Abstract Spermatogonial stem cells (SSCs) possess the capacity for spontaneous reprogramming during in vitro culture, while the underlying mechanisms remain poorly understood, especially why the addition of epidermal growth factor (EGF), leukemia inhibitory factor (LIF) remarkably enhanced transition efficiency. Here we employed a multi-omics approach, integrating transcriptomics, metabolomics, and DNA methylation analyses to focus on the interplay between exogenous growth factors, metabolic pathways, and signaling cascades, particularly the role of SMAD3 in these networks. Our findings reveal that SSC reprogramming is contingent upon a metabolic shift from the tricarboxylic acid (TCA) cycle to aerobic glycolysis, modulated by fluctuating SMAD3 levels. SMAD3 downregulation activates HIF-1α, inducing aerobic glycolysis to supply energy and substrates for reprogramming. Subsequent SMAD3 reactivation promotes rapid cell proliferation, facilitating successful reprogramming. This study elucidates the pivotal role of SMAD3 in modulating glycometabolic pathways driving SSC transformation, emphasizing the necessity of aerobic glycolysis following SMAD3 fluctuations for effective reprogramming, which provides novel insights into the intricate interplay between energy metabolism and stem cell plasticity and potential applications in regenerative medicine and fertility treatments. Supplementary Information The online version contains supplementary material available at 10.1186/s13287-025-04541-w. Keywords: Aerobic glycolysis, Cell fate, Cellular plasticity, Germline-to-pluripotent transition, HIF-1A activation, Metabolic reprogramming, SMAD3 signaling Introduction Spermatogonial stem cells (SSCs), derived from primordial germ cells (PGCs), robustly generate sperm under the regulatory effects of the testicular niche [[38]18]. In the last century, it was discovered that PGCs could spontaneously transition to a pluripotent state during in vitro culture [[39]29, [40]34], highlighting their inherent pluripotency. Similarly, SSCs, traditionally viewed as unipotent, have been found capable of abrogating lineage commitment and spontaneously converting to pluripotent cells after long-term in vitro culture. These reprogrammed cells share many features with embryonic stem cells (ESCs) derived from the inner cell mass (ICM), including the ability to induce teratoma formation and contribute to chimeric animals [[41]13, [42]35]. This unique reprogramming event distinguishes SSCs from induced pluripotent stem (iPS) cells, as it occurs without the introduction of exogenous genes or gene products, suggesting a unique potential in cellular therapy with minimal risk. However, controlling the efficiency of SSC transformation remains challenging, as the mechanisms underlying their spontaneous reprogramming are largely unknown. Notably, the loss of p53 remarkably accelerates SSC transformation [[43]13], potentially linked to methylation changes mediated by DNMT1 [[44]39]. Our recent studies have analyzed chromatin accessibility changes triggered by p53 deletion, revealing an increase in the accessibility of the SMAD3 binding domain during the initial phase of cell transformation, alongside a notable decrease in SMAD3 protein levels. As the culture period extends, SMAD3 expression begins to rise, and the cell proliferation rate starts to accelerate, culminating in pluripotent transformation [[45]22]. Additionally, we found that SMAD3 can directly bind to the promoter of the key pluripotency gene, Nanog [[46]22]. Consistent with these findings, exogenous activation of SMAD3 in the early stage of SSC culture (germline commitment) led to cell death, whereas its activation of SMAD3 in transforming SSCs (with a pluripotent cell fate) increased transformation efficiency [[47]22]. This oscillatory change in SMAD3 expression—initially decreasing and subsequently increasing—suggests that inhibiting SMAD3 expression in the early stage of transformation might be a prerequisite for spontaneous reprogramming of SSCs. However, the molecular underpinnings of SMAD3 fluctuations and cell fate transformation remain unclear. A more recent study from our laboratory reported that addition of EGF/LIF to SSC culture medium significantly improved the transformation efficiency of SSCs maintained on fresh mouse embryonic fibroblast (MEF) feeder layers, particularly noting that EGF signaling activates Smad3 expression through RAC1 during SSC transformation [[48]44]. Moreover, transcriptomic analysis revealed significant changes in the expression of