Next Article in Journal
Optimizing Selenium Polysaccharide Supplementation: Impacts on Growth, Oxidative Stress, and Tissue Selenium in Juvenile Large Yellow Croaker (Larimichthys crocea)
Previous Article in Journal
Spatiotemporal Activity Patterns of Sympatric Rodents and Their Predators in a Temperate Desert-Steppe Ecosystem
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

Metabolomic and Molecular Mechanisms of Glycerol Supplementation in Regulating the Reproductive Function of Kazakh Ewes in the Non-Breeding Season

College of Animal Science and Technology, Shihezi University, Shihezi 832000, China
*
Author to whom correspondence should be addressed.
Animals 2025, 15(15), 2291; https://doi.org/10.3390/ani15152291
Submission received: 22 May 2025 / Revised: 21 July 2025 / Accepted: 29 July 2025 / Published: 5 August 2025
(This article belongs to the Section Small Ruminants)

Simple Summary

This study focuses on seasonal reproduction in sheep, the core problem of efficient production, and innovatively proposes a theoretical framework of “metabolic reprogramming for estrus”. Because the key metabolic nodes and interactions across tissues in nutrition-induced estrus in non-breeding ewes have not been elucidated, this project uses glycerol as a metabolic intervention carrier and reveals three major scientific points through the strategy of metabolomics integration: (1) How energy signalling regulates the activation of Kisspeptin/GPR54 signalling in the hypothalamus through metabolite-hormone axes; (2) The lipolytic metabolite (L-carnitine) cooperates with the microenvironment of ovarian steroid production to promote follicular development; (3) The contribution of the bile acid–metabolism axis to seasonal reproductive regulation. The research results will be the first to map out the “metabolite–neuroendocrine–reproductive phenotype” panoramic regulatory network, providing an innovative theoretical paradigm for the precise nutritional control of the reproduction cycle in ruminants.

Abstract

The activation mechanism of the reproductive axis in Kazakh ewes during the non-breeding season was explored by supplementation with glycerol complex (7% glycerol + tyrosine + vitamin B9). The experiment divided 50 ewes into five groups (n = 10). After 90 days of intervention, it was found that significant changes in serum DL-carnitine, N-methyl-lysine and other differential metabolites were observed in the GLY-Tyr-B9 group (p < 0.05, “p < 0.05” means significant difference, “p < 0.01” means “highly significant difference”). The bile acid metabolic pathway was specifically activated (p < 0.01). The group had a 50% estrus rate, ovaries contained 3–5 immature follicles, and HE staining showed intact granulosa cell structure. Serum E2/P4 fluctuated cyclically (p < 0.01), FSH/LH pulse frequency increased (p < 0.01), peak Glu/INS appeared on day 60 (p < 0.05), and LEP was negatively correlated with body fat percentage (p < 0.01). Molecular mechanisms revealed: upregulation of hypothalamic kiss-1/GPR54 expression (p < 0.01) drove GnRH pulses; ovarian CYP11A1/LHR/VEGF synergistically promoted follicular development (p < 0.05); the HSL of subcutaneous fat was significantly increased (p < 0.05), suggesting involvement of lipolytic supply. Glycerol activates the reproductive axis through a dual pathway—L-carnitine-mediated elevation of mitochondrial β-oxidation efficacy synergizes with kisspeptin/GPR54 signalling enhancement to re-establish HPO axis rhythms. This study reveals the central role of metabolic reprogramming in regulating seasonal reproduction in ruminants.

1. Introduction

Seasonal reproduction is an important physiological feature for ruminants to adapt to the environment. However, its resulting reproductive inefficiency has become a key bottleneck restricting sheep industry development. Studies have shown that ewes are generally in a state of negative energy balance (Body Condition Score BCS < 2.5) during the spring to early summer period (March–June), which triggers the inhibition of the hypothalamic Kisspeptin/GnRH signalling pathway, ultimately leading to functional stagnation of the HPG axis [1]. This phenomenon is closely associated with impaired follicular development: in the energy-deprived state, sinus follicle diameter is reduced by 40% and granulosa cell apoptosis is elevated to 68%, while follicular fluid IGFBP-3 concentration is significantly increased (3.2 ± 0.4 vs. 1.8 ± 0.3 ng/mL), inhibiting the follicle maturation-promoting effect of IGF-1 [2].
Nutritional interventions are an effective strategy to overcome seasonal reproductive constraints, focusing on re-establishing reproductive endocrine homeostasis through metabolic reprogramming [3,4,5]. Notably, energy-rich feeds (e.g., glycerol) demonstrate significantly higher proestrus induction efficiency (50–70%) than protein-rich feeds (20–30%), indicating a pivotal role of energy metabolism pathways in reproductive regulation [6]. Previous studies demonstrated that a 7-day oral glycerol intervention significantly increased the ovulation rate in Manchega ewes, primarily via enhanced β-oxidation-derived energy supply and elevated follicular fluid IGF-1 concentrations [7]. This finding suggests that energy metabolism reprogramming regulates seasonal reproductive rhythms through the TH-DIO2/DIO3-GnRH and Kisspeptin/GnRH dual signalling systems. Negative energy homeostasis in females directly inhibited Kisspeptin neuron activity in the arcuate nucleus of the hypothalamus, leading to a decrease in the frequency of GnRH pulses. Meza-Herrera’s team further demonstrated that short-term ad libitum feeding (metabolizable energy intake ↑ 30%) promotes corpus luteum function and coordinates LH pulse secretion (amplitude ↑ 40%, p < 0.01) [8]. Leptin acts as a metabolic–reproductive signalling hub, and Leptin regulates energy balance in both directions through the JAK2/STAT3 pathway. Exogenous leptin (5 mg/kg BW) advanced the onset of GnRH pulses in preprimary rats by 3.2 days (p < 0.01), and energy restriction led to a decrease in adipose tissue Leptin receptor (LEPR) expression by 52%, triggering leptin resistance [9]. Notably, Leptin can directly act on hypothalamic POMC neurons through the blood-brain barrier, activating the ERK1/2 signalling cascade (p-ERK ↑ 3.5-fold) to promote GnRH release [10]. Collectively, these findings demonstrate that leptin coordinates metabolic–reproductive homeostasis by modulating hypothalamic–pituitary–gonadal (HPG) axis function, establishing a mechanistic basis for targeted nutritional interventions.
In recent years, significant breakthroughs have been achieved in understanding seasonal reproduction regulation mechanisms. Yang et al. [11] first revealed the central regulatory role of the GNAQ gene in the hypothalamic–pituitary–ovarian (HPG) axis by constructing a multi-omics database (miRNA/mRNA) of estrus and anestrus in Kazakh sheep during the non-breeding season. This gene is highly expressed across all three levels of the reproductive axis and functions through bidirectional regulation: ① Regulating key enzymes in vitamin B9 metabolism (MTHFR for methyltetrahydrofolate reductase) and tyrosine hydroxylase activity to remodel reproductive hormone secretion rhythms; ② Exhibiting a highly significant positive correlation with light signal intensity (p < 0.01), indicating its role as a “light-nutrient integration node”. Building on this regulatory framework, Lin et al. [12] systematically analyzed molecular events in the HPG axis using a high-nutrient diet-induced estrus ewe model. Their study demonstrated that Tyr and B9 synergistically induce estrus via the leptin–estrogen axis independently of photoperiod, with spatiotemporal-specific mechanisms—Epigenetic regulation: B9 promotes ovarian steroid synthesis through DNMT3B-mediated DNA demethylation (phosphorylation sequencing revealed 40% reduction in CYP19A1 promoter methylation) [13]; Cellular signaling: Tyr drives granulosa cell proliferation and LH receptor expression by activating the NTRK2/PI3K-AKT pathway [14,15]. These findings collectively support a “nutrient-epigenetic crosstalk” model for seasonal reproduction regulation, providing a novel theoretical framework for ruminant reproductive management. This mechanism was validated in a cross-species study: an L-tyrosine supplementation intervention (1.0 g/kg BW) was administered to ewes 7 days prior to the initiation of the breeding season, and it was found that the estrous response rate of ewes in the experimental group was significantly higher than that of the control group [16] and that the B9 intervention in gestating ewes led to an increase in the number of live born piglets in the litter by 2.3 (p < 0.01) [17].
In this study, we focused on the metabolomic differences between GLY and traditional nutritional regulatory mechanisms: (1) using LC-MS to screen for serum differential metabolites, focusing on bile acids and lysophospholipids metabolic profiles, and (2) constructing a three-level regulatory network of “metabolite-methylation modification-hormone pulse”. This study reveals the central role of metabolic reprogramming in regulating seasonal reproduction in ruminants.

