Abstract
Graphical abstract
Abstract
Objective
Emerging evidence links prenatal androgen excess to altered pubertal timing, yet the neuroendocrine mechanisms mediating this effect in male offspring remain poorly characterized. This study aimed to investigate the effects of prenatal androgen exposure on the timing of puberty onset in male offspring and the role of KNDy neurons in this process.
Methods
Eight-week-old pregnant Sprague–Dawley rats (n = 16) were randomized into control (olive oil) and prenatal androgen (PNA, testosterone injection) groups (n = 8 per group). Hypothalamic samples of six male offspring rats at postnatal day (PND) 21 (n = 3 per group) were collected for transcriptome analysis. The time of puberty onset was recorded in 36 male offspring (n = 18 per group). Serum samples and hypothalamic tissue from 12 male offspring rats (n = 6 per group) were collected, and GnRH, LH, FSH, and kisspeptin 1 protein levels were measured by ELISA. The mRNA levels of Kiss1, Tac3, and Pdyn were measured by real-time qPCR. In the brains of six male offspring rats (n = 3 per group), protein levels of kisspeptin, neurokinin B (NKB), and dynorphin (Dyn) in the arcuate nucleus (ARC) were measured using immunohistochemistry.
Results
Compared to controls, PNA male offspring rats showed significantly earlier puberty onset (P < 0.001). At PND21, Tac3 (P < 0.01) and Pdyn (P < 0.05) expression levels increased significantly in PNA male offspring rats. At puberty, ARC kisspeptin protein levels increased (P < 0.01), while dynorphin protein levels decreased (P < 0.01) in PNA male offspring rats.
Conclusion
Prenatal androgen exposure accelerates puberty onset in male offspring rats, likely by activating Tac3 expression in early life and reducing ARC dynorphin inhibition of kisspeptin neurons at puberty.
Keywords: prenatal, testosterone, offspring, puberty onset, KNDy neurons
Introduction
Puberty is a critical phase of physical and psychological development during which adolescents experience the emergence of secondary sexual characteristics, sexual maturation, and personality formation. With socioeconomic development, the trend toward earlier puberty onset has become increasingly apparent (1). Studies have shown that the age of pubertal onset in girls, based on breast development, has advanced by nearly 3 months per decade (2). Similarly, data from Europe indicate a trend of earlier puberty onset in boys, assessed by testicular volume and genital staging (3). However, the early onset of puberty has been associated with higher risks of developing type 2 diabetes, cardiovascular diseases, obesity, and psychological conditions such as depression (4, 5, 6, 7), thus becoming a significant public health concern.
The onset of puberty is a complex process influenced by genetics, environment, nutrition, and hormones (8, 9, 10, 11). It involves the reactivation of the hypothalamic–pituitary–gonadal (HPG) axis. This axis is briefly active at birth, becomes suppressed during childhood, and is reactivated at the onset of puberty (12). Upon activation of GnRH neurons, gonadotropin-releasing hormone (GnRH) is released in pulses into the pituitary portal system and transported to the pituitary gland, stimulating the synthesis and release of gonadotropins (13). Recent studies have shown that hypothalamic GnRH secretion is regulated by various factors, including neuropeptide Y, pro-opiomelanocortin, gamma-aminobutyric acid neurons, and glutamate (14, 15, 16). However, the precise mechanisms by which HPG axis activation triggers puberty onset remain unclear. In recent years, KNDy neurons (kisspeptin/neurokinin B (NKB)/dynorphin (Dyn)-expressing neurons), which co-express kisspeptin, NKB, and Dyn in the arcuate nucleus (ARC), have been identified as key players in this process (17).
The co-expression of KNDy peptides in the ARC has been confirmed in various mammals, including sheep, mice, and rats (18, 19, 20). Previous studies have demonstrated that KNDy neurons act as the primary regulators of GnRH pulses (21, 22). KNDy neurons release NKB (encoded by Tac3), which recruits other KNDy neurons via the NKB receptor (NK3R) to secrete kisspeptin around GnRH neuron dendrites. Kisspeptin binds to its receptor, G protein-coupled receptor 54 (GPR54), activating GnRH neurons. Subsequently, KNDy neurons release dynorphin (encoded by Pdyn), which binds to kappa-opioid receptors (KOR) to terminate GnRH pulses. Animal studies have demonstrated that transfecting the Kiss1 gene into the KNDy neurons of infertile female rats with Kiss1 gene knockout can restore normal pulsatile gonadotropin release and folliculogenesis (23). In addition, Kiss1 mRNA levels in the monkey hypothalamus increased with puberty (24). Administration of NK3R antagonists has been shown to modestly delay vaginal opening and reduce luteinizing hormone (LH) levels (25). These experimental findings suggest that KNDy neurons may play a critical role in regulating puberty.
Polycystic ovary syndrome (PCOS), a complex endocrine disorder characterized in part by hyperandrogenism, affects 5–20% of women of reproductive age worldwide (26). With the rising prevalence of PCOS, elevated androgen levels among women of childbearing age have become more common (27). Progress has been made in the study of the mechanisms by which androgen levels during pregnancy affect the offspring. When fetuses are exposed to high levels of androgens in utero, they face an increased risk of perinatal mortality, more congenital anomalies, and metabolic disorders (28). Prenatal androgen levels are associated with the development of girls (29, 30). In animal experiments, existing research has found that androgens during pregnancy advance the onset of puberty in male offspring by affecting the leptin–NPY pathway (31). Although epidemiological evidence indicates that excessive androgen exposure during pregnancy may disrupt the timing of puberty onset in offspring, the underlying neuroendocrine mechanisms remain incompletely understood. Notably, the role of hypothalamic KNDy neurons in this process represents a knowledge gap in the field. In the present study, we aimed to explore whether androgen exposure during pregnancy affects pubertal development by influencing the expression of KNDy neurons in the offspring and thus the role and mechanism of pubertal development. Therefore, we established a prenatal androgen (PNA) exposure rat model to determine the role of these neurons in the onset of puberty in offspring rats. This study provides scientific evidence for understanding the relationship between elevated prenatal androgen levels and pubertal development in male offspring.
