammatory factors play a synergistic role in the regulation of reproduction in male mice through hypothalamic kisspeptin-mediated energy balance. Reprod Biol Endocrinol 2021;19:12. 10.1186/s12958-021-00698-0 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.Gouhier C, Pons-Rejraji H, Dollet S, et al. Freezing Does Not Alter Sperm Telomere Length despite Increasing DNA Oxidation and Fragmentation. Genes (Basel) 2023;14:1039. 10.3390/genes14051039 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.Fleming SD, Thomson LK. The Oxidative Stress of Human Sperm Cryopreservation. Antioxidants (Basel) 2025;14:402. 10.3390/antiox14040402 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Shokri S, Ebrahimi SM, Ziaeipour S, et al. Effect of insulin on functional parameters of human cryopreserved sperms. Cryobiology 2019;87:68-73. 10.1016/j.cryobiol.2019.02.002 [DOI] [PubMed] [Google Scholar]
  • 32.Yannasithinon S, Chaimontri C, Sawatpanich T, et al. Dolichandrone serrulata flower extract ameliorates male reproductive damages in type 2 diabetic rats. Andrologia 2021;53:e13911. 10.1111/and.13911 [DOI] [PubMed] [Google Scholar]
  • 33.Rezazadeh-Reyhani Z, Razi M, Malekinejad H, et al. Cytotoxic effect of nanosilver particles on testicular tissue: Evidence for biochemical stress and Hsp70-2 protein expression. Environ Toxicol Pharmacol 2015;40:626-38. 10.1016/j.etap.2015.08.024 [DOI] [PubMed] [Google Scholar]
  • 34.Aeeni M, Razi M, Alizadeh A, et al. The molecular mechanism behind insulin protective effects on testicular tissue of hyperglycemic rats. Life Sci 2021;277:119394. 10.1016/j.lfs.2021.119394 [DOI] [PubMed] [Google Scholar]
  • 35.Minas A, Talebi H, Taravat Ray M, et al. Insulin treatment to type 1 male diabetic rats protects fertility by avoiding testicular apoptosis and cell cycle arrest. Gene 2021;799:145847. 10.1016/j.gene.2021.145847 [DOI] [PubMed] [Google Scholar]
  • 36.Shivaprakash P, Beeraka NM, Madhunapantula SRV, et al. Metformin Effects on SHIP2, AMPKs and Gut Microbiota: Recent Updates on Pharmacology. Curr Med Chem 2025;32:1732-54. 10.2174/0109298673289342240213040144 [DOI] [PubMed] [Google Scholar]
  • 37.Rasouli N, Younes N, Ghosh A, et al. Longitudinal Effects of Glucose-Lowering Medications on β-Cell Responses and Insulin Sensitivity in Type 2 Diabetes: The GRADE Randomized Clinical Trial. Diabetes Care 2024;47:580-8. 10.2337/dc23-1070 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.Leisegang K, Almaghrawi W, Henkel R. The effect of Nigella sativa oil and metformin on male seminal parameters and testosterone in Wistar rats exposed to an obesogenic diet. Biomed Pharmacother 2021;133:111085. 10.1016/j.biopha.2020.111085 [DOI] [PubMed] [Google Scholar]
  • 39.Liu CY, Chang TC, Lin SH, et al. Metformin Ameliorates Testicular Function and Spermatogenesis in Male Mice with High-Fat and High-Cholesterol Diet-Induced Obesity. Nutrients 2020;12:1932. 10.3390/nu12071932 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.Naghibi M, Tayefi Nasrabadi H, Soleimani Rad J, et al. The effects of metformin and forskolin on sperm quality parameters and sexual hormones in type II diabetic male rats. Andrologia 2022;54:1605-17. 10.1111/and.14426 [DOI] [PubMed] [Google Scholar]
  • 41.Malaekeh-Nikouei A, Shokri-Naei S, Karbasforoushan S, et al. Metformin beyond an anti-diabetic agent: A comprehensive and mechanistic review on its effects against natural and chemical toxins. Biomed Pharmacother 2023;165:115263. 10.1016/j.biopha.2023.115263 [DOI] [PubMed] [Google Scholar]
  • 42.Zhao J, Yang PC, Yang H, et al. Dietary supplementation with metformin improves testis function and semen quality and increases antioxidants and autophagy capacity in goats. Theriogenology 2022;188:79-89. 10.1016/j.theriogenology.2022.05.030 [DOI] [PubMed] [Google Scholar]