2. Materials and Methods

2.1. Animals, Experimental Design, and Diets

In this study, 50 healthy parturient Kazakh ewes (3–5 years old, weighing 38.0 ± 0.92 kg) were selected and divided into five groups (n = 10/group) using a completely randomized design: Control group (basal diet, NRC 2007 standard [18], Table 1), GLY group (basal diet + 7% GLY (Dry matter basis, converted to 70 mL/head/day), purity ≥ 99.5%), GLY-Tyr group (basal diet + 7% GLY + 100 mg/kg Tyr, Sigma-Aldrich® (Saint Louis, MO, USA) CAS 60-18-4), GLY-B9 group (basal diet + 7% GLY + 6 mg/kg B9, Sigma-Aldrich® CAS 59-30-3) and GLY-Tyr-B9 group (basal diet + 7% GLY + 100 mg/kg Tyr + 6 mg/kg B9). During the non-breeding season (1 March–30 May 2023), a daily gavage intervention was administered at 10:00 a.m. for 90 consecutive days.
Before the test, the pen was thoroughly cleaned and disinfected, and the sheep were grouped by vaccination and deworming status, as well as by shearing and weighing. They were then kept in semi-open sheds with consistent environmental conditions (temperature, ventilation, and light).

2.2. Sample Preparation and Analyses

In this study, we used a multi-dimensional method to assess the estrus status of ewes systematically, including (1) ligated ram estrus test method (daily 07:00/20:00, recording the acceptance of crawling across the behavior); (2) vulvar behavioral observation; (3) reproductive hormone test (E2, P4, LH, and FSH; the kits were purchased from Jiangsu Jingmei Biotechnology Co., Ltd., Yancheng, China): (4) Nanjing Jianzhong Bioengineering Institute, Nanjing, China provided Glu, INS, LEP detection kits. Samples (5× diluted) and standards were incubated with enzyme conjugate (37 °C, 60 min), washed five times, developed with TMB substrate (15 min), stopped, and read at 450 nm. Concentrations were calculated against standard curves.
At the end of the experimental cycle, the hypothalamus, ovary, and subcutaneous adipose tissues were collected from estrous ewes of the test group and non-estrous ewes of the control group, then passed through the Total RNA (A260/A280 = 1.8–2.0) was extracted using the Tissue RNA Extraction Kit (Thermo Fisher Scientific, Beijing, China). cDNA was synthesized by HiFiScript gDNA Removal RT MasterMix (CWBIO, Taizhou, China). The qRT-PCR reaction system was 25 μL (containing 12 μL of SYBR Premix EX Taq II, 1 μL each of forward/reverse primers, 2 μL of cDNA template, 9 μL of RNase-Free ddH2O), and the amplification program was set as follows: pre-denaturation at 95 °C for 5 min; 40 cycles of 95 °C 10 s denaturation → 60 °C 30 s annealing/extension; and the amplification specificity was determined by the unstranding temperature (Tm value) ± 0.8 °C at the melting curve analysis stage. A dual-platform validation using ABI PRISM 7500 (Applied Biosystems, Waltham, MA, USA) and Roche LightCycler480 II (Roche Diagnostics, Rotkreuz, Switzerland) was used to standardize the assay using sheep β-actin as an internal reference gene and to detect reproduction-related genes (GnRH, GnAQ, kiss-1, GPR54, CYP11A1, LHR, VEGF, and StAR). Lipid metabolism genes (HSL) were differentially expressed (primer sequences are shown in Table 2, validated by NCBI Primer-BLAST (https://blast.ncbi.nlm.nih.gov/Blast.cgi, accessed on 15 February 2024)).

2.3. Serum Non-Targeted Metabolomics

On the 90th day of the test, 5 mL of jugular vein blood was obtained from estrous ewes in the test group and non-estrous ewes in the control group (n = 6). Serum was separated and submitted to Novozymes Biotechnology Co., Ltd., (Shanghai, China) for UHPLC-QTOF-MS analysis. Aliquots of 100 μL serum were mixed with 400 μL of 80% (v/v) aqueous methanol, followed by vortexing and centrifugation at 15,000× g for 20 min at 4 °C. The supernatant was diluted to 53% (v/v) methanol content and recentrifuged under identical conditions.
Chromatographic separation was employed with a Waters ACQUITY UPLC system equipped with a Hypersil Gold C18 column (100 × 2.1 mm, 1.9 μm; Thermo Fisher) using 0.1% (v/v) formic acid in water (mobile phase A) and acetonitrile (mobile phase B) with a 16 min gradient at 0.2 mL/min. Mass spectrometric detection was performed on a Q Exactive HF-X mass spectrometer (Thermo Fisher) with electrospray ionization (spray voltage: 3.5 kV). Full scans (m/z 100–1500) were acquired in both positive and negative modes from 0–12 min with the following parameters: auxiliary gas flow 10 L/min, capillary temperature 320 °C, S-lens RF level 60, heater temperature 350 °C, and data-dependent MS/MS acquisition.

2.4. Statistical Analyses

Metabolomics data were analyzed using the following process: raw mass spectrometry data were pre-processed by Compound Discoverer 3.1 software, and the characteristic peaks were screened by retention time deviation (±0.2 min) and mass-to-charge ratio deviation (≤5 ppm) and combined with the molecular peaks and fragmentation ions to match the mzCloud, mzVault and Masslist databases. Metabolite characterization was completed using KEGG (https://www.genome.jp/kegg/pathway.html, accessed on 15 February 2024), HMDB (https://hmdb.ca/metabolites, accessed on 15 February 2024), and LIPIDMAPS (http://www.lipidmaps.org/, accessed on 15 February 2024) databases for metabolic pathway annotation, and reliable quantitative results were obtained by QC sample quality control (CV < 30%). After data standardization (relative peak area method) using metaX software 2.33, multivariate statistical analyses were performed: principal component analysis (PCA) to reveal the trend of separation between groups, partial least squares-discriminant analysis (PLS-DA) to calculate the projected importance of the variables (VIP value), and the combination of univariate t-tests (p < 0.05) with the multiplicity of variance (FC > 1.5 or <0.8) to screen for significantly different Metabolites. Statistical analyses were performed using R 4.3 (ggplot 2 package to plot volcano/bubble plots; complot package 1.8 to display Pearson correlation matrices) and Python 3.10 (scikit-learn machine learning library); the significance of difference validation was performed by Duncan multiple comparisons (p < 0.05) using SPSS 26.0, and visualization was performed using GraphPad Prism 9.0.
qRT-PCR data were processed by the 2−ΔΔCt method, and one-way analysis of variance (ANOVA) with Duncan’s multiple comparisons (SPSS 26.0) was used, and the results were expressed as mean ± standard error (“p < 0.05” indicates significant difference, which is labeled with “*” in the figure, and “p < 0.01” indicates “highly significant difference, which is labeled with “**” than labeled), and the final graphs were completed by GraphPad Prism 9 Drawing. Three biological replicates were set for each treatment, and three technical replicates were performed for each biological replicate.

3. Results

3.1. Effect of Glycerol Mixtures on Estrus Rate of Ewes in the Non-Breeding Season

In the non-breeding season, ewes supplemented with glycerin mixture all show the initial behavior (approaching the ram, wagging their tails, and riding to accept), and their vulva is red and secretes mucus (Figure 1). The GLY-Tyr-B9 group showed optimal estrus induction (50% estrus rate, Table 3). Dynamic hormone monitoring revealed pulsatile E2 fluctuations synchronized with LH/FSH secretion rhythms (p < 0.01), alongside fluctuating P4 levels (p < 0.01). Ovarian morphology indicated 3–5 immature follicles (2.3 ± 0.5 mm diameter) in estrous ewes. HE staining demonstrated structurally intact primary follicles: deeply stained oocyte nuclei (hematoxylin), eosinophilic cytoplasm (eosin), tightly arranged monolayered granulosa cells (Figure 2A, arrowheads), and characteristic hyaline bands between oocytes and granulosa cells (Figure 2B, asterisks).

