Next Article in Journal
Spatiotemporal Distribution Shifts of Zelkova schneideriana Under Climate Change: A Biomod2-Driven Modeling Framework
Previous Article in Journal
Off-Season Reproductive Performance of Tsurcana Ewes Under Five Estrous Induction Protocols with Different Hormonal Profiles
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Choline-Mediated Regulation of Follicular Growth: Interplay Between Steroid Synthesis, Epigenetics, and Oocyte Development

College of Animal Science and Technology, Yangzhou University, Yangzhou 225009, China
*
Author to whom correspondence should be addressed.
Biology 2025, 14(9), 1220; https://doi.org/10.3390/biology14091220
Submission received: 25 July 2025 / Revised: 3 September 2025 / Accepted: 5 September 2025 / Published: 8 September 2025

Simple Summary

This review delves into the impact of choline, a vital feed additive, on follicular development and reproductive performance. By blending animal nutrition, epigenetics, and reproductive endocrinology, this study uncovers how choline regulates follicular hierarchy and maturation. Adequate dietary choline is shown to enhance follicle maturation and boost reproductive success. Conversely, both insufficient and excessive choline intake can negatively affect follicular development, leading to issues like increased follicle atresia. Additionally, the research highlights the importance of establishing optimal choline supply systems. It suggests that future studies should focus on determining precise choline requirements and understanding the related metabolic and epigenetic mechanisms. Overall, this work provides valuable insights into improving animal reproduction through better choline nutrition.

Abstract

This review article focuses on the role of choline in ovarian follicular development, regulated by nutrient–epigenetic interactions. Choline, a key feed additive, participates in DNA methylation and steroid hormone synthesis via its methyl donor function. However, its role in follicular hierarchy and maturation is unclear. Research lacks an understanding of species-specific choline metabolism, follicular fluid methylation dynamics, and toxicity thresholds. This study combines animal nutrition, epigenetics, and reproductive endocrinology. Using in vitro follicle culture models, metabolomics analysis, and cytochrome P450 family 19 subfamily A member 1 (CYP19a1) methylation site screening, it reveals that choline regulates follicle hierarchy through the betaine-S-adenosylmethionine (SAM) pathway. Proper dietary choline reduces homocysteine (HCY) and boosts CYP19a1 demethylation, enhancing theca cell estradiol (E2) production and accelerating follicle maturation. In contrast, inadequate or excessive choline causes mesoderm-specific transcript (MEST) gene methylation abnormalities or trimethylamine N-oxide (TMAO)-mediated β-oxidation inhibition, increasing follicle atresia. A phenomenon of steroidogenic factor 1 (SF-1) methylation has been observed in poultry, showing that choline affects offspring egg-laying persistence by altering the adrenal–ovarian axis DNA methylation imprint. Future research should establish a precise choline supply system based on the HCY/TMAO ratio in follicular fluid and the CYP19a1 methylation map to improve animal reproduction.

1. Introduction

Reproductive dysfunction poses a common challenge in both clinical medicine and animal husbandry, with follicular health and developmental quality being key determinants [1,2]. Follicular development not only affects oocyte formation but also directly influences fertilization rates and embryonic survival, thereby determining overall reproductive efficiency [3]. As a crucial component of cell membranes and a key player in one-carbon metabolism, choline is essential for normal follicular development. Studies indicate choline deficiency may impair membrane synthesis and elevate homocysteine (HCY) levels, consequently disrupting follicular growth and function [4,5]. In addition, the role of long non-coding RNAs (lncRNAs) in reproductive health has gained attention in recent studies, with LncRNA Gm2044 identified as a key regulator in germ cell development. Choline’s involvement in regulating such lncRNAs could represent a critical pathway influencing folliculogenesis [6]. Moreover, choline participates in DNA methylation and gene expression regulation—indispensable biological processes during folliculogenesis [7,8]. Recent advancements have highlighted the importance of epigenetic modifications in regulating reproductive health, and choline’s role in these processes, particularly through its involvement in super-enhancers and transcription factor regulation, is gaining attention [9].
Although existing research has explored choline’s general effects on animal growth and health, its specific roles in follicular development remain insufficiently investigated. As an essential nutrient, choline’s involvement in the regulation of epigenetic mechanisms and homocysteine metabolism has been observed in several animal models, suggesting its broader influence on folliculogenesis [10]. Additionally, environmental stressors, including heat stress, have been shown to negatively affect choline metabolism and, consequently, follicular development. Understanding the intricate relationship between choline and reproductive health, especially under environmental stress, remains an area that warrants further research [10,11].
This review systematically examines choline’s functions in folliculogenesis, analyzing how it influences follicular development through one-carbon metabolism, homocysteine regulation, and epigenetic modulation pathways.

2. Literature Review Methodology

This manuscript is a mini-review aimed at summarizing the effects of choline on follicular development. The literature search and selection followed strict criteria to ensure comprehensive coverage of relevant studies.

2.1. Literature Search Timeframe and Databases

The literature search covered studies from 1971 to 2025, using several databases for retrieval, including PubMed, Web of Science, and Google Scholar. These databases are widely used in the life sciences and medical fields, ensuring the breadth and reliability of the literature.

2.2. Search Keywords

The following keywords were used in the search: “choline”, “follicular development”, “DNA methylation”, “epigenetics”, “reproductive performance”, “follicle maturation”, etc. These keywords were combined to select the most recent studies relevant to choline and follicular development.

2.3. Literature Selection and Evaluation Criteria

The selection criteria included experimental studies that explored the direct or indirect effects of choline on follicular development, research involving various species (e.g., poultry, mammals), and studies employing in vitro or animal models. Review articles and irrelevant studies were excluded to maintain a focus on primary experimental data and observations.

2.4. Literature Assessment

All included articles were independently reviewed by two researchers, who assessed the quality and relevance of each article. Any doubts were discussed further to ensure the integrity of the selected literature.

3. Choline Structure, Absorption, and Metabolism

Choline (trimethyl-β-hydroxyethylammonium) is a positively charged quaternary ammonium compound that serves as a critical one-carbon metabolite in cellular methylation, acetylcholine synthesis, and phosphatidylcholine (PC) biosynthesis [12,13], consisting of a nitrogen atom bonded to four alkyl or other groups with excellent water solubility.
Dietary choline primarily exists as PC > 90%, with <10% present as free choline or sphingomyelin [14]. Choline absorption mainly occurs in the jejunum and ileum, facilitated by specialized transporters [15]. Dietary conjugated choline forms free choline through deacetylation catalyzed by pancreatic lipase and mucosal lipase with assistance from intestinal epithelial transporters [16]. Quantitatively, pancreatic lipase dominates choline digestion [17]. However, approximately 50% of dietary PC escapes intestinal enzymatic digestion, entering lymphatic vessels intact. The digestive process converts most ingested PC into lysophosphatidylcholine (LPC) for absorption. Within intestinal wall cells, LPC can either deacylate into glycerophosphocholine or re-acylate into PC [18]. Choline absorption occurs in the jejunum and ileum via energy- and sodium-dependent transporters in the intestinal epithelium [19]. In guinea pigs, ileal cells absorb choline approximately three times faster than jejunal cells [20], as PC primarily hydrolyzes into choline in proximal and middle small intestine segments [21]. Choline absorption does not compete with other nutrients; only one-third of dietary choline is absorbed intact, while the remaining two-thirds undergo microbial metabolism into betaine and trimethylamine (TMA) [22,23]. Additionally, choline can cross membranes via passive diffusion. Absorption efficiency depends on individual nutritional status, dietary choline intake, and the presence of other intestinal substances like fats [24]. After absorption, choline transports into lymphatic circulation as PC–chylomicron complexes and then distributes to tissues primarily as phospholipids bound to plasma lipoproteins [25,26].
Choline plays a critical role in membrane synthesis and methylation pathways. It is primarily involved in the production of phosphatidylcholine (PC) and phosphatidylethanolamine (PE), crucial for cellular function [27,28,29,30,31,32]. Being the primary source of membrane phospholipids, newly absorbed choline rapidly converts into PC via the cytidine diphosphate (CDP)–choline pathway catalyzed by choline kinase α [33]. All cells express at least one of three protein families for choline transport, except erythrocytes and the blood–brain barrier, which use facilitated diffusion [34,35], high-affinity choline transporter 1, medium-affinity choline transporter-like proteins (CTL), and low-affinity polyspecific organic cation transporters [36,37]. Once formed, betaine cannot revert to choline. After donating one methyl group to HCY, betaine generates methionine (MET) and dimethylglycine (DMG) [38,39]. DMG loses one methyl group, forming sarcosine, and then another, forming glycine. A minor choline fraction acetylates into acetylcholine for neural signaling and muscle control [40], catalyzed by choline acetyltransferase, the activity of which depends on choline availability and acetyl-CoA levels [41,42,43]. Choline is metabolized by intestinal microbiota into trimethylamine (TMA), which is further oxidized into trimethylamine N-oxide (TMAO), a compound associated with cardiovascular risk [44,45]. These studies demonstrate choline’s complex metabolic roles.