Materials and methods
Animals and prenatally androgenized
Eight-week-old female and male Sprague–Dawley rats were purchased from Jinan Pengyue Laboratory Animal Breeding Co. and housed under controlled conditions with a 12 h light/12 h darkness cycle and a constant temperature of 25 ± 2°C. The rats had access to food and water ad libitum. After a 1 week acclimatization period, females and males were mated overnight. Female rats were monitored daily for the presence of post-copulatory plugs, and the day a plug was observed was designated as gestational day 1. Pregnant rats were randomly assigned to one of two groups: control or prenatal androgen (PNA), with eight rats in each group. On gestational days 15, 17, 19, and 21, the PNA group received subcutaneous injections of 2 mL of a 2.5 mg/mL testosterone solution at the nape of the neck (31, 32, 33). The control group received equivalent volumes of olive oil following the same schedule and procedure.
Determination of onset of puberty and samples collection
Three male offspring at postnatal day 21 (PND21) from each of the control and PNA groups were randomly selected and anesthetized with 2% sodium pentobarbital (3 mL/kg), their hypothalamus was rapidly removed on ice, immediately placed in liquid nitrogen, and stored at −80°C for transcriptome sequencing and bioinformatics analysis. Preputial separation was used as an external marker of puberty onset in male offspring rats. Starting from PND21, 36 male offspring rats (18 in the control group and 18 in the PNA group) were monitored daily for signs of puberty onset. On the day of puberty onset, 12 male offspring rats (control:PNA = 6:6) were anesthetized as described above, and blood samples were collected, centrifuged, and stored at −80°C for subsequent analysis; hypothalamic samples were collected for real-time qPCR analysis. While six male offspring rats were anesthetized as described above, and the brains were fixed overnight in 4% formaldehyde for immunohistochemical evaluation.
RNA sequencing
Total RNA was extracted from hypothalamic tissues of male offspring in both the control and PNA groups at PND21 using TRIzol reagent (Ambion/Invitrogen, USA). RNA integrity and quantity were assessed. mRNA with poly(A) tails was enriched, fragmented in Fragmentation Buffer, and reverse transcribed into cDNA. The resulting cDNA fragments were purified using the AMPure XP system (Beckman Coulter, USA) to select fragments between 370 and 420 bp in length. PCR amplification was performed using Phusion High-Fidelity DNA Polymerase, Universal PCR Primers, and Index (X) Primers. The PCR products were purified, and library quality was assessed using the Agilent Bioanalyzer 2100 system. Sequencing was carried out on the Illumina NovaSeq 6000 platform (Illumina, USA). Fluorescence signals captured by the sequencer were converted into sequencing reads, and the fragment sequences were obtained through computer analysis.
Bioinformatics analysis
To ensure data quality and reliability, clean reads were obtained by removing reads containing adapters, poly-N, and low-quality bases from the raw data. The Q20, Q30, and GC content of the clean data were calculated, and all subsequent analyses were conducted using this high-quality dataset. The reference genome and gene model annotation files were downloaded from a genome database, and the reference genome index was constructed using Hisat2 v2.0.5. Paired-end clean reads were aligned to the reference genome using Hisat2 v2.0.5, and gene read counts were quantified using featureCounts v1.5.0-p3. Differential expression analysis between groups was performed using the DESeq2 R package (v1.20.0), with genes showing a P < 0.05 considered differentially expressed. The Benjamini–Hochberg method was applied to adjust P-values and control the false discovery rate. Gene ontology (GO) enrichment analysis and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment analysis of differentially expressed genes (DEGs) were conducted using the clusterProfiler R package, with GO or KEGG terms having an adjusted P-value (Padj) < 0.05 considered significantly enriched. Sequencing and bioinformatics analyses were carried out by Novogene Bioinformatics Technology Co. Ltd (China).
Hormone assay
Enzyme-linked immunosorbent assay (ELISA) kits were used to quantify serum concentrations of GnRH (CSB-E08037r; Cusabio, China), LH (CSB-E12654r; Cusabio, China), FSH (CSB-E06869r; Cusabio, China), and kisspeptin 1 (CSB-E13434r; Cusabio, China). The detection ranges were 2–150 pg/mL for GnRH, 0.3–60 mlU/mL for LH, 0.17–10 mlU/mL for FSH, and 0.156–10 ng/mL for kisspeptin 1.
Real-time qPCR
Total RNA was extracted from hypothalamic tissues using TRIzol reagent (Wuhan Servicebio Technology CO., LTD, China) and quantified with a NanoDrop 2000 spectrophotometer (Thermo Fisher Scientific, USA). The extracted RNA was reverse-transcribed into first-strand cDNA using SweScript All-in-One RT SuperMix (Wuhan Servicebio Technology CO., LTD, China). Quantitative PCR assays were performed on a real-time PCR system (Bio-Rad, USA). The PCR system consisted of 10 μL SYBR Green master mix, 1 μL 10 μM primer mix, 7 μL ddH2O, and 2 μL cDNA. Gapdh was used as the endogenous control, and mRNA expression is presented as 2−ΔΔCT. Primer sequences are provided in Table 1.
Table 1.