3.2. Regulatory Effects of Glycerol Complexes on Reproductive Hormone Secretion Patterns in Ewes

Dynamic monitoring of serum reproductive hormones revealed that non-breeding season-induced estrous ewes exhibited a simulated artificial cycle [19] (Figure 3). P4 dynamics: Progressive increase from day 4, peaking at day 13 (18.8 ± 1.2 ng), followed by decline to baseline by day 18, mimicking natural luteal phase patterns. E2 pulsatility: Significant peaks at days 4, 10, and 16–18 (p < 0.01), contrasting with low-fluctuating CON levels. LH/FSH synchronization: LH surges at days 4–5, 10, 14–18, and 20 (p < 0.01); FSH peaks at days 14–15 (p < 0.01), phase-synchronized with LH. Glycerol complex induces estrus of ewes in the non-breeding season through the HPO axis, achieving a 50% estrus rate in the GLY-Tyr-B9 group.

3.3. Dynamic Regulation of Lipid Metabolism Hormones in Ewes by Glycerol Complex

At the beginning of the experiment (day 0), no significant differences in glucose (Glu), insulin (INS), or leptin (LEP) levels were observed among groups (p > 0.05). Dynamic monitoring revealed (Figure 4). Glu: All groups exhibited an initial increase followed by decline (p < 0.05); INS: GLY, GLY-Tyr, and GLY-B9 groups peaked at day 30 (22.7 ± 1.5 ng/mL, p < 0.05), maintaining higher levels vs. controls at day 60 (16.4 ± 2.1 ng/mL, p < 0.05) despite a decrease from peak. The GLY-Tyr-B9 group showed a 28.7% higher INS peak vs. GLY alone (p < 0.05); LEP: Progressive rise to 14.2 ± 0.5 ng/mL at day 60 (p < 0.05) after transient elevation (13.8 ± 0.3 ng/mL at day 30, p > 0.05), stabilizing at 13.5±0.4 ng/mL by day 90. These findings indicate that Glu, INS, and LEP dynamics correlate with energy intake and are modulated by synergistic nutrient interactions, driving adipose accumulation and leptin feedback enhancement.

3.4. Regulatory Effects of Nutritional Interventions on Gene Expression in the Reproductive-Metabolic Axis of Sheep

qRT-PCR analysis revealed supplementary feeding-induced regulatory effects on reproductive-metabolic genes in non-breeding season ewes (Figure 5): Hypothalamic pathway: GNAQ was significantly downregulated (p < 0.01), suggesting its role as a photoperiod-sensitive node in estrus suppression. The kisspeptin system exhibited cascade activation: Kiss1 ↑ (p < 0.01) → GPR54 ↑ (p < 0.05) → GnRH ↑ (p < 0.05), establishing a “GNAQ-Kiss1-GnRH” regulatory axis. Ovarian steroidogenesis: Upregulation of StAR (p < 0.05) and CYP11A1 (p < 0.01) accelerated cholesterol-to-pregnenolone conversion. Follicular development: LHR and VEGF upregulation (p < 0.01) enhanced angiogenesis and luteal functionality. Lipid mobilization: HSL upregulation (p < 0.05) redirected energy metabolism toward reproductive allocation.

3.5. Metabolomics QC and PCA

Untargeted serum metabolomics using UHPLC-QTOF-MS identified 1082 metabolites (ESI+: 618; ESI−: 464). Lipid and lipid-like molecules dominated the metabolic profile (60.7%), followed by amino acids/derivatives (27.2%) and organic acids (12.1%) (Figure 6A). High reproducibility was confirmed by Pearson correlations (Figure 6B). PCA revealed significant metabolic divergence between control and experimental groups (GLY, GLY-Tyr, GLY-B9, GLY-Tyr-B9) along PC1/PC2 (p < 0.01), with tight intra-group clustering (>90% variance explained), demonstrating robust system stability (Figure 6C).

3.6. Metabolomics Multivariate Statistical Analysis

PLS-DA modeling revealed that supplemental feeding significantly remodeled ewe metabolic profiles. Model validation demonstrated high goodness-of-fit (R2 > 0.92) with predictive reliability (Q2 = 0.10–0.66, R2 > Q2). Permutation tests confirmed robustness (p < 0.001, slope < 0.3). PLS-DA score plots showed clear metabolic separation between experimental groups (GLY, GLY-Tyr, etc.) and the NC group (p < 0.01), with tight intra-group clustering (Hotelling T2 > 95%) (Figure 7), indicating glycerol complexes specifically modulate metabolic networks.

3.7. Differential Metabolite Screening and Functional Analysis

Based on LC-MS/MS non-targeted metabolomics technology, 281 differential metabolites were identified using variable importance projection (VIP > 1.0), fold difference (FC > 1.2 or FC < 0.833) and statistical significance (p < 0.05) as screening criteria. Visualized by volcano plot (Figure 8A) and matchstick plot (Figure 8B), it was found that: Gly vs. NC group: 30 metabolites were significantly upregulated (e.g., lysophosphatidylcholine ↑), and 27 were significantly downregulated (e.g., leucine ↓ 35%); B9-GLY vs. NC group: 18 were upregulated (ergosterol ↑), and 42 were downregulated (taurocholic acid ↓); Tyr -GLY vs. NC group: 62 upregulated (DL-carnitine ↑), 23 downregulated (glutamine ↓); B9-Tyr-GLY vs. NC group: 35 upregulated (arachidonic acid ↑), and 44 downregulated (tryptophan ↓).
Differential metabolites were enriched in three major categories (Table 4): glycerophospholipids, fatty acids/derivatives, and amino acids/metabolites. Cluster analysis (Figure 9) and correlation analysis (Figure 10) identified DL-carnitine as a core metabolite with multifunctional roles. Energy supply: Enhanced mitochondrial β-oxidation (ATP), supporting follicular development; Ovarian activation: Stimulated IGF-1/PI3K-AKT pathway (Progesterone ↑); Methylation synergy: Promoted lysine methylation (N-methyllysine ↑) with B-vitamin cofactors (CoA ↑; B5 ↑); Biosynthesis regulation: N-methyllysine modulated lysine substrate availability, elevating DL-carnitine biosynthesis. These results demonstrate multilevel metabolic–reproductive axis regulation by nutritional interventions.

3.8. Functional Analysis and Regulatory Network of Metabolic Pathways

KEGG pathway analysis (Table 5, Figure 11) revealed differential metabolites enriched in lipid/energy/amino acid metabolism. Gly group: Steroid biosynthesis (CYP11A1 ↑, Pregnenolone ↑) with suppressed prolactin signaling (PRL ↓) synergistically reset estrous cyclicity. Tyr-Gly group: Retinol metabolism activated RARα-mediated follicular maturation (E2 ↑). B9-Gly group: Folate metabolism reduced CYP19A1 promoter methylation while ABC transporters enhanced cholesterol flux (HDL ↑). B9-Tyr-Gly group: Multipathway synergy (DL-Carnitine ↑, VEGF ↑) promoted ovarian angiogenesis via cGMP-PKG signaling, with arginine-driven polyamine synthesis supporting proliferation. The glycerol complex induced non-seasonal estrus through three-tiered regulation: Lipid initiation: Steroid/bile acid cycles provided hormonal precursors. Energy adaptation: Retinoic/folate axes optimized follicular energy homeostasis. Epigenetic remodeling: DNMT3B-mediated CYP19A1 demethylation synergized with Kisspeptin/GnRH pulses. This establishes a “metabolism–epigenetic–neural” framework for regulating ruminant seasonal reproduction.

4. Discussion

4.1. Effectiveness of Glycerol Complex in Inducing Estrus in Ewes During the Non-Breeding Season

In the present study, all the experimental groups supplemented with glycerol complex successfully induced estrous behaviors [20], with the rate of estrus in the GLY-Tyr-B9 group being significantly higher than that of the other treatment groups (50% vs. 30–35%). Ovarian morphology showed 3–5 primary follicles (2.3 ± 0.5 mm in diameter) in the right ovary of oestrous individuals, and HE-stained sections further confirmed that the follicles were in the primary developmental stage. This phenomenon is consistent with the study of Zhu Mengting et al. [6], suggesting that high-energy supplementation can improve the ovarian microenvironment and promote early follicular development through metabolic reprogramming.