4. Impact of Choline on Follicular Development

4.1. Deficiency

In non-pregnant women with a daily choline intake of 480 mg, the CDP–choline pathway is prioritized for PC biosynthesis, while the betaine synthesis pathway is systemically suppressed [31]. Adequate PC is vital for cell membrane integrity and cell survival. Choline transport into cells is inhibited by caspase-3-dependent and -independent mechanisms, which lead to an overall suppression of choline incorporation into the CDP–choline pathway [46]. During follicular development, PC accumulation is critical for follicular development, and its depletion can significantly impact follicular development and follicular cavity formation [47]. Both the CDP–choline pathway, which uses choline as a substrate, and the PEMT-PC pathway, which uses SAM as a methyl donor, have high metabolic priority [48,49]. SAM is primarily used for PEMT-PC synthesis. Pregnant and lactating women with low choline intake show increased CDP-PC synthesis of different genotypes and a weakened PEMT-PC pathway, which may lead to methyl donor deficiency and affect the body’s methylation level. Under choline deficiency and the absence of MET, folic acid, vitamin B6, and vitamin B12, there are significant functional impairments in follicular growth and oocyte maturation [50,51,52]. This can reduce embryo developmental potential and cause stage-specific developmental arrest. These phenomena are attributed to impaired regulation of the methylation process of key maternal imprinting genes (e.g., mesoderm-specific transcript, MEST) during folliculogenesis [53,54]. Adding high-dose SAM to culture media causes hypermethylation of genes encoding key DNA methylation factors, including DNA methyltransferases 3 beta (DNMT3B), in cells [55]. Polycystic ovary syndrome (PCOS), a metabolic disorder, can alter serum levels of various metabolites, thereby reducing fertility in patients [56]. PCOS patients exhibit significantly reduced plasma and follicular fluid choline levels, lower choline and phosphocholine in follicular fluid, and abnormal cleavage dynamics [57,58]. This suggests that choline may play a role in oocyte competence development, potentially involving the metabolic process of 1-(5Z, 8Z, 11Z, 14Z, 17Z-eicosapentaenoic)-cn-glycerol-3-phosphocholine [59].

4.2. Toxicity

Excessive free choline intake can result in a fishy odor, vomiting, sweating, salivation, hypotension, and liver toxicity. It has been demonstrated that choline consumption can increase the production of TMAO [60,61]. In oocytes, inhibition of β-oxidation can cause insufficient adenosine triphosphate (ATP) synthesis, which in turn can induce cytoplasmic maturation defects and meiotic arrest [62,63]. Therefore, elevated TMAO levels and decreased acetoacetic acid levels can serve as potential indicators of oocyte quality. Studies have shown that in women with repeated implantation failure and PCOS, increased choline levels can promote successful pregnancy, and TMAO levels are negatively correlated with low fertilization rates and poor-quality embryos [64], which is consistent with the findings of a previous study [65]. Research has also indicated that TMAO is present in large amounts in follicular fluid. However, dietary precursors of TMAO, choline, and L-carnitine are not directly related to oocyte and embryo quality. Moreover, high-quality embryos are linked to lower TMAO and γ-butyrobetaine levels in follicular fluid. This suggests that TMAO is associated with poor reproductive outcomes [64]. In summary, these findings indicate that excessive choline metabolites, such as TMAO, can enter the follicular fluid and potentially affect oocyte development.

4.3. Optimal Choline Levels for Follicular Health

In Japanese quail (Coturnix japonica), the optimal dietary choline requirement appears to vary depending on the evaluated trait. Evidence indicates that a dietary choline level of approximately 0.126% (1260 mg/kg) can effectively improve egg weight and feed conversion efficiency [66]. In contrast, another experiment demonstrated that 1500 mg/kg of choline is sufficient to maintain laying performance and egg quality, whereas a higher level of 3500 mg/kg is necessary to markedly enhance yolk antioxidant capacity. These findings suggest that the optimal choline level may differ according to the specific performance or quality parameter considered [67]. In laying geese, dietary choline supplementation within the range of 784–913 mg/kg has been shown to enhance reproductive performance, egg production, and sex hormone levels, suggesting this range as optimal for promoting ovarian development and hormone synthesis [68]. For gestating sows, a dietary choline level of approximately 1910 mg/kg has been shown to optimize feed intake, improve birth weight and litter uniformity, and enhance antioxidant capacity and gut microbiota composition [69]. In pigs, dietary supplementation with 500–1000 mg/kg of choline has been shown to improve ovarian function, promote corpus luteum formation, and regulate the expression of reproduction-related genes, thereby enhancing reproductive performance [70]. In transition Holstein cows, supplementation with 60 g/day of choline (ReaShure®, Balchem Corporation, New Hampton, NY, USA) was shown to reduce inflammatory gene expression in follicular cells, suggesting that this level supports a healthier follicular environment during early postpartum [71]. In ewes, daily supplementation with 1.60 g/ewe of rumen-protected choline during the periconceptional and late gestation periods effectively reduced embryonic loss and increased lamb birth weight, suggesting this as the most suitable level [72]. In lactating women, an intake of 930 mg/day of choline has been shown to increase the concentration of PEMT-derived choline metabolites in breast milk, supporting enhanced choline delivery to the infant [73]. In pregnant women, prenatal supplementation with 550 mg/day of choline has been shown to enhance plasma concentrations of choline metabolites and promote fetal phospholipid synthesis via the PEMT pathway, making it a recommended intake level to support maternal–fetal health during gestation [74]. Collectively, these findings highlight that choline is not only essential for maintaining basic productivity but also plays pivotal roles in reproduction, growth, and transgenerational health. Therefore, the optimal level should be determined by species, physiological stage, and the targeted trait of interest. A summary of the optimal dietary choline levels across species and their associated reproductive or physiological effects is provided in Table 1.

5. Mechanism of Choline in Follicular Development

SAM plays a key role in DNA methylation and is central to reproductive and metabolic regulation [75,76]. Choline, through epigenetic modifications, directly affects key factors in follicular development, such as follicle-stimulating hormone receptor (FSHR) and cytochrome P450 family 19 subfamily A member 1 (CYP19a1), thereby coordinating oocyte maturation and granulosa cell function [77,78]. SAM also regulates steroidogenic factor 1 (SF-1) gene promoter methylation, influencing steroidogenesis in the adrenal–ovarian axis [79]. In PCOS, choline deficiency exacerbates the hypomethylation of genes like CYP19a1 [80,81], causing hormonal imbalances [82]. Choline supplementation can restore methylation patterns, thereby stabilizing metabolism and endocrine function. Choline and betaine metabolism, linked to folate metabolism, modulates HCY levels and DNMT activity [83,84,85]. This interaction affects reproductive function by regulating the methylation of genes such as FSHR, AR, and Cyp11a1 [86,87,88]. Overall, these mechanisms form a choline-mediated metabolic–epigenetic network, suggesting that choline supplementation may enhance follicular development.

5.1. Choline and DNA Methylation

As a co-factor for DNMTs and histone methyltransferases, SAM directly regulates the promoter methylation of reproduction-related genes. During folliculogenesis, global DNA methylation reprogramming occurs. Choline intake regulates DNMT1 expression. In rats, choline deficiency causes a 1.5-fold upregulation of DNMT1, leading to global DNA hypermethylation [89]. Moreover, in the livers of E17 fetuses with choline deficiency, the differentially methylated region 2 of the insulin-like growth factor 2 (Igf2) gene becomes hypermethylated, and the methylation level is linked to DNMT1 expression. Choline deficiency may activate a compensatory mechanism that upregulates DNMT1 to maintain or enhance DNA methylation, affecting specific gene expression. A similar mechanism applies to poultry. From E7 to E18 in chicken embryos, DNA methylation levels change significantly, with hypermethylated regions concentrated in the promoters of development-related genes [90]. In Langshan chickens, whole-genome bisulfite sequencing (WGBS) identified 5948 differentially methylated regions (DMRs) in the hypothalamus and 4593 in the ovary. These DMRs are directly related to reduced fertility, causing a 20% decrease in reproductive performance [91]. In cattle, adding 1.8 mM choline to in vitro cultures increased postnatal body weight and survival rate by 8%, attributed to the hypermethylation of imprinted genes [92]. Similar to poultry, dynamic methylation changes in chicken embryos during organogenesis indicate that choline can alleviate the “reproductive bottleneck” [90,93]. In the Langshan chicken, ovarian DMRs in the hypothalamus and ovary are negatively correlated with fertilization rates [91]. However, most data come from rats or cattle, like in the choline deficiency model [89], and there is a lack of poultry-specific research [93]. For example, the expression pattern of chicken DNA methyltransferase DNMT3A differs from that in mammals, suggesting that choline dosage may not be universally applicable [90].

5.2. DNA Methylation and Folliculogenesis

Avian follicular development is a complex and tightly regulated process that spans the entire reproductive cycle. It consists of two phases: prehierarchical and hierarchical follicles, both strictly controlled to guarantee optimal egg-laying performance [94]. With the onset of sexual maturity, the ovarian primordial follicle pool is activated, embarking on the growth phase and progressing to primary and mature follicles in preparation for ovulation [95]. Follicular development adheres to a strict hierarchy, gradually maturing from small to large [96]. Prehierarchical follicles are prone to atresia before entering the differentiation stage. In birds, hierarchical follicles usually consist of 3–6 follicles over a species-specific size threshold: chicken (>9 mm) [97], goose (>12 mm) [98], quail (>8 mm) [99].
In birds, from their formation to the early embryonic development after fertilization, germ cells experience significant epigenetic changes [100,101]. During epigenetic reprogramming, avian primordial germ cells only undergo limited CpG demethylation and retain numerous imprinted genes [101,102]. However, when developing into functional gametes, they face extensive epigenetic reprogramming [103]. The specific mechanisms regulating genomic methylation and remethylation in avian oocytes, particularly during follicle selection, warrant further investigation.

5.3. Choline Regulation of Steroid Hormone Synthesis and Follicular Development

Ovarian steroid hormones include progesterone, estrogens, and androgens. Current research indicates that estrogens, particularly estradiol, have the most significant impact on follicular development [104]. Neonatal mice were injected with 20 µg of E2 daily for 3 days starting at 1 day postpartum (1 dpp). This treatment significantly inhibited primordial follicle (PF) formation and reduced the number of PFs in ovaries at 4 dpp. E2 treatment also decreased cell proliferation, as indicated by the downregulation of Ki67 and Top2a. In 21 dpp ovaries, multiple oocyte follicles (MOFs) appeared, indicating disrupted PF formation. In vitro, ovarian cells from 4 dpp E2-treated mice were analyzed by single-cell RNA sequencing (scRNA-seq), revealing that E2 treatment disrupted the differentiation of pre-granulosa cells (PG) and altered mitochondrial activity in oocytes [105]. Estradiol is also a crucial factor for follicular development, differentiation, and survival, especially in the growth of antral follicles [106]. Estradiol increases the sensitivity of granulosa cells to FSH and LH, thereby promoting granulosa cell proliferation and follicular development [107]. Estrogens exert their effects after binding to their respective receptors. Two types of estrogen receptors, estrogen receptor alpha (ERα) and estrogen receptor beta (ERβ), have been identified in the ovary, with ERβ being expressed at significantly higher levels than ERα [108]. Knockout mice lacking ERβ exhibit infertility reduced follicular development, and lower ovulation rates [109,110,111], indicating that estradiol primarily promotes follicular development through the ERβ pathway. In a study investigating additions of 0, 200, 400, 600, 800, and 1000 mg/kg choline to the diet of laying geese, the content of E2 in serum and the number of small white follicles were significantly increased after adding 600 mg/kg choline, and the expression of the ERα gene in the ovaries was the highest in the 1000 mg/kg group, while the expression of ERβ was lowest in 1000 mg/kg group [68]. It can be seen from the above that appropriate choline supplementation affects the expression of ERα and ERβ through the hypothalamic–pituitary–gonadal axis, enhances the binding of E2 to its receptor, and promotes follicular development, thereby improving the reproductive performance of geese.
Cyp19a1 encodes aromatase, which converts testosterone to E2. High temperatures cause hypermethylation of multiple CpG sites in the Cyp19a1 promoter region of red-eared slider turtle embryos’ gonads. When this hypermethylation reduces aromatase activity, the E2/T ratio in the gonadal microenvironment becomes imbalanced. ERα can enhance Cyp19a1 transcription by binding to the estrogen response element upstream of Cyp19a1, forming a positive feedback loop. This activates the SRY-Box transcription factor 9/doublesex and mab-3 related transcription factor 1 (SOX9/DMRT1) signaling pathway and induces testis differentiation [112,113,114,115]. In PCOS patients’ ovarian granulosa cells, Cyp19a1 promoter methylation is significantly downregulated, and this downregulation is negatively correlated with gene expression [80]. Cyp19a1 promoter methylation is negatively related to the E2/androstenedione ratio in follicular fluid, indicating it may suppress aromatase activity and increase androgen synthesis in granulosa cells [116]. Current research implies that choline could regulate Cyp19a1 methylation by reducing the methylation level of CpG islands, removing the inhibition of Cyp19a1 transcription, and restoring the function of aromatase, promoting androgen-to-estrogen conversion and alleviating PCOS-related hyperandrogenism. Mice aged 21–24 days were injected with equine chorionic gonadotropin to stimulate follicular growth, followed by human chorionic gonadotropin (hCG) injection 48 h later to induce ovulation and luteinization. Ovarian granulosa cells were collected at 0, 1, 2, 4, 8, and 12 h after hCG injection. The study found that hCG induction led to high H3K4me3 and low H3K9me3 and H3K27me3 at the Star promoter region. This increased C/EBPβ binding to the Star promoter, upregulating Star expression. For Cyp19a1, the promoter region saw low H3K4me3 and high H3K27me3, reducing cyclic adenosine monophosphate response element binding and downregulating Cyp19a1 expression [117]. Also, reducing anti-müllerian hormone receptor type 2 methylation may prevent excessive AMH-induced suppression of primordial follicle recruitment, thus preserving ovarian reserve [118,119,120]. Stimulating immortalized non-luteinized human granulosa cells (HGrC1) with bone morphogenetic protein 15 (BMP15) induces FSHR expression in human granulosa cells via Smad and non-Smad pathways and increases Cyp19a1 mRNA expression and estradiol production [121]. Lowered BMP15 levels may disrupt follicular development [122,123]. Choline can reduce follicle atresia and improve oocyte survival by blocking BMP15-mediated atresia and gene expression chaos from DNA hypomethylation [124,125]. Also, choline shortage can upset the competitive binding of SF-1 and chicken ovalbumin upstream promoter transcription factor [126], possibly increasing the regulation of aromatase promoter I.4 by pro-inflammatory factors like interleukin-1 beta, thus promoting estrogen-dependent conditions such as endometriosis and uterine fibroids [127,128]. By discussing choline’s roles in these processes, this review provides theoretical foundations for future research and novel approaches to improve animal reproductive performance, contributing to the broader field of reproductive health [129,130]. To provide a clearer understanding, a schematic diagram of the proposed mechanisms of choline in follicular development is presented in Figure 1.

6. Conclusions

This review highlights the essential role of choline in ovarian follicular development. Adequate choline intake supports methylation balance, steroid hormone synthesis, and oocyte maturation, whereas deficiency or excess impairs follicular health through homocysteine accumulation or TMAO production. Overall, maintaining optimal choline levels is critical for ensuring a favorable follicular microenvironment and reproductive performance.

Author Contributions

W.L.: Writing—original draft, Writing—Review and Editing, Investigation. X.Z.: Writing—Review and Editing. H.Y.: Writing—Review and Editing, Investigation. Z.W.: Methodology, Writing—Review and Editing, Conceptualization, Supervision, Project Administration. All authors have read and agreed to the published version of the manuscript.

Funding

The authors declare that no funds, grants, or other support were received during the preparation of this manuscript.

Informed Consent Statement

Not applicable.

Data Availability Statement

No new data were created or analyzed in this study. Data sharing is not applicable to this article.

Conflicts of Interest

The authors declare no competing interests.