Primer sequences used for RT-PCR.
| Targeted genes | Primer sequence (5′→3′) |
|---|---|
| Kiss1 | F: TCTCGCTGGCTTCTTGGC |
| R: GGGTTCAGGGTTCACCACA | |
| Tac3 | F: TAGCGTGGGACCGAAGGAGA |
| R: TCAACCACATCAGCGGGAGT | |
| Pdyn | F: CGGAGGAGTGGGAGACAT |
| R: GACGCTGGTAAGGAGTTGG | |
| Gapdh | F: CTGGAGAAACCTGCCAAGTATG |
| R: GGTGGAAGAATGGGAGTTGCT |
Immunohistochemistry
Coronal sections, 3 μm thick, containing the ARC were prepared from the fixed brain tissue. The sections were sequentially deparaffinized, hydrated, and subjected to antigen retrieval, followed by incubation in 3% hydrogen peroxide solution (Sinopharm Chemical Reagent Co., Ltd, China) for 25 min to block endogenous peroxidase activity. After washing, the slides were treated with a serum-blocking solution (Servicebio, China) and incubated overnight at 4°C with primary antibodies: KISS1 antibody (1:20, Affinity-DF7133, China), rabbit anti-neurokinin B (NKB) polyclonal antibody (1:200, Beijing Biosynthesis Biotechnology Co., Ltd-bs-0070R, China), and rabbit anti-PDYN polyclonal antibody (1:100, Absin-abs116888, China). The slides were then washed and incubated with species-specific secondary antibodies (Servicebio, China) for 50 min at room temperature. Following three rinses in phosphate-buffered saline (PBS) (Servicebio, China), the sections were air-dried slightly, and freshly prepared DAB (3,3′-diaminobenzidine) solution was applied dropwise. The color development process was monitored under a microscope, and staining was terminated by rinsing with tap water. After counterstaining the nuclei with hematoxylin, the sections were dehydrated and mounted. Images of the ARC region were captured using CaseViewer software 2.0 (3DHISTECH Ltd, Hungary), and the average optical density of the immunohistochemical staining was measured using ImageJ image processing software (NIH, USA).
Statistical analysis
Statistical analyses were performed using SPSS 23.0 (IBM Corp., USA), GraphPad Prism 9.0 (GraphPad Software, USA), and R software 4.3.3. The timing of preputial separation between the control and PNA groups was compared using the log-rank test. Differences in serum hormone levels, mRNA expression, and immunohistochemical indices at puberty onset between the two groups were analyzed using Student’s t-test. Hierarchical multiple regression, adjusted for puberty onset timing, was applied to assess the effects of prenatal testosterone intervention on serum hormone levels and mRNA expression in male offspring. Mixed linear models were employed to analyze differences in immunohistochemical indices between the control and PNA groups. Differences were considered statistically significant when P < 0.05.
Results
Effect of PNA on the onset of puberty in male offspring
As shown in Fig. 1, the onset of puberty in male offspring from the PNA group occurred significantly earlier than in the control group (P < 0.001). The first instance of preputial separation in the PNA group was observed on PND41, compared to PND44 in the control group. In addition, over 50% of male rats in the PNA group had undergone preputial separation by PND42, whereas none of the male rats in the control group had exhibited this marker of puberty onset.
Figure 1.

Cumulative percentage of male rats with preputial separation in control and PNA groups. Statistical significance was determined by the log-rank test. ***P < 0.001.
Transcriptomic analysis of the hypothalamus in male offspring at PND21
In the comparison of PND21 male offspring between the PNA and control groups, 1,315 genes were differentially expressed (P < 0.05, |log2 fold change| >0), including 648 up-regulated and 667 down-regulated genes. In the hypothalamus of PND21 male offspring from the PNA group, the expression of Tac3 and Pdyn was significantly higher compared to controls (P < 0.01 for Tac3, and P < 0.05 for Pdyn), while changes in Kiss1 expression were not significant (Fig. 2A, Supplementary Tables 1 and 2 (see section on Supplementary materials given at the end of the article)). The expressions of Agrp and Pomc genes, which are associated with sexual maturation, were also significantly increased (P < 0.05) (Supplementary Table 1).
Figure 2.
Transcriptome differences in the hypothalamus of PND21 male offspring between PNA and control groups. (A) Volcano plot of all detected genes. (B and C) GO enrichment analysis of up- and down-regulated genes. (D and E) KEGG pathway analysis of up- and down-regulated genes.
GO enrichment analysis showed that up-regulated genes were primarily enriched in the molecular function categories of ‘heme binding’ and ‘tetrapyrrole binding’. In the cellular component category, DEGs were mainly located in the extracellular region (Fig. 2B). Conversely, down-regulated genes were enriched in the biological process of ‘dephosphorylation’ and were involved in molecular functions such as ‘cytoskeletal protein binding’, ‘protein tyrosine/serine/threonine phosphatase activity’, ‘phosphoprotein phosphatase activity’ and ‘microtubule binding’ (Fig. 2C).
KEGG pathway analysis revealed that up-regulated genes were enriched in pathways related to ‘African trypanosomiasis’, ‘tyrosine metabolism’, and ‘drug metabolism-cytochrome P450’ (Fig. 2D). Down-regulated genes were mainly enriched in pathways associated with ‘Human papillomavirus infection’, ‘PI3K–Akt signaling pathway’, ‘microRNAs in cancer’, and ‘Kaposi sarcoma-associated herpesvirus infection’ (Fig. 2E).
Notably, the differential expression of Tac3 and Pdyn was enriched in the biological processes of the ‘G-protein-coupled receptor signaling pathway’ and the ‘neuroactive ligand–receptor interaction’ pathway. Although these enrichments were not statistically significant (P > 0.05), they may provide valuable insights into the potential mechanisms underlying the biological significance of this study.