  • 43.Zhang M, Li H, Ma J, et al. Effects of Zinc Combined with Metformin on Zinc Homeostasis, Blood-Epididymal Barrier, and Epididymal Absorption in Male Diabetic Mice. Biol Trace Elem Res 2025;203:291-304. 10.1007/s12011-024-04171-y [DOI] [PubMed] [Google Scholar]
  • 44.Li H, Zhang M, Ma J, et al. Zinc Combined with Metformin Corrects Zinc Homeostasis and Improves Steroid Synthesis and Semen Quality in Male Type 2 Diabetic Mice by Activating PI3K/AKT/mTOR Pathway. Biol Trace Elem Res 2025;203:4659-70. 10.1007/s12011-025-04518-z [DOI] [PubMed] [Google Scholar]
  • 45.Fu G, Dai J, Li Z, et al. The role of STAT3/p53 and PI3K-Akt-mTOR signaling pathway on DEHP-induced reproductive toxicity in pubertal male rat. Toxicol Appl Pharmacol 2020;404:115151. 10.1016/j.taap.2020.115151 [DOI] [PubMed] [Google Scholar]
  • 46.Rindone GM, Dasso ME, Centola CL, et al. Effect of Metformin on Sertoli Cell Fatty Acid Metabolism and Blood-Testis Barrier Formation. Biology (Basel) 2024;13:330. 10.3390/biology13050330 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47.Liu Q, Zhu C, Ma Y, et al. Metformin improves fish sperm quality by regulating glucose uptake capacity during in vitro storage. J Anim Sci 2023;101:skad152. 10.1093/jas/skad152 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.Bertoldo MJ, Guibert E, Tartarin P, et al. Effect of metformin on the fertilizing ability of mouse spermatozoa. Cryobiology 2014;68:262-8. 10.1016/j.cryobiol.2014.02.006 [DOI] [PubMed] [Google Scholar]
  • 49.Usuga A, Vergara AK, Tobón MC, et al. Metformin and rosiglitazone affect motility, lipid peroxidation and mitochondrial activity of thawed equine spermatozoa. J Equine Vet Sci 2025;149:105570. 10.1016/j.jevs.2025.105570 [DOI] [PubMed] [Google Scholar]
  • 50.Grandhaye J, Partyka A, Ligocka Z, et al. Metformin Improves Quality of Post-Thaw Canine Semen. Animals (Basel) 2020;10:287. 10.3390/ani10020287 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51.Wensink MJ, Lu Y, Tian L, et al. Preconception Antidiabetic Drugs in Men and Birth Defects in Offspring: A Nationwide Cohort Study. Ann Intern Med 2022;175:665-73. 10.7326/M21-4389 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52.Rotem RS, Weisskopf MG, Huybrechts KF, et al. Paternal Use of Metformin During the Sperm Development Period Preceding Conception and Risk for Major Congenital Malformations in Newborns. Ann Intern Med 2024;177:851-61. 10.7326/M23-1405 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53.McPherson NO, Lane M. Metformin treatment of high-fat diet-fed obese male mice restores sperm function and fetal growth, without requiring weight loss. Asian J Androl 2020;22:560-8. 10.4103/aja.aja_141_19 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54.Berisha F, Blankenberg S, Nikolaev VO. Live Cell Monitoring of Phosphodiesterase Inhibition by Sulfonylurea Drugs. Biomolecules 2024;14:985. 10.3390/biom14080985 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55.Capozzi ME, Bouslov D, Sargsyan A, et al. β Cell Gαs signaling is critical for physiological and pharmacological enhancement of insulin secretion. J Clin Invest. 2025;135:e183741. 10.1172/JCI183741 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56.Shokri A, Pourheydar B, Hossein Farjah G, et al. Effects of glibenclamide and troxerutin on the sperm parameters and histopathological changes of testis in streptozotocin-induced diabetic male rats: An experimental study. Int J Reprod Biomed 2023;21:123-38. 10.18502/ijrm.v21i2.12803 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57.Rabbani SI, Devi K, Khanam S. Inhibitory effect of glimepiride on nicotinamide-streptozotocin induced nuclear damages and sperm abnormality in diabetic Wistar rats. Indian J Exp Biol 2009;47:804-10. [PubMed] [Google Scholar]
  • 58.Rabbani SI, Devi K, Khanam S. Protective role of glibenclamide against nicotinamide-streptozotocin induced nuclear damage in diabetic Wistar rats. J Pharmacol Pharmacother 2010;1:18-23. 10.4103/0976-500X.64531 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59.Uzomba CG, Ezemagu UK, Ofoegbu MS, et al. Edible mushroom (Pleurotus cornucopiae) extract vs. glibenclamide on alloxan induced diabetes: sub-acute in vivo study of Nrf2 expression and renal toxicity. Anat Cell Biol 2024;57:446-58. 10.5115/acb.24.054 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60.Elmalí E, Altan N, Bukan N. Effect of the sulphonylurea glibenclamide on liver and kidney antioxidant enzymes in streptozocin-induced diabetic rats. Drugs R D 2004;5:203-8. 10.2165/00126839-200405040-00003 [DOI] [PubMed] [Google Scholar]
  • 61.Rabbani SI, Devi K, Khanam S. Role of Pioglitazone with Metformin or Glimepiride on Oxidative Stress-induced Nuclear Damage and Reproductive Toxicity in Diabetic Rats. Malays J Med Sci 2010;17:3-11. [PMC free article] [PubMed] [Google Scholar]
  • 62.Öztaş E, Yılmaz TE, Güzel E, et al. Gliclazide alone or in combination with atorvastatin ameliorated reproductive damage in streptozotocin-induced type 2 diabetic male rats. Saudi Pharm J 2019;27:422-31. 10.1016/j.jsps.2019.01.003 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63.Alyami NM, Alnakhli ZA, Alshiban NM, et al. Oral administration of proniosomal glibenclamide formulation protects testicular tissue from hyperglycemia fluctuations and ROS via Nrf2/HO-1 pathway. Heliyon 2024;10:e31283. 10.1016/j.heliyon.2024.e31283 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 64.Dutta S, Sengupta P, Slama P, et al. Oxidative Stress, Testicular Inflammatory Pathways, and Male Reproduction. Int J Mol Sci 2021;22:10043. 10.3390/ijms221810043 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65.Acevedo JJ, Mendoza-Lujambio I, de la Vega-Beltrán JL, et al. KATP channels in mouse spermatogenic cells and sperm, and their role in capacitation. Dev Biol 2006;289:395-405. 10.1016/j.ydbio.2005.11.002 [DOI] [PubMed] [Google Scholar]
  • 66.Kumar N, Jain S, Gupta A, et al. Spermicidal activity of sulfonylureas and meglitinide analogues: role of intrasperm Ca2+ elevation. J Pharm Pharmacol 2008;60:323-30. 10.1211/jpp.60.3.0007 [DOI] [PubMed] [Google Scholar]
  • 67.Li S, Huang N, Wang M, et al. GLP-1R in diabetes mellitus: from basic discovery to therapeutics development. Front Pharmacol 2025;16:1610512. 10.3389/fphar.2025.1610512 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68.Drucker DJ. Mechanisms of Action and Therapeutic Application of Glucagon-like Peptide-1. Cell Metab 2018;27:740-56. 10.1016/j.cmet.2018.03.001 [DOI] [PubMed] [Google Scholar]
  • 69.Pourheydar M, Hasanzadeh S, Razi M, et al. Effects of liraglutide on sperm characteristics and fertilization potential following experimentally induced diabetes in mice. Vet Res Forum 2021;12:109-16. 10.30466/vrf.2019.96822.2315 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 70.Yin D, Li F, Xia L, et al. GLP-1 receptor agonists show no detrimental effect on sperm quality in mouse models and cell lines. Endocrine 2025;89:614-26. 10.1007/s12020-025-04245-4 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 71.Qi R, Liang Y, Yu J, et al. Liraglutide improved the reproductive function of obese mice by upregulating the testicular AC3/cAMP/PKA pathway. Reprod Biol Endocrinol 2024;22:31. 10.1186/s12958-024-01202-0 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72.Andersen E, Juhl CR, Kjøller ET, et al. Sperm count is increased by diet-induced weight loss and maintained by exercise or GLP-1 analogue treatment: a randomized controlled trial. Hum Reprod 2022;37:1414-22. 10.1093/humrep/deac096 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 73.Gregorič N, Šikonja J, Janež A, et al. Semaglutide improved sperm morphology in obese men with type 2 diabetes mellitus and functional hypogonadism. Diabetes Obes Metab 2025;27:519-28. 