References

  1. Crowe, M.A.; Hostens, M.; Opsomer, G. Reproductive Management in Dairy Cows-the Future. Ir. Vet. J. 2018, 71, 1. [Google Scholar] [CrossRef] [PubMed]
  2. Costa, K.A.; Marques, D.B.D.; De Campos, C.F.; Saraiva, A.; Guimarães, J.D.; Guimarães, S.E.F. Nutrition Influence on Sow Reproductive Performance and Conceptuses Development and Survival: A Review about l-Arginine Supplementation. Livest. Sci. 2019, 228, 97–103. [Google Scholar] [CrossRef]
  3. Leroy, J.L.M.R.; Rizos, D.; Sturmey, R.; Bossaert, P.; Gutierrez-Adan, A.; Hoeck, V.V.; Valckx, S.; Bols, P.E.J. Intrafollicular Conditions as a Major Link between Maternal Metabolism and Oocyte Quality: A Focus on Dairy Cow Fertility. Reprod. Fertil. Dev. 2011, 24, 1–12. [Google Scholar] [CrossRef]
  4. Liu, Y.; Morita, T.; Sugiyama, K. Effects of Betaine Supplementation and Choline Deficiency on Folate Deficiency-Induced Hyperhomocysteinemia in Rats. Pubmed 2012, 58, 69–77. [Google Scholar] [CrossRef][Green Version]
  5. Berker, B.; Kaya, C.; Aytac, R.; Satiroglu, H. Homocysteine Concentrations in Follicular Fluid Are Associated with Poor Oocyte and Embryo Qualities in Polycystic Ovary Syndrome Patients Undergoing Assisted Reproduction. Hum. Reprod. 2009, 24, 2293–2302. [Google Scholar] [CrossRef]
  6. Zhu, Q.; Sun, J.; An, C.; Li, X.; Xu, S.; He, Y.; Zhang, X.; Liu, L.; Hu, K.; Liang, M. Mechanism of LncRNA Gm2044 in Germ Cell Development. Front. Cell Dev. Biol. 2024, 12, 1410914. [Google Scholar]
  7. Krzysztof Blusztajn, J.; Mellott, J.T. Choline Nutrition Programs Brain Development via DNA and Histone Methylation. Cent. Nerv. Syst. Agents Med. Chem. 2012, 12, 82–94. [Google Scholar] [CrossRef]
  8. Jiang, X.; Greenwald, E.; Jack-Roberts, C. Effects of Choline on DNA Methylation and Macronutrient Metabolic Gene Expression in in Vitro Models of Hyperglycemia. Nutr. Metab. Insights 2016, 9, 11. [Google Scholar] [CrossRef] [PubMed]
  9. Qian, F.-C.; Zhou, L.-W.; Li, Y.-Y.; Yu, Z.-M.; Li, L.-D.; Wang, Y.-Z.; Xu, M.-C.; Wang, Q.-Y.; Li, C.-Q. SEanalysis 2.0: A Comprehensive Super-Enhancer Regulatory Network Analysis Tool for Human and Mouse. Nucleic Acids Res. 2023, 51, W520–W527. [Google Scholar] [CrossRef]
  10. Gao, Y.; Wang, C.; Wang, K.; He, C.; Hu, K.; Liang, M. The Effects and Molecular Mechanism of Heat Stress on Spermatogenesis and the Mitigation Measures. Syst. Biol. Reprod. Med. 2022, 68, 331–347. [Google Scholar] [CrossRef] [PubMed]
  11. Chang, H.; Wang, D.; Xia, W.; Pan, X.; Huo, W.; Xu, S.; Li, Y. Epigenetic Disruption and Glucose Homeostasis Changes Following Low-Dose Maternal Bisphenol a Exposure. Toxicol. Res. 2016, 5, 1400–1409. [Google Scholar] [CrossRef] [PubMed]
  12. Chen, X.; Qiu, H.; Wang, C.; Yuan, Y.; Tickner, J.; Xu, J.; Zou, J. Molecular Structure and Differential Function of Choline Kinases CHKα and CHKβ in Musculoskeletal System and Cancer. Cytokine Growth Factor. Rev. 2017, 33, 65–72. [Google Scholar] [CrossRef] [PubMed]
  13. Estrada-Cortés, E.; Ortiz, W.G.; Chebel, R.C.; Jannaman, E.A.; Moss, J.I.; De Castro, F.C.; Zolini, A.M.; Staples, C.R.; Hansen, P.J. Embryo and Cow Factors Affecting Pregnancy per Embryo Transfer for Multiple-Service, Lactating Holstein Recipients. Transl. Anim. Sci. 2019, 3, 60–65. [Google Scholar] [CrossRef]
  14. McDowell, L.R. Vitamins in Animal and Human Nutrition, 1st ed.; Wiley: Ames, IA, USA, 2000; ISBN 978-0-8138-2630-1. [Google Scholar]
  15. Richmond, G.S.; Smith, T.K. Phospholipases A1. Int. J. Mol. Sci. 2011, 12, 588–612. [Google Scholar] [CrossRef]
  16. Zeisel, S.H.; Blusztajn, J.K. Choline and Human Nutrition. Annu. Rev. Nutr. 1994, 14, 269–296. [Google Scholar] [CrossRef] [PubMed]
  17. Zeisel, S.H. Choline Deficiency. J. Nutr. Biochem. 1990, 1, 332–349. [Google Scholar] [CrossRef]
  18. Murota, K. Digestion and Absorption of Dietary Glycerophospholipids in the Small Intestine: Their Significance as Carrier Molecules of Choline and n-3 Polyunsaturated Fatty Acids. Biocatal. Agric. Biotechnol. 2020, 26, 101633. [Google Scholar] [CrossRef]
  19. Kamath, A.V.; Darling, I.M.; Morris, M.E. Choline Uptake in Human Intestinal Caco-2 Cells Is Carrier-Mediated. J. Nutr. 2003, 133, 2607–2611. [Google Scholar] [CrossRef]
  20. Hegazy, E.; Schwenk, M. Choline Uptake by Isolated Enterocytes of Guinea Pig. J. Nutr. 1984, 114, 2217–2220. [Google Scholar] [CrossRef]
  21. Sanford, P.A.; Smyth, D.H. Intestinal Transfer of Choline in Rat and Hamster. J. Physiol. 1971, 215, 769–788. [Google Scholar] [CrossRef]
  22. Arias, N.; Arboleya, S.; Allison, J.; Kaliszewska, A.; Higarza, S.G.; Gueimonde, M.; Arias, J.L. The Relationship between Choline Bioavailability from Diet, Intestinal Microbiota Composition, and Its Modulation of Human Diseases. Nutrients 2020, 12, 2340. [Google Scholar] [CrossRef]
  23. Luo, T.; Guo, Z.; Liu, D.; Guo, Z.; Wu, Q.; Li, Q.; Lin, R.; Chen, P.; Ou, C.; Chen, M. Deficiency of PSRC1 Accelerates Atherosclerosis by Increasing TMAO Production via Manipulating Gut Microbiota and Flavin Monooxygenase 3. Gut Microbes 2022, 14, 2077602. [Google Scholar] [CrossRef] [PubMed]
  24. Chakraborty, N.; Menze, M.A.; Elmoazzen, H.; Hand, S.C.; Toner, M. Choline Chloride Improves the Desiccation Tolerance of Chinese Hamster Ovary Cells. In Proceedings of the ASME 2010 Summer Bioengineering Conference, Parts A and B, Naples, FL, USA, 16–19 June 2010; American Society of Mechanical Engineers: Naples, FL, USA, 2010; pp. 993–994. [Google Scholar]
  25. Fernando-Warnakulasuriya, G.J.; Eckerson, M.L.; Clark, W.A.; Wells, M.A. Lipoprotein Metabolism in the Suckling Rat: Characterization of Plasma and Lymphatic Lipoproteins. J. Lipid Res. 1983, 24, 1626–1638. [Google Scholar] [CrossRef] [PubMed]
  26. Li, Z.; Agellon, L.B.; Allen, T.M.; Umeda, M.; Jewell, L.; Mason, A.; Vance, D.E. The Ratio of Phosphatidylcholine to Phosphatidylethanolamine Influences Membrane Integrity and Steatohepatitis. Cell Metab. 2006, 3, 321–331. [Google Scholar] [CrossRef]
  27. Glunde, K.; Bhujwalla, Z.M.; Ronen, S.M. Choline Metabolism in Malignant Transformation. Nat. Rev. Cancer 2011, 11, 835–848. [Google Scholar] [CrossRef] [PubMed]
  28. Celemin Sarmiento, A.; Bradford, B.J.; Mamedova, L.K.; Zhou, G.; Estes, K.A.; Swartz, T.H. Effects of Dietary Rumen-Protected Choline Supplementation on Genome-Wide DNA Methylation in Mammary Tissue from Postpartum Dairy Cows. JDS Commun. 2025, 6, 333–338. [Google Scholar] [CrossRef]
  29. Sato, N.; Mori, N.; Hirashima, T.; Moriyama, T. Diverse Pathways of Phosphatidylcholine Biosynthesis in Algae as Estimated by Labeling Studies and Genomic Sequence Analysis. Plant J. 2016, 87, 281–292. [Google Scholar] [CrossRef]
  30. DeLong, C.J.; Hicks, A.M.; Cui, Z. Disruption of Choline Methyl Group Donation for Phosphatidylethanolamine Methylation in Hepatocarcinoma Cells. J. Biol. Chem. 2002, 277, 17217–17225. [Google Scholar] [CrossRef]
  31. Ganz, A.B.; Shields, K.; Fomin, V.G.; Lopez, Y.S.; Mohan, S.; Lovesky, J.; Chuang, J.C.; Ganti, A.; Carrier, B.; Yan, J.; et al. Genetic Impairments in Folate Enzymes Increase Dependence on Dietary Choline for Phosphatidylcholine Production at the Expense of Betaine Synthesis. FASEB J. 2016, 30, 3321–3333. [Google Scholar] [CrossRef]
  32. Gibellini, F.; Smith, T.K. The Kennedy Pathway-De Novo Synthesis of Phosphatidylethanolamine and Phosphatidylcholine. IUBMB Life 2010, 62, 414–428. [Google Scholar] [CrossRef]
  33. Sanchez-Lopez, E.; Zhong, Z.; Stubelius, A.; Sweeney, S.R.; Booshehri, L.M.; Antonucci, L.; Liu-Bryan, R.; Lodi, A.; Terkeltaub, R.; Lacal, J.C.; et al. Choline Uptake and Metabolism Modulate Macrophage IL-1β and IL-18 Production. Cell Metab. 2019, 29, 1350–1362.e7. [Google Scholar] [CrossRef]
  34. Okuda, T.; Haga, T. Functional Characterization of the Human High-affinity Choline Transporter. FEBS Lett. 2000, 484, 92–97. [Google Scholar] [CrossRef]
  35. Koepsell, H.; Schmitt, B.M.; Gorboulev, V. Organic Cation Transporters. In Reviews of Physiology, Biochemistry and Pharmacology; Springer: Berlin/Heidelberg, Germany, 2004; Volume 150, pp. 36–90. ISBN 978-3-540-20214-1. [Google Scholar]
  36. Taylor, A.; Grapentine, S.; Ichhpuniani, J.; Bakovic, M. Choline Transporter-like Proteins 1 and 2 Are Newly Identified Plasma Membrane and Mitochondrial Ethanolamine Transporters. J. Biol. Chem. 2021, 296, 100604. [Google Scholar] [CrossRef] [PubMed]
  37. Nakamura, T.; Fujiwara, R.; Ishiguro, N.; Oyabu, M.; Nakanishi, T.; Shirasaka, Y.; Maeda, T.; Tamai, I. Involvement of Choline Transporter-like Proteins, CTL1 and CTL2, in Glucocorticoid-Induced Acceleration of Phosphatidylcholine Synthesis via Increased Choline Uptake. Biol. Pharm. Bull. 2010, 33, 691–696. [Google Scholar] [CrossRef]
  38. Imbard, A.; Smulders, Y.M.; Barto, R.; Smith, D.E.; Kok, R.M.; Jakobs, C.; Blom, H.J. Plasma Choline and Betaine Correlate with Serum Folate, Plasma S-Adenosyl-Methionine and S-Adenosyl-Homocysteine in Healthy Volunteers. Clin. Chem. Lab. Med. 2013, 51, 683–692. [Google Scholar] [CrossRef]
  39. Robinson, J.L.; McBreairty, L.E.; Brunton, J.A.; Bertolo, R.F. The Effects of Folate, Choline and Betaine on Transmethylation in a Methionine Restricted Piglet. FASEB J. 2013, 27, 1077.24. [Google Scholar] [CrossRef]
  40. Ziganshin, A.U.; Khairullin, A.E.; Hoyle, C.H.V.; Grishin, S.N. Modulatory Roles of ATP and Adenosine in Cholinergic Neuromuscular Transmission. Int. J. Mol. Sci. 2020, 21, 6423. [Google Scholar] [CrossRef] [PubMed]
  41. Sastry, B.V.R. Human Placental Cholinergic System. Biochem. Pharmacol. 1997, 53, 1577–1586. [Google Scholar] [CrossRef] [PubMed]
  42. Dey, S.; Ray, K. Cholinergic Activity Is Essential for Maintaining the Anterograde Transport of Choline Acetyltransferase in Drosophila. Sci. Rep. 2018, 8, 8028. [Google Scholar] [CrossRef]
  43. Yan, M.; Man, S.; Sun, B.; Ma, L.; Guo, L.; Huang, L.; Gao, W. Gut Liver Brain Axis in Diseases: The Implications for Therapeutic Interventions. Signal Transduct. Target. Ther. 2023, 8, 443. [Google Scholar] [CrossRef]
  44. Satheesh Babu, A.K.; Petersen, C.; Iglesias-Carres, L.; Paz, H.A.; Wankhade, U.D.; Neilson, A.P.; Anandh Babu, P.V. Blueberry Intervention Mitigates Detrimental Microbial Metabolite Trimethylamine N-oxide by Modulating Gut Microbes. BioFactors 2024, 50, 392–404. [Google Scholar] [CrossRef]
  45. Jaworska, K.; Bielinska, K.; Gawrys-Kopczynska, M.; Ufnal, M. TMA (Trimethylamine), but Not Its Oxide TMAO (Trimethylamine-Oxide), Exerts Haemodynamic Effects: Implications for Interpretation of Cardiovascular Actions of Gut Microbiome. Cardiovasc. Res. 2019, 115, 1948–1949. [Google Scholar] [CrossRef] [PubMed]
  46. Morton, C.C.; Aitchison, A.J.; Gehrig, K.; Ridgway, N.D. A Mechanism for Suppression of the CDP-Choline Pathway during Apoptosis. J. Lipid Res. 2013, 54, 3373–3384. [Google Scholar] [CrossRef]
  47. Lai, F.; Liu, X.; Li, N.; Zhang, R.; Zhao, Y.; Feng, Y.; Nyachoti, C.M.; Shen, W.; Li, L. Phosphatidylcholine Could Protect the Defect of Zearalenone Exposure on Follicular Development and Oocyte Maturation. Aging 2018, 10, 3486–3506. [Google Scholar] [CrossRef]
  48. Noga, A.A.; Vance, D.E. Insights into the Requirement of Phosphatidylcholine Synthesis for Liver Function in Mice. J. Lipid Res. 2003, 44, 1998–2005. [Google Scholar] [CrossRef]
  49. West, A.A.; Yan, J.; Jiang, X.; Perry, C.A.; Innis, S.M.; Caudill, M.A. Choline Intake Influences Phosphatidylcholine DHA Enrichment in Nonpregnant Women but Not in Pregnant Women in the Third Trimester123. Am. J. Clin. Nutr. 2013, 97, 718–727. [Google Scholar] [CrossRef]
  50. Anckaert, E.; Romero, S.; Adriaenssens, T.; Smitz, J. Effects of Low Methyl Donor Levels in Culture Medium during Mouse Follicle Culture on Oocyte Imprinting Establishment. Biol. Reprod. 2010, 83, 377–386. [Google Scholar] [CrossRef] [PubMed][Green Version]
  51. Bokor, S.; Vass, R.A.; Funke, S.; Ertl, T.; Molnár, D. Epigenetic Effect of Maternal Methyl-Group Donor Intake on Offspring’s Health and Disease. Life 2022, 12, 609. [Google Scholar] [CrossRef]
  52. Ren, Y.; Zeng, Y.; Wu, Y.; Zhang, Q.; Xiao, X. Maternal Methyl Donor Supplementation: A Potential Therapy for Metabolic Disorder in Offspring. J. Nutr. Biochem. 2024, 124, 109533. [Google Scholar] [CrossRef] [PubMed]
  53. Velker, B.A.M.; Denomme, M.M.; Krafty, R.T.; Mann, M.R.W. Maintenance of Mest Imprinted Methylation in Blastocyst-Stage Mouse Embryos Is Less Stable than Other Imprinted Loci Following Superovulation or Embryo Culture. Environ. Epigenet. 2017, 3, dvx015. [Google Scholar] [CrossRef]
  54. Fang, S.; Chang, K.; Lefebvre, L. Roles of Endogenous Retroviral Elements in the Establishment and Maintenance of Imprinted Gene Expression. Front. Cell Dev. Biol. 2024, 12, 1369751. [Google Scholar] [CrossRef]
  55. Shojaei Saadi, H.A.; Gagné, D.; Fournier, É.; Baldoceda Baldeon, L.M.; Sirard, M.-A.; Robert, C. Responses of Bovine Early Embryos to S-Adenosyl Methionine Supplementation in Culture. Epigenomics 2016, 8, 1039–1060. [Google Scholar] [CrossRef]
  56. Patel, J.; Chaudhary, H.; Chudasama, A.; Panchal, J.; Trivedi, A.; Panchal, S.; Joshi, T.; Joshi, R. Comparing the metabolomic landscape of polycystic ovary syndrome within urban and rural environments. Commun. Med. 2025, 5, 253. [Google Scholar] [CrossRef]
  57. Sun, L.; Hu, W.; Liu, Q.; Hao, Q.; Sun, B.; Zhang, Q.; Mao, S.; Qiao, J.; Yan, X. Metabonomics Reveals Plasma Metabolic Changes and Inflammatory Marker in Polycystic Ovary Syndrome Patients. J. Proteome Res. 2012, 11, 2937–2946. [Google Scholar] [CrossRef]
  58. Ersahin, A.A.; Çalışkan, E. Clomiphene Citrate Changes Metabolite Content of Follicular Fluid of PCOS Women. Eur. Rev. Med. Pharmacol. Sci. 2018, 22, 4359–4362. [Google Scholar] [CrossRef] [PubMed]
  59. Zhan, X.; Fletcher, L.; Dingle, S.; Baracuhy, E.; Wang, B.; Huber, L.-A.; Li, J. Choline Supplementation Influences Ovarian Follicular Development. Front. Biosci. Landmark 2021, 26, 1525–1536. [Google Scholar] [CrossRef] [PubMed]
  60. Lee, G.; Choi, S.; Chang, J.; Choi, D.; Son, J.S.; Kim, K.; Kim, S.M.; Jeong, S.; Park, S.M. Association of L-α Glycerylphosphorylcholine with Subsequent Stroke Risk after 10 Years. JAMA Netw. Open 2021, 4, e2136008. [Google Scholar] [CrossRef] [PubMed]
  61. Zeisel, S.H.; Warrier, M. Trimethylamine N-Oxide, the Microbiome, and Heart and Kidney Disease. Annu. Rev. Nutr. 2017, 37, 157–181. [Google Scholar] [CrossRef]
  62. Downs, S.M.; Mosey, J.L.; Klinger, J. Fatty Acid Oxidation and Meiotic Resumption in Mouse Oocytes. Mol. Reprod. Dev. 2009, 76, 844–853. [Google Scholar] [CrossRef]