metabolism-associated genes, especially those related to carbohydrate and energy metabolism, during the initiation stage of SSC transformation [[49]44]. Increased glycolytic activity, a hallmark of rapid proliferation in both tumor cells and stem cells, is characterized by a preference for glycolysis even in the presence of adequate oxygen—a phenomenon known as aerobic glycolysis or the Warburg effect. This dysregulation of glycolysis provides cells with essential nutrients and ATP, restructuring the microenvironment to facilitate rapid growth and metastasis by acidifying the extracellular milieu and disrupting the extracellular matrix [[50]11, [51]23]. These observations imply some potential connections between SMAD3 and energy metabolism in SSCs spontaneous reprogramming. SMAD3 is a key transcription factor downstream of classical TGF-β signaling and plays a pivotal role in the regulation of cell proliferation, apoptosis, differentiation, epithelial–mesenchymal transition, tumor suppression and cancer promotion [[52]7, [53]42]. Accumulating evidence suggests that SMAD3 activity is closely associated with metabolites, for example, the reduced de novo fatty acid synthesis and increased fatty acid β-oxidation lead to intracellular acetyl-CoA accumulation, which promotes SMAD3 acetylation and entry into the nucleus to regulate endoderm differentiation [[54]47]. Based on these observations, we proposed that SMAD3 might function as a molecule that connects energy metabolism and cell fate determination in the SSCs' spontaneous reprogramming. Notably, the shift from TCA to aerobic glycolysis entails a reprogramming of glucometabolic gene expression, orchestrated by Hypoxia-inducible factor (HIF)-1α, a transcription factor with a helix-loop-helix structure that is activated in response to hypoxia and homeostasis. HIF-1α directly or indirectly regulates the expression of several genes involved in cell differentiation, including Ldha (lactate dehydrogenase-A), Vegf (vascular endothelial growth factor), Flt1 (FMS-associated tyrosine kinase 1), Pdgf-β (platelet-derived growth factor-β), bFgf (basic fibroblast growth factor), and other genes that affect glycolysis [[55]17, [56]33], suggesting that it plays a role in the regulation of glycolytic activity. More importantly, HIF-1α can bind to the MH2 domain of phosphorylated SMAD3 and switch the function of TGF-β to promoting glycolysis under hypoxic conditions by altering its SMAD binding partner [[57]10]. In addition, in tumor cells, increasing evidence has revealed the connections among HIF-1α, TGF-β/SMAD3 signaling and glycolysis: TGF-β signaling inhibits glycolysis under normoxic conditions but significantly promotes glycolysis under hypoxic conditions in tumor cells in vitro and in vivo, and SMAD3 plays a key role in the metabolic reprogramming associated with tumor progression, including tumor glycolysis [[58]46], glutaminolysis [[59]2], and lipid metabolism [[60]20]. These findings imply that HIF-1α plays a role in SMAD3-mediated energy metabolism reprogramming during SSC transformation. In this study, we aim to delineate the cooperative roles of SMAD3 and HIF-1α in driving metabolic reprogramming toward aerobic glycolysis and to establish this transition as a prerequisite for the spontaneous pluripotency acquisition in SSCs. By integrating transcriptomic, metabolomic, and functional analyses, we further elucidate how metabolic-epigenetic crosstalk orchestrates SSC fate determination. Our study provides new insights into the mechanism of spontaneous reprogramming of SSCs, offering valuable information for the treatment of reproductive diseases and for future advancements in stem cell research and animal cloning technology. Results Induction of SSC pluripotency via an optimized culture system To induce spontaneous reprogramming, a long-term cultured SSCs cell line was established. SSCs, isolated from the testes of neonatal mice, were purified and cultured on MEFs for 4–5 days (Fig. [61]1A). The resulting colonies exhibited a grape-like cluster morphology, characteristic of SSCs, and could be stably maintained on a MEF feeder layer for more than 30 passages (Fig. [62]1B), exhibiting a robust proliferation rate throughout the culture process (Fig. [63]1C). To confirm that the obtained cells were SSCs, total RNA was extracted and subjected to RT‒PCR analysis, which revealed the expression of key germline markers, including Id4, Plzf, Mvh, Gfrα1, Itgα6, Itgβ1, and c-Kit (Fig. [64]1D). Immunofluorescence (IF) assays further confirmed the expression of germ cell marker MVH, undifferentiated spermatogonia marker PLZF, and SSCs marker ID4 (Figure [65]S1). The percentages of PLZF-positive and GFRA1-positive cells were 92.38 ± 1.46% and 88.53 ± 1.86%, respectively (Fig. [66]1E), suggesting that SSCs were efficiently enriched in the obtained cells. Fig. 1. [67]Fig. 1 [68]Open in a new tab Characterization and Pluripotent Transition of Spermatogonial Stem Cells (SSCs). (A) Schematic of SSC isolation and culture on mouse embryonic fibroblast (MEF) feeder layer. (B) Clonal morphology across different passages of SSCs. (C) Growth kinetics of SSCs. (D) Identification of SSC-specific markers by RT-PCR. (E) Quantitative analysis of the proportion of PLZF + and GFRA1 + cells. (F) Observation of a transforming state in SSCs during prolonged culture. (G) Representative colonies of transformed GSPCs from long-term SSC cultures. (H) Assessment of germline and pluripotency marker expression by Western blot. (I, J) IF assays revealing pluripotency marker expression in GSPCs (I: NANOG, PLZF, DAPI, merged; J: SOX2, SSEA1, DAPI, merged). Data are presented as mean ± SD (*p < 0.05; **p < 0.01). Scale bars, 20 μm As described in a previous study [[69]44], SSCs cultured on fresh MEF feeder layers for approximately 5–6 generations could be transformed into pluripotent state cells after 2–3 generations exposed to 10 ng/mL LIF and 20 ng/mL EGF (Fig. [70]1F). These cells were identified as being in an intermediate state of transformation [[71]44]. After sustained culture for an additional 5 generations, all colonies adopted an epithelium-like phenotype (Fig. [72]1G). These transformed cells highly expressed the pluripotency markers SOX2, OCT4, and NANOG but expressed very low levels of the germ cell marker MVH and the SSC marker PLZF (Fig. [73]1H). IF assays corroborated these findings, revealing nearly ubiquitous expression of NANOG, SOX2 and SSEA1 but no expression of PLZF (Fig. [74]1I and J). In summary, these data indicate that SSCs undergo a transition to a pluripotent state upon the addition of LIF and EGF; these cells are termed germline stem cell-derived pluripotent stem cells (GSPCs) [[75]44]. The proportion of GSPCs progressively increased with continued culture and completed transformation after approximately 5 passages, accompanying with metabolic changes [[76]44], while the mechanism remained unclear. Metabolic reprogramming and changes in energy flux during the transformation of SSCs to GSPCs To explore the role of metabolic changes during SSC transformation, especially the connection of transcriptomic alterations such as Ras/Rac1/p38, TGF-β and JAK-STAT signaling pathways and energy metabolisms, we harvested stable cultured SSCs (p10 in culture medium), intermediate-state cells (p10 SSCs cultured in transformation medium for 2 passages, referred to as In state cells), and stable GSPCs (p10 SSCs cultured in transformation medium for 12 passages) for metabolomics analysis (Figure [77]S2A). In total, 42 metabolites involved in energy metabolism were detected by using LC‒MS/MS and a self-built metabolic database from the Maiwei Company. The results were subjected to principal component analysis (PCA) and cluster analysis (Figure [78]S2B), which revealed the pattern of differences in metabolite abundance among the 3 groups (Fig. [79]2A, Table [80]S3). Fig. 2. [81]Fig. 2 [82]Open in a new tab Metabolic reprogramming dynamics observed during SSC transformation. (A) Sample cluster diagram. (B) Volcanic map of SSC p10 vs. GSPCs p2 differential metabolites. (C) KEGG enrichment analysis of upregulated metabolites. (D) Bubble chart of changes in the content of pyruvate and downstream metabolites. (E) Radar chart of changes in the content of some amino acids and nucleotides (The values in the figure represent Fold Change). (F) Comparison of L-Glutamic acid and glutamine content with D-Glucose-6-phosphate content. (G) Cluster analysis of metabolites in SSC p10 and GSPCs p12. (H) KEGG enrichment analysis of up-regulated metabolites in GSPCs p12. (I-K) Overall trends of different metabolic pathways during transformation: change trend of metabolites involved in glycolysis pathway (I), change trend of metabolites involved in PPP (J), and change trend of metabolites involved in TCA (K). The metabolites displayed in this figure were specifically selected based on their significantly elevated expression profiles observed in GSPCs through targeted metabolomics screening Metabolic changes during the initial stage of SSC pluripotent transformation Previous study revealed that after