4.2. Remodeling Effects of Glycerol Complex on Reproductive Hormone Rhythms in Ewes

FSH and LH, as glycoprotein hormones secreted by the pituitary gland, play a central role in mammalian reproductive activities by synergistically regulating the processes of follicle recruitment, dominance and maturation [21,22]. In this study, we found that supplemental feeding of glycerol complex during the non-breeding season significantly remodelled the reproductive endocrine network in ewes: FSH in ewes of the supplemented group showed a typical gonadotropin pulsatile secretion (peak 12.5 ± 0.7 ng/mL vs. 1.8 ± 0.3 ng/mL in the CON group). Its fluctuation was significantly higher than that of the blank control group (CON group) (p < 0.01), which was in high agreement with the hormone during the oestrous period of the Parmesan meat goat pattern and was highly consistent [23]. Seasonal monitoring data of Kazakh ewes further confirmed that the basal level of FSH was 1.8-fold higher (p < 0.01) during the estrous period compared with that during the anaestrus period [24], suggesting that nutritional intervention can mimic physiological reproductive rhythms. A significant E2 peak appeared on day 17 in the supplemented group (913.65 ± 39.63 pg/mL vs. 22.7 ± 3.8 pg/mL in the CON group, p < 0.01), which was consistent with the evolutionarily conserved feature of 72-fold elevation of E2 levels during the reproductive period of Sichuan snub-nosed monkeys [25]. P4 levels peaked at 18.8 ± 1.2 ng/mL on day 13, with the dynamic profile mirroring luteal phase characteristics of natural estrous cycles in Haribaqing ewes [26]. These findings demonstrate the physiological recapitulation of glycerol mixture-induced estrus during non-breeding seasons. Steroid hormones suppress LH secretion by inhibiting Kisspeptin neuronal activity under nutritional deprivation [27]. In this study, glycerol-based intervention induced hyperglycemia to stimulate pancreatic β-cell insulin secretion. Promoting free fatty acid (FFA) esterification into triglycerides, thereby triggering lipid accumulation and subsequent LEP elevation [28]. This cascade re-established hypothalamic–pituitary–gonadal (HPG) axis activation (GnRH pulsatility), overcoming “metabolic–reproductive decoupling” in seasonal breeding. Our findings propose a novel nutritional strategy for off-season estrus induction in ruminants via metabolic reprogramming of energy allocation.

4.3. Molecular Mechanism of Synergistic Regulation of the Lipid Metabolism-Reproduction Axis in Ewes by Glycerol Complex

Previous studies demonstrated that glycerol absorbed via the rumen preferentially enters hepatic portal circulation, activating the AMPK/mTOR pathway to upregulate PEPCK (↑ 2.1-fold) and drive gluconeogenesis [29]. At the ovarian level, follicles possess an insulin-glucose-IGF-1 system modulated by short-term nutritional interventions, where glucose (Glu) serves as the primary metabolic fuel for fertility [30,31]. Berlinguer et al. in vitro follicular culture confirmed that high-glucose microenvironments (6.5 mmol/L) enhance granulosa cell mitochondrial membrane potential (Δψm ↑ 40%) and glucose uptake (↑ 55%) via GLUT4 translocation, improving oocyte maturation [32]. In this study, serum INS peaked at 22.7 ± 1.5 ng/mL on day 30 post-supplementation, potentially linked to ovarian IGF-1/PI3K-AKT pathway activation. Supplemental glycerol reshaped lipid metabolism balance, with Glu levels showing biphasic trends (peak: 4.2 ± 0.5 mmol/L at day 30), correlating with adipose tissue deposition-mobilization phases. These findings align with Sutton-McDowall’s report on glycogen-mediated oocyte quality enhancement; Rodent overfeeding models where sustained hyperglycemia stimulates LEP secretion via INS-LPL-FFA esterification, accelerating fat synthesis. Critically, glycerol supplementation induced phase-specific LEP fluctuations (decrease → increase), reducing leptin resistance and activating hypothalamic Kisspeptin/GnRH neurons to promote LH/FSH secretion [33]. Collectively, cyclic changes in circulating Glu, INS, and LEP establish a systemic metabolic environment conducive to hypothalamic–pituitary–ovarian axis reactivation during non-breeding seasons.

4.4. Mechanisms of Glycerol Complex Regulation of the Reproduction-Lipid Metabolism Axis in Ewes

Kisspeptin encoded by the kiss-1 gene drives pulsatile GnRH secretion through activation of the membrane receptor GPR54 in hypothalamic neurons, thereby promoting pituitary LH/FSH release (↑ 40% LH peak amplitude) [34,35,36]. This study demonstrated that glycerol supplementation significantly downregulated GnAQ expression (p < 0.01), alleviating its transcriptional repression on kiss-1 to activate the hypothalamic–pituitary–gonadal (HPG) axis (GnRH ↑, p < 0.05). These findings align with Zhou et al.’s report on high-energy diets promoting LHR expression [37]. In our study, LHR was significantly upregulated (p < 0.01), confirming the universality of energy-reproductive axis regulation. Synergistic actions of StAR (mediating mitochondrial cholesterol transport, p < 0.05) and CYP11A1 (catalyzing cholesterol side-chain cleavage, p < 0.01) enhanced pregnenolone synthesis efficiency, with serum P4 peaking on day 13 in synchrony with follicular luteinization. Concurrently, VEGF upregulation (p < 0.01) increased ovarian vascular density to support folliculogenesis [38]. The intervention promoted dual-pathway lipid mobilization: subcutaneous adipose HSL upregulation (p < 0.05) hydrolyzed triglycerides, while hepatic FFA oxidation provided cholesterol precursors for ovarian steroidogenesis (StAR ↑, CYP19A1 ↑). These metabolic reprogramming effects align with the “metabolism-reproductive coupling axis” theory in rodents [39], confirming glycerol’s tripartite regulatory network in non-seasonal estrus induction: Hepatic glucose homeostasis; Lipolysis-steroid synthesis coupling via cholesterol precursor flux; and LEP-Kisspeptin-GnRH neuroendocrine rhythm reset.
Since the original hypothalamic–ovarian tissues have been fully utilized for RNA extraction and histological analyses, the regulatory mechanisms of LHR, StAR, and CYP11A1 are currently based on mRNA-level data. Future studies should validate these mechanisms at the protein level using western blot or immunohistochemistry. In subsequent investigations, we will establish a primary cell model of the ovine hypothalamus/ovary to systematically examine the transcription-translation regulatory relationships of key genes.

4.5. Serum Metabolomic Profiling and Key Metabolite Resolution

Non-targeted metabolomics analysis based on LC-MS technology showed significant metabolic differences between the test groups (GLY, Tyr-GLY, B9-Tyr-GLY) and the control group (VIP > 1.5, FC > 1.2, p < 0.05). The differential metabolites were dominated by lipids and lipid-like molecules (68.3%), followed by organic acids and their derivatives (19.5%) and organic oxides (12.2%), which aligns with energy metabolism trends in ruminants.
Previous studies indicate that DL-carnitine mediates long-chain fatty acid transport to mitochondria via carnitine palmitoyltransferase I, promoting β-oxidation and enhancing energy metabolism efficiency [40,41,42,43]. Notably, Samir’s study demonstrated that DL-carnitine intervention elevated total antioxidant capacity, E2 and P4 in ewes [44], consistent with the significant DL-carnitine elevation (p < 0.001) and improved reproductive performance observed here. Glycerol phospholipid-like molecules were significantly enriched (p < 0.01) in the GLY group (vs. NC), suggesting glycerol regulates fat mobilization through phospholipid metabolism remodeling. In the Tyr-GLY and B9-Tyr-GLY groups, N-stearoyl taurine was significantly upregulated (p < 0.01) by Asp-Phe. Crucially, Asp-Phe (a prostaglandin analog) synchronizes estrous cycles via luteolysis, mirroring bovine estrus synchronization mechanisms reported by Yizengaw L [13]. This provides a theoretical basis for precision nutritional interventions in seasonal reproductive disorders.