Effect of PNA on serum hormone levels in male offspring at the onset of puberty
As shown in Fig. 3, at the onset of puberty, the differences in serum levels of GnRH, LH, FSH, and kisspeptin 1 between male offspring in the PNA group and the control group were not statistically significant (P > 0.05). The impact of PNA on serum hormone levels in male offspring was further analyzed using hierarchical multiple regression, adjusting for the time of puberty onset. The results, presented in Table 2, indicate a positive correlation between the time of puberty onset and serum kisspeptin 1 levels (P < 0.05). However, the remaining models did not reach statistical significance (P > 0.05).
Figure 3.
Effect of PNA on serum levels of GnRH, LH, FSH, and kisspeptin 1 in male offspring at the onset of puberty. No significant differences were observed between groups (P > 0.05).
Table 2.
Hierarchical multiple regression analysis of serum hormone levels in male offspring at the onset of puberty.
| GnRH (pg/mL) | LH (mlU/mL) | FSH (mlU/mL) | Kisspeptin 1 (ng/mL) | |||||||||
|---|---|---|---|---|---|---|---|---|---|---|---|---|
| B | SE | P | B | SE | P | B | SE | P | B | SE | P | |
| Timing of onset of puberty | 1.195 | 3.353 | 0.730 | 0.755 | 0.757 | 0.345 | 0.163 | 0.292 | 0.091 | 0.041* | 0.016 | 0.033 |
| Groups | −10.541 | 15.755 | 0.520 | −5.156 | 3.557 | 0.181 | −2.786 | 1.374 | 0.073 | 0.018 | 0.077 | 0.823 |
| R2 | 0.070 | 0.293 | 0.356 | 0.414 | ||||||||
| ΔR2 | 0.046 | 0.165 | 0.294 | 0.003 | ||||||||
| F | 0.340 | 1.863 | 2.488 | 3.184 | ||||||||
Groups: 0, control; 1, PNA. SE, standard error.
P < 0.05.
Effect of PNA on hypothalamic mRNA expression in male offspring at the onset of puberty
Regarding hypothalamic mRNA expression, an outlier was identified in the hypothalamic Kiss1 mRNA levels of male offspring in the PNA group. This outlier was excluded, and the Kiss1 mRNA data from five male offspring in the PNA group were ultimately used for statistical analysis. As shown in Fig. 4, the differences in hypothalamic Kiss1, Tac3, and Pdyn mRNA levels between the PNA group and the control group were not statistically significant (P > 0.05). The effect of PNA on hypothalamic gene expression in male offspring was further analyzed using hierarchical multiple regression, adjusted for the time of puberty onset, as presented in Table 3. However, none of the models reached statistical significance (P > 0.05).
Figure 4.
Effect of PNA on hypothalamic mRNA expression in male offspring at the onset of puberty. No significant differences were observed between groups (P > 0.05).
Table 3.
Hierarchical multiple regression analysis of hypothalamic mRNA expression in male offspring at the onset of puberty.
| Kiss1 | Tac3 | Pdyn | |||||||
|---|---|---|---|---|---|---|---|---|---|
| B | SE | P | B | SE | P | B | SE | P | |
| Timing of onset of puberty | −0.142 | 0.114 | 0.249 | 0.104 | 0.090 | 0.276 | −0.088 | 0.097 | 0.389 |
| Groups | −0.971 | 0.540 | 0.110 | −0.398 | 0.424 | 0.372 | −0.178 | 0.454 | 0.704 |
| R2 | 0.354 | 0.230 | 0.089 | ||||||
| ΔR2 | 0.261 | 0.075 | 0.016 | ||||||
| F | 2.191 | 1.348 | 0.438 | ||||||
Groups: 0, control; 1, PNA. SE, standard error.
Effect of PNA on protein expression in the ARC of male offspring at the onset of puberty
Figure 5 illustrates the expression levels of kisspeptin, NKB, and Dyn in the ARC of male offspring from both the control and PNA groups. The results were analyzed using mixed linear models, with group as a fixed effect variable and puberty onset time as a random effect variable. The findings, summarized in Table 4, show a positive correlation between puberty onset time and kisspeptin expression. Adjusted for the time of puberty onset, the effect of PNA on the expression of kisspeptin and Dyn in the ARC was significant (P < 0.01). Male offspring in the PNA group may have exhibited significantly higher kisspeptin expression in the ARC compared to the control group (P < 0.01). In contrast, their expression of Dyn in the ARC was significantly lower than that of the control group (P < 0.01).
Figure 5.
Expression of kisspeptin, NKB, and Dyn in the ARC of male offspring at the onset of puberty. *P < 0.05; **P < 0.01.
Table 4.
Mixed linear model analysis of kisspeptin, NKB, and Dyn expression in the ARC of male offspring at the onset of puberty.
| Target protein | Variable | Estimated coefficient | SE | t | P |
|---|---|---|---|---|---|
| Kisspeptin | Intercept | −0.714 | 0.165 | −4.33 | <0.001 |
| Timing of onset of puberty | 0.020 | 0.003 | 5.73 | <0.001 | |
| Groups | 0.054 | 0.016 | 3.48 | 0.001 | |
| NKB | Intercept | 0.010 | 0.059 | 0.17 | 0.866 |
| Timing of onset of puberty | 0.001 | 0.001 | 0.69 | 0.494 | |
| Groups | −0.009 | 0.006 | −1.67 | 0.105 | |
| Dyn | Intercept | 0.380 | 0.133 | 2.85 | 0.007 |
| Timing of onset of puberty | −0.003 | 0.003 | −1.24 | 0.223 | |
| Groups | −0.044 | 0.013 | −3.50 | 0.001 |
Groups: 0, control; 1, PNA. SE, standard error.
Discussion
Exposure to excessive androgens during early life has been linked to various chronic and malignant diseases (33, 34, 35). In this study, we investigated the effect of androgen excess during late pregnancy on puberty onset in male offspring using a hyperandrogenic pregnant rat model. Our findings indicate that in utero exposure to elevated androgen levels led to an earlier onset of puberty in male offspring. Moreover, male offspring in the PNA group exhibited upregulation of Tac3, a key gene involved in HPG axis activation, during the juvenile stage. Further analysis of the effects of PNA on male offspring at puberty onset revealed significant changes in the ARC, specifically in the expression of kisspeptin and dynorphin, proteins closely linked to puberty onset. These results suggest a potential mechanism by which hyperandrogenic exposure during late pregnancy may influence the timing of puberty onset by altering the secretion of KNDy neurons in the offspring.
Puberty onset occurs when the pulsatile secretion of gonadotropin-releasing hormone (GnRH) is reactivated, following suppression during childhood (12). Premature pulsatile GnRH secretion from the hypothalamus may result from an increase in excitatory or a decrease in inhibitory signaling within the HPG axis during childhood, leading to premature reactivation of the axis and early puberty onset. The hypothalamus regulates GnRH pulses through a complex network of neurons and glia, among which KNDy neurons are of particular interest (36). Mutations in the Kiss1 gene have been linked to hypogonadotropic hypogonadism and pubertal developmental disorders in humans (37). Similarly, mutations in Tac3 or Tacr3, which encode NKB and its receptor, have been associated with this phenotype (38). Our results show that male offspring in the PNA group exhibited significantly higher expression of Tac3 and Pdyn in the hypothalamus during the juvenile stage compared to controls. Although Kiss1 expression was elevated, the difference was not statistically significant. These findings suggest that exposure to hyperandrogenism during late pregnancy may affect the onset of puberty in male offspring by altering KNDy neurons, influencing the timing of puberty. Specifically, PNA exposure led to the upregulation of Tac3 at PND21, a key gene in activating the HPG axis during the juvenile stage. However, this activation was mitigated by elevated Pdyn, resulting in a less pronounced increase in Kiss1 expression. Consequently, the HPG axis may not have been fully reactivated during this period. Consistent with our findings, a previous study on hypothalamic mRNA levels in PNA-induced female offspring at 4 weeks of age also reported no significant difference in Kiss1 mRNA expression between the PNA and control groups, but a significant increase in hypothalamic Tac3 mRNA expression in PNA-treated females (39). In addition, we found that the DEGs Tac3 and Pdyn were enriched in biological processes related to the ‘G-protein-coupled receptor signaling pathway’ and the ‘neuroactive ligand–receptor interaction’ pathway. Although these enrichment results were not statistically significant, they suggest a potential pathway through which PNA may influence pubertal development. G protein-coupled receptor 54 (GPR54), a key receptor for kisspeptin, is a major regulator of GnRH and gonadotropin secretion (40), and plays a crucial role in the regulation of pubertal development (41). Notably, transcriptomic results from this study revealed that the expression of Agrp and Pomc genes associated with sexual maturation was significantly elevated. Studies indicate that Pomc-derived α-MSH can activate MC4R receptors within KNDy neurons via the melanocortin signaling pathway, promoting the release of NKB and kisspeptin to drive GnRH pulse secretion (42). Conversely, Agrp neurons may inhibit GnRH pulses by suppressing MC4R signaling or enhancing dynorphin release within KNDy neurons (43). This is consistent with our core findings, suggesting that Pomc and Agrp may act as upstream regulators of KNDy neurons to participate in the regulatory process of pubertal timing.
Our results showed that androgen exposure during late pregnancy significantly advanced the onset of puberty in male offspring. To further elucidate the mechanism by which PNA causes the early onset of puberty in male offspring, we aimed to validate and extend the primary findings from the RNA-seq analysis conducted during the juvenile stage in male offspring at the onset of puberty. ELISA and qPCR analyses demonstrated no statistically significant differences in serum concentrations of GnRH, LH, FSH, and kisspeptin 1, or hypothalamic mRNA expression levels of Kiss1, Tac3, and Pdyn between the PNA group and control (P > 0.05). These findings suggest that the effects of prenatal androgen exposure on circulating hormone levels may diminish progressively during development. This observation, combined with transcriptomic evidence from hypothalamic tissue at PND21 (Tac3 upregulation), indicates that critical developmental programming was likely established during earlier stages (PND21). Furthermore, prenatal androgen exposure may lower the intrinsic stimulation threshold required for HPG axis activation in male offspring. Given that post-transcriptional modifications or variations in the regulation and modification of the translational process can result in discrepancies between protein and mRNA expression, we used immunohistochemistry to localize the ARC in male offspring at the onset of puberty and detect target protein levels in this region. Our results demonstrated a significant increase in kisspeptin expression and a significant decrease in dynorphin expression in the ARC of the PNA group compared to controls, after adjusting for the time of puberty onset. KNDy neurons located in the ARC regulate NKB and dynorphin through synaptic connections to fine-tune the release of kisspeptin and GnRH (44). NKB stimulates kisspeptin release by binding to NK3R on KNDy neurons, with kisspeptin subsequently binding to GPR54 to activate GnRH neurons. The release of dynorphin by KNDy neurons terminates the GnRH pulse. Previous studies have demonstrated that bisphenol A exposure promotes early puberty onset by increasing Kiss1 expression in the ARC of juvenile female rats (45). In addition, chronic central administration of Kiss1 peptide in immature female rats advanced the timing of vaginal opening and significantly increased LH levels (46). Makorin ring finger protein 3 (MKRN3) has also been shown to play an important role in puberty onset, and deletion of MKRN3 in female mice led to earlier puberty and was associated with increased NKB levels in the ARC at PND25 (47). Administration of insulin-like growth factor 1 (IGF-1) in female rats also resulted in earlier puberty onset, accompanied by increased NKB expression and decreased dynorphin expression (48). These findings suggest that PNA may reduce dynorphin expression in the ARC of male offspring, diminishing its inhibitory effect. This could lead to increased kisspeptin secretion in the ARC, ultimately resulting in premature activation of GnRH neurons and earlier onset of puberty. However, the differences in kisspeptin, serum GnRH, LH, and FSH levels between the PNA group at puberty onset and controls were not statistically significant (P > 0.05). This may be attributed to the pulsatile nature of GnRH release, which causes fluctuations in serum LH and FSH concentrations.