10.1111/dom.16042 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 74.Salvio G, Ciarloni A, Ambo N, et al. Effects of glucagon-like peptide 1 receptor agonists on testicular dysfunction: A systematic review and meta-analysis. Andrology 2025. [Epub ahead of print]. doi: . 10.1111/andr.70022 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 75.Lengsfeld S, Probst L, Emara Y, et al. Effects of the glucagon-like peptide-1 receptor agonist dulaglutide on sexuality in healthy men: a randomised, double-blind, placebo-controlled crossover study. EBioMedicine 2024;107:105284. 10.1016/j.ebiom.2024.105284 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 76.Cannarella R, Calogero AE, Condorelli RA, et al. Is there a role for glucagon-like peptide-1 receptor agonists in the treatment of male infertility? Andrology 2021;9:1499-503. 10.1111/andr.13015 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 77.Li S, Lu MM, Zhou D, et al. GLP-1: a novel zinc finger protein required in somatic cells of the gonad for germ cell development. Dev Biol 2007;301:106-16. 10.1016/j.ydbio.2006.07.048 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 78.Han L, Qu Q, Aydin D, et al. Structure and mechanism of the SGLT family of glucose transporters. Nature 2022;601:274-9. 10.1038/s41586-021-04211-w [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 79.Dana P, Hayati Roodbari N, Yaghmaei P, et al. Effects of empagliflozin on the expression of kisspeptin gene and reproductive system function in streptozotocin-induced diabetic male rats. Front Endocrinol (Lausanne) 2022;13:1059942. 10.3389/fendo.2022.1059942 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 80.Kiani M, Mehranjani MS, Shariatzadeh MA. Empagliflozin improves sperm quality in diabetic rats by lowering oxidative stress and reducing apoptosis-related genes expression. Reprod Biol 2025;25:100971. 10.1016/j.repbio.2024.100971 [DOI] [PubMed] [Google Scholar]
  • 81.Luo ZC, Jin ZR, Jiang YF, et al. The protective effects and underlying mechanisms of dapagliflozin on diabetes-induced testicular dysfunction. Asian J Androl 2023;25:331-8. 10.4103/aja202242 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 82.Albekairi NA, Al-Hamamah MA, Alshamrani AA, et al. Dapagliflozin Mitigated Elevated Disomic and Diploid Sperm in a Mouse Model of Diabetes and Recover the Disrupted Ogg1, Parp1, and P53 Gene Expression. Biomedicines 2023;11:2980. 10.3390/biomedicines11112980 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 83.Jin Z, Cao Y, Wen Q, et al. Dapagliflozin ameliorates diabetes-induced spermatogenic dysfunction by modulating the adenosine metabolism along the gut microbiota-testis axis. Sci Rep 2024;14:641. 10.1038/s41598-024-51224-2 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 84.Ribeiro JC, Martins AD, Jarak I, et al. Exenatide and Dapagliflozin Combination Enhances Sertoli Cell Secretion of Key Metabolites for Spermatogenesis. Biomedicines 2022;10:1115. 10.3390/biomedicines10051115 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 85.Alshamrani AA, Al-Hamamah MA, Albekairi NA, et al. Impacts of the DPP-4 Inhibitor Saxagliptin and SGLT-2 Inhibitor Dapagliflozin on the Gonads of Diabetic Mice. Biomedicines 2023;11:2674. 10.3390/biomedicines11102674 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 86.Zhu X, Xia L, Yin D, et al. Evaluating the Safety and Efficacy of Sodium-Glucose Co-transporter 2 Inhibitors in Subjects with Prediabetes: A Protocol for a Randomized Controlled Trial. Diabetes Ther 2024;15:1231-44. 10.1007/s13300-024-01560-3 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 87.Chang YC, Hsieh ML, Lee HL, et al. Identification of PTGR2 inhibitors as a new therapeutic strategy for diabetes and obesity. EMBO Mol Med 2025;17:938-66. 10.1038/s44321-025-00216-4 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 88.Wang Y, Zhang R, Chen Q, et al. PPARγ Agonist Pioglitazone Prevents Hypoxia-induced Cardiac Dysfunction by Reprogramming Glucose Metabolism. Int J Biol Sci 2024;20:4297-313. 