5. Conclusions

This study confirmed that glycerol complexes restore the estrous cycle in ewes during the non-breeding season through a tripartite synergistic mechanism: (1) Metabolic–Energy Axis Regulation: The glycerol mixture activates Kisspeptin-GnRH neuronal activity in the hypothalamic arcuate nucleus, driving pulsatile LH/FSH secretion. Simultaneously, it enhances hepatic DL-carnitine synthesis efficiency via carnitine palmitoyltransferase I-mediated β-oxidation of long-chain fatty acids, providing energy support for follicular development. (2) Ovarian Function Optimization: DL-carnitine reduces mitochondrial oxidative stress, promotes granulosa cell proliferation, and stimulates steroid hormone synthesis (E2 ↑, P4 ↑). This approach provides a novel nutritional intervention strategy for seasonal reproductive regulation in ruminants.

Author Contributions

Conceptualization, Y.N. and Z.Z.; Methodology, Y.N., B.J. and X.Q.; Software, Y.N. and X.Q.; Validation, Y.N., B.J. and X.Q.; Investigation, Y.N., B.J., C.Y., M.X. and X.Q.; Resources, Z.Z.; Data curation, Y.N.; Writing—original preparation, Y.N., B.J. and X.Q.; Funding acquisition, Z.Z.; Supervision, Z.Z. and C.Y.; Editing, Z.Z. and C.Y. All authors have read and agreed to the published version of the manuscript.

Funding

This study was supported by the National Natural Science Foundation of China (No. 32160770) and the Production and Construction Corps 2023 Graduate Student Innovation Program.

Institutional Review Board Statement

All animal manipulations were performed by the Guide for the Care and Use of Laboratory Animals of Shihezi University, China, and approved by the Animal Ethics Committee of the Institute of Animal Science, Shihezi University, Agricultural Sciences (Ethics License IAS2021-227).

Informed Consent Statement

Informed consent was obtained from all participants involved in the study.

Data Availability Statement

The data presented in this study are available upon request from the corresponding author. The data are not publicly available to preserve privacy.

Acknowledgments

The authors would like to thank the College of Animal Science and Technology, Shihezi University, Xinjiang, China, for providing important equipment and the staff of the experimental station at the College of Animal Science and Technology, Shihezi University, Xinjiang, China, for their help.

Conflicts of Interest

The authors declare no conflicts of interest.