two passages in the presence of EGF/LIF, SSCs enter the initial stage of the transition to a pluripotent state, probably accompanying with metabolic reprogramming, especially changes in energy metabolism, as a critical event in reprogramming. Therefore, we compared and analyzed the altered metabolic patterns in SSCs and In state cells. Metabolomic analysis revealed 25 differentially abundant metabolites between SSCs and In state cells, of which 20 showed increased abundance (p ≤ 0.05, fold change ≥ 1.5) and 5 showed decreased abundance (p ≤ 0.05, fold change ≤ 0.5) (Fig. [83]2B). The upregulated metabolites included glucose 6-phosphate, phosphoenolpyruvic acid, pyruvate, D-5-ribulose phosphate, and 7-sedoheptulose phosphate. We performed metabolic pathway enrichment analysis of the 20 metabolites with increased abundance by using the SMPDB database and MetaboAnalyst 5.0, and a total of 43 metabolic pathways were enriched. There were 6 pathways for which P was ≤ 0.05: glycolysis, gluconeogenesis, the pentose phosphate pathway, lactose synthesis, carnitine synthesis and ribonucleotide metabolism (Fig. [84]2C). This indicates that during the early stage of transformation, the glucose metabolism patterns of the cells changed significantly, and the activities of the glycolysis and pentose phosphate pathways were markedly increased. ATP, Inline graphic -D-fructofuranose 1,6-bisphosphate, 6-phospho-D-gluconic acid, L-citrulline and adenine exhibited decreased abundance (Fig. [85]2A, Table [86]S3). The observed decrease in the concentration of ATP, the main intracellular energy carrier, further proves that an increase in glycolytic activity leads to a decrease in the efficiency of intracellular energy production. We noted a substantial increase in pyruvate concentrations, with a fold change of 5.28, during the initial phase of SSC pluripotent transformation (Table [87]S3). Given the critical role of pyruvate in maintaining stem cell pluripotency [[88]32], we examined alterations in the downstream metabolic products of pyruvate (Fig. [89]2D). Alanine, which can be directly synthesized from pyruvate via a transamination reaction, displayed a negligible fold change of 0.97 (Table [90]S3). Although pyruvate predominantly enters the mitochondrial TCA cycle under aerobic conditions, only the succinate level was significantly upregulated, with a fold change of 12.04. Neither α-ketoglutarate (fold change = 1.04) nor citrate (fold change = 0.98) exhibited notable changes in abundance (Table [91]S3). This observation aligns with prior studies indicating a reduction in the protein level of dihydrolipoyl transacetylase within the pyruvate dehydrogenase complex in GSPCs. Lactate, an anaerobic byproduct of pyruvate metabolism, exhibited a fold change of 1.65, but this change was not statistically significant (p value = 0.12) (Table [92]S3). Moreover, phosphoenolpyruvate, a key intermediate in gluconeogenesis, exhibited a fold change of 2.45 (Table [93]S3). Metabolomic analyses revealed augmented nucleotide and amino acid biosynthesis activity, necessitating precursor availability and reducing flux through both the glycolysis and pentose phosphate pathways (Fig. [94]2E). These findings suggest that the increase in pyruvate abundance, attributed to enhanced glycolytic flux, does not contribute to oxidative metabolism via the TCA cycle but rather facilitates glucose homeostasis through gluconeogenesis and lactate clearance. Subsequently, we checked on the concentration of glutamine during the early dedifferentiation stages of SSCs, since it’s essential for amino acids, nucleotides, and fatty acids and intracellular protein synthesis [[95]28], as well as stemness maintenance [[96]51]. In the In state cells, glutamine and glutamate exhibited the highest absolute concentrations among all assessed metabolites, which were 146-fold and 56-fold greater than that of 6-phosphogluconate, respectively. The fold changes in glutamine and glutamate relative to the baseline levels were 1.5 and 2.7, respectively (Fig. [97]2F). These data indicate an increased cellular preference for glutamine during the initial stages of pluripotent transformation, facilitating increased biosynthetic activity within the cell. Metabolic changes during SSC-to-GSPC transformation Following the addition of growth factors and subsequent culture through the 12th generation, the SSCs transitioned to pluripotent stem cells. Thus, the associated changes in metabolic patterns reflect the metabolic preferences of pluripotent cells, providing insights into the