The secular trend toward earlier puberty onset has emerged as a global public health challenge (49). Our findings suggest that prenatal hyperandrogenic exposure may act as a critical contributing factor to this phenomenon. Specifically, KNDy neurons in the ARC play a pivotal role in mediating the accelerated puberty onset observed in male offspring exposed to elevated testosterone levels during gestation. Targeted interventions modulating the KNDy signaling pathway hold promise for disrupting this adverse developmental trajectory.
Conclusion
Our study demonstrates that KNDy neurons in the ARC play a crucial role in the onset of puberty in male offspring exposed to elevated testosterone levels during late pregnancy. Specifically, prenatal androgen exposure may lead to a reduction in Dyn–KOR signaling in the ARC, which enhances kisspeptin–GPR54 signaling. This, in turn, results in the premature activation of GnRH neurons and the early onset of puberty.
Supplementary materials
Declaration of interest
The authors declare that this study was conducted in the absence of any commercial or financial relationships that could be construed as potential conflicts of interest.
Funding
This work was supported by the General Program of Natural Incubation of Bengbu Medical University (2022byfy001); the 512 Talent Training Program of Bengbu Medical University (by51201204); the Overseas Visiting and Training Programs for Outstanding, Young Core Talents in Universities (gxgwfx2020042); Anhui Provincial University Scientific Research Project (2023AH040285); and the Longhu Talent Project of Bengbu Medical University (LH250203002).
Author contribution statement
Runfei Ge and Yun Zhang contributed to formal analysis, visualization, original draft writing, and investigation. Yongting Yuan, Tingting Li, and GuiYu Qiu were responsible for data curation, investigation, methodology, and validation. Shuaijun Guo contributed to supervision, validation, and project administration. Lianguo Fu was responsible for conceptualization, data curation, funding acquisition, methodology, as well as writing, review, and editing.
Data availability
The RNA-seq data generated in this study have been deposited in the NCBI Gene Expression Omnibus (GEO) under accession number PRJNA1263749. The data used to support the findings of this study are available from the authors upon reasonable request.
Ethics statement
All experimental procedures were approved by the Ethics Committee of Bengbu Medical University (Approval No. (2018) 032).
References
- 1.Bräuner EV, Busch AS, Eckert-Lind C, et al. Trends in the incidence of central precocious puberty and normal variant puberty among children in Denmark, 1998 to 2017. JAMA Netw Open 2020. 3 e2015665. ( 10.1001/jamanetworkopen.2020.15665) [DOI] [PMC free article] [PubMed] [Google Scholar]
- 2.Eckert-Lind C, Busch AS, Petersen JH, et al. Worldwide secular trends in age at pubertal onset assessed by breast development among girls: a systematic review and meta-analysis. JAMA Pediatr 2020. 174 e195881. ( 10.1001/jamapediatrics.2019.5881) [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3.Sørensen K, Aksglaede L, Petersen JH, et al. Recent changes in pubertal timing in healthy Danish boys: associations with body mass index. J Clin Endocrinol Metab 2010. 95 263–270. ( 10.1210/jc.2009-1478) [DOI] [PubMed] [Google Scholar]
- 4.Cheng TS, Day FR, Lakshman R, et al. Association of puberty timing with type 2 diabetes: a systematic review and meta-analysis. PLoS Med 2020. 17 e1003017. ( 10.1371/journal.pmed.1003017) [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5.O’Kelly AC, Michos ED, Shufelt CL, et al. Pregnancy and reproductive risk factors for cardiovascular disease in women. Circ Res 2022. 130 652–672. ( 10.1161/CIRCRESAHA.121.319895) [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6.Copeland WE, Worthman C, Shanahan L, et al. Early pubertal timing and testosterone associated with higher levels of adolescent depression in girls. J Am Acad Child Adolesc Psychiatry 2019. 58 1197–1206. ( 10.1016/j.jaac.2019.02.007) [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7.Fuhrman BJ, Moore SC, Byrne C, et al. Association of the age at menarche with site-specific cancer risks in pooled data from nine cohorts. Cancer Res 2021. 81 2246–2255. ( 10.1158/0008-5472.CAN-19-3093) [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8.Brito VN, Canton APM, Seraphim CE, et al. The congenital and acquired mechanisms implicated in the etiology of central precocious puberty. Endocr Rev 2023. 44 193–221. ( 10.1210/endrev/bnac020) [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9.Malin IA, Rahman A, Johansson ALV, et al. Early life environmental exposure to cadmium, lead, and arsenic and age at menarche: a longitudinal mother-child cohort study in Bangladesh. Environ Health Perspect 2023. 131 27003. ( 10.1289/EHP11121) [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10.Lam BYH, Williamson A, Finer S, et al. MC3R links nutritional state to childhood growth and the timing of puberty. Nature 2021. 599 436–441. ( 10.1038/s41586-021-04088-9) [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11.Wang G, Radovick S, Buckley JP, et al. Plasma insulin concentration in newborns and children and age at menarche. Diabetes Care 2023. 46 1231–1238. ( 10.2337/dc22-2017) [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12.Abreu AP & Kaiser UB. Pubertal development and regulation. Lancet Diabetes Endocrinol 2016. 4 254–264. ( 10.1016/S2213-8587(15)00418-0) [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13.Maggi R, Cariboni AM, Marelli MM, et al. GnRH and GnRH receptors in the pathophysiology of the human female reproductive system. Hum Reprod Update 2016. 22 358–381. ( 10.1093/humupd/dmv059) [DOI] [PubMed] [Google Scholar]
