Next Article in Journal
Effects of Four Weeks of Alternate-Day Fasting with or Without Protein Supplementation—A Randomized Controlled Trial
Previous Article in Journal
The New Orleans Food System and COVID-19: A Case Study in Strengthening Food System Resiliency to Facilitate Healthy Eating
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Nutrition-Based Paternal Influence on Gynecological Diseases in Female Offspring via Epigenetic Mechanisms

by
Titilayomi J. Durojaye
1,
Sebanti Ganguly
1,
Yuanyuan Li
2 and
Trygve O. Tollefsbol
1,3,4,5,*
1
Department of Biology, University of Alabama at Birmingham, Birmingham, AL 35294, USA
2
Department of Nutrition and Food Science, University of Maryland, College Park, MD 20742, USA
3
O’Neal Comprehensive Cancer Research, University of Alabama at Birmingham, Birmingham, AL 35294, USA
4
Nutrition Obesity Research Center, University of Alabama at Birmingham, Birmingham, AL 35294, USA
5
Comprehensive Diabetes Center, University of Alabama at Birmingham, Birmingham, AL 35294, USA
*
Author to whom correspondence should be addressed.
Nutrients 2025, 17(23), 3690; https://doi.org/10.3390/nu17233690
Submission received: 20 October 2025 / Revised: 19 November 2025 / Accepted: 20 November 2025 / Published: 25 November 2025
(This article belongs to the Section Nutrition in Women)

Abstract

Studies have widely indicated that the composition of maternal nutrition and diets might affect offspring health later in life. Studies on paternal contribution to the offspring’s disease are relatively scarce but are an important subject to the field. Recent research has suggested that paternal factors influenced by nutrition have been implicated in the transgenerational heritage of health and diseases through epigenetic mechanisms. This review aims to explore the current state of knowledge on nutrition-based paternal impacts on gynecological disease through epigenetics, focusing on the transmission of cancer and metabolic diseases from father to female offspring. We will explore the various mechanisms by which epigenetic landmarks, such as DNA methylation, histone modifications, and non-coding RNAs, are passed on through sperm and reprogrammed in the embryo, influencing offspring development and health. We will discuss the impacts of preconception paternal nutrition on two common cancer such as breast cancer and ovarian cancer in female offspring. Additionally, paternal overweight or obesity has been associated with increased risk of obesity in the offspring and compromised metabolic health, which may link to reproductive conditions such as infertility. Understanding the molecular mechanisms underlying non-genetic inheritance is crucial for elucidating the nutrition-mediated developmental origins of health and disease. This review highlights the mechanistic correlation between preconception paternal nutrition and female offspring gynecological health. Furthermore, it emphasizes the need for additional research to establish evidence-based paternal nutrition consultation and guidelines aimed at optimizing reproductive health and pregnancy outcomes in couples planning to conceive.

Graphical Abstract

1. Introduction

Epigenetics has a compounded role in cancer, affecting different facets of tumor biology [1,2]. Processes such as histone modification, DNA methylation, nucleosome remodeling, and non-coding RNA expression play pivotal roles in many biological mechanisms essential for cancer development that alter gene expression without changing the DNA sequence itself [3,4]. Also, epigenetic mechanisms are key in modulating cell cycle and steering gene expression which are important factors in cancer development [5]. Epigenetic alterations contribute to the onward motion and spread of cancer by influencing several cellular functions [6]. For instance, they can impact how pathogens recognize receptors or lead to mis-regulation of imprinted genes which promote tumor development by allowing uncontrolled cell growth [6,7]. Ovarian cancer is among the most fatal and common gynecological malignancies. It is distinguished by challenges in early diagnosis, high recurrence rates, and resistance to existing treatment [8,9]. The tumor microenvironment plays a vital role in its growth and metastasis, with peritoneal metastasis being facilitated by the interaction between tumor cells and other cells such as tumor-associated macrophages [10,11]. Hormonal factors, specifically estrogens, are implicated in its development and play a role in its tumor biology [12]. Recent advancements in therapies include targeting these macrophages and employing immunotherapy and ferroptosis, a form of programmed cell death showing promise in suppressing tumor growth [13,14]. Breast cancer is a prime global health issue and the most common cause of cancer deaths among women [15]. Many risk factors contribute to its development including age, genetics, family history, lifestyle, and hormonal factors [15,16]. Breast cancer shows a complex molecular biology with signaling pathways like ER, HER2, and Wnt playing essential roles in its progression [17]. Gene mutations such as BRCA1 and BRCA2 significantly elevate risk levels, highlighting the need for targeted genetic therapies [17].
The outstanding nature of nutrition-mediated paternal epigenetic impact on the health and development of offspring is becoming extensively acknowledged and is now seen as a critical element in transgenerational inheritance [18,19,20]. Environmental exposure, lifestyle factors such as diet, stress, toxicants, and father’s age can affect the epigenome of sperm cells [21,22,23] which can then influence the development of offspring through epigenetic mechanisms, like histone modification, DNA methylation, and non-coding RNAs [24,25,26]. Importantly, paternal nutritional choices in the diet have been shown to have effects on sperm which can affect the health of the offspring [27].
Traditionally, research has focused on maternal influence on offspring, but recent evidence indicates that paternal factors also substantially alter offspring traits through epigenetic processes [18,19]. This review focuses on the role of paternal dietary factors in influencing breast and ovarian cancer risk in offspring through epigenetic mechanisms. We explore how paternal intake of macro- and micronutrients, as well as bioactive compounds, can induce epigenetic changes in sperm DNA methylation, histone modifications, and non-coding RNAs. By synthesizing current evidence and identifying knowledge gaps, we aim to stimulate further investigation into the transgenerational effects of paternal diet on female cancer risk in offspring and potential interventions to lower these risks.

2. Mechanisms of Paternal Epigenetics and Its Role in Gynecological Cancers

2.1. DNA Methylation, Paternal Transmission, and Nutritional Influence

DNA methylation is crucial in the development and progression of cancer (Table 1), acting as an epigenetic modification that impacts gene expression and stabilizes the genome [28,29]. Aberrant methylations, such as hypermethylation of tumor suppressor genes and hypomethylation of oncogenes, are essential for the survival of abnormal cells and responsible for cell cycle, apoptosis, proliferation, drug resistance, metastasis, and intracellular signaling [30,31,32]. Paternal transmission of DNA methylation changes can influence cancer susceptibility in offspring, and the diet and body composition of fathers can modify male germline epigenetically, which affects the prospect of breast cancer in their daughters [33,34]. DNA methylation also plays a vital role in the growth and progression of ovarian cancer by affecting the expression of critical genes. Studies show that aberrant DNA methylation, characterized by global hypomethylation and region-specific hypermethylation, is common in tumor cells. Methylation changes can lead to chromosomal instability and inactivation of tumor suppressor genes, thereby contributing to ovarian cancer [35,36,37]. Methylation patterns of specific non-X-linked promoter CpG islands (CGIs) differ between tissues and their implications in normal development and cancer [38]. Some CGIs are heavily methylated in normal somatic tissues but unmethylated in germline cells, leading to gene silencing in somatic tissues while these genes are expressed in testis and sperm. These genes include ANKRD30A, FLJ40201, INSL6, SOHLH2, FTMT, C12orf12, and DPPA [38].
In cancer, these genes often lose their methylation and become abnormally expressed, with the extent of this hypomethylation varying among different genes in cancer cell lines [39,40]. DNA methyltransferase inhibitors can reactivate these silenced genes in cancer cells, and the hypomethylation and expression of these genes in cancer may trigger an immune response.
Additionally, other types of abnormal methylation in cancer include de novo methylation by DNMT3A/3B enzymes, and mutations in DNMT3B are associated with certain conditions [41,42]. DNA methylation plays a critical role in determining nucleosome occupancy, particularly in the 5′-CpG islands of tumor suppressor genes, demonstrating a bidirectional relationship where hypermethylated CpG islands are tightly linked to nucleosome presence [39,40]. Inducing DNA hypomethylation through genetic or pharmacological approaches leads to nucleosome eviction from previously hypermethylated CpG islands of tumor suppressor genes. This process is reversible, as demonstrated by the reoccupation of nucleosomes in de novo methylated CpG islands upon restoration of DNA methyltransferase activity [40,43]. Genomically, DNA hypermethylation and dense nucleosome occupancy consistently correlate with gene silencing, while hypomethylation and nucleosome eviction correspond to gene expression [40,44]. In cancer, promoter CpG island hypermethylation of tumor suppressor genes is a common epigenetic feature associated with transcriptional silencing (Figure 1), suggesting a link between hypermethylation of specific genes and cancer development [45,46].
Table 1. Role of DNA methylation and their functional consequences in cancer.
Table 1. Role of DNA methylation and their functional consequences in cancer.
Type of MethylationFunctionEffectGenes AffectedReference
HypomethylationOccurs in tumor cells at the repetitive sequences residing in satellite regionsChromosome breakageRepetitive sequences in satellite or pericentromeric region[47,48]
HypomethylationThis leads to the activation of silenced genes and affects global DNA and specific genesGene activationSilenced genes can activate proto-oncogenes and destabilize the genome promoting cancer progression[47,49]
HypermethylationOften occurs at specific regulatory sites in the promoter regions or repetitive sequencesTumor specificityGenes involved in DNA repair and apoptosis, such as tumor suppressor genes[50,51]
HypermethylationThe heavy density of cytosine methylation in the CpG islands of the tumor suppressor gene promotersTranscription blockTumor suppressor genes[50]
Aberrant Promoter MethylationLeads to transcriptional silencing of tumor suppressors and metastasis inhibitor genesMalignant and metastastic phenotypeTumor suppressor[51]

2.2. Histone Modifications in Paternal Transmission

Histone modifications (Table 2) have been implicated in carcinogenesis and also play a significant role in the regulation of gene expression and are primarily localized at the amino-terminal and carboxy-terminal tails of histones [52]. One of the extensively studied modifications is histone acetyltransferase (HATs), which catalyzes the addition of acetyl groups and promotes an “open” chromatin conformation, which means enhancing accessibility to transcriptional machinery. Histone deacetylases (HDACs) [53], which catalyze the removal of acetyl groups, result in chromatin compaction and transcriptional repression, in which, when aberrantly expressed, they can result in various hematological malignancies and solid tumors [54,55,56]. Furthermore, specific histone modifications can serve as a prognostic measure of cancer and the development of histone deacetylase (HDAC) inhibitors can alter the acetylation state of various proteins [57,58], such as histones, thereby influencing gene expression and potentially exerting anti-tumor effects [59,60,61]. Lower global levels of histone modifications have been correlated with more aggressive cancer subtypes in breast cancer [62,63]. The overexpression of HDAC2 is prevailing in breast cancer and may be a factor in the abnormal histone acetylation patterns discerned in malignancy [64]. Generally, reduced levels of histone modification such as H3K4me2 and H3K18ac are associated with defective prognosis across various types of cancer, including breast cancer [65,66,67]. A study emphasizes that alterations in histone modifications contribute to ovarian carcinogenesis by silencing tumor suppressor genes and facilitating the expression of oncogenes [68]. In ovarian cancer, histone modifications are being targeted to develop therapeutic interventions. Histone deacetylase inhibitors and other epigenetic therapies have shown potential in altering disease progression and improving outcomes when used in combination with other therapies [69,70]
Epigenetics changes in the germline, specifically in sperm, are now recognized as primary intermediaries in facilitating epigenetic transgenerational inheritance, which includes the transmission of environmentally induced epimutations to future generations without direct exposure [25]. In sperm, the active mark H3K4me3 and the repressive mark H3K27me3 have attracted significant attention. Research in mouse models reveal that overexpressing the histone demethylase KDM1A in the male germline can cause widespread loss of H3K4me3 at transcription start sites of developmental genes in sperm, leading to infertility, developmental abnormalities, and traits that persist for at least two subsequent generations [71,72]. Simultaneously, rodent studies show that while vinclozolin or DDT exposure usually leaves H3K27me3 levels largely unchanged, they prompt differential histone retention sites (DHRs) in the F3 generation, demonstrating that histone retention, rather than modification, may influence certain transgenerational contexts, with exposure-specific patterns [73,74]. Remarkably, H3K4me3 modifications in sperm bypass post-fertilization epigenetic reprogramming, remain in the early embryo, and act on genes involved in morphogenesis, metabolism, and pathways often dysregulated in cancer. Another contributing factor is the histone demethylase Kdm6a (Utx), a potential tumor suppressor, whose deletion in the paternal germline develop to extensive H3K27me3 redistribution, enhancer hypermethylation, and altered transcription factor binding in offspring somatic tissues, correlating with increased tumor incidence and reduced survival effects that are amplified by successive generations of Kdm6a loss [75].
Table 2. Major histone modification and their epigenetic roles.
Table 2. Major histone modification and their epigenetic roles.
Histone ModificationDescriptionEpigenetic RoleLocationReference
H3 lysine 9 methylation (H3K9me3)The addition of three methyl groups to the 9th lysine residue of histone H3Associated with transcriptional silencing by recruiting HP1 (heterochromatin protein 1) to initiate and maintain heterochromatin formationConstitutive heterochromatin[76]
H3 Lysine 4 methylation (H3K4me3)The addition of three methyl groups to the 4th lysine residue of histone H3Associated with transcriptional activation and euchromatic regionsPromoter regions of active genes[77]
H3 Lysine 27 methylation (H3K27me3)The addition of three methyl groups to the 27th lysine residue of histone H3.Associated with transcriptional repression particularly through Polycomb (Pc) group proteinGene repressed by Polycomb group proteins[78,79]
Histone acetylationHistone acetyl transferases (HATs) add acetyl groups to histone tailsFacilitates gene transcription [76]
DeacetylationHistone deacetylases (HDACs) remove acetyl groups from histone tailsInhibits gene transcription [80,81]
MethylationHistone methyltransferases add methyl groups to histone tailsRegulation of gene expression [80,81]
PhosphorylationKinases add phosphate groups to histone tailsRegulation of gene expression, DNA repair, and chromosome condensation [81]