10.7150/ijbs.98387 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 89.Collodel G, Moretti E, Marcucci C, et al. PPARγ Expression in Human Spermatozoa and Its Relationship with Seminal F(2)-Isoprostanes and Resolvin D1 in the Presence of Varicocele and Urogenital Infections. Biology (Basel) 2025;14:137. 10.3390/biology14020137 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 90.Rabbani SI, Devi K, Khanam S. Pioglitazone, a PPAR-gamma ligand inhibited the nicotinamide-streptozotocin induced sperm abnormalities in type-2 diabetic Wistar rats. Pak J Pharm Sci 2010;23:326-31. [PubMed] [Google Scholar]
  • 91.Pourazadi L, Sharafi M, Torshizi MAK, et al. Modulatory effects of pioglitazone as a ligand for the peroxisome proliferator-activated receptor on semen quality and fertility potential of broiler breeder roosters. Poult Sci 2022;101:101795. 10.1016/j.psj.2022.101795 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 92.Feinstein DL, Spagnolo A, Akar C, et al. Receptor-independent actions of PPAR thiazolidinedione agonists: is mitochondrial function the key? Biochem Pharmacol 2005;70:177-88. 10.1016/j.bcp.2005.03.033 [DOI] [PubMed] [Google Scholar]
  • 93.Alfarhan MW, Al-Hussaini H, Kilarkaje N. Role of PPAR-γ in diabetes-induced testicular dysfunction, oxidative DNA damage and repair in leptin receptor-deficient obese type 2 diabetic mice. Chem Biol Interact 2022;361:109958. 10.1016/j.cbi.2022.109958 [DOI] [PubMed] [Google Scholar]
  • 94.Mullins EA, Rodriguez AA, Bradley NP, et al. Emerging Roles of DNA Glycosylases and the Base Excision Repair Pathway. Trends Biochem Sci 2019;44:765-81. 10.1016/j.tibs.2019.04.006 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 95.Mehdipour M, Daghigh-Kia H, Najafi A, et al. Protective effect of rosiglitazone on microscopic and oxidative stress parameters of ram sperm after freeze-thawing. Sci Rep 2022;12:13981. 10.1038/s41598-022-18298-2 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 96.Zhang Y, Yan S, Gao X, et al. Metabonomic investigation on the protective effects of rosiglitazone and caloric restriction for renal senescence in a rat model. Aging Clin Exp Res 2012;24:430-8. 10.3275/8409 [DOI] [PubMed] [Google Scholar]
  • 97.Ortiz-Rodriguez JM, Balao da Silva C, Masot J, et al. Rosiglitazone in the thawing medium improves mitochondrial function in stallion spermatozoa through regulating Akt phosphorylation and reduction of caspase 3. PLoS One 2019;14:e0211994. 10.1371/journal.pone.0211994 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 98.Boeri L, Capogrosso P, Ventimiglia E, et al. Undiagnosed prediabetes is highly prevalent in primary infertile men - results from a cross-sectional study. BJU Int 2019;123:1070-7. 10.1111/bju.14558 [DOI] [PubMed] [Google Scholar]
  • 99.Salama N, Tsuji M, Tamura M, et al. Transforming growth factor (beta1) in testes of aged and diabetic rats: correlation with testicular function. Arch Androl 2001;47:217-26. 10.1080/014850101753145933 [DOI] [PubMed] [Google Scholar]
  • 100.Abbasihormozi S, Kouhkan A, Shaverdi A, et al. Nuclear Factor Kappa-B Protein Levels in Sperm of Obese Men with and without Diabetes; Cellular Approach in Male Infertility. Cell J 2023;25:17-24. 10.22074/cellj.2022.557547.1065 [DOI] [PMC free article] [PubMed] [Google Scholar]

Associated Data

    This section collects any data citations, data availability statements, or supplementary materials included in this article.

    Supplementary Materials

    The article’s supplementary files as

    tau-14-10-3387-rc.pdf (94.9KB, pdf)
    DOI: 10.21037/tau-2025-440
    tau-14-10-3387-coif.pdf (225.7KB, pdf)
    DOI: 10.21037/tau-2025-440
    DOI: 10.21037/tau-2025-440

    Articles from Translational Andrology and Urology are provided here courtesy of AME Publications

    RESOURCES