- 14.Chen X, Xiao Z, Cai Y, et al. Hypothalamic mechanisms of obesity-associated disturbance of hypothalamic–pituitary–ovarian axis. Trends Endocrinol Metab 2022. 33 206–217. ( 10.1016/j.tem.2021.12.004) [DOI] [PubMed] [Google Scholar]
- 15.Silva MSB, Desroziers E, Hessler S, et al. Activation of arcuate nucleus GABA neurons promotes luteinizing hormone secretion and reproductive dysfunction: implications for polycystic ovary syndrome. EBioMedicine 2019. 44 582–596. ( 10.1016/j.ebiom.2019.05.065) [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16.Sánchez-Garrido MA, García-Galiano D & Tena-Sempere M. Early programming of reproductive health and fertility: novel neuroendocrine mechanisms and implications in reproductive medicine. Hum Reprod Update 2022. 28 346–375. ( 10.1093/humupd/dmac005) [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17.Navarro VM. Metabolic regulation of kisspeptin – the link between energy balance and reproduction. Nat Rev Endocrinol 2020. 16 407–420. ( 10.1038/s41574-020-0363-7) [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18.Uenoyama Y, Inoue N, Nakamura S, et al. Central mechanism controlling pubertal onset in mammals: a triggering role of Kisspeptin. Front Endocrinol 2019. 10 312. ( 10.3389/fendo.2019.00312) [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19.Han SY, Morris PG, Kim JC, et al. Mechanism of kisspeptin neuron synchronization for pulsatile hormone secretion in male mice. Cell Rep 2023. 42 111914. ( 10.1016/j.celrep.2022.111914) [DOI] [PubMed] [Google Scholar]
- 20.Navarro VM, Castellano JM, McConkey SM, et al. Interactions between kisspeptin and neurokinin B in the control of GnRH secretion in the female rat. Am J Physiol Endocrinol Metab 2011. 300 E202–E210. ( 10.1152/ajpendo.00517.2010) [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21.Moore AM, Coolen LM & Lehman MN. In vivo imaging of the GnRH pulse generator reveals a temporal order of neuronal activation and synchronization during each pulse. Proc Natl Acad Sci U S A 2022. 119 e2117767119. ( 10.1073/pnas.2117767119) [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22.Panda SP, Kesharwani A, Singh GD, et al. Impose of KNDy/GnRH neural circuit in PCOS, ageing, cancer and Alzheimer’s disease: StAR actions in prevention of neuroendocrine dysfunction. Ageing Res Rev 2023. 92 102086. ( 10.1016/j.arr.2023.102086) [DOI] [PubMed] [Google Scholar]
- 23.Nagae M, Uenoyama Y, Okamoto S, et al. Direct evidence that KNDy neurons maintain gonadotropin pulses and folliculogenesis as the GnRH pulse generator. Proc Natl Acad Sci U S A 2021. 118 e2009156118. ( 10.1073/pnas.2009156118) [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24.Shahab M, Mastronardi C, Seminara SB, et al. Increased hypothalamic GPR54 signaling: a potential mechanism for initiation of puberty in primates. Proc Natl Acad Sci U S A 2005. 102 2129–2134. ( 10.1073/pnas.0409822102) [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25.Navarro VM, Ruiz-Pino F, Sánchez-Garrido MA, et al. Role of neurokinin B in the control of female puberty and its modulation by metabolic status. J Neurosci 2012. 32 2388–2397. ( 10.1523/JNEUROSCI.4288-11.2012) [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26.Azziz R, Carmina E, Chen Z, et al. Polycystic ovary syndrome. Nat Rev Dis Primers 2016. 2 16057. ( 10.1038/nrdp.2016.57) [DOI] [PubMed] [Google Scholar]
- 27.Yang R, Li Q, Zhou Z, et al. Changes in the prevalence of polycystic ovary syndrome in China over the past decade. Lancet Reg Health West Pac 2022. 25 100494. ( 10.1016/j.lanwpc.2022.100494) [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28.Doherty DA, Newnham JP, Bower C, et al. Implications of polycystic ovary syndrome for pregnancy and for the health of offspring. Obstet Gynecol 2015. 125 1397–1406. ( 10.1097/AOG.0000000000000852) [DOI] [PubMed] [Google Scholar]
- 29.Li T, Meng Y, Yao R, et al. The associations between left-hand digit ratio (2D:4D) and puberty characteristics among Chinese girls. Early Hum Dev 2019. 130 22–26. ( 10.1016/j.earlhumdev.2019.01.007) [DOI] [PubMed] [Google Scholar]
- 30.Lunddorf LLH, Arendt LH, Ernst A, et al. Maternal polycystic ovarian syndrome and pubertal development in daughters and sons: a population-based cohort study. Hum Reprod 2022. 37 2623–2634. ( 10.1093/humrep/deac197) [DOI] [PubMed] [Google Scholar]
- 31.Liu J, Yuan Y, Peng X, et al. Mechanism of leptin-NPY on the onset of puberty in male offspring rats after androgen intervention during pregnancy. Front Endocrinol 2023. 14 1090552. ( 10.3389/fendo.2023.1090552) [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32.DeChick A, Hetz R, Lee J, et al. Increased skeletal muscle fiber cross-sectional area, muscle phenotype shift, and altered insulin signaling in rat hindlimb muscles in a prenatally androgenized rat model for polycystic ovary syndrome. Int J Mol Sci 2020. 21 7918. ( 10.3390/ijms21217918) [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33.Noroozzadeh M, Raoufy MR, Bidhendi Yarandi R, et al. The effects of prenatal androgen exposure on cardiac function and tolerance to ischemia/reperfusion injury in male and female rats during adulthood. Life Sci 2019. 229 251–260. ( 10.1016/j.lfs.2019.05.044) [DOI] [PubMed] [Google Scholar]
- 34.Manti M, Fornes R, Pironti G, et al. Maternal androgen excess induces cardiac hypertrophy and left ventricular dysfunction in female mice offspring. Cardiovasc Res 2020. 116 619–632. ( 10.1093/cvr/cvz180) [DOI] [PubMed] [Google Scholar]
- 35.Sanchez-Garrido MA & Tena-Sempere M. Metabolic dysfunction in polycystic ovary syndrome: pathogenic role of androgen excess and potential therapeutic strategies. Mol Metabol 2020. 35 100937. ( 10.1016/j.molmet.2020.01.001) [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36.Ikegami K, Watanabe Y, Nakamura S, et al. Cellular and molecular mechanisms regulating the KNDy neuronal activities to generate and modulate GnRH pulse in mammals. Front Neuroendocrinol 2022. 64 100968. ( 10.1016/j.yfrne.2021.100968) [DOI] [PubMed] [Google Scholar]
- 37.Topaloglu AK, Tello JA, Kotan LD, et al. Inactivating KISS1 mutation and hypogonadotropic hypogonadism. N Engl J Med 2012. 366 629–635. ( 10.1056/NEJMoa1111184) [DOI] [PubMed] [Google Scholar]
- 38.Topaloglu AK, Reimann F, Guclu M, et al. TAC3 and TACR3 mutations in familial hypogonadotropic hypogonadism reveal a key role for neurokinin B in the central control of reproduction. Nat Genet 2009. 41 354–358. ( 10.1038/ng.306) [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39.Yan X, Yuan C, Zhao N, et al. Prenatal androgen excess enhances stimulation of the GNRH pulse in pubertal female rats. J Endocrinol 2014. 222 73–85. ( 10.1530/JOE-14-0021) [DOI] [PubMed] [Google Scholar]
- 40.Messager S, Chatzidaki EE, Ma D, et al. Kisspeptin directly stimulates gonadotropin-releasing hormone release via G protein-coupled receptor 54. Proc Natl Acad Sci U S A 2005. 102 1761–1766. ( 10.1073/pnas.0409330102) [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41.Popa SM, Clifton DK & Steiner RA. The role of kisspeptins and GPR54 in the neuroendocrine regulation of reproduction. Annu Rev Physiol 2008. 70 213–238. ( 10.1146/annurev.physiol.70.113006.100540) [DOI] [PubMed] [Google Scholar]
- 42.Aerts EG, Griesgraber MJ, Thomson AL, et al. Activity of KNDy neurons and the melanocortin system during pubertal development in female sheep. Biol Reprod 2025. ioaf074. ( 10.1093/biolre/ioaf074) [DOI] [PubMed] [Google Scholar]
- 43.Si R, Pan D, Wang Z, et al. Regulation of the central melanocortin system on energy balance in mammals and birds. Neuropeptides 2022. 95 102267. ( 10.1016/j.npep.2022.102267) [DOI] [PubMed] [Google Scholar]
- 44.Uenoyama Y, Nagae M, Tsuchida H, et al. Role of KNDy neurons expressing kisspeptin, neurokinin B, and dynorphin A as a GnRH pulse generator controlling mammalian reproduction. Front Endocrinol 2021. 12 724632. ( 10.3389/fendo.2021.724632) [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45.Dong W, He J, Wang J, et al. Bisphenol A exposure advances puberty onset by changing Kiss1 expression firstly in arcuate nucleus at juvenile period in female rats. Reprod Toxicol 2022. 110 141–149. ( 10.1016/j.reprotox.2022.04.005) [DOI] [PubMed] [Google Scholar]
- 46.Navarro VM, Fernández-Fernández R, Castellano JM, et al. Advanced vaginal opening and precocious activation of the reproductive axis by KiSS-1 peptide, the endogenous ligand of GPR54. J Physiol 2004. 561 379–386. ( 10.1113/jphysiol.2004.072298) [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47.Naulé L, Mancini A, Pereira SA, et al. MKRN3 inhibits puberty onset via interaction with IGF2BP1 and regulation of hypothalamic plasticity. JCI insight 2023. 8 e164178. ( 10.1172/jci.insight.164178) [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48.Dees WL, Hiney JK & Srivastava VK. Regulation of prepubertal dynorphin secretion in the medial basal hypothalamus of the female rat. J Neuroendocrinol 2019. 31 e12810. ( 10.1111/jne.12810) [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49.Bräuner EV, Busch AS, Eckert-Lind C, et al. Trends in the incidence of central precocious puberty and normal variant puberty among children in Denmark, 1998 to 2017. JAMA Netw Open 2020. 3 e2015665. ( 10.1001/jamanetworkopen.2020.15665) [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
Data Availability Statement
The RNA-seq data generated in this study have been deposited in the NCBI Gene Expression Omnibus (GEO) under accession number PRJNA1263749. The data used to support the findings of this study are available from the authors upon reasonable request.

This work is licensed under a 