2.3. Non-Coding RNAs (ncRNAs) in Paternal Transmission

The concept of paternal inheritance of disease risk via epigenetic imprinting is supported by several historical epidemiological studies of human cohorts from famine-affected regions worldwide. Such epigenetic imprints have been noted to be often sex-specific and pervasive through multiple generations [82,83]. During embryogenesis, parental DNA methylation marks undergo genome-wide reprogramming involving sequential erasure and re-establishment. While this reestablishment partially conserves parental methylation patterns, biparental inheritance and developmental programming generate a distinct methylomic landscape in the offspring that retains selective parental epigenetic signatures while establishing novel, lineage-specific methylation profiles [84].
This phenomenon necessitates investigation of epigenetic inheritance mediated by small non-coding RNAs (ncRNAs), including microRNAs (miRNAs), PIWI-interacting RNAs (piRNAs), tRNA-derived fragments (tRFs), long non-coding RNAs (lncRNAs), and similar regulatory RNA species that potentially escape global reprogramming and transmit epigenetic information across generations (Table 3). An emerging body of evidence supports the hypothesis that paternal exposure to environmental chemicals [85], psychosocial stressors [86], and dietary factors [87] directly influences epigenetic modifications in offspring, often at embryonic level via epigenetic reprogramming, potentially altering disease susceptibility, including cancer. Research in this area remains limited, with even fewer studies specifically investigating cancer-related mechanisms. In a 2018 study, da Cruz et al. demonstrated that low protein diet resulted in increased cancer risk in next generation female offsprings that is mainly mediated by aberrant modulation of small ncRNAs such as miR-28a, miR-92a, miR-200c, miR-451a, miR-191, miR-15b, and sperm-specific small RNA variants- tRF5-Gly-CCC, tRF5-Val-TAC [87]. Mounting evidence has shown that a vast majority (~60%) of protein coding mRNAs in humans have conserved seed sequence complementary to miRNAs [88], which are one of the common factors passed on to next generations through sperm. These ncRNAs are responsible for regulating expression levels of maternally contributed mRNA within a zygote [89]. Importantly, only a selected fraction of ncRNAs generated during spermatogenesis within the seminiferous tubules, and not the epididymis, becomes incorporated into the mature spermatozoon and subsequently transmitted to the next generation. This selective packaging occurs due to the highly specialized architecture of the sperm head, which undergoes extensive cytoplasmic reduction and chromatin compaction, limiting the retention of RNA species to those specifically associated with the condensed paternal genome or residual cytoplasm [85]. In a small cohort study conducted by Vaz et al. in 2021 [90], it was revealed that even a short-term change in the intake of micronutrients such as vitamin D, olive oil, and omega-3 fatty acids over 6 weeks can significantly modulate the expression of 112 piRNAs, 8 different categories of tRFs, and 15 miRNAs. These in turn dysregulate the expressions of genes downstream such as ACAA2, ACSL1, CPT1A, ELOVL5, HADH, OXSM, PECR, and SCD [90]. In other independent studies the genes ACAA2, ACSL1, CPT1A, ELOVL5, HADH, OXSM, PECR, and SCD have shown to sustain hallmarks of ovarian and breast cancer [91,92,93,94,95,96,97]. Such piRNAs include piR-004054, piR-004656, and piR001152, miRNAs include miR-513c-3p, mir-136-3p, and miR-4760-5ap, and tRFs include trf5b-TyrGTA, tir5-CysGCA, and trf5b-AlaAGC [91,92,93,94,95,96,97].
Table 3. Paternal microRNAs (miRNAs) related to breast cancer risk in vivo (mouse). Upward arrows indicate an increase, and downward arrows indicate a decrease. ** These genes have been found to be relevant in the context of stress response. These genes are also relevant to cancer progression. ‡ The corresponding cited study includes comprehensive catalog of ncRNAs relevant to cancer. Only a few relevant to cancer have been enlisted here.
Table 3. Paternal microRNAs (miRNAs) related to breast cancer risk in vivo (mouse). Upward arrows indicate an increase, and downward arrows indicate a decrease. ** These genes have been found to be relevant in the context of stress response. These genes are also relevant to cancer progression. ‡ The corresponding cited study includes comprehensive catalog of ncRNAs relevant to cancer. Only a few relevant to cancer have been enlisted here.
Differentially Regulated Paternal miRNAExpressionTarget Pathway/mRNACauseOrganismReference
miR-28a, miR-92a, miR-200c, miR-451a, miR-191, and miR-15bAMP-activated protein kinase pathway (Prkaa2, Cab39), mammalian target of rapamycin (mTOR) signaling pathwayPaternal malnutrition, low protein dietMouse[87]
miRNA-1896, miRNA-874 and miRNA-296-5pHypoxia signaling, insulin receptor signaling, NANOG pathway, CDK5 signaling, epithelial–mesenchymal transition pathway, ERK/MAPK pathway, SAPK/JNK signaling, estrogen receptor signaling, April mediated signaling, axonal guidance signalingPaternal obesityMouse[33]
miR-29c, miR-30a, miR-30c, miR-32, miR-193-5p, miR-204, miR-375, miR-5323p, and miR-698Sirt1, Ube3a, Srsf2, IL6st, Ncl, Aara, Agfg1, and Ralbp1 **Chronic paternal stressMouse[89]
miRNA- let-7d-5p, miR-10a-5p, miR-138-1-3p, miR-221-3p, miR-222-3p ‡ Exposure to toxicants (herbicide- Vinclozolin, fungicide, jet fuel, pesticide-DDT)Rat[85]
miR-30c, miR-30e, miR-124, miR-145, miR-361,miR-762 ‡Apoptosis, myogenesis, tumor suppression, immune responsePaternal exposure to radiationMouse[98]
miR-29c, miR-134, miR-181a ‡Bax, Bcl, PTEN, stem cell survival
Differentially regulated paternal tRFExpressionTarget pathway/mRNACauseOrganismReference
tRF-Gly-GCC, tRF-Gly-CCC, tRF-Val-CAC, tRF-Gly-TCC, tRF-Lys-CTT, and tRF-His-GTGDub3, Ddr2, Tcstv3Paternal low protein dietMouse[99,100,101]
tRF5-Gly-CCC, tRF5-Val-TAC, tRF5-Pro-AGG and tRF5-Ser-CGAWnt/β-cateninPaternal low protein dietMouse[87]
tRF5-Ile-TAT, tRF5-Arg-ACG, and tRF5-SeC-TCA
tRNA-Pro-AGG-1-2, tRNA-Pro-TGG-1-4, tRNA-Pro-AGG-1-M8↓ (Potentially)Trim7, Ccdc136Exposure to toxicants (herbicide- Vinclozolin, fungicide, jet fuel, pesticide-DDT)Rat[85,102,103]

2.4. RNA Modification

Spermatozoa contribute not only to genetic DNA and retained histones but also to diverse modified RNAs, specifically small tRNA fragments (stRNAs) which are rich in chemical marks such as 5-methylcytidine (m5C) and N2-methylguanosine [21]. Beyond histone changes, DNA methylation, and small non-coding RNAs, RNA modification such as epitranscriptome represents a significant aspect of paternal epigenetic inheritance [21]. These modifications elevate RNA stability and modify their regulatory potential during the early stages of embryonic development. Study indicates that paternal nutritional stress, including diets high in fat or low in protein, alters both the quantity and modification profiles of sperm stRNAs. Strikingly, injecting stRNAs from diet-affected males into control zygotes can induce metabolic dysfunction in the F1 generation, showing a direct role for epitranscriptomic signals in paternal inheritance. Further insights from C. elegans reveal that sperm-specific paternal epigenetic inheritance (PEI) granules—containing Argonaute proteins like WAGO-3 bound to modified 22G-RNAs—are required for passing on epigenetically programmed responses across generations [104]. The conservation of PEI-like proteins (e.g., human BTBD7) suggests that comparable RNA-modification-dependent pathways may also exist in mammals [104].
Epitranscriptomic regulation play a vital role in gynecology-related cancer risk, especially in breast cancer, where the imbalance of RNA “writers” (e.g., METTL3), “readers” (e.g., YTHDF1/3), and “erasers” (e.g., FTO, ALKBH5) contribute to tumor development, cancer stem cell maintenance, metastasis, and resistance to treatment [105]. For example, overexpression of METTL3 enhances cell proliferation and resistance to chemotherapy, while ALKBH5-driven demethylation of pluripotency genes promotes breast cancer stemness. Remarkably, FTO, an m6A/m6Am RNA demethylase initially identified through its association with obesity, is associated with breast cancer-related SNPs and mechanistically connects metabolic status with cancer susceptibility [105]. Since YTHDC2 and ALKBH5 also regulate spermatogenesis, m6A modification is positioned at the crossroad of germline integrity, fertility, and transgenerational epigenetic communication. Dietary exposures like high-fat diets, miRNAs from bovine milk exosomes, and altered glucose metabolism can affect the expression or activity of epitranscriptomic regulators such as METTL3 and FTO [105,106]. This creates a conceptual framework in which paternal diet-induced RNA-modification changes in sperm and obesity-linked epitranscriptomic dysregulation in women coincide on shared RNA-based mechanisms that influence reproductive health and gynecological cancer risk [107].

2.5. Epigenetics Regulations in Breast and Ovarian Cancer

In ovarian cancer (OC), the disruption of histone acetylation—regulated by the balance between histone acetyltransferases (HATs) and histone deacetylases (HDACs)—is significant, with 37 out of 40 acetylation-related genes showing different expression levels in OC compared to normal tissues. An eight-gene histone acetylation signature (SIRT5, BRD4, OGA, SIRT2, HDAC4, NCOA3, HDAC1, and HDAC11) has been identified as an independent prognostic marker, where seven genes are linked to poorer outcomes, while SIRT5 is associated with a favorable prognosis [108]. These changes affect pathways like Wnt signaling, leading to platinum resistance and reduced anti-tumor immunity. Specific disrupted “histone codes” in OC include OGA, an O-GlcNAcase that modulates histone acetylation indirectly via O-GlcNAcylation crosstalk (influences p53 stability), NCOA3 (a co-activator linked to platinum resistance), BRD4 (a reader that enhances transcription and can be targeted by inhibitors), and erasers such as SIRT2, HDAC1, HDAC4, and HDAC11, whose altered activities influence drug sensitivity, tumor progression, and survival [108]. Similar disruptions occur in breast cancer, where NCOA3, OGA, HDAC1, and HDAC11 exhibit cancer-type-specific prognostic effects. Dietary compounds such as curcumin have been shown to affect histone acetylation [109]. Curcumin, a component found in turmeric, exerts protective effects against cancer by activating tumor suppressor genes and inhibiting oncogenes [109]. Overall, these findings underscore that abnormal histone acetylation and methylation in germline and somatic cells not only contribute to ovarian cancer pathogenesis but may also serve as inheritable risk factors and actionable therapeutic targets [108]. H3K4me2 and H3K27me3 play a part in transgenerational inheritance by preserving the developmental gene expression state in sperm [110,111]. This involves Trithorax MLL family and Polycomb (PRC2/EZH2) functioning as writers, EED acting as a reader, and LSD1/KDM1 as an eraser. Dysregulation of these marks can elevate the risk of cancer in offspring [112]. In breast cancer, altered histone codes are essential to tumor initiation, progression, resistance to therapy, and metastasis. Writers (HATs, HMTs like CARM1, and the MLL family) add activating or repressive marks, readers (e.g., PELP1 recognizing H3R17me2a/H3R26me2a) interpret these modifications and recruit co-regulators, and erasers (HDACs, KDM1/LSD1, HDAC3) remove marks to silence or reprogram transcriptional states [113,114] (Figure 2). Writers, readers, and erasers not only contribute to tumorigenesis within an individual but may also enable the heritage transmission of oncogenic across generations [115].