  63. Li, J.; Zhang, Z.; Wei, Y.; Zhu, P.; Yin, T.; Wan, Q. Metabonomic Analysis of Follicular Fluid in Patients with Diminished Ovarian Reserve. Front. Endocrinol. 2023, 14, 1132621. [Google Scholar] [CrossRef]
  64. Nagy, R.A.; Homminga, I.; Jia, C.; Liu, F.; Anderson, J.L.C.; Hoek, A.; Tietge, U.J.F. Trimethylamine-N-Oxide Is Present in Human Follicular Fluid and Is a Negative Predictor of Embryo Quality. Hum. Reprod. 2020, 35, 81–88. [Google Scholar] [CrossRef]
  65. Wallace, M.; Cottell, E.; Gibney, M.J.; McAuliffe, F.M.; Wingfield, M.; Brennan, L. An Investigation into the Relationship between the Metabolic Profile of Follicular Fluid, Oocyte Developmental Potential, and Implantation Outcome. Fertil. Steril. 2012, 97, 1078–1084.e8. [Google Scholar] [CrossRef] [PubMed]
  66. Júnior, D.N.; Pereira, A.A.; Júnior, G.M.O.; de Almeida, V.V.; da Silva, E.F.; da Silva, E.F.; da Silva, W.A.; de Alcântara, R.S.; da Silva, W.A.; Júnior, G.S.C.; et al. Choline and Digestible Methionine + Cystine Supplementation for Quail in the Laying Phase. Semin. Ciênc. Agrár. 2020, 41, 305–312. [Google Scholar] [CrossRef]
  67. Olgun, O.; Gül, E.T.; Kılınç, G.; Yıldız, A.; Çolak, A.; Sarmiento-García, A. Performance, Egg Quality, and Yolk Antioxidant Capacity of the Laying Quail in Response to Dietary Choline Levels. Animals 2022, 12, 3361. [Google Scholar] [CrossRef]
  68. Liu, W.; Wang, J.; Wang, J.; Ji, R.; Wang, Z.; Yang, H. Effects of Choline on Reproductive Performance, Egg Quality, and Ovarian Morphological Development in Laying Geese. Anim. Feed. Sci. Technol. 2025, 324, 116288. [Google Scholar] [CrossRef]
  69. Zhong, W.; Hu, L.; Zhao, Y.; Li, Z.; Zhuo, Y.; Jiang, X.; Li, J.; Zhao, X.; Che, L.; Feng, B.; et al. Effects of Dietary Choline Levels During Pregnancy on Reproductive Performance, Plasma Metabolome and Gut Microbiota of Sows. Front. Vet. Sci. 2022, 8, 771228. [Google Scholar] [CrossRef]
  70. Zhan, X. The Effect of Choline Supplement and miR-29b-3p in Porcine Ovarian Follicular Development. Ph.D. Thesis, University of Guelph, Guelph, ON, Canada, 2023. [Google Scholar]
  71. Acosta, D.A.V.; Rivelli, M.I.; Skenandore, C.; Zhou, Z.; Keisler, D.H.; Luchini, D.; Corrêa, M.N.; Cardoso, F.C. Effects of Rumen-Protected Methionine and Choline Supplementation on Steroidogenic Potential of the First Postpartum Dominant Follicle and Expression of Immune Mediators in Holstein Cows. Theriogenology 2017, 96, 1–9. [Google Scholar] [CrossRef]
  72. Kotsampasi, B.; Tsiplakou, E.; Karatzia, M.-A.; Oikonomou, S.; Mitsiopoulou, C.; Kalogiannis, D.; Dovolou, E.; Lymperopoulos, A.; Sotirakoglou, K.; Anastasiadou, M.; et al. Effects of Rumen-Protected Methionine, Choline, and Betaine Supplementation on Ewes’ Pregnancy and Reproductive Outcomes. Vet. Sci. 2025, 12, 723. [Google Scholar] [CrossRef]
  73. Davenport, C.; Yan, J.; Taesuwan, S.; Shields, K.; West, A.A.; Jiang, X.; Perry, C.A.; Malysheva, O.V.; Stabler, S.P.; Allen, R.H.; et al. Choline Intakes Exceeding Recommendations during Human Lactation Improve Breast Milk Choline Content by Increasing PEMT Pathway Metabolites. J. Nutr. Biochem. 2015, 26, 903–911. [Google Scholar] [CrossRef]
  74. Taesuwan, S.; McDougall, M.Q.; Malysheva, O.V.; Bender, E.; Nevins, J.E.H.; Devapatla, S.; Vidavalur, R.; Caudill, M.A.; Klatt, K.C. Choline Metabolome Response to Prenatal Choline Supplementation across Pregnancy: A Randomized Controlled Trial. FASEB J. 2021, 35, e22063. [Google Scholar] [CrossRef]
  75. Zhao, D.; Gao, Y.; Su, Y.; Zhou, Y.; Yang, T.; Li, Y.; Wang, Y.; Sun, Y.; Chen, L.; Zhang, F.; et al. Oroxylin a Regulates cGAS DNA Hypermethylation Induced by Methionine Metabolism to Promote HSC Senescence. Pharmacol. Res. 2023, 187, 106590. [Google Scholar] [CrossRef]
  76. Ferré, V.M.; Brousseau, J.; Charpentier, C.; Péré, H. Nouveaux Marqueurs Développés Pour Le Dépistage Des Cancers Liés Aux HPV. Rev. Francoph. Lab. 2024, 2024, 46–58. [Google Scholar] [CrossRef]
  77. Yang, G.; Li, S.; Cai, S.; Zhou, J.; Ye, Q.; Zhang, S.; Chen, F.; Wang, F.; Zeng, X. Dietary Methionine Supplementation during the Estrous Cycle Improves Follicular Development and Estrogen Synthesis in Rats. Food Funct. 2024, 15, 704–715. [Google Scholar] [CrossRef]
  78. Usman, M.; Li, A.; Wu, D.; Qinyan, Y.; Yi, L.X.; He, G.; Lu, H. The Functional Role of lncRNAs as ceRNAs in Both Ovarian Processes and Associated Diseases. Non-Coding RNA Res. 2024, 9, 165–177. [Google Scholar] [CrossRef] [PubMed]
  79. Annisa, N.G.; Febri, R.R.; Darmawi; Kinasih, T.; Muharam, R. Asmarinah Analysis of the Methylation Profiles of the Steroidogenic Factor-1 (SF-1) Gene in Peritoneal and Ovarian Endometriosis. J. Phys. Conf. Ser. 2018, 1073, 32080. [Google Scholar] [CrossRef]
  80. Yu, Y.; Sun, C.; Liu, Y.; Li, Y.; Wang, L.; Zhang, W. Promoter Methylation of CYP19A1 Gene in Chinese Polycystic Ovary Syndrome Patients. Gynecol. Obstet. Investig. 2013, 76, 209–213. [Google Scholar] [CrossRef] [PubMed]
  81. Hosseini, E.; Shahhoseini, M.; Afsharian, P.; Karimian, L.; Ashrafi, M.; Mehraein, F.; Afatoonian, R. Role of Epigenetic Modifications in the Aberrant CYP19A1 Gene Expression in Polycystic Ovary Syndrome. Arch. Med. Sci. 2019, 15, 887–895. [Google Scholar] [CrossRef]
  82. Bulun, S.E.; Takayama, K.; Suzuki, T.; Sasano, H.; Yilmaz, B.; Sebastian, S. Organization of the Human Aromatase P450 (CYP19) Gene. Semin. Reprod. Med. 2004, 22, 5–9. [Google Scholar] [CrossRef]
  83. Ding, Y.; Long, C.; Liu, X.; Chen, X.; Guo, L.; Xia, Y.; He, J.; Wang, Y. 5-Aza-2’-Deoxycytidine Leads to Reduced Embryo Implantation and Reduced Expression of DNA Methyltransferases and Essential Endometrial Genes. PLoS ONE 2012, 7, e45364. [Google Scholar] [CrossRef]
  84. Geng, Y.; Gao, R.; Chen, X.; Liu, X.; Liao, X.; Li, Y.; Liu, S.; Ding, Y.; Wang, Y.; He, J. Folate Deficiency Impairs Decidualization and Alters Methylation Patterns of the Genome in Mice. Mol. Hum. Reprod. 2015, 21, 844–856. [Google Scholar] [CrossRef] [PubMed]
  85. Long, J.; Li, W.; Chen, M.; Ding, Y.; Chen, X.; Tong, C.; Li, N.; Liu, X.; He, J.; Peng, C.; et al. Uterine Deficiency of Dnmt3b Impairs Decidualization and Causes Consequent Embryo Implantation Defects. Cell Biol. Toxicol. 2023, 39, 1077–1098. [Google Scholar] [CrossRef]
  86. Kumar, P.; Sait, S.F. Luteinizing Hormone and Its Dilemma in Ovulation Induction. J. Hum. Reprod. Sci. 2011, 4, 2–7. [Google Scholar] [CrossRef] [PubMed]
  87. Xia, Y.; Shen, S.; Zhang, X.; Deng, Z.; Xiang, Z.; Wang, H.; Yi, L.; Gao, Q.; Wang, Y. Epigenetic Pattern Changes in Prenatal Female Sprague-Dawley Rats Following Exposure to Androgen. Reprod. Fertil. Dev. 2015, 28, 1414–1423. [Google Scholar] [CrossRef] [PubMed]
  88. Zhu, G.; Mao, Y.; Zhou, W.; Jiang, Y. Dynamic Changes in the Follicular Transcriptome and Promoter DNA Methylation Pattern of Steroidogenic Genes in Chicken Follicles throughout the Ovulation Cycle. PLoS ONE 2015, 10, e0146028. [Google Scholar] [CrossRef]
  89. Kovacheva, V.P.; Mellott, T.J.; Davison, J.M.; Wagner, N.; Lopez-Coviella, I.; Schnitzler, A.C.; Blusztajn, J.K. Gestational Choline Deficiency Causes Global and Igf2 Gene DNA Hypermethylation by Up-Regulation of Dnmt1 Expression. J. Biol. Chem. 2007, 282, 31777–31788. [Google Scholar] [CrossRef]
  90. Li, S.; Zhu, Y.; Zhi, L.; Han, X.; Shen, J.; Liu, Y.; Yao, J.; Yang, X. DNA Methylation Variation Trends during the Embryonic Development of Chicken. PLoS ONE 2016, 11, e0159230. [Google Scholar] [CrossRef] [PubMed]
  91. Han, W.; Xue, Q.; Li, G.; Yin, J.; Zhang, H.; Zhu, Y.; Xing, W.; Cao, Y.; Su, Y.; Wang, K.; et al. Genome-Wide Analysis of the Role of DNA Methylation in Inbreeding Depression of Reproduction in Langshan Chicken. Genomics 2020, 112, 2677–2687. [Google Scholar] [CrossRef]
  92. Haimon, M.L.J.; Estrada-Cortés, E.; Amaral, T.F.; Martin, H.; Jeensuk, S.; Block, J.; Heredia, D.; Venturini, M.; Rojas, C.S.; Gonella-Diaza, A.M.; et al. Provision of Choline Chloride to the Bovine Preimplantation Embryo Alters Postnatal Body Size and DNA Methylation. Biol. Reprod. 2024, 111, 567–579. [Google Scholar] [CrossRef]
  93. Ju, X.; Wang, Z.; Cai, D.; Bello, S.F.; Nie, Q. DNA Methylation in Poultry: A Review. J. Anim. Sci. Biotechnol. 2023, 14, 138. [Google Scholar] [CrossRef]
  94. Zhong, C.; Liu, Z.; Li, D.; Kang, L.; Jiang, Y. Long-Read Sequencing Reveals the Effect of Follicle-Stimulating Hormone on the mRNA Profile of Chicken Granulosa Cells from Prehierarchical Follicles. Poult. Sci. 2023, 102, 102600. [Google Scholar] [CrossRef]
  95. Zhang, T.; He, M.; Zhao, L.; Qin, S.; Zhu, Z.; Du, X.; Zhou, B.; Yang, Y.; Liu, X.; Xia, G.; et al. HDAC6 Regulates Primordial Follicle Activation through mTOR Signaling Pathway. Cell Death Dis. 2021, 12, 559. [Google Scholar] [CrossRef]
  96. Lin, X.; Liu, X.; Guo, C.; Liu, M.; Mi, Y.; Zhang, C. Promotion of the Prehierarchical Follicle Growth by Postovulatory Follicles Involving PGE2-EP2 Signaling in Chickens. J. Cell. Physiol. 2018, 233, 8984–8995. [Google Scholar] [CrossRef]
  97. Shen, M.; Wu, P.; Li, T.; Wu, P.; Chen, F.; Chen, L.; Xie, K.; Wang, J.; Zhang, G. Transcriptome Analysis of circRNA and mRNA in Theca Cells during Follicular Development in Chickens. Genes. 2020, 11, 489. [Google Scholar] [CrossRef] [PubMed]
  98. Yang, Y.; Yao, Y.; Cao, Z.F.; Gu, T.; Xu, Q.; Chen, G. Histological Characteristics of Follicles and Reproductive Hormone Secretion during Ovarian Follicle Development in Laying Geese. Poult. Sci. 2019, 98, 6063–6070. [Google Scholar] [CrossRef]
  99. Sreesujatha, R.M.; Jeyakumar, S.; Kundu, A.; Balasundaram, C. Use of Transcutaneous Ultrasonography to Characterize Ovarian Status, Size Distribution, and Hierarchical Status of Follicles in Japanese Quail (Coturnix Coturnix Japonica). Theriogenology 2016, 86, 1231–1239. [Google Scholar] [CrossRef]
  100. Li, C.; Guo, S.; Zhang, M.; Guo, Y.; Gao, J. DNA Methylation and Histone Modification Patterns during the Late Embryonic and Early Postnatal Development of Chickens. Poult. Sci. 2015, 94, 706–721. [Google Scholar] [CrossRef]
  101. Kress, C.; Jouneau, L.; Pain, B. Reinforcement of Repressive Marks in the Chicken Primordial Germ Cell Epigenetic Signature: Divergence from Basal State Resetting in Mammals. Epigenet. Chromatin 2024, 17, 11. [Google Scholar] [CrossRef] [PubMed]
  102. Jang, H.-J.; Seo, H.W.; Lee, B.R.; Yoo, M.; Womack, J.E.; Han, J.Y. Gene Expression and DNA Methylation Status of Chicken Primordial Germ Cells. Mol. Biotechnol. 2013, 54, 177–186. [Google Scholar] [CrossRef] [PubMed]
  103. Yu, M.; Li, D.F.; Cao, W.; Chen, X.; Du, W. Effects of Ten-Eleven Translocation 1 (Tet1) on DNA Methylation and Gene Expression in Chicken Primordial Germ Cells. Reprod. Fertil. Dev. 2019, 31, 509–520. [Google Scholar] [CrossRef]
  104. Xu, R.; Wen, D.; Yin, L.; Tang, Y.; Lu, S.; Gao, Y.; Pan, M.-H.; Han, B.; Ma, B. Estrogen Influences the Transzonal Projection Assembly of Cumulus-Oocyte Complexes through G Protein-Coupled Estrogen Receptor during Goat Follicle Development. Mol. Reprod. Dev. 2024, 91, e23763. [Google Scholar] [CrossRef]
  105. Yan, Y.; Zhang, H.; Xu, R.; Luo, L.; Yin, L.; Wu, H.; Zhang, Y.; Li, C.; Lu, S.; Tang, Y.; et al. Single-Cell Sequencing Reveals the Transcriptional Alternations of 17β-Estradiol Suppressing Primordial Follicle Formation in Neonatal Mouse Ovaries. Cell Prolif. 2024, 57, e13713. [Google Scholar] [CrossRef]
  106. Gupta, R.K.; Singh, J.M.; Leslie, T.C.; Meachum, S.; Flaws, J.A.; Yao, H.H.-C. Di-(2-Ethylhexyl) Phthalate and Mono-(2-Ethylhexyl) Phthalate Inhibit Growth and Reduce Estradiol Levels of Antral Follicles In Vitro. Toxicol. Appl. Pharmacol. 2010, 242, 224–230. [Google Scholar] [CrossRef]
  107. Orisaka, M.; Hattori, K.; Fukuda, S.; Mizutani, T.; Miyamoto, K.; Sato, T.; Tsang, B.K.; Kotsuji, F.; Yoshida, Y. Dysregulation of Ovarian Follicular Development in Female Rat: LH Decreases FSH Sensitivity during Preantral-Early Antral Transition. Endocrinology 2013, 154, 2870–2880. [Google Scholar] [CrossRef] [PubMed]
  108. Oliveira, A.G.; Dornas, R.A.P.; Mahecha, G.A.B.; Oliveira, C.A. Occurrence and Cellular Distribution of Estrogen Receptors ERα and ERβ in the Testis and Epididymal Region of Roosters. Gen. Comp. Endocrinol. 2011, 170, 597–603. [Google Scholar] [CrossRef]
  109. Krege, J.H.; Hodgin, J.B.; Couse, J.F.; Enmark, E.; Warner, M.; Mahler, J.F.; Sar, M.; Korach, K.S.; Gustafsson, J.-Å.; Smithies, O. Generation and Reproductive Phenotypes of Mice Lacking Estrogen Receptor β. Proc. Natl. Acad. Sci. USA 1998, 95, 15677–15682. [Google Scholar] [CrossRef]
  110. Cheng, G.; Weihua, Z.; Mäkinen, S.; Mäkelä, S.; Saji, S.; Warner, M.; Gustafsson, J.-A.; Hovatta, O. A Role for the Androgen Receptor in Follicular Atresia of Estrogen Receptor Beta Knockout Mouse Ovary. Biol. Reprod. 2002, 66, 77–84. [Google Scholar] [CrossRef] [PubMed]
  111. Emmen, J.M.; Couse, J.F.; Elmore, S.A.; Yates, M.M.; Kissling, G.E.; Korach, K.S. In Vitro Growth and Ovulation of Follicles from Ovaries of Estrogen Receptor (ER) α and ERβ Null Mice Indicate a Role for ERβ in Follicular Maturation. Endocrinology 2005, 146, 2817–2826. [Google Scholar] [CrossRef]
  112. Ramsey, M.; Crews, D. Steroid Signaling System Responds Differently to Temperature and Hormone Manipulation in the Red-Eared Slider Turtle (Trachemys Scripta Elegans), a Reptile with Temperature-Dependent Sex Determination. Sex. Dev. 2007, 1, 181–196. [Google Scholar] [CrossRef] [PubMed]
  113. Ru, W.; Hua, L.; Wei, Y.; Li, W.; Cao, D.; Ge, Y.; Chen, H.; Lan, X.; Gong, S. The Effect of the Cyp19a1 Gene Methylation Modification on Temperature-Dependent Sex Determination of Reeves’ Turtle (Mauremys reevesii). Asian Herpetol. Res. 2017, 8, 213–220. [Google Scholar] [CrossRef]
  114. Ge, C.; Ye, J.; Zhang, H.; Zhang, Y.; Sun, W.; Sang, Y.; Capel, B.; Qian, G. Dmrt1 Induces the Male Pathway in a Turtle with Temperature-Dependent Sex Determination. Development 2017, 144, dev.152033. [Google Scholar] [CrossRef]
  115. Shen, J.; Sun, W.; Wu, K.; Zhuang, T.; Lei, J.; Ma, Q.; Xiao, L.; Ge, C. Loss-and Gain-of-Function Analyses Reveal the Essential Role of Cyp19a1 in Ovarian Determination of the Red-Eared Slider Turtle. Genetics 2023, 224, iyad041. [Google Scholar] [CrossRef]
  116. Chen, Y.; Sun, Y.; Zhao, A.; Cai, X.; Yu, A.; Xu, Q.; Wang, P.; Yao, J.; Wang, Q.; Wang, W. Arsenic Exposure Diminishes Ovarian Follicular Reserve and Induces Abnormal Steroidogenesis by DNA Methylation. Ecotoxicol. Environ. Saf. 2022, 241, 113816. [Google Scholar] [CrossRef] [PubMed]
  117. Lee, L.; Asada, H.; Kizuka, F.; Tamura, I.; Maekawa, R.; Taketani, T.; Sato, S.; Yamagata, Y.; Tamura, H.; Sugino, N. Changes in Histone Modification and DNA Methylation of the StAR and Cyp19a1 Promoter Regions in Granulosa Cells Undergoing Luteinization during Ovulation in Rats. Endocrinology 2013, 154, 458–470. [Google Scholar] [CrossRef]