References

  1. Kiyma, Z.; Alexander, B.M.; Van, K.E.A.; Murdoch, W.J.; Hallford, D.M.; Moss, G.E. Effects of feed restriction on reproductive and metabolic hormones in ewes. J. Anim. Sci. 2004, 82, 2548–2557. [Google Scholar] [CrossRef]
  2. Ma, L.W.; Hou, S.Z.; Wang, Z.Y.; Zhang, J.X. Effects of Supplementing Concentrate Feeds with Different Energy Levels on Estrus and Conception Rates of Grazing Tibetan Ewes. Jiangsu Agric. Sci. 2015, 43, 123–128. [Google Scholar] [CrossRef]
  3. Andrade, M.A.M.d.M.; Alves, J.P.M.; Galvão, I.T.O.M.; Cavalcanti, C.M.; Silva, M.R.L.; Conde, A.J.H.; Rondina, D. Glycerin supplementation strategies for three or seven days affects oxidative stress, follicle dynamics and ovulatory response in Morada Nova sheep. Anim. Reprod. 2022, 19, e20200025. [Google Scholar] [CrossRef] [PubMed]
  4. Rodrigues, F.V.; Silva, C.M.G.; Lima, I.M.T.; Silva, A.M.; Rondina, D. Effect of oral drenching of glycerin as a source of pre-mating energetic supplementation on reproductive response in goats. Anim. Reprod. 2015, 12, 890–898. [Google Scholar]
  5. Tyl, R.W.; Bechtel, D.H. One-generation reproduction study of esterified propoxylated glycerol (EPG) administered in the feed to CD®(Sprague-Dawley) rats. Regul. Toxicol. Pharmacol. 2014, 70, S114–S122. [Google Scholar] [CrossRef] [PubMed]
  6. Zhu, M.T.; Li, H.; Peng, F.; Yang, H.; Zhao, Z.S. The influence of glycerol on seasonal estrus and serum reproductive hormone levels in sheep. Heilongjiang Anim. Husb. Vet. Med. 2017. [Google Scholar] [CrossRef]
  7. Letelier, C.; Mallo, F.; Encinas, T.; Ros, J.; Gonzalez-Bulnes, A. Glucogenic supply increases ovulation rate by modifying follicle recruitment and subsequent development of preovulatory follicles without effects on ghrelin secretion. Reproduction 2008, 136, 65–72. [Google Scholar] [CrossRef]
  8. Meza-Herrera, C.A.; Ross, T.; Hallford, D.; Hawkins, D.; Gonzalez-Bulnes, A. Effects of body condition and protein supplementation on LH secretion and luteal function in sheep. Reprod. Domest. Anim. 2010, 42, 461–465. [Google Scholar] [CrossRef]
  9. Liefers, S.C.; Veerkamp, R.F.; Pas, M.F.W.T.; Delavaud, C.; Chilliard, Y.; Lende, T.V.D. Leptin concentrations in relation to energy balance, milk yield, intake, live weight, and estrus in dairy cows. J. Dairy Sci. 2003, 86, 799–807. [Google Scholar] [CrossRef]
  10. Moschos, S.; Chan, J.L.; Mantzoros, C.S. Leptin and reproduction: A review. Fertil. Steril. 2002, 77, 433–444. [Google Scholar] [CrossRef]
  11. Yang, H.; Liu, X.; Hu, G.; Xie, Y.; Lin, S.; Zhao, Z.; Chen, J. Identification and analysis of microRNAs-mRNAs pairs associated with nutritional status in seasonal sheep. Biochem. Biophys. Res. Commun. 2018, 499, 321–327. [Google Scholar] [CrossRef]
  12. Lin, S. Screening of Micrornas and Functional Verification of Target Genes Related to Estrus in Non-Breeding Seasons Induced by Nutrition in Sheep. Master's Thesis, Shihezi University, Shihezi, China, 2015. [Google Scholar]
  13. Yizengaw, L. Review on estrus synchronization and its application in cattle. Int. J. Adv. Res. Biol. Sci. 2017, 4, 67–76. [Google Scholar] [CrossRef]
  14. Rettmer, I.; Goodband, R.; Stevenson, J.; Davis, D.; Tokach, M.; Rozeboom, D.; Pettigrew, J.E.; Johnston, L.J.; Rust, J.W.; Chester-Jones, H. Failure of dietary amino acid supplementation at weaning to influence reproductive traits of sows. J. Anim. Sci. 1993, 71, 2511–2515. [Google Scholar] [CrossRef]
  15. Jia, Y.; Liu, Y.; Wang, P.; Liu, Z.; Zhang, R.; Chu, M.; Zhao, A. NTRK2 Promotes Sheep Granulosa Cells Proliferation and Reproductive Hormone Secretion and Activates the PI3K/AKT Pathway. Animals 2024, 14, 1465. [Google Scholar] [CrossRef]
  16. Abu, A.A.; El-Gohary, E.S.; Abdel-Samee, A.M. Productive and reproductive performance of goats as affected by l-tyrosine supplement. 1-Sexual activity and reproductive performance. Egypt. J. Sheep Goats Sci. 2011, 6, 1–10. [Google Scholar]
  17. Hong, J. Effects of Dietary Arginine, Lysine, and Energy Levels on Physiological Responses and Reproductive Performance in Sows and Growth of Their Progeny. Ph.D. Dissertation, Seoul National University Graduate School, Seoul, Republic of Korea, 2018. [Google Scholar]
  18. Council, N.R. Nutrient Requirements of Small Ruminants; The National Academies Press: Washington, DC, USA, 2007. [Google Scholar] [CrossRef]
  19. Ebling, F.J.P. Photoperiodic regulation of puberty in seasonal species. Mol. Cell Endocrinol. 2010, 324, 95–101. [Google Scholar] [CrossRef] [PubMed]
  20. Zhang, J.N.; Zhang, F.C.; Zhang, J.; Wang, A.J.; Liao, L.W.; Zhang, F.L. Study on the preferred program for inducing estrus in ewes during the non-breeding season. Heilongjiang Anim. Husb. Vet. Med. 1999, 12–13. [Google Scholar] [CrossRef]
  21. Guerra, D.D.; Bok, R.; Cari, E.L.; Nicholas, C.; Orlicky, D.J.; Johnson, J.; Hurt, K.J. Effect of neuronal nitric oxide synthase serine-1412 phosphorylation on hypothalamic–pituitary–ovarian function and leptin response. Biol. Reprod. 2020, 102, 1281–1289. [Google Scholar] [CrossRef] [PubMed]
  22. Bruun, T.; Lyderik, K.; Dall, J.; Strathe, A. Effect of feeding sows a combination of extra vitamin B2, vitamin B6, vitamin B12, and folic acid during the first 45 days of gestation on piglet birthweight. Livest. Sci. 2024, 282, 105434. [Google Scholar] [CrossRef]
  23. Qi, Y.X.; He, L.; Liu, X.F.; Wu, J.H.; Rong, W.H.; Liu, Y.B. Analysis of the Variation patterns of serum FSH and LH concentrations in Bamei Sheep during estrus and their relationship with the number of lambs born. Heilongjiang Anim. Sci. Vet. Med. 2014. [Google Scholar] [CrossRef]
  24. Tian, Z.W.; Zhao, Z.S.; Lin, S.; Zhai, M.J.; Yu, Y.S.; Lei, X.P. The changing patterns of reproductive hormones in Xinjiang Kazakh sheep during the estrus and hypoestrus periods. Jiangsu Agric. Sci. 2016, 44, 256. [Google Scholar]
  25. Mondal, M.; Rajkhowa, C.; Prakash, B.S. Relationship of plasma estradiol-17β, total estrogen, and progesterone to estrus behavior in mithun (Bos frontalis) cows. Horm. Behav. 2006, 49, 626–633. [Google Scholar] [CrossRef] [PubMed]
  26. Sogorescu, E.; Zamfirescu, S.; Anghel, A.H.; Nadolu, D.; Rosoiu, N. Seasonal Variations of Progesterone Level and Characteristics of Breeding Season and Anoestrus Period on Carpathian Goats. J. Anim. Vet. Adv. 2012, 11, 1472–1477. [Google Scholar] [CrossRef]
  27. Lehman, M.N.; Merkley, C.M.; Coolen, L.M.; Goodman, R.L. Anatomy of the kisspeptin neural network in mammals. Brain Res. 2010, 1364, 90–102. [Google Scholar] [CrossRef]
  28. Jiang, B.H.; Nan, Y.; Xie, M.T.; Qi, X.D.; Ye, C.F.; Nuerli, A.; Zhao, Z. Effects of glycerol supplementation at different doses on apparent digestibility of nutrients, ruminal environment, and volatile fatty acid absorption in empty ewes. J. Anim. Nutr. 2024, 36, 2541–2550. [Google Scholar] [CrossRef]
  29. Nielsen, N.I.; Ingvartsen, K.L. Propylene glycol for dairy cows: A review of the metabolism of propylene glycol and its effects on physiological parameters, feed intake, milk production and risk of ketosis. Anim. Feed Sci. Technol. 2004, 115, 191–213. [Google Scholar] [CrossRef]
  30. Donkin, S.; Koser, S.; White, H.; Doane, P.; Cecava, M. Feeding value of glycerol as a replacement for corn grain in rations fed to lactating dairy cows. J. Dairy Sci. 2009, 92, 5111–5119. [Google Scholar] [CrossRef]
  31. Scaramuzzi, R.; Brown, H.M.; Dupont, J. Nutritional and metabolic mechanisms in the ovary and their role in mediating the effects of diet on folliculogenesis: A perspective. Reprod. Domest. Anim. 2010, 45, 32–41. [Google Scholar] [CrossRef]
  32. Berlinguer, F.; Gonzalez-Bulnes, A.; Contreras-Solis, I.; Spezzigu, A.; Torres-Rovira, L.; Succu, S.; Naitana, S.; Leoni, G.G. Glucogenic supply increases oocyte developmental competence in sheep. Reprod. Fertil. Dev. 2012, 24, 1055–1062. [Google Scholar] [CrossRef]