3. Nutrition Impacts on Sperm Epigenetics and Offspring’s Gynecological Cancers

Recent studies demonstrate that specific bioactive food compounds derived from obesogenic diets can cause stable yet dynamic epigenetics modifications in sperm which in turn affect offspring health and disease susceptibility. Epigenetic modifications such as DNA methylation, histone modification, and small non-coding RNAs are heritable through fertilization thereby supporting the Paternal Origins of Health and Disease (POHaD). Paternal undernutrition and overnutrition have been linked to breast cancer risk in daughters [116], and paternal obesity has been connected with enhanced susceptibility to breast cancer in daughters. Paternal malnutrition has also resulted in a greater occurrence of mammary cancer in female offspring, with these tumors materializing earlier and growing more rapidly compared to controls [87]. Emanated evidence shows that comparing unhealthy fast foods (e.g., fries, pizza) with nutrient dense diets like fruit, nuts, whole grains, and vegetables convey contrasting sperm epigenetic patterns. Regular ingestion of fast food is linked to altered methylation at imprinted gene loci (e.g., IGF2, MEG3-IG), lower sperm motility (directionality of methylation change vary by locus), and increased risk of transferring adverse metabolic phenotype to offspring [117]. Implied mechanisms include oxidative stress from high fat and carbohydrates, acrylamide exposure including DNA damage, trans-fatty acid accumulation impairing Sertoli cell lipid metabolism, and endocrine-disrupting chemicals like BPA, phthalates altering sperm methylation [117]. These paternal epigenetic alterations are connected to offspring predisposition to chronic metabolic disorders, cancer, obesity, diabetes, and cardiovascular dysfunction, even across multiple generations. Contrarily, consuming vegetables, whole grains, fruits, nuts, and polyunsaturated fatty acids is associated with positive sperm outcomes, such as reduced methylation at NNAT, IGF2, and MEG3 loci, higher total motile count, and improved semen volume [117,118]. Such provident diets amplify sperm epigenetic integrity, enhance reproductive success, and boost antioxidant defenses. This protective effect extends to offspring, leading to lower risk of metabolic dysfunction and better fetal growth [118].
Micronutrients and B vitamins, specifically folate, play a crucial role through one carbon metabolism and methyl group donation [119]. Folate deficiency in fathers alters sperm methylation and histone marks (H3K4, H3K9), leading to developmental abnormalities in offspring, placental defects, increased pregnancy loss, and a higher risk of chronic diseases such as cancer and diabetes [119,120]. Human studies also show that disruptions in folate metabolism, particularly due to MTHFR polymorphisms, are linked with idiopathic male infertility and abnormal sperm methylation [118,119]. Importantly, both folate deficiency and excessive supplementation can negatively influence the sperm epigenome and offspring outcomes, emphasizing the importance of balanced intake [118]. Animal and human studies also underline the effects of high fat diets and high sugar diets. High fat diets induced abnormal DNA methylation at imprinted genes, histone mark changes (H3K27me3), and altered sperm small ncRNAs such as miRNA, tsRNA, piRNAs which are transmitted to offspring, leading to glucose intolerance, insulin resistance, hypoandrogenism, obesity, and pro-inflammatory phenotypes across F1 and F2 generations. Similarly, short-term high sugar diet (HSD) alters sperm small non-coding RNA (sncRNA) composition, particularly mitochondria tsRNAs, linking paternal dietary sugar intake with sperm motility, oxidative stress, and potentially altered embryonic gene regulation [118,119,120].

3.1. Macronutrient on Sperm Epigenetics

Imbalanced paternal macronutrient intake such as diets high in fat and high sugar Western diet, low in protein, or excessive high protein processed meats (bodybuilder foods), along with high caloric intake and early dietary exposures play a vital role in influencing sperm epigenetics and offspring health (Figure 3). Studies show that dietary imbalances disrupt sperm through epigenetic mechanisms, impacting not only fertility but also the developmental trajectory and long-term disease risk of descendants. High sugar diet impairs sperm motility and fertilization, contributes to adverse embryonic and offspring phenotypes, and intensifies oxidative stress [121], which works in hand with high fat diet [121]. Hyperglycemia and high-glycemic load exposure aggravate ROS and epigenetic instability, linking diet-induced glucose dysregulation to male infertility and intergenerational metabolic issues [121]. Similarly, high fat diet consistently induces significant changes in sperm DNA methylation, including global hypomethylation and imprinting errors (notably at IGF2 DMRs—Differentially Methylated Region) [122], also affecting histone acetylation and the exchange of histones for protamine, leading to less condensed chromatin that is susceptible to oxidative damage. These diets alter small RNA content, tsRNAs, miRNAs, lncRNA, and RNA modifications such as m6A. Paternal high fat diets increase reactive oxygen species (ROS) and DNA fragmentation [123], alter the composition and microbiota of seminal plasma, and disrupt the blood–testis barrier [122,123]. The consequences range from reduced sperm motility [123] and viability to delayed embryo development, fewer blastocyst cells, and implantation failure [123]. In offspring, metabolic syndrome traits such as insulin resistance, β-cell dysfunction, and obesity manifest alongside reproductive subfertility, neurobehavioral changes like increased anxiety and impaired learning, cardiovascular disease, and elevated cancer susceptibility. Many of these outcomes persist into the F2 generation, supporting the concept of “inherited metabolic memory [124].”.
High protein diets dominated by processed red meat (e.g., bodybuilder meals) indirectly play a part in infertility by reducing sperm count, testicular volume, testosterone, and motility. Mechanisms likely involve endocrine disruption from fat residues, preservatives, and hormone contaminants, along with increased oxidative stress. Additionally, low protein diets affect genome-wide sperm DNA hypomethylation and disrupt one carbon metabolism, downregulating Dnmt1, Dnmt3L, and folate-cycle enzymes [123]. They alter sperm and seminal plasma tRNA content, impair spermatogonia stem cell populations, and lower serum testosterone. Through altered seminal plasma signaling, low protein diets influence uterine gene expression and early embryonic development, resulting in cardiometabolic dysfunction, hypertension [123], vascular abnormalities, smaller fetuses, stalled blastocyst development, and altered lipid metabolism in offspring [125]. Many of these effects are dependent, with male offspring particularly vulnerable, and some persist into the F2 generation.

3.2. Micronutrients and Sperm Epigenetics

Sufficient levels of micronutrients are increasingly recognized as critical for preserving the epigenetic integrity of sperm [126]. Deficiency in folate and other B vitamins such as B2, B6, and B12 as well as minerals like iron [119,126], magnesium, iodine, selenium, manganese, and zinc have been shown to affect paternal germline programming [127]. One-carbon vitamins are crucial for regulating the availability of S-adenosylmethionine (SAM), a universal methyl donor essential for DNA and histone methylation, meaning that a lack of folate or B12 deficiency can obstruct the methylation imprinted genes, global methylation balance, and spermatogenesis [128]. Animal studies have shown that paternal folate deficiency can alter sperm methylation patterns, reduce sperm count, and impact offspring with developmental issues such as growth restriction and higher metabolic risk [129]. Additionally, both low and excessively high folic acid intake have been associated with changes in seminal DNA methylation and altered embryonic growth trajectories in human studies [130,131]. Roles of these vitamins in sperm are shown in Table 4. Minerals influence various pathways, including chromatin stability, oxidative stress, antioxidant defense, and endocrine regulation. Calcium, magnesium, phosphorus, zinc, iron, and potassium are indispensable for spermatogenesis and sperm function. Potassium, alongside sodium, maintains osmotic balance and regulates ion channels critical for sperm motility and hyperpolarization, with imbalances impairing spermatogenesis [127]. Phosphorus, a fundamental component of nucleotides (AMP/ADP/ATP), drives energy transfer and supports DNA/RNA structure, linking deficiency to reduced fertility [127]. Calcium regulates capacitation, motility, and acrosome reaction, with both deficiency and excess impairing fertilization competence and increasing oxidative stress. Zinc, abundant in seminal plasma, aids in protamine cross-linking, antioxidant defense [132], and chromatin packaging, and deficiency in zinc can lead to low sperm count and motility, chromatin destabilization, and increased oxidative damage [129]. Notably, zinc supplementation can restore sperm DNA integrity and reduce negative metabolic programming in offspring, as observed in rodent models. Additionally, selenium is crucial for the activity of selenoproteins, such as glutathione peroxidase, which protects sperm DNA from reactive oxygen species (ROS) [132]. Importantly, a lack of selenium in fathers has been shown to alter mammary gland development and increase breast cancer risk in female offspring, thereby providing an example of a direct link between micronutrients, epigenetics, and cancer risk in offspring.
The significant limitations that are evident throughout all the references in Table 4 are that they lack well-controlled human trials and difficulties in applying results from animal studies to humans’ reproductive biology. Short-term interventions might enhance specific sperm or embryo parameters but often do not address the underlying metabolic dysfunction. Additionally, current genetic analyses often ignore compound allele interaction, and the ideal nutrient concentrations and supplementation thresholds are yet to be determined.

3.3. Influence of Paternal Obesity on Female Reproductive Health

Paternal nutrition, especially when it leads to obesity, has significant implications for female infertility and the broader reproductive health of future generations (Figure 4). Increasing evidence has highlighted the role of paternal obesity in influencing offspring health, not only through genetic inheritance but also via epigenetic and environmental factors [136]. Obesity can alter sperm quality through different pathways such as hormonal imbalances, oxidative stress, and epigenetic modifications [137]. These factors can lead to low sperm viability [138] and DNA damage, contributing to developmental issues in offspring and indirectly affecting female fertility [130,137]. Reduced sperm quality due to obesity can impair fertilization processes, leading to suboptimal embryonic development and higher miscarriage rates [139,140]. Animal studies have shown that paternal obesity, often induced by a high-fat diet (HFD), affects the reproductive health of at least two subsequent generations. In mice, obesity in fathers led to diminished reproductive functions in both male and female offspring over two generations, suggesting a transgenerational impact of paternal metabolic state on fertility [141]. The offspring of obese fathers exhibit metabolic and reproductive challenges, highlighting the importance of paternal health at the time of conception [141]. Additionally, paternal obesity can cause endocrine disruptions that alter luteinizing hormone and testosterone levels, intensifying reproductive difficulties in female offspring when exposed to a similar high-fat environment after birth [142], obesity-related hormonal imbalance, such as hypogonadotropic hypogonadism [142], and elevated pro-inflammatory markers in semen, including IL-8 [137], may further impair the hypothalamic–pituitary–ovarian axis, a critical regulator of female reproductive health. In addition, obesity-associated changes in seminal fluid composition and sperm DNA methylation can further compromise reproductive outcomes [142]. Studies in rodents reveal that a father’s metabolic condition can reprogram the zygote by altering sperm DNA methylation, histone modifications, and small non-coding RNAs (miRNAs, tsRNAs) [117,143], along with causing oxidative DNA damage [144], hence influencing developmental pathways [145,146]. In female offspring (F1), these paternal influences result in impaired oocyte quality [147], diminished meiotic competence, elevated ROS, delayed embryo development (at the 2- and 8-cell stages and caused lower blastocyst quality) [130,147]. Additional alterations include disrupted ovarian metabolism-characterized by increased lipid accumulation and GLUT4-expression in cumulus-oocyte complexes-and mitochondria/redox imbalances such as reduced mtDNA copy number, downregulated PGC-1α/TFAM/NRF1 and AMPK [148], and diminished TAC and Nfe2l2/NRF2 activity, alongside decreased miR-149 expression [130].
Exercise, nutrition, and lifestyle changes have been proposed to mitigate the negative effects of paternal obesity. For instance, engaging in exercise prior to conception has shown potential in improving sperm quality and, subsequently, offspring health [130]. Therefore, addressing paternal obesity through nutrition management may enhance reproductive outcome and aid in managing infertility issues not only in women but also in future generations.