  118. Durlinger, A.L.L.; Gruijters, M.J.G.; Kramer, P.; Karels, B.; Ingraham, H.A.; Nachtigal, M.W.; Uilenbroek, J.T.J.; Grootegoed, J.A.; Themmen, A.P.N. Anti-Müllerian Hormone Inhibits Initiation of Primordial Follicle Growth in the Mouse Ovary. Endocrinology 2002, 143, 1076–1084. [Google Scholar] [CrossRef]
  119. van Rooij, I.A.J.; Broekmans, F.J.M.; te Velde, E.R.; Fauser, B.C.J.M.; Bancsi, L.F.J.M.M.; de Jong, F.H.; Themmen, A.P.N. Serum Anti-Müllerian Hormone Levels: A Novel Measure of Ovarian Reserve. Hum. Reprod. 2002, 17, 3065–3071. [Google Scholar] [CrossRef]
  120. Holt, J.E.; Jackson, A.; Roman, S.D.; Aitken, R.J.; Koopman, P.; McLaughlin, E.A. CXCR4/SDF1 Interaction Inhibits the Primordial to Primary Follicle Transition in the Neonatal Mouse Ovary. Dev. Biol. 2006, 293, 449–460. [Google Scholar] [CrossRef]
  121. Shimizu, K.; Nakamura, T.; Bayasula; Nakanishi, N.; Kasahara, Y.; Nagai, T.; Murase, T.; Osuka, S.; Goto, M.; Iwase, A.; et al. Molecular Mechanism of FSHR Expression Induced by BMP15 in Human Granulosa Cells. J. Assist. Reprod. Genet. 2019, 36, 1185–1194. [Google Scholar] [CrossRef]
  122. Wei, L.; Huang, R.; Li, L.; Fang, C.; Li, Y.; Liang, X. Reduced and Delayed Expression of GDF9 and BMP15 in Ovarian Tissues from Women with Polycystic Ovary Syndrome. J. Assist. Reprod. Genet. 2014, 31, 1483–1490. [Google Scholar] [CrossRef]
  123. Cao, L.; Zhang, Z.; Liu, P.; Xu, D.; Tang, L.; Fan, L.; Sun, X.; Li, J.; Wu, Q.; Li, Z.; et al. Aberrant BMP15/HIF-1α/SCF Signaling Pathway in Human Granulosa Cells Is Involved in the PCOS Related Abnormal Follicular Development. Taylor Fr. 2022, 38, 971–977. [Google Scholar] [CrossRef] [PubMed]
  124. Bestor, T.H. The DNA Methyltransferases of Mammals. Hum. Mol. Genet. 2000, 9, 2395–2402. [Google Scholar] [CrossRef] [PubMed]
  125. Gehring, M.; Reik, W.; Henikoff, S. DNA Demethylation by DNA Repair. Trends Genet. TIG 2009, 25, 82–90. [Google Scholar] [CrossRef]
  126. Zeitoun, K.; Takayama, K.; Michael, M.D.; Bulun, S.E. Stimulation of Aromatase P450 Promoter (II) Activity in Endometriosis and Its Inhibition in Endometrium Are Regulated by Competitive Binding of Steroidogenic Factor-1 and Chicken Ovalbumin Upstream Promoter Transcription Factor to the Same Cis-Acting Element. Mol. Endocrinol. 1999, 13, 239–253. [Google Scholar] [CrossRef][Green Version]
  127. Shozu, M.; Sumitani, H.; Segawa, T.; Yang, H.-J.; Murakami, K.; Kasai, T.; Inoue, M. Overexpression of Aromatase P450 in Leiomyoma Tissue Is Driven Primarily through Promoter I.4 of the Aromatase P450 Gene (CYP19). J. Clin. Endocrinol. Metab. 2002, 87, 2540–2548. [Google Scholar] [CrossRef] [PubMed][Green Version]
  128. Luo, J.; Song, Z.; Zhang, T.; Chu, K.; Li, J.; Zhou, J.; Lin, J. Upregulation of H-TERT and Ki-67 in Ectopic Endometrium Is Associated with Recurrence of Endometriosis. Pubmed 2022, 23, 158–163. [Google Scholar] [CrossRef] [PubMed]
  129. Zhou, J.; Guo, Z.; Peng, X.; Wu, B.; Meng, Q.; Lu, X.; Feng, L.; Guo, T. Chrysotoxine Regulates Ferroptosis and the PI3K/AKT/mTOR Pathway to Prevent Cervical Cancer. J. Ethnopharmacol. 2025, 338, 119126. [Google Scholar] [CrossRef] [PubMed]
  130. Zhang, G.; Song, C.; Yin, M.; Liu, L.; Zhang, Y.; Li, Y.; Zhang, J.; Guo, M.; Li, C. TRAPT: A Multi-Stage Fused Deep Learning Framework for Predicting Transcriptional Regulators Based on Large-Scale Epigenomic Data. Nat. Commun. 2025, 16, 3611. [Google Scholar] [CrossRef]
Figure 1. Proposed mechanisms of choline in follicular development. Choline participates in multiple pathways, including serving as a methyl donor through one-carbon metabolism, contributing to phosphatidylcholine synthesis for cell membrane integrity, and supporting acetylcholine production. These processes collectively influence DNA methylation, steroid hormone biosynthesis, and granulosa cell function, thereby regulating follicular growth and maturation.
Figure 1. Proposed mechanisms of choline in follicular development. Choline participates in multiple pathways, including serving as a methyl donor through one-carbon metabolism, contributing to phosphatidylcholine synthesis for cell membrane integrity, and supporting acetylcholine production. These processes collectively influence DNA methylation, steroid hormone biosynthesis, and granulosa cell function, thereby regulating follicular growth and maturation.
Biology 14 01220 g001
Table 1. Optimal dietary choline levels across species and their main reproductive or physiological effects.
Table 1. Optimal dietary choline levels across species and their main reproductive or physiological effects.
SpeciesOptimal Choline LevelMain Effects
Japanese quail (Coturnix japonica)0.126% (≈1260 mg/kg) diet; 1500 mg/kg to maintain laying performance and egg quality; 3500 mg/kg to enhance yolk antioxidant capacityImproved egg weight and feed conversion; maintained egg quality; increased yolk antioxidant activity [66,67]
Laying geese784–913 mg/kg dietEnhanced reproductive performance, egg production, and sex hormone levels; promoted ovarian development and hormone synthesis [68]
Gestating sows≈1910 mg/kg dietIncreased feed intake; improved piglet birth weight and litter uniformity; enhanced maternal antioxidant capacity and gut microbiota composition [69]
Pig500–1000 mg/kg (diet)Improves ovarian function, promotes corpus luteum formation, regulates reproduction-related gene expression, and enhances reproductive performance [70]
Transition Holstein cows60 g/day rumen-protected choline (ReaShure®)Reduced inflammatory gene expression in follicular cells; supported a healthier follicular environment during early postpartum [71]
Ewes (periconceptional and late gestation)1.60 g/ewe/day rumen-protected cholineReduced embryonic loss; increased lamb birth weight; improved maternal antioxidant status [72]
Lactating women930 mg/dayIncreased concentrations of PEMT-derived choline metabolites in breast milk; enhanced infant choline supply [73]
Pregnant women550 mg/dayIncreased maternal plasma choline metabolites; promoted fetal phospholipid synthesis via the PEMT pathway; supported maternal–fetal health [74]
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

Liu, W.; Zheng, X.; Yang, H.; Wang, Z. Choline-Mediated Regulation of Follicular Growth: Interplay Between Steroid Synthesis, Epigenetics, and Oocyte Development. Biology 2025, 14, 1220. https://doi.org/10.3390/biology14091220

AMA Style

Liu W, Zheng X, Yang H, Wang Z. Choline-Mediated Regulation of Follicular Growth: Interplay Between Steroid Synthesis, Epigenetics, and Oocyte Development. Biology. 2025; 14(9):1220. https://doi.org/10.3390/biology14091220

Chicago/Turabian Style

Liu, Wenfeng, Xucheng Zheng, Haiming Yang, and Zhiyue Wang. 2025. "Choline-Mediated Regulation of Follicular Growth: Interplay Between Steroid Synthesis, Epigenetics, and Oocyte Development" Biology 14, no. 9: 1220. https://doi.org/10.3390/biology14091220

APA Style

Liu, W., Zheng, X., Yang, H., & Wang, Z. (2025). Choline-Mediated Regulation of Follicular Growth: Interplay Between Steroid Synthesis, Epigenetics, and Oocyte Development. Biology, 14(9), 1220. https://doi.org/10.3390/biology14091220

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