  33. Beretta, E. Long-term suppression of weight gain, adiposity, and serum insulin by central leptin gene therapy in prepubertal rats: Effects on serum ghrelin and appetite-regulating genes. Pediatr. Res. 2002, 52, 189–198. [Google Scholar] [CrossRef]
  34. Bern, H.A. From fish tail to human brain: Preface. Peptides 2008, 29, 649–650. [Google Scholar] [CrossRef] [PubMed]
  35. Golshan, M.; Alavi, S.M.H.; Hatef, A.; Kazori, N.; Socha, M.; Milla, S.; Sokołowska-Mikołajczyk, M.; Unniappan, S.; Butts, I.A.E. Impact of absolute food deprivation on the reproductive system in male goldfish exposed to sex steroids. J. Comp. Physiol. B Biochem. Syst. Environ. Physiol. 2024, 194, 411–426. [Google Scholar] [CrossRef] [PubMed]
  36. Smith, J.T. Sex steroid control of hypothalamic Kiss1 expression in sheep and rodents: Comparative aspects. Peptides 2009, 30, 94–102. [Google Scholar] [CrossRef] [PubMed]
  37. Zhou, X.; Yu, M.; Liu, L.; Yi, K.; Li, C.; Chen, L.; Sun, Y. Effects of dietary energy level on ovarian expression of mRNA s for luteinizing hormone receptor and follicle-stimulating hormone receptor in prepubertal gilts. Chin. J. Vet. Sci. 2009, 97–105. [Google Scholar]
  38. Horan, C.J.; Williams, S.A. Oocyte stem cells: Fact or fantasy? Reproduction 2017, 154, R23–R35. [Google Scholar] [CrossRef]
  39. Gao, L.S.; Wu, J.P.; Song, S.Z.; Li, H.; Lang, X.; Wei, Y.B.; Gong, X.; Liu, L. The influence of different energy levels on serum lipid indices and fat deposition in Altay sheep. Feed Ind. 2020, 41, 14–23. [Google Scholar] [CrossRef]
  40. Fathi, E.; Farahzadi, R. Application of L-carnitine as nutritional supplement in veterinary medicine. Rom. J. Biochem. 2014, 1, 31–41. [Google Scholar]
  41. Gerber, L.K.; Aronow, B.J.; Matlib, M.A. Activation of a novel long-chain free fatty acid generation and export system in mitochondria of diabetic rat hearts. Am. J. Physiol. Cell Physiol. 2006, 291, C1198–C1207. [Google Scholar] [CrossRef]
  42. Crayhon, R. The Carnitine Miracle: The Supernutrient Program That Promotes High Energy, Fat Burning, Heart Health, Brain Wellness and Longevity; Rowman Littlefield: Washington, DC, USA, 2001. [Google Scholar]
  43. Koohpeyma, F.; Siri, M.; Allahyari, S.; Mahmoodi, M.; Saki, F.; Dastghaib, S. The effects of L-carnitine on renal function and gene expression of caspase-9 and Bcl-2 in monosodium glutamate-induced rats. BMC Nephrol. 2021, 22, 162. [Google Scholar] [CrossRef]
  44. Samir, H.; Swelum, A.A.; Abdelnaby, E.A.; El-Sherbiny, H.R. Incorporation of L-Carnitine in the OvSynch protocol enhances the morphometrical and hemodynamic parameters of the ovarian structures and uterus in ewes under summer climatic conditions. BMC Vet. Res. 2023, 19, 246. [Google Scholar] [CrossRef]
Figure 1. Vulva of a spent ewe (A), vulva of an estrous ewe (B).
Figure 1. Vulva of a spent ewe (A), vulva of an estrous ewe (B).
Animals 15 02291 g001
Figure 2. Ovary of an estrous ewe (A) and HE-stained section (B).
Figure 2. Ovary of an estrous ewe (A) and HE-stained section (B).
Animals 15 02291 g002
Figure 3. Hormone levels in the estrous cycle of ewes. (A) P4 hormone level, (B) E2 hormone level, (C) LH hormone level, and (D) FSH hormone level. Note: “p < 0.05” indicates a significant difference, which is labeled with “*” in the figure, and “p < 0.01” indicates “highly significant difference, which is labeled with “**” in the figure.
Figure 3. Hormone levels in the estrous cycle of ewes. (A) P4 hormone level, (B) E2 hormone level, (C) LH hormone level, and (D) FSH hormone level. Note: “p < 0.05” indicates a significant difference, which is labeled with “*” in the figure, and “p < 0.01” indicates “highly significant difference, which is labeled with “**” in the figure.
Animals 15 02291 g003
Figure 4. Hormonal changes in lipid metabolism. (A) Glu levels. (B) INS levels. (C) LEP levels. “p < 0.05” indicates a significant difference, which is labeled with “*” in the figure.
Figure 4. Hormonal changes in lipid metabolism. (A) Glu levels. (B) INS levels. (C) LEP levels. “p < 0.05” indicates a significant difference, which is labeled with “*” in the figure.
Animals 15 02291 g004
Figure 5. Gene expression quantity. (A) Oestrus gene expression in hypothalamic tissue; (B) Oestrus gene expression in ovarian tissue; (C) Oestrus gene expression in subcutaneous adipose tissue. “p < 0.05” indicates a significant difference, which is labeled with “*” in the figure, and “p < 0.01” indicates “highly significant difference, which is labeled with “**” in the figure.
Figure 5. Gene expression quantity. (A) Oestrus gene expression in hypothalamic tissue; (B) Oestrus gene expression in ovarian tissue; (C) Oestrus gene expression in subcutaneous adipose tissue. “p < 0.05” indicates a significant difference, which is labeled with “*” in the figure, and “p < 0.01” indicates “highly significant difference, which is labeled with “**” in the figure.
Animals 15 02291 g005
Figure 6. (A) Metabolite classification. (B) QC sample correlation analysis. (C) Total sample PCA analysis.
Figure 6. (A) Metabolite classification. (B) QC sample correlation analysis. (C) Total sample PCA analysis.
Animals 15 02291 g006
Figure 7. (A) PLS-DA Score Scatter Plot. (B) Ranked Validation Plot.
Figure 7. (A) PLS-DA Score Scatter Plot. (B) Ranked Validation Plot.
Animals 15 02291 g007
Figure 8. (A) Volcano diagram. (B) Matchstick diagram.
Figure 8. (A) Volcano diagram. (B) Matchstick diagram.
Animals 15 02291 g008aAnimals 15 02291 g008b
Figure 9. Clustering heat map of differential metabolites.
Figure 9. Clustering heat map of differential metabolites.
Animals 15 02291 g009
Figure 10. Correlation diagram of differential metabolites.
Figure 10. Correlation diagram of differential metabolites.
Animals 15 02291 g010
Figure 11. KEGG enrichment bubble plot.
Figure 11. KEGG enrichment bubble plot.
Animals 15 02291 g011
Table 1. Composition and nutritional components of the basic diet (of DM, %).
Table 1. Composition and nutritional components of the basic diet (of DM, %).
ItemContent, %Nutrient Content 2Content, %
Ingredients (MJ/kg)10.96
Alfalfa hay40DM83.375
Alfalfa silage35CP17.5
Corn Stover5Ash8.63
Soybean Meal7.9EE2.9
Corn8.5Ca0.49
NaHCO31.5P0.41
premix 11NDF44.94
NaCl0.8ADF35.59
Lys0.3
consider100
Note: 1 Each kilogram of premix contains 400,000 IU of vitamin A, 40,000 IU of vitamin D, 0.8 million IU of vitamin E, 3600 mg of iron, 4000 mg of zinc, 400 mg of copper, 2000 mg of manganese, 20 mg of Na, 20 mg of drill, and 40 mg of iodine. 2 Maintenance net energy values are calculated values, and the remaining nutrients are measured values. DM (Dry Matter); CP (Crude Protein); Ash (Crude Ash); EE (Ether Extract); Ca (Calcium); P (Phosphorus); NDF (Neutral Detergent Fiber); ADF (Acid Detergent Fiber).
Table 2. qRT-PCR primers (with 90–110% amplification efficiency).
Table 2. qRT-PCR primers (with 90–110% amplification efficiency).
GeneSequence (5′→3′)Temperature/°CProduct Length/bp
GnAQF: GGACAGGAGAGAGTGGCAAG
R: GGCCGTGAAGATGTTCTGAT
57112
kiss-1F: ATGAACGTGCTGCTTTCCT
R: TCCGAGCTGCGAGCCTGTG
58117
GPR54F: CCTTCACCGCTCTGCTCTAC
R: ACCGAGACCTGCTGGATGTA
5888
GnRHF: CTTAGGTTCTACTGGCTGAT
R: TCCTGCTGACTTTCTGTG
58112
CYP11A1F: CGAGGGATCCTACCCACAGA
R: GTTCTGGAGGGAGGTTGAGC
55422
LHRF: TGCGGCCTTTAATCGTTCCT
R: ATACTACTGGGCCTGGGTGT
58216
VEGFF: GCACCGTCTTTTTGTCCCTC
R: TCTTCCCAAAAGCAGGCCAA
58103
StARF: GGCAGAGATGAGCCACACTT
R: CCTGTGCCCCTTACCTTGAG
58584
β-actinF: AGAGCAAGAGAGGCATCC
R: TCGTTGTAGAAGGTGTGGT
50~60108
HSLF: AGCACTACAAACGAACGA
R: CTGAATGATCCGCTCAAACT
58207
GAPDHF: GTCGGAGTGAACGGATTTGG
R: CATTGATGACGAGCTTCCCG
50~60196
Table 3. Number of estrus ewes in each group and ovarian development of estrous ewes in the experimental group.
Table 3. Number of estrus ewes in each group and ovarian development of estrous ewes in the experimental group.
GroupEwe in HeatNon-Estrus EwesEstrus RateNumber of Follicles/mm Follicle Diameter/mmFollicle Thickness/mm
Control1910%---
GLY3730%3 ± 11.8 ± 0.320.95 ± 0.25
GLY-Tyr4640%3 ± 21.9 ± 0.180.85 ± 0.32
GLY-B93730%3 ± 12.2 ± 0.271.01 ± 0.43
GLY-Tyr-B95550%4 ± 22.4 ± 0.261.08 ± 0.53
Table 4. Distribution of differential metabolites in each comparison group (Partial).
Table 4. Distribution of differential metabolites in each comparison group (Partial).
GroupMetabolite NameFormulaRT [min]m/zFCp-ValueVIPTrend
GLY.vs.NCDL-CarnitineC7 H15 N O31.330162.1122.6550.0002.215
4-HydroxyisoleucineC6 H13 N O31.832148.0971.4560.0021.915
Asp-PheC13 H16 N2 O54.852281.1131.7510.0032.131
PC O-17:0C25 H52 N O7 P9.249510.3541.4390.0121.259
LPC O-20:5C28 H50 N O6 P9.301528.3441.7410.0121.393
5-Hydroxyindole-2-carboxylic acidC9 H7 N O32.834178.0500.6630.0161.654
Meperidine-d5C15 H16 H5 N O26.694253.1950.7000.0211.309
gamma-GlutamyltyrosineC14 H18 N2 O64.940311.1231.5940.0241.558
N-MethyllysineC7 H16 N2 O21.243161.1282.4540.0271.820
OrnithineC5 H12 N2 O21.179133.0971.5300.0421.532
D-Erythro-sphingosine 1-phosphateC18 H38 N O5 P8.438380.2550.8070.0441.333
morpholino(quinolin-6-yl)methanoneC14 H14 N2 O25.431243.1100.5500.0481.668
ErgosterolC28 H44 O7.929397.3462.2990.0481.662
Tyr-GLY.vs.NCHomoarginineC7 H16 N4 O21.310189.13413.4400.0042.145
ErgosterolC28 H44 O7.929397.3462.9750.0042.029
3-(4-hydroxy-3-methoxyphenyl)propanoic acidC10 H12 O45.587219.0630.6430.0042.192
Asp-PheC13 H16 N2 O54.852281.1131.5220.0051.428
N2-Acetyl-L-ornithineC7 H14 N2 O31.415175.1080.6270.0071.631
DL-CarnitineC7 H15 N O31.330162.1122.8020.0122.058
PC 17:0_17:0C42 H84 N O8 P11.127762.5950.4900.0271.680
Arachidonoyl amideC20 H33 N O8.730304.2632.5750.0271.658
L-HomocitrullineC7 H15 N3 O31.414190.1183.5350.0271.795
DL-ArginineC6 H14 N4 O21.280175.1190.7720.0291.408
Vitamin AC20 H30 O8.739304.2632.7960.0321.663
PC 36:2C44 H84 N O8 P9.883808.5810.4470.0342.037
N-MethyllysineC7 H16 N2 O21.243161.1282.4960.0351.552
CreatinineC4 H7 N3 O1.383114.0661.4520.0401.408
D-Erythro-sphingosine 1-phosphateC18 H38 N O5 P8.438380.2550.7880.0431.326
B9-GLYvs.NCN-MethyllysineC7 H16 N2 O21.243161.1282.5471.3490.010
L-HomocitrullineC7 H15 N3 O31.414190.1183.3671.7510.011
Methyl palmitateC17 H34 O26.740288.2892.2261.1550.021
N2-Acetyl-L-ornithineC7 H14 N2 O31.415175.1080.647−0.6270.022
1-MethylhistidineC7 H11 N3 O21.293170.0921.7250.7870.023
2-hydroxy-6-[(8Z,11Z)-pentadeca-8,11,14-trien-1-yl]benzoic acidC22 H30 O38.180343.2221.7380.7980.024
Glycerophospho-N-palmitoyl ethanolamineC21 H44 N O7 P8.941454.2920.730−0.4540.033
LPC O-16:1C24 H50 N O6 P9.533480.3440.714−0.4860.042
CystamineC4 H12 N2 S21.245153.0520.766−0.3840.043
16-Heptadecyne-1,2,4-triolC17 H32 O37.801307.2241.3580.4410.043
DeoxycytidineC9 H13 N3 O41.409250.0790.593−0.7540.043
CytosineC4 H5 N3 O1.880112.0510.577−0.7930.043
Lysopc 20:4C28 H50 N O7 P8.979544.3400.574−0.8000.045
B9-Tyr-GLYvs.NCDL-CarnitineC7 H15 N O31.330162.1122.3561.2360.000
N-FormylkynurenineC11 H12 N2 O44.888237.0872.7441.4560.000
PC O-38:5C46 H84 N O7 P11.299794.6040.501−0.9980.001
N-Stearoyl taurineC20 H41 N O4 S11.437392.2820.631−0.6640.002
Asp-PheC13 H16 N2 O54.852281.1131.5070.5910.003
CholineC5 H13 N O1.314104.1070.723−0.4680.005
gamma-GlutamyltyrosineC14 H18 N2 O64.940311.1231.9460.9610.006
LPC 19:1-SN1C27 H54 N O7 P9.797536.3701.7880.8380.007
N-AcetylvalineC7 H13 N O34.967182.0790.505−0.9850.009
Methyl palmitateC17 H34 O26.740288.2892.1261.0880.012
AcetylcholineC7 H15 N O21.395146.1171.4130.4980.014
Gly-PheC11 H14 N2 O35.234223.1071.9560.9680.014
N2-Acetyl-L-ornithineC7 H14 N2 O31.415175.1080.615−0.7020.018
All trans-RetinalC20 H28 O6.453285.2211.6740.7430.038
PhenylacetylglycineC10 H11 N O35.460194.0811.6330.7080.042
OrnithineC5 H12 N2 O21.179133.0971.5520.6340.046
Table 5. Enrichment results of KEGG Pathway for differential metabolites.
Table 5. Enrichment results of KEGG Pathway for differential metabolites.
GroupMapIDMapTitlep-ValuexynN
GLY.vs.NCmap04022cGMP-PKG signaling pathway0.0711692
map04740Olfactory transduction0.0711692
map04744Phototransduction0.0711692
map00100Steroid biosynthesis0.1312692
map01523Antifolate resistance0.1312692
map04742Taste transduction0.1312692
map04917Prolactin signaling pathway0.1312692
map00760Nicotinate and nicotinamide metabolism0.1913692
map01100Metabolic pathways0.33669692
map04913Ovarian steroidogenesis0.3416692
map04977Vitamin digestion and absorption0.3416692
map00230Purine metabolism0.4318692
map00140Steroid hormone biosynthesis0.58112692
Tyr-GLY.vs.NCmap00830Retinol metabolism0.02221392
map04977Vitamin digestion and absorption0.20261392
map00100Steroid biosynthesis0.26121392
map00220Arginine biosynthesis0.37131392
map00760Nicotinate and nicotinamide metabolism0.37131392
map00330Arginine and proline metabolism0.46141392
map00590Arachidonic acid metabolism0.46141392
map04726Serotonergic synapse0.46141392
map05215Prostate cancer0.46141392
map01100Metabolic pathways0.5111691392
map00360Phenylalanine metabolism0.54151392
map012102-Oxocarboxylic acid metabolism0.54151392
map05200Pathways in cancer0.54151392
map00380Tryptophan metabolism0.632101392
map00260Glycine, serine and threonine metabolism1.00161392
map04913Ovarian steroidogenesis1.00161392
B9-GLY.vs.NCmap00240Pyrimidine metabolism0.0045892
map01523Antifolate resistance0.1712892
map00220Arginine biosynthesis0.2413892
map012102-Oxocarboxylic acid metabolism0.3715892
map00260Glycine, serine, and threonine metabolism0.4316892
map00340Histidine metabolism0.4817892
map01230Biosynthesis of amino acids0.5819892
map02010ABC transporters0.5819892
map00380Tryptophan metabolism1.00110892
Tyr-B9-GLY.vs.NCmap01523Antifolate resistance0.01221192
map04022cGMP-PKG signaling pathway0.12111192
map04740Olfactory transduction0.12111192
map04744Phototransduction0.12111192
map05230Central carbon metabolism in cancer0.12111192
map00410beta-Alanine metabolism0.23121192
map04742Taste transduction0.23121192
map04917Prolactin signaling pathway0.23121192
map00230Purine metabolism0.24281192
map04974Protein digestion and absorption0.24281192
map01100Metabolic pathways0.2810691192
map01230Biosynthesis of amino acids0.29291192
map00220Arginine biosynthesis0.32131192
map00380Tryptophan metabolism0.342101192
map00240Pyrimidine metabolism0.48151192
map00360Phenylalanine metabolism0.48151192
map00400Phenylalanine, tyrosine and tryptophan biosynthesis0.48151192
map012102-Oxocarboxylic acid metabolism0.48151192
map04913Ovarian steroidogenesis0.54161192
map00970Aminoacyl-tRNA biosynthesis1.00181192
map04976Bile secretion1.00181192
map00340Histidine metabolism1.00171192
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Nan, Y.; Jiang, B.; Qi, X.; Ye, C.; Xie, M.; Zhao, Z. Metabolomic and Molecular Mechanisms of Glycerol Supplementation in Regulating the Reproductive Function of Kazakh Ewes in the Non-Breeding Season. Animals 2025, 15, 2291. https://doi.org/10.3390/ani15152291

AMA Style

Nan Y, Jiang B, Qi X, Ye C, Xie M, Zhao Z. Metabolomic and Molecular Mechanisms of Glycerol Supplementation in Regulating the Reproductive Function of Kazakh Ewes in the Non-Breeding Season. Animals. 2025; 15(15):2291. https://doi.org/10.3390/ani15152291

Chicago/Turabian Style

Nan, Ying, Baihui Jiang, Xingdong Qi, Cuifang Ye, Mengting Xie, and Zongsheng Zhao. 2025. "Metabolomic and Molecular Mechanisms of Glycerol Supplementation in Regulating the Reproductive Function of Kazakh Ewes in the Non-Breeding Season" Animals 15, no. 15: 2291. https://doi.org/10.3390/ani15152291

APA Style

Nan, Y., Jiang, B., Qi, X., Ye, C., Xie, M., & Zhao, Z. (2025). Metabolomic and Molecular Mechanisms of Glycerol Supplementation in Regulating the Reproductive Function of Kazakh Ewes in the Non-Breeding Season. Animals, 15(15), 2291. https://doi.org/10.3390/ani15152291

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

Article Metrics

Back to TopTop