3.4. Phytochemicals Influence Sperm Epigenetics

Dietary phytochemicals consumed by fathers, particularly bioactive compounds derived from plants like sulforaphane found in broccoli sprouts (SFN), epigallocatechin-3-gallate (EGCG) from green tea polyphenols, carotenoids, flavonoids, and resveratrol influentially impact sperm health (Figure 5), epigenetic programming, and the cancer susceptibility of their offspring [149,150]. Research has shown that when the father consumes SFN-rich broccoli sprouts and EGCG-rich green tea polyphenols, there is significant suppression of estrogen receptor-negative (ER-) mammary tumor development in female offspring, as seen in transgenic mouse models [149]. This protective effect is synergistic when these compounds are combined, leading to a lower tumor incidence, smaller tumor size, and delayed tumor latency [141]. These effects are driven by germline epigenetic inheritance, where the paternal diet alters sperm molecular profiles, transmitting these changes to the next generation. In sperm, the presence of SFN and EGCG from the paternal diet altered the transcriptome, with 271 genes showing differential expression, including those linked to spermatogenesis and breast cancer progression, and 467 differentially methylated regions (DMRs) associated with cancer-related pathways. Notably, changes in sperm DNA methylation alone did not fully account for gene expression regulation, suggesting a complex interaction of multiple epigenetic markers [150,151]. These modifications were mirrored in the mammary tumors of offspring, where paternal phytochemicals reduced HDAC enzymatic activity, elevated global H3K4 methylation, decreased H3K27 methylation, and increased global DNA 5-methylcytosine levels [149]. Tumor suppressor proteins (p16, p53) were upregulated, while oncogenic HDACs (HDAC1, HDAC3, HDAC8) and BMI1 were downregulated [149].
The Mediterranean diet is a model “epigenetic diet,” rich in whole grains, vegetables, fruits, legumes, olive oil, fish, and polyphenols, with proven inverse associations with cancer, cardiovascular, and metabolic diseases. Beyond SFN and EGCG, nutraceutical components such as curcumin, lycopene, quercetin, and resveratrol modulate DNA methylation, miRNA expression [150], and histone acetylation, functioning as neutral epigenetic regulators that suppress oncogenes, reactivate tumor suppressor genes, and attenuate inflammation and oxidative stress [152]. Additionally, it has been shown that quercetin, rutin, genistein, luteolin, apigenin, hesperetin, morin, daidzein, anthocyanidins, and resveratrol have positive effects on male reproductive health and sperm integrity, specifically under environmental pollutant stress [152]. These compounds exert their effects through antioxidants, chelation, anti-inflammatory, and epigenetic pathways. Epidemiological evidence demonstrates that adherence to the Mediterranean diet reduces cancer mortality and incidence. Paternal diet can also counteract the transgenerational carcinogenic risks posed by endocrine disruptors and pollutants, thereby preserving male fertility and potentially reducing cancer risk in offspring [150,151,152].

4. Conclusions

The review presented here indicates that paternal epigenetic inheritance plays a critical, though often overlooked, role in influencing breast and ovarian cancer risk in offspring. The paternal diet, particularly intake of macro- and micronutrients as well as bioactive compounds, can induce epigenetic changes in sperm DNA methylation, histone modifications, and non-coding RNAs. These alterations in sperm are capable of bypassing reprogramming during embryonic development and influence gene expression patterns in female offspring, potentially modulating cancer susceptibility. Key findings include the impact of paternal folate deficiency on sperm DNA methylation, the role of paternal nutrition in increasing breast cancer risk in daughters, and the protective effects of paternal intake of phytochemicals like sulforaphane against mammary tumors in offspring. Overall, this review underscores the need to consider paternal nutrition as an important factor in female cancer prevention strategies.

5. Limitation and Future Directions

One major limitation of the current research in this field is the reliance on animal models, particularly rodents, for many of the mechanistic studies. While these models provide valuable insights, the translation of findings to humans requires caution due to differences in reproductive biology and epigenetic reprogramming between species. Additionally, most human studies are observational and cannot establish causality. The long-term nature of transgenerational effects also poses challenges for conducting controlled human studies. Furthermore, while maternal factors have been extensively studied, the impact of paternal lifestyle and nutrition in modulating gynecological cancer susceptibility across generations is an emerging area that requires further exploration.
Future research should focus on validating findings from animal models in human cohorts through long-term epidemiological studies and clinical trials. Mechanistic studies are needed to show how specific dietary components influence sperm epigenetic marks and how these are maintained during embryonic reprogramming. Advanced sequencing technologies should be employed to comprehensively profile the sperm epigenome and identify robust biomarkers of paternal dietary exposures. The potential for dietary interventions in men to reduce gynecological cancer risk in offspring warrants further investigation. Additionally, the interaction between paternal and maternal dietary factors in modulating offspring cancer risk should be explored. Finally, the development of non-invasive methods to assess sperm epigenetic profiles could facilitate larger-scale human studies and potentially inform personalized preconception care strategies aimed at reducing breast and ovarian cancer risk in future generations.

Author Contributions

T.J.D. conceived of the review paper with the guidance of T.O.T. The manuscript was drafted and edited by T.J.D., S.G., Y.L. and T.O.T. All authors have read and agreed to the published version of the manuscript.

Funding

This work was supported in part by a grant from the National Institute of Health (NCI R01CA178441) to TOT and the United States Department of Agriculture, National Institute of Food and Agriculture (USDA NIFA, 2021-67017-39063 to YL).

Data Availability Statement

No new data were created or analyzed in this study.

Conflicts of Interest

The authors declare no conflicts of interest.

Abbreviations

ANKRD30AAnkyrin Repeat Domain 30
ACAA2Acetyl-CoA Acyltransferase
ACSL1Acyl-CoA Synthetase Long Chain Family Member 1
Agfg1ArfGAP with FG repeats 1
AMPKAdenosine Monophosphate-activated Protein Kinase
BRCA1 and BRCA2Breast Cancer Susceptibility gene
BaxBCL2-Associated X
BclB Cell Lymphoma Protein
BMI1Polycomb Ring Finger 1
BPABisphenol A
BRD4Bromodomain Containing 4
Ccdc136Coiled-Coil Domain Containing 136
CARM1Coactivator-Associated Arginine Methyltransferase 1
CpGCytosine Phosphate Guanine
CGICpG Island
CDK5Cyclin-Dependent Kinase 5
CPT1ACarnitine Palmitoyltransferase 1A
Cab39Calcium Binding Protein 39
C12orf12Coiled-coil Glutamate Rich Protein 1
DDTDichlorodiphenyltrichloroethane
DHRsDifferential Histone retention
DMRDifferentially Methylated Regions
DPPADevelopmental Pluripotency Associated gene
DNMTDNA Methyltransferase protein
EGCGEpigallocatechin Gallate
ELOVL5Fatty Acid Elongase 5
ERKExtracellular Regulated Kinase
EREstrogen Receptor
EZH2Enhancer of Zeste Homolog 2
FLJ40201Ciliary Microtubule-associated Protein 2
FTMTFerritin Mitochondrial gene
GLUT4Glucose Transporter 4
GPx-1Glutathione Peroxidase 1
HER2Human Epidermal Growth Factor 2
HADH3-hydroxyacyl-CoA Dehydrogenase
HATHistone Acetyltransferase
HMTHistone Acetyltransferase
HDACHistone Deacetylase
H3K4me2Histone 3 lysine 4 dimethylation
H3K18acHistone 3 lysine 18 acetylation
H3K27me3Histone 3 lysine 27 trimethylation
H3K9meHistone 3 lysine 9 methylation
HFDHigh Fat Diet
IGF2Insulin-like Growth Factor 2
INSL6Insulin Like 6 gene
IL6stGlycoprotein 130 (includes Interleukin 6)
IL8Interleukin 8
JNKc-Jun N-terminal Kinase Family Protein
KDM1/LSD1Lysine-specific Demethylase 1
Kdm6aLysine-specific Demethylase 6A
lncRNALong non-coding RNAs
miRNAsMicroRNA
mTORMammalian Target of Rapamycin
MAPKMitogen Activated Kinase-like Protein
MEG3Maternally Expressed 3 (long non-coding RNA)
MEG3-IGMaternally Expressed Gene 3-intergenic Differentially Methylated Region
m6AN6-methyladenosine
MLLMixed-lineage Leukemia
MTHFRMethylenetetrahydrofolate Reductase
NANOGHomeobox Transcription Factor
NNATNeuronatin
NclNucleolin
NCOA3Nuclear Receptor Coactivator 3
NRF1Nuclear Respiratory Factor 1
Nfe2l2Nuclear Factor Erythroid 2-related Factor 2
ncRNANon-coding RNA
OGAO-GlcNAcase protein
OXSM3-oxoacyl-ACP Synthase
PRC2Polycomb Repressive Complex 2
piRNAPIWI-interacting
PECRPeroxisomal Trans-2-Enoyl-CoA Reductase
PTENPhosphatase and Tensin Homolog
PI3KPhosphoinositide 3-Kinase
PRProgesterone Receptor
PR+Progesterone Receptor-Positive
Prkaa2Alpha-2 Catalytic Subunit of AMP-activated Protein Kinase
PELP1Proline-, Glutamic acid-, and Leucine-rich Protein 1
PGC-1αPeroxisome Proliferator-activated Receptor-gamma Coactivator 1-alpha
POHaDPaternal Origins of Health and Disease
RNAPIIRNA Polymerase II
Ralbp1RalA Binding Protein 1
ROSReactive Oxygen Species
SOHLH2Spermatogenesis and oogenesis specific basic helix-loop-helix 2
SAMS-adenosylmethionin
SAPKStress-Activated Protein Kinase
SCDStearoyl-CoA Desaturase
SirtSirtuin
SFNSulforaphane
Srsf2Serine and arginine rich splicing factor 2
TFAMMitochondrial Transcription Factor A
tRFstRNA-derived fragments
tsRNAtRNA-derived small RNAs
tRNATransfer RNA
TACTotal Antioxidant Capacity
Trim7Tripartite Motif Containing 7
TMCTotal Motile Count
Ube3aUbiquitin Protein Ligase E3A (E6-AP) Protein
UtxUbiquitously Transcribed Tetratricopeptide Repeat on Chromosome X
WntWingless-related integration site

References

  1. Bhat, S.A.; Majid, S.; Wani, H.A.; Rashid, S. Diagnostic utility of epigenetics in breast cancer—A review. Cancer Treat. Res. Commun. 2019, 19, 100125. [Google Scholar] [CrossRef]
  2. Chen, Q.W.; Zhu, X.Y.; Li, Y.Y.; Meng, Z.Q. Epigenetic regulation and cancer (Review). Oncol. Rep. 2014, 31, 523–532. [Google Scholar] [CrossRef] [PubMed]
  3. Dawson, M.A.; Kouzarides, T. Cancer Epigenetics: From Mechanism to Therapy. Cell 2012, 150, 12–27. [Google Scholar] [CrossRef] [PubMed]
  4. Youness, E. Overview on Epigenetics and Cancer. Clin. Med. Rev. Rep. 2020, 2, 1–6. [Google Scholar] [CrossRef] [PubMed]
  5. Muyrers-Chen, I.; Paro, R. Epigenetics: Unforeseen regulators in cancer. Biochim. Biophys. Acta (BBA)—Rev. Cancer 2001, 1552, 15–26. [Google Scholar] [CrossRef]
  6. Larsson, L. Current Concepts of Epigenetics and Its Role in Periodontitis. Curr. Oral Health Rep. 2017, 4, 286–293. [Google Scholar] [CrossRef]
  7. Berger, S.L.; Kouzarides, T.; Shiekhattar, R.; Shilatifard, A. An operational definition of epigenetics: Figure 1. Genes Dev. 2009, 23, 781–783. [Google Scholar] [CrossRef]
  8. Guo, K.; Lu, M.; Bi, J.; Yao, T.; Gao, J.; Ren, F.; Zhu, L. Ferroptosis: Mechanism, immunotherapy and role in ovarian cancer. Front. Immunol. 2024, 15, 1410018. [Google Scholar] [CrossRef]
  9. Sharbatoghli, M.; Vafaei, S.; Aboulkheyr Es, H.; Asadi-Lari, M.; Totonchi, M.; Madjd, Z. Prediction of the treatment response in ovarian cancer: A ctDNA approach. J. Ovarian Res. 2020, 13, 124. [Google Scholar] [CrossRef]
  10. Ghoneum, A.; Afify, H.; Salih, Z.; Kelly, M.; Said, N. Role of tumor microenvironment in ovarian cancer pathobiology. Oncotarget 2018, 9, 22832–22849. [Google Scholar] [CrossRef]
  11. Gupta, V.; Yull, F.; Khabele, D. Bipolar Tumor-Associated Macrophages in Ovarian Cancer as Targets for Therapy. Cancers 2018, 10, 366. [Google Scholar] [CrossRef]
  12. Kozieł, M.J.; Piastowska-Ciesielska, A.W. Estrogens, Estrogen Receptors and Tumor Microenvironment in Ovarian Cancer. Int. J. Mol. Sci. 2023, 24, 14673. [Google Scholar] [CrossRef]
  13. Zhang, W.; Torres-Rojas, C.; Yue, J.; Zhu, B.-M. Adipose-derived stem cells in ovarian cancer progression, metastasis, and chemoresistance. Exp. Biol. Med. 2021, 246, 1810–1815. [Google Scholar] [CrossRef]
  14. An, Y.; Yang, Q. Tumor-associated macrophage-targeted therapeutics in ovarian cancer. Int. J. Cancer 2020, 149, 21–30. [Google Scholar] [CrossRef]
  15. Fakhri, N.; Chad, M.A.; Lahkim, M.; Houari, A.; Dehbi, H.; Belmouden, A.; El Kadmiri, N. Risk factors for breast cancer in women: An update review. Med. Oncol. 2022, 39, 197. [Google Scholar] [CrossRef] [PubMed]
  16. Sahu, R.; Pattanayak, S.P. Strategic Developments & Future Perspective on Gene Therapy for Breast Cancer: Role of mTOR and Brk/ PTK6 as Molecular Targets. Curr. Gene Ther. 2020, 20, 237–258. [Google Scholar] [CrossRef] [PubMed]
  17. Ghosh, A.; Gopinath, S.C.B. Molecular Mechanism of Breast Cancer and Predisposition of Mouse Mammary Tumor Virus Propagation Cycle. Curr. Med. Chem. 2025, 32, 2330–2348. [Google Scholar] [CrossRef] [PubMed]
  18. Soubry, A.; Hoyo, C.; Jirtle, R.L.; Murphy, S.K. A paternal environmental legacy: Evidence for epigenetic inheritance through the male germ line. BioEssays 2014, 36, 359–371. [Google Scholar] [CrossRef]
  19. Soubry, A. Epigenetics as a Driver of Developmental Origins of Health and Disease: Did We Forget the Fathers? BioEssays 2018, 40, 1700113. [Google Scholar] [CrossRef]
  20. Tabuchi, T.M.; Rechtsteiner, A.; Jeffers, T.E.; Egelhofer, T.A.; Murphy, C.T.; Strome, S. Caenorhabditis elegans sperm carry a histone-based epigenetic memory of both spermatogenesis and oogenesis. Nat. Commun. 2018, 9, 4310. [Google Scholar] [CrossRef]
  21. Champroux, A.; Cocquet, J.; Henry-Berger, J.; Drevet, J.R.; Kocer, A. A Decade of Exploring the Mammalian Sperm Epigenome: Paternal Epigenetic and Transgenerational Inheritance. Front. Cell Dev. Biol. 2018, 6, 50. [Google Scholar] [CrossRef]
  22. Curley, J.P.; Mashoodh, R.; Champagne, F.A. Epigenetics and the origins of paternal effects. Horm. Behav. 2011, 59, 306–314. [Google Scholar] [CrossRef] [PubMed]
  23. Lismer, A.; Kimmins, S. Emerging evidence that the mammalian sperm epigenome serves as a template for embryo development. Nat. Commun. 2023, 14, 2142. [Google Scholar] [CrossRef] [PubMed]
  24. Pang, T.Y.; Short, A.K.; Bredy, T.W.; Hannan, A.J. Transgenerational paternal transmission of acquired traits: Stress-induced modification of the sperm regulatory transcriptome and offspring phenotypes. Curr. Opin. Behav. Sci. 2017, 14, 140–147. [Google Scholar] [CrossRef] [PubMed]
  25. Kaneshiro, K.R.; Rechtsteiner, A.; Strome, S. Sperm-inherited H3K27me3 impacts offspring transcription and development in C. elegans. Nat. Commun. 2019, 10, 1271. [Google Scholar] [CrossRef]
  26. Feinberg, J.I.; Schrott, R.; Ladd-Acosta, C.; Newschaffer, C.J.; Hertz-Picciotto, I.; Croen, L.A.; Daniele Fallin, M.; Feinberg, A.P.; Volk, H.E. Epigenetic changes in sperm are associated with paternal and child quantitative autistic traits in an autism-enriched cohort. Mol. Psychiatry 2024, 29, 43–53. [Google Scholar] [CrossRef]
  27. Sengupta, P.; Dutta, S.; Liew, F.F.; Dhawan, V.; Das, B.; Mottola, F.; Slama, P.; Rocco, L.; Roychoudhury, S. Environmental and Genetic Traffic in the Journey from Sperm to Offspring. Biomolecules 2023, 13, 1759. [Google Scholar] [CrossRef]
  28. Hao, X.; Luo, H.; Krawczyk, M.; Wei, W.; Wang, W.; Wang, J.; Flagg, K.; Hou, J.; Zhang, H.; Yi, S.; et al. DNA methylation markers for diagnosis and prognosis of common cancers. Proc. Natl. Acad. Sci. USA 2017, 114, 7414–7419. [Google Scholar] [CrossRef]
  29. Mehrmohamadi, M.; Mentch, L.K.; Clark, A.G.; Locasale, J.W. Integrative modelling of tumour DNA methylation quantifies the contribution of metabolism. Nat. Commun. 2016, 7, 13666. [Google Scholar] [CrossRef]
  30. Aryee, M.J.; Liu, W.; Engelmann, J.C.; Nuhn, P.; Gurel, M.; Haffner, M.C.; Esopi, D.; Irizarry, R.A.; Getzenberg, R.H.; Nelson, W.G.; et al. DNA Methylation Alterations Exhibit Intraindividual Stability and Interindividual Heterogeneity in Prostate Cancer Metastases. Sci. Transl. Med. 2013, 5, ra110–ra169. [Google Scholar] [CrossRef]
  31. Brocato, J.; Costa, M. Basic mechanics of DNA methylation and the unique landscape of the DNA methylome in metal-induced carcinogenesis. Crit. Rev. Toxicol. 2013, 43, 493–514. [Google Scholar] [CrossRef] [PubMed]
  32. Klutstein, M.; Nejman, D.; Greenfield, R.; Cedar, H. DNA Methylation in Cancer and Aging. Cancer Res. 2016, 76, 3446–3450. [Google Scholar] [CrossRef] [PubMed]
  33. Fontelles, C.C.; Carney, E.; Clarke, J.; Nguyen, N.M.; Yin, C.; Jin, L.; Cruz, M.I.; Ong, T.P.; Hilakivi-Clarke, L.; De Assis, S. Paternal overweight is associated with increased breast cancer risk in daughters in a mouse model. Sci. Rep. 2016, 6, 28602. [Google Scholar] [CrossRef] [PubMed]
  34. Illum, L.R.H.; Bak, S.T.; Lund, S.; Nielsen, A.L. DNA methylation in epigenetic inheritance of metabolic diseases through the male germ line. J. Mol. Endocrinol. 2018, 60, R39–R56. [Google Scholar] [CrossRef]
  35. Wei, S.H.; Balch, C.; Paik, H.H.; Kim, Y.-S.; Baldwin, R.L.; Liyanarachchi, S.; Li, L.; Wang, Z.; Wan, J.C.; Davuluri, R.V.; et al. Prognostic DNA Methylation Biomarkers in Ovarian Cancer. Clin. Cancer Res. 2006, 12, 2788–2794. [Google Scholar] [CrossRef]
  36. Samudio-Ruiz, S.L.; Hudson, L.G. Increased DNA methyltransferase activity and DNA methylation following epidermal growth factor stimulation in ovarian cancer cells. Epigenetics 2012, 7, 216–224. [Google Scholar] [CrossRef]
  37. Fu, M.; Deng, F.; Chen, J.; Fu, L.; Lei, J.; Xu, T.; Chen, Y.; Zhou, J.; Gao, Q.; Ding, H. Current data and future perspectives on DNA methylation in ovarian cancer (Review). Int. J. Oncol. 2024, 64, 62. [Google Scholar] [CrossRef]
  38. Shen, L.; Kondo, Y.; Guo, Y.; Zhang, J.; Zhang, L.; Ahmed, S.; Shu, J.; Chen, X.; Waterland, R.A.; Issa, J.-P.J. Genome-Wide Profiling of DNA Methylation Reveals a Class of Normally Methylated CpG Island Promoters. PLoS Genet. 2007, 3, e181. [Google Scholar] [CrossRef]
  39. Uhm, K.-O.; Lee, E.S.; Lee, Y.M.; Kim, H.S.; Park, Y.-N.; Park, S.-H. Aberrant Promoter CpG Islands Methylation of Tumor Suppressor Genes in Cholangiocarcinoma. Oncol. Res. Featur. Preclin. Clin. Cancer Ther. 2008, 17, 151–157. [Google Scholar] [CrossRef]
  40. Portela, A.; Liz, J.; Nogales, V.; Setién, F.; Villanueva, A.; Esteller, M. DNA methylation determines nucleosome occupancy in the 5′-CpG islands of tumor suppressor genes. Oncogene 2013, 32, 5421–5428. [Google Scholar] [CrossRef]
  41. Sharma, S.; De Carvalho, D.D.; Jeong, S.; Jones, P.A.; Liang, G. Nucleosomes Containing Methylated DNA Stabilize DNA Methyltransferases 3A/3B and Ensure Faithful Epigenetic Inheritance. PLoS Genet. 2011, 7, e1001286. [Google Scholar] [CrossRef]
  42. Shen, L.; Gao, G.; Zhang, Y.; Zhang, H.; Ye, Z.; Huang, S.; Huang, J.; Kang, J. A single amino acid substitution confers enhanced methylation activity of mammalian Dnmt3b on chromatin DNA. Nucleic Acids Res. 2010, 38, 6054–6064. [Google Scholar] [CrossRef] [PubMed]
  43. Shiraishi, M.; Sekiguchi, A.; Terry, M.J.; Oates, A.J.; Miyamoto, Y.; Chuu, Y.H.; Munakata, M.; Sekiya, T. A comprehensive catalog of CpG islands methylated in human lung adenocarcinomas for the identification of tumor suppressor genes. Oncogene 2002, 21, 3804–3813. [Google Scholar] [CrossRef] [PubMed]
  44. Lopez-Serra, L.; Ballestar, E.; Fraga, M.F.; Alaminos, M.; Setien, F.; Esteller, M. A Profile of Methyl-CpG Binding Domain Protein Occupancy of Hypermethylated Promoter CpG Islands of Tumor Suppressor Genes in Human Cancer. Cancer Res. 2006, 66, 8342–8346. [Google Scholar] [CrossRef] [PubMed]
  45. Samy, M.D.; Yavorski, J.M.; Mauro, J.A.; Blanck, G. Impact of SNPs on CpG Islands in the MYC and HRAS oncogenes and in a wide variety of tumor suppressor genes: A multi-cancer approach. Cell Cycle 2016, 15, 1572–1578. [Google Scholar] [CrossRef]
  46. Fukushima, N.; Sato, N.; Ueki, T.; Rosty, C.; Walter, K.M.; Wilentz, R.E.; Yeo, C.J.; Hruban, R.H.; Goggins, M. Aberrant Methylation of Preproenkephalin and p16 Genes in Pancreatic Intraepithelial Neoplasia and Pancreatic Ductal Adenocarcinoma. Am. J. Pathol. 2002, 160, 1573–1581. [Google Scholar] [CrossRef]
  47. Smith, G.; Carey, F.A.; Beattie, J.; Wilkie, M.J.V.; Lightfoot, T.J.; Coxhead, J.; Garner, R.C.; Steele, R.J.C.; Wolf, C.R. Mutations in APC, Kirsten-ras, and p53—Alternative genetic pathways to colorectal cancer. Proc. Natl. Acad. Sci. USA 2002, 99, 9433–9438. [Google Scholar] [CrossRef]
  48. Zheng, H.; Momeni, A.; Cedoz, P.-L.; Vogel, H.; Gevaert, O. Whole slide images reflect DNA methylation patterns of human tumors. NPJ Genom. Med. 2020, 5, 11. [Google Scholar] [CrossRef]
  49. Kim, M.S.; Lee, J.; Sidransky, D. DNA methylation markers in colorectal cancer. Cancer Metastasis Rev. 2010, 29, 181–206. [Google Scholar] [CrossRef]
  50. Jemal, A.; Murray, T.; Ward, E.; Samuels, A.; Tiwari, R.C.; Ghafoor, A.; Feuer, E.J.; Thun, M.J. Cancer Statistics, 2005. CA A Cancer J. Clin. 2005, 55, 10–30. [Google Scholar] [CrossRef]
  51. Parkin, D.M.; Pisani, P.; Ferlay, J. Global cancer statistics. CA A Cancer J. Clin. 1999, 49, 33–64. [Google Scholar] [CrossRef]
  52. Munshi, A.; Shafi, G.; Aliya, N.; Jyothy, A. Histone modifications dictate specific biological readouts. J. Genet. Genom. 2009, 36, 75–88. [Google Scholar] [CrossRef]
  53. Jenuwein, T.; Allis, C.D. Translating the Histone Code. Science 2001, 293, 1074–1080. [Google Scholar] [CrossRef]
  54. Antoniou, A.C.; Spurdle, A.B.; Sinilnikova, O.M.; Healey, S.; Pooley, K.A.; Schmutzler, R.K.; Versmold, B.; Engel, C.; Meindl, A.; Arnold, N.; et al. Common Breast Cancer-Predisposition Alleles Are Associated with Breast Cancer Risk in BRCA1 and BRCA2 Mutation Carriers. Am. J. Hum. Genet. 2008, 82, 937–948. [Google Scholar] [CrossRef]
  55. Sun, Y.-S.; Zhao, Z.; Yang, Z.-N.; Xu, F.; Lu, H.-J.; Zhu, Z.-Y.; Shi, W.; Jiang, J.; Yao, P.-P.; Zhu, H.-P. Risk Factors and Preventions of Breast Cancer. Int. J. Biol. Sci. 2017, 13, 1387–1397. [Google Scholar] [CrossRef]
  56. Pedroza, D.A.; Subramani, R.; Tiula, K.; Do, A.; Rashiraj, N.; Galvez, A.; Chatterjee, A.; Bencomo, A.; Rivera, S.; Lakshmanaswamy, R. Crosstalk between progesterone receptor membrane component 1 and estrogen receptor α promotes breast cancer cell proliferation. Lab. Investig. 2021, 101, 733–744. [Google Scholar] [CrossRef]
  57. Bernstein, B.E.; Mikkelsen, T.S.; Xie, X.; Kamal, M.; Huebert, D.J.; Cuff, J.; Fry, B.; Meissner, A.; Wernig, M.; Plath, K.; et al. A Bivalent Chromatin Structure Marks Key Developmental Genes in Embryonic Stem Cells. Cell 2006, 125, 315–326. [Google Scholar] [CrossRef]
  58. Lin, J.C.; Jeong, S.; Liang, G.; Takai, D.; Fatemi, M.; Tsai, Y.C.; Egger, G.; Gal-Yam, E.N.; Jones, P.A. Role of Nucleosomal Occupancy in the Epigenetic Silencing of the MLH1 CpG Island. Cancer Cell 2007, 12, 432–444. [Google Scholar] [CrossRef]
  59. Schotta, G.; Lachner, M.; Sarma, K.; Ebert, A.; Sengupta, R.; Reuter, G.; Reinberg, D.; Jenuwein, T. A silencing pathway to induce H3-K9 and H4-K20 trimethylation at constitutive heterochromatin. Genes Dev. 2004, 18, 1251–1262. [Google Scholar] [CrossRef]
  60. Li, B.; Carey, M.; Workman, J.L. The Role of Chromatin during Transcription. Cell 2007, 128, 707–719. [Google Scholar] [CrossRef]
  61. Heintzman, N.D.; Stuart, R.K.; Hon, G.; Fu, Y.; Ching, C.W.; Hawkins, R.D.; Barrera, L.O.; Van Calcar, S.; Qu, C.; Ching, K.A.; et al. Distinct and predictive chromatin signatures of transcriptional promoters and enhancers in the human genome. Nat. Genet. 2007, 39, 311–318. [Google Scholar] [CrossRef]
  62. Heintzman, N.D.; Hon, G.C.; Hawkins, R.D.; Kheradpour, P.; Stark, A.; Harp, L.F.; Ye, Z.; Lee, L.K.; Stuart, R.K.; Ching, C.W.; et al. Histone modifications at human enhancers reflect global cell-type-specific gene expression. Nature 2009, 459, 108–112. [Google Scholar] [CrossRef]
  63. Jones, P.L.; Veenstra, G.J.C.; Wade, P.A.; Vermaak, D.; Kass, S.U.; Landsberger, N.; Strouboulis, J.; Wolffe, A.P. Methylated DNA and MeCP2 recruit histone deacetylase to repress transcription. Nat. Genet. 1998, 19, 187–191. [Google Scholar] [CrossRef]
  64. Cedar, H.; Bergman, Y. Linking DNA methylation and histone modification: Patterns and paradigms. Nat. Rev. Genet. 2009, 10, 295–304. [Google Scholar] [CrossRef]
  65. Sasaki, H.; Matsui, Y. Epigenetic events in mammalian germ-cell development: Reprogramming and beyond. Nat. Rev. Genet. 2008, 9, 129–140. [Google Scholar] [CrossRef]
  66. Kriaucionis, S.; Heintz, N. The Nuclear DNA Base 5-Hydroxymethylcytosine Is Present in Purkinje Neurons and the Brain. Science 2009, 324, 929–930. [Google Scholar] [CrossRef]
  67. Illingworth, R.S.; Bird, A.P. CpG islands—‘A rough guide’. FEBS Lett. 2009, 583, 1713–1720. [Google Scholar] [CrossRef]
  68. Kwon, M.J.; Kim, S.-S.; Choi, Y.-L.; Jung, H.S.; Balch, C.; Kim, S.-H.; Song, Y.-S.; Marquez, V.E.; Nephew, K.P.; Shin, Y.K. Derepression of CLDN3 and CLDN4 during ovarian tumorigenesis is associated with loss of repressive histone modifications. Carcinogenesis 2010, 31, 974–983. [Google Scholar] [CrossRef]
  69. Maldonado, L.; Hoque, M.O. Epigenomics and Ovarian Carcinoma. Biomark. Med. 2010, 4, 543–570. [Google Scholar] [CrossRef]
  70. Matei, D.; Nephew, K.P. Epigenetic Attire in Ovarian Cancer: The Emperor’s New Clothes. Cancer Res. 2020, 80, 3775–3785. [Google Scholar] [CrossRef]
  71. Siklenka, K.; Erkek, S.; Godmann, M.; Lambrot, R.; McGraw, S.; Lafleur, C.; Cohen, T.; Xia, J.G.; Suderman, M.; Hallett, M.; et al. Disruption of histone methylation in developing sperm impairs offspring health transgenerationally. Science 2015, 350, aab2006. [Google Scholar] [CrossRef]
  72. Lismer, A.; Siklenka, K.; Lafleur, C.; Dumeaux, V.; Kimmins, S. Sperm histone H3 lysine 4 trimethylation is altered in a genetic mouse model of transgenerational epigenetic inheritance. Nucleic Acids Res. 2020, 48, 11380–11393. [Google Scholar] [CrossRef]
  73. Ben Maamar, M.; Sadler-Riggleman, I.; Beck, D.; Skinner, M.K. Epigenetic Transgenerational Inheritance of Altered Sperm Histone Retention Sites. Sci. Rep. 2018, 8, 5308. [Google Scholar] [CrossRef]
  74. Ben Maamar, M.; Sadler-Riggleman, I.; Beck, D.; McBirney, M.; Nilsson, E.; Klukovich, R.; Xie, Y.; Tang, C.; Yan, W.; Skinner, M.K. Alterations in sperm DNA methylation, non-coding RNA expression, and histone retention mediate vinclozolin-induced epigenetic transgenerational inheritance of disease. Environ. Epigenetics 2018, 4, dvy010. [Google Scholar] [CrossRef]
  75. Lesch, B.J.; Tothova, Z.; Morgan, E.A.; Liao, Z.C.; Bronson, R.T.; Ebert, B.L.; Page, D.C. Intergenerational epigenetic inheritance of cancer susceptibility in mammals. Elife 2019, 8, e39380. [Google Scholar] [CrossRef]
  76. Kondo, Y. Epigenetic Cross-Talk between DNA Methylation and Histone Modifications in Human Cancers. Yonsei Med. J. 2009, 50, 455. [Google Scholar] [CrossRef]
  77. Taberlay, P.C.; Jones, P.A. DNA Methylation and Cancer. In Epigenetics and Disease; Springer: Basel, Switzerland, 2011; pp. 1–23. [Google Scholar]
  78. Haun, W.J.; Springer, N.M. Maternal and paternal alleles exhibit differential histone methylation and acetylation at maize imprinted genes. Plant J. 2008, 56, 903–912. [Google Scholar] [CrossRef]
  79. Rahman, M.M.; Brane, A.C.; Tollefsbol, T.O. MicroRNAs and Epigenetics Strategies to Reverse Breast Cancer. Cells 2019, 8, 1214. [Google Scholar] [CrossRef]
  80. Kim, A.; Mo, K.; Kwon, H.; Choe, S.; Park, M.; Kwak, W.; Yoon, H. Epigenetic Regulation in Breast Cancer: Insights on Epidrugs. Epigenomes 2023, 7, 6. [Google Scholar] [CrossRef]
  81. Connolly, R.; Stearns, V. Epigenetics as a Therapeutic Target in Breast Cancer. J. Mammary Gland. Biol. Neoplasia 2012, 17, 191–204. [Google Scholar] [CrossRef]
  82. González-Rodríguez, P.; Füllgrabe, J.; Joseph, B. The hunger strikes back: An epigenetic memory for autophagy. Cell Death Differ. 2023, 30, 1404–1415. [Google Scholar] [CrossRef]
  83. Painter, R.C.; De Rooij, S.R.; Bossuyt, P.M.M.; Osmond, C.; Barker, D.J.P.; Bleker, O.P.; Roseboom, T.J. A possible link between prenatal exposure to famine and breast cancer: A preliminary study. Am. J. Hum. Biol. 2006, 18, 853–856. [Google Scholar] [CrossRef]
  84. Wilkinson, A.L.; Zorzan, I.; Rugg-Gunn, P.J. Epigenetic regulation of early human embryo development. Cell Stem Cell 2023, 30, 1569–1584. [Google Scholar] [CrossRef]
  85. McSwiggin, H.; Magalhães, R.; Nilsson, E.E.; Yan, W.; Skinner, M.K. Epigenetic transgenerational inheritance of toxicant exposure-specific non-coding RNA in sperm. Environ. Epigenetics 2024, 10, dvae014. [Google Scholar] [CrossRef]
  86. Zheng, X.; Li, Z.; Wang, G.; Wang, H.; Zhou, Y.; Zhao, X.; Cheng, C.Y.; Qiao, Y.; Sun, F. Sperm epigenetic alterations contribute to inter- and transgenerational effects of paternal exposure to long-term psychological stress via evading offspring embryonic reprogramming. Cell Discov. 2021, 7, 101. [Google Scholar] [CrossRef]
  87. Da Cruz, R.S.; Carney, E.J.; Clarke, J.; Cao, H.; Cruz, M.I.; Benitez, C.; Jin, L.; Fu, Y.; Cheng, Z.; Wang, Y.; et al. Paternal malnutrition programs breast cancer risk and tumor metabolism in offspring. Breast Cancer Res. 2018, 20, 99. [Google Scholar] [CrossRef]
  88. Robles, V.; Valcarce, D.G.; Riesco, M.F. Non-coding RNA regulation in reproduction: Their potential use as biomarkers. Non-Coding RNA Res. 2019, 4, 54–62. [Google Scholar] [CrossRef]
  89. Rodgers, A.B.; Morgan, C.P.; Leu, N.A.; Bale, T.L. Transgenerational epigenetic programming via sperm microRNA recapitulates effects of paternal stress. Proc. Natl. Acad. Sci. USA 2015, 112, 13699–13704. [Google Scholar] [CrossRef]
  90. Vaz, C.; Kermack, A.J.; Burton, M.; Tan, P.F.; Huan, J.; Yoo, T.P.X.; Donnelly, K.; Wellstead, S.J.; Fisk, H.L.; Houghton, F.D.; et al. Short-term diet intervention alters the small non-coding RNA (sncRNA) landscape of human sperm. bioRxiv 2021. [Google Scholar] [CrossRef]
  91. Zhang, Q.; Li, N.; Deng, L.; Jiang, X.; Zhang, Y.; Lee, L.T.O.; Zhang, H. ACSL1-induced ferroptosis and platinum resistance in ovarian cancer by increasing FSP1 N-myristylation and stability. Cell Death Discov. 2023, 9, 83. [Google Scholar] [CrossRef]
  92. Zhu, Y.; Chen, S.; Su, H.; Meng, Y.; Zang, C.; Ning, P.; Hu, L.; Shao, H. CPT1A-mediated MFF succinylation promotes stemness maintenance in ovarian cancer stem cells. Commun. Biol. 2025, 8, 250. [Google Scholar] [CrossRef]
  93. Zhao, G.; Tan, Y.; Cardenas, H.; Vayngart, D.; Wang, Y.; Huang, H.; Keathley, R.; Wei, J.-J.; Ferreira, C.R.; Orsulic, S.; et al. Ovarian cancer cell fate regulation by the dynamics between saturated and unsaturated fatty acids. Proc. Natl. Acad. Sci. USA 2022, 119, e2203480119. [Google Scholar] [CrossRef]
  94. Thomas, R.; Al-Rashed, F.; Akhter, N.; Al-Mulla, F.; Ahmad, R. ACSL1 Regulates TNFα-Induced GM-CSF Production by Breast Cancer MDA-MB-231 Cells. Biomolecules 2019, 9, 555. [Google Scholar] [CrossRef]
  95. Das, M.; Giannoudis, A.; Sharma, V. The role of CPT1A as a biomarker of breast cancer progression: A bioinformatic approach. Sci. Rep. 2022, 12, 16441. [Google Scholar] [CrossRef]
  96. Kieu, T.-L.-V.; Pierre, L.; Derangère, V.; Perrey, S.; Truntzer, C.; Jalil, A.; Causse, S.; Groetz, E.; Dumont, A.; Guyard, L.; et al. Downregulation of Elovl5 promotes breast cancer metastasis through a lipid-droplet accumulation-mediated induction of TGF-β receptors. Cell Death Dis. 2022, 13, 758. [Google Scholar] [CrossRef]
  97. Nikulin, S.; Zakharova, G.; Poloznikov, A.; Raigorodskaya, M.; Wicklein, D.; Schumacher, U.; Nersisyan, S.; Bergquist, J.; Bakalkin, G.; Astakhova, L.; et al. Effect of the Expression of ELOVL5 and IGFBP6 Genes on the Metastatic Potential of Breast Cancer Cells. Front. Genet. 2021, 12, 662843. [Google Scholar] [CrossRef]
  98. Paris, L.; Giardullo, P.; Leonardi, S.; Tanno, B.; Meschini, R.; Cordelli, E.; Benassi, B.; Longobardi, M.G.; Izzotti, A.; Pulliero, A.; et al. Transgenerational inheritance of enhanced susceptibility to radiation-induced medulloblastoma in newborn Ptch1+/− mice after paternal irradiation. Oncotarget 2015, 6, 36098–36112. [Google Scholar] [CrossRef]
  99. Sharma, U.; Conine, C.C.; Shea, J.M.; Boskovic, A.; Derr, A.G.; Bing, X.Y.; Belleannee, C.; Kucukural, A.; Serra, R.W.; Sun, F.; et al. Biogenesis and function of tRNA fragments during sperm maturation and fertilization in mammals. Science 2016, 351, 391–396. [Google Scholar] [CrossRef]
  100. Liu, D.; Wu, C.; Wang, J.; Zhang, L.; Sun, Z.; Chen, S.; Ding, Y.; Wang, W. Transfer RNA-derived fragment 5′tRF-Gly promotes the development of hepatocellular carcinoma by direct targeting of carcinoembryonic antigen-related cell adhesion molecule 1. Cancer Sci. 2022, 113, 3476–3488. [Google Scholar] [CrossRef]
  101. Chen, F.; Song, C.; Meng, F.; Zhu, Y.; Chen, X.; Fang, X.; Ma, D.; Wang, Y.; Zhang, C. 5′-tRF-GlyGCC promotes breast cancer metastasis by increasing fat mass and obesity-associated protein demethylase activity. Int. J. Biol. Macromol. 2023, 226, 397–409. [Google Scholar] [CrossRef]
  102. Hernandez-Alias, X.; Benisty, H.; Schaefer, M.H.; Serrano, L. Translational efficiency across healthy and tumor tissues is proliferation-related. Mol. Syst. Biol. 2020, 16, e9275. [Google Scholar] [CrossRef] [PubMed]
  103. Yang, M.; Mo, Y.; Ren, D.; Liu, S.; Zeng, Z.; Xiong, W. Transfer RNA-derived small RNAs in tumor microenvironment. Mol. Cancer 2023, 22, 32. [Google Scholar] [CrossRef] [PubMed]
  104. Schreier, J.; Dietz, S.; Boermel, M.; Oorschot, V.; Seistrup, A.-S.; de Jesus Domingues, A.M.; Bronkhorst, A.W.; Nguyen, D.A.H.; Phillis, S.; Gleason, E.J.; et al. Membrane-associated cytoplasmic granules carrying the Argonaute protein WAGO-3 enable paternal epigenetic inheritance in Caenorhabditis elegans. Nat. Cell Biol. 2022, 24, 217–229. [Google Scholar] [CrossRef] [PubMed]
  105. Petri, B.J.; Klinge, C.M. m6A readers, writers, erasers, and the m6A epitranscriptome in breast cancer. J. Mol. Endocrinol. 2023, 70, e220110. [Google Scholar] [CrossRef]
  106. Kumari, K.; Groza, P.; Aguilo, F. Regulatory roles of RNA modifications in breast cancer. NAR Cancer 2021, 3, zcab036. [Google Scholar] [CrossRef]
  107. Benak, D.; Benakova, S.; Plecita-Hlavata, L.; Hlavackova, M. The role of m6A and m6Am RNA modifications in the pathogenesis of diabetes mellitus. Front. Endocrinol. 2023, 14, 1223583. [Google Scholar] [CrossRef]
  108. Dai, Q.; Ye, Y. Development and Validation of a Novel Histone Acetylation-Related Gene Signature for Predicting the Prognosis of Ovarian Cancer. Front. Cell Dev. Biol. 2022, 10, 793425. [Google Scholar] [CrossRef]
  109. Gao, Y.; Tollefsbol, T.O. Impact of Epigenetic Dietary Components on Cancer through Histone Modifications. Curr. Med. Chem. 2015, 22, 2051–2064. [Google Scholar] [CrossRef]
  110. Lapierre, M.; Linares, A.; Dalvai, M.; Duraffourd, C.; Bonnet, S.; Boulahtouf, A.; Rodriguez, C.; Jalaguier, S.; Assou, S.; Orsetti, B.; et al. Histone deacetylase 9 regulates breast cancer cell proliferation and the response to histone deacetylase inhibitors. Oncotarget 2016, 7, 19693–19708. [Google Scholar] [CrossRef]
  111. Liu, Z.; Zhou, S.; Liao, L.; Chen, X.; Meistrich, M.; Xu, J. Jmjd1a Demethylase-regulated Histone Modification Is Essential for cAMP-response Element Modulator-regulated Gene Expression and Spermatogenesis. J. Biol. Chem. 2010, 285, 2758–2770. [Google Scholar] [CrossRef]
  112. Brykczynska, U.; Hisano, M.; Erkek, S.; Ramos, L.; Oakeley, E.J.; Roloff, T.C.; Beisel, C.; Schübeler, D.; Stadler, M.B.; Peters, A.H.F.M. Repressive and active histone methylation mark distinct promoters in human and mouse spermatozoa. Nat. Struct. Mol. Biol. 2010, 17, 679–687. [Google Scholar] [CrossRef] [PubMed]
  113. Mann, M.; Cortez, V.; Vadlamudi, R. PELP1 oncogenic functions involve CARM1 regulation. Carcinogenesis 2013, 34, 1468–1475. [Google Scholar] [CrossRef] [PubMed]
  114. Messier, T.L.; Gordon, J.A.R.; Boyd, J.R.; Tye, C.E.; Browne, G.; Stein, J.L.; Lian, J.B.; Stein, G.S. Histone H3 lysine 4 acetylation and methylation dynamics define breast cancer subtypes. Oncotarget 2016, 7, 5094–5109. [Google Scholar] [CrossRef]
  115. Falahi, F.; van Kruchten, M.; Martinet, N.; Hospers, G.; Rots, M.G. Current and upcoming approaches to exploit the reversibility of epigenetic mutations in breast cancer. Breast Cancer Res. 2014, 16, 412. [Google Scholar] [CrossRef]
  116. Fontelles, C.C.; Da Cruz, R.S.; Gonsiewski, A.K.; Barin, E.; Tekmen, V.; Jin, L.; Cruz, M.I.; Loudig, O.; Warri, A.; De Assis, S. Paternal obesity and epigenetic inheritance of breast cancer: The role of systemic effects and transmission to the second generation. bioRxiv 2020. [Google Scholar] [CrossRef]
  117. Soubry, A.; Murphy, S.K.; Vansant, G.; He, Y.; Price, T.M.; Hoyo, C. Opposing Epigenetic Signatures in Human Sperm by Intake of Fast Food Versus Healthy Food. Front. Endocrinol. 2021, 12, 625204. [Google Scholar] [CrossRef]
  118. Akhatova, A.; Jones, C.; Coward, K.; Yeste, M. How do lifestyle and environmental factors influence the sperm epigenome? Effects on sperm fertilising ability, embryo development, and offspring health. Clin. Epigenetics 2025, 17, 7. [Google Scholar] [CrossRef]
  119. Pascoal, G.F.L.; Geraldi, M.V.; Maróstica, M.R., Jr.; Ong, T.P. Effect of Paternal Diet on Spermatogenesis and Offspring Health: Focus on Epigenetics and Interventions with Food Bioactive Compounds. Nutrients 2022, 14, 2150. [Google Scholar] [CrossRef]
  120. Lambrot, R.; Xu, C.; Saint-Phar, S.; Chountalos, G.; Cohen, T.; Paquet, M.; Suderman, M.; Hallett, M.; Kimmins, S. Low paternal dietary folate alters the mouse sperm epigenome and is associated with negative pregnancy outcomes. Nat. Commun. 2013, 4, 2889. [Google Scholar] [CrossRef]
  121. Skoracka, K.; Eder, P.; Łykowska-Szuber, L.; Dobrowolska, A.; Krela-Kaźmierczak, I. Diet and Nutritional Factors in Male (In)fertility-Underestimated Factors. J. Clin. Med. 2020, 9, 1400. [Google Scholar] [CrossRef]
  122. Bodden, C.; Hannan, A.J.; Reichelt, A.C. Diet-Induced Modification of the Sperm Epigenome Programs Metabolism and Behavior. Trends Endocrinol. Metab. 2020, 31, 131–149. [Google Scholar] [CrossRef]
  123. Eid, N. Defining the Links Between Paternal Diet, Metabolic Health, and Reproductive Fitness in Mice. Ph.D. Thesis, University of Nottingham (United Kingdom), Nottingham, UK, 2023. [Google Scholar]
  124. Crisóstomo, L.; Jarak, I.; Rato, L.P.; Raposo, J.F.; Batterham, R.L.; Oliveira, P.F.; Alves, M.G. Inheritable testicular metabolic memory of high-fat diet causes transgenerational sperm defects in mice. Sci. Rep. 2021, 11, 9444. [Google Scholar] [CrossRef] [PubMed]
  125. Jahan-Mihan, A.; Leftwich, J.; Berg, K.; Labyak, C.; Nodarse, R.R.; Allen, S.; Griggs, J. The Impact of Parental Preconception Nutrition, Body Weight, and Exercise Habits on Offspring Health Outcomes: A Narrative Review. Nutrients 2024, 16, 4276. [Google Scholar] [CrossRef] [PubMed]
  126. Almujaydil, M.S. The Role of Dietary Nutrients in Male Infertility: A Review. Life 2023, 13, 519. [Google Scholar] [CrossRef] [PubMed]
  127. Tvrdá, E.; Sikeli, P.; Lukáčová, J.; Massányi, P.; Lukáč, N. Mineral Nutrients and Male Fertility. J. Microbiol. Biotechnol. Food Sci. 2013, 3, 1–14. [Google Scholar]
  128. Hoek, J.; Koster, M.P.H.; Schoenmakers, S.; Willemsen, S.P.; Koning, A.H.J.; Steegers, E.A.P.; Steegers-Theunissen, R.P.M. Does the father matter? The association between the periconceptional paternal folate status and embryonic growth. Fertil. Steril. 2019, 111, 270–279. [Google Scholar] [CrossRef]
  129. Ames, B.N. Micronutrient deficiencies. A major cause of DNA damage. Ann. N. Y. Acad. Sci. 1999, 889, 87–106. [Google Scholar] [CrossRef]
  130. Billah, M.M.; Khatiwada, S.; Morris, M.J.; Maloney, C.A. Effects of paternal overnutrition and interventions on future generations. Int. J. Obes. 2022, 46, 901–917. [Google Scholar] [CrossRef]
  131. Du Toit, E.W. Impact of Micro-Nutrient Supplementation on Semen Parameters. Ph.D. Thesis, University of the Free State, Bloemfontein, South Africa, 2016. [Google Scholar]
  132. Erdoğan, K.; Sanlier, N.T.; Sanlier, N. Are epigenetic mechanisms and nutrition effective in male and female infertility? J. Nutr. Sci. 2023, 12, e103. [Google Scholar] [CrossRef]
  133. Singh, K.; Jaiswal, D. One-carbon metabolism, spermatogenesis, and male infertility. Reprod. Sci. 2013, 20, 622–630. [Google Scholar] [CrossRef]
  134. Ames, B.N.; Wakimoto, P. Are vitamin and mineral deficiencies a major cancer risk? Nat. Rev. Cancer 2002, 2, 694–704. [Google Scholar] [CrossRef]
  135. McPherson, N.O.; Shehadeh, H.; Fullston, T.; Zander-Fox, D.L.; Lane, M. Dietary Micronutrient Supplementation for 12 Days in Obese Male Mice Restores Sperm Oxidative Stress. Nutrients 2019, 11, 2196. [Google Scholar] [CrossRef]
  136. Palmer, N.O.; Bakos, H.W.; Fullston, T.; Lane, M. Impact of obesity on male fertility, sperm function and molecular composition. Spermatogenesis 2012, 2, 253–263. [Google Scholar] [CrossRef] [PubMed]
  137. Raad, G.; Hazzouri, M.; Bottini, S.; Trabucchi, M.; Azoury, J.; Grandjean, V. Paternal obesity: How bad is it for sperm quality and progeny health? Basic Clin. Androl. 2017, 27, 20. [Google Scholar] [CrossRef]
  138. Larqué, C.; Lugo-Martínez, H.; Mendoza, X.; Nochebuena, M.; Novo, L.; Vilchis, R.; Sánchez-Bringas, G.; Ubaldo, L.; Velasco, M.; Escalona, R. Paternal Obesity Induced by High-Fat Diet Impairs the Metabolic and Reproductive Health of Progeny in Rats. Metabolites 2023, 13, 1098. [Google Scholar] [CrossRef] [PubMed]
  139. Zheng, L.; Yang, L.; Guo, Z.; Yao, N.; Zhang, S.; Pu, P. Obesity and its impact on female reproductive health: Unraveling the connections. Front. Endocrinol. 2024, 14, 1326546. [Google Scholar] [CrossRef] [PubMed]
  140. Craig, J.R.; Jenkins, T.G.; Carrell, D.T.; Hotaling, J.M. Obesity, male infertility, and the sperm epigenome. Fertil. Steril. 2017, 107, 848–859. [Google Scholar] [CrossRef]
  141. Fullston, T.; Palmer, N.O.; Owens, J.A.; Mitchell, M.; Bakos, H.W.; Lane, M. Diet-induced paternal obesity in the absence of diabetes diminishes the reproductive health of two subsequent generations of mice. Hum. Reprod. 2012, 27, 1391–1400. [Google Scholar] [CrossRef]
  142. Sanchez-Garrido, M.A.; Ruiz-Pino, F.; Velasco, I.; Barroso, A.; Fernandois, D.; Heras, V.; Manfredi-Lozano, M.; Vazquez, M.J.; Castellano, J.M.; Roa, J.; et al. Intergenerational Influence of Paternal Obesity on Metabolic and Reproductive Health Parameters of the Offspring: Male-Preferential Impact and Involvement of Kiss1-Mediated Pathways. Endocrinology 2017, 159, 1005–1018. [Google Scholar] [CrossRef]
  143. Sultan, S.; Patel, A.G.; El-Hassani, S.; Whitelaw, B.; Leca, B.M.; Vincent, R.P.; le Roux, C.W.; Rubino, F.; Aywlin, S.J.B.; Dimitriadis, G.K. Male Obesity Associated Gonadal Dysfunction and the Role of Bariatric Surgery. Front. Endocrinol. 2020, 11, 408. [Google Scholar] [CrossRef]
  144. Venigalla, G.; Ila, V.; Dornbush, J.; Bernstein, A.; Loloi, J.; Pozzi, E.; Miller, D.; Ramasamy, R. Male obesity: Associated effects on fertility and the outcomes of offspring. Andrology 2025, 13, 64–71. [Google Scholar] [CrossRef]
  145. Barbouni, K.; Jotautis, V.; Metallinou, D.; Diamanti, A.; Orovou, E.; Liepinaitienė, A.; Nikolaidis, P.; Karampas, G.; Sarantaki, A. When Weight Matters: How Obesity Impacts Reproductive Health and Pregnancy-A Systematic Review. Curr. Obes. Rep. 2025, 14, 37. [Google Scholar] [CrossRef] [PubMed]
  146. Fullston, T.; McPherson, N.O.; Zander-Fox, D.; Lane, M. The most common vices of men can damage fertility and the health of the next generation. J. Endocrinol. 2017, 234, F1–F6. [Google Scholar] [CrossRef] [PubMed]
  147. McPherson, N.O.; Owens, J.A.; Fullston, T.; Lane, M. Preconception diet or exercise intervention in obese fathers normalizes sperm microRNA profile and metabolic syndrome in female offspring. Am. J. Physiol. Endocrinol. Metab. 2015, 308, E805–E821. [Google Scholar] [CrossRef]
  148. Ramadan, A.G.; Abdel-Rehim, W.M.; El-Tahan, R.A.; Elblehi, S.S.; Kamel, M.A.; Shaker, S.A. Maternal and paternal obesity differentially reprogram the ovarian mitochondrial biogenesis of F1 female rats. Sci. Rep. 2023, 13, 15480. [Google Scholar] [CrossRef] [PubMed]
  149. Li, S.; Wu, H.; Chen, M.; Tollefsbol, T.O. Paternal Combined Botanicals Contribute to the Prevention of Estrogen Receptor-Negative Mammary Cancer in Transgenic Mice. J. Nutr. 2023, 153, 1959–1973. [Google Scholar] [CrossRef]
  150. D’Angelo, S.; Scafuro, M.; Meccariello, R. BPA and Nutraceuticals, Simultaneous Effects on Endocrine Functions. Endocr. Metab. Immune Disord. Drug Targets 2019, 19, 594–604. [Google Scholar] [CrossRef]
  151. Divella, R.; Daniele, A.; Savino, E.; Paradiso, A. Anticancer Effects of Nutraceuticals in the Mediterranean Diet: An Epigenetic Diet Model. Cancer Genom. Proteom. 2020, 17, 335–350. [Google Scholar] [CrossRef]
  152. Montano, L.; Maugeri, A.; Volpe, M.G.; Micali, S.; Mirone, V.; Mantovani, A.; Navarra, M.; Piscopo, M. Mediterranean Diet as a Shield against Male Infertility and Cancer Risk Induced by Environmental Pollutants: A Focus on Flavonoids. Int. J. Mol. Sci. 2022, 23, 1568. [Google Scholar] [CrossRef]
Figure 1. CpG island hypermethylation and cancer development. Promoter hypermethylation leads to increased nucleosome occupancy and silencing of tumor suppressor genes, driving cancer progression. Me is methyl group, SAM is S-adenosylmethionine, PRC2 is Polycomb Repressive Complex 2, and RNAPII is RNA Polymerase II. The bold arrow indicates the directional flow of the epigenetic changes leading to cancer development. Created in https://BioRender.com (accessed on 11 September 2025).
Figure 1. CpG island hypermethylation and cancer development. Promoter hypermethylation leads to increased nucleosome occupancy and silencing of tumor suppressor genes, driving cancer progression. Me is methyl group, SAM is S-adenosylmethionine, PRC2 is Polycomb Repressive Complex 2, and RNAPII is RNA Polymerase II. The bold arrow indicates the directional flow of the epigenetic changes leading to cancer development. Created in https://BioRender.com (accessed on 11 September 2025).
Nutrients 17 03690 g001
Figure 2. Altered histone codes in ovarian and breast cancer. Epigenetic regulation of chromatin involves three classes of protein. Writers (protein that add modifications), readers (proteins that recognize and interpret these modifications), and erasers (enzymes that remove them). Dysregulation of these enzymes alters gene expression and contributes to breast and ovarian cancer. The arrows indicate the sequential progression of the epigenetic regulations. Created in https://BioRender.com (accessed on 11 September 2025).
Figure 2. Altered histone codes in ovarian and breast cancer. Epigenetic regulation of chromatin involves three classes of protein. Writers (protein that add modifications), readers (proteins that recognize and interpret these modifications), and erasers (enzymes that remove them). Dysregulation of these enzymes alters gene expression and contributes to breast and ovarian cancer. The arrows indicate the sequential progression of the epigenetic regulations. Created in https://BioRender.com (accessed on 11 September 2025).
Nutrients 17 03690 g002
Figure 3. Paternal macronutrient imbalance such high-fat diet, high-sugar, and low protein diets induce changes in sperm through epigenetic mechanisms. Altered DNA methylation, histone modification, and small ncRNAs signals reprogram early embryonic development and pass to F1–F2, leading to metabolic dysfunction, cancer, and developmental effects. The bold arrow represents the directional flow from paternal diet sperm epigenetic alteration. Created in https://BioRender.com (accessed on 11 September 2025).
Figure 3. Paternal macronutrient imbalance such high-fat diet, high-sugar, and low protein diets induce changes in sperm through epigenetic mechanisms. Altered DNA methylation, histone modification, and small ncRNAs signals reprogram early embryonic development and pass to F1–F2, leading to metabolic dysfunction, cancer, and developmental effects. The bold arrow represents the directional flow from paternal diet sperm epigenetic alteration. Created in https://BioRender.com (accessed on 11 September 2025).
Nutrients 17 03690 g003
Figure 4. Mechanistic schema on how paternal obesity influences female reproductive health. These molecular alterations are passed on during fertilization, leading to compromised early embryonic development such as delayed cell cycles, and subsequent intergenerational health risks. 8-OHdG is 8-hydroxy-2-deoxyguanosine, an oxidized form of DNA that is biomarker for oxidative stress and DNA damage. Upward arrows indicate an increase, and downward arrows indicate a decrease or low in the female phenotype. The arrow outside the box indicates directional flow of the figure. Created in https://BioRender.com (accessed on 11 September 2025).
Figure 4. Mechanistic schema on how paternal obesity influences female reproductive health. These molecular alterations are passed on during fertilization, leading to compromised early embryonic development such as delayed cell cycles, and subsequent intergenerational health risks. 8-OHdG is 8-hydroxy-2-deoxyguanosine, an oxidized form of DNA that is biomarker for oxidative stress and DNA damage. Upward arrows indicate an increase, and downward arrows indicate a decrease or low in the female phenotype. The arrow outside the box indicates directional flow of the figure. Created in https://BioRender.com (accessed on 11 September 2025).
Nutrients 17 03690 g004
Figure 5. Paternal intake of phytochemical influence on sperm epigenetics. This includes changes in DNA methylation, histone modifications, enzyme activity, transcriptome, and sperm quality. These germline changes contribute to a reduced risk of breast cancer in their offspring by decreasing the incidence and tumor volume, delaying latency, and modulating genes associated with tumors. Upward arrows indicate an increase, and downward arrows indicate a decrease in the outcome. Created in https://BioRender.com (accessed on 11 September 2025).
Figure 5. Paternal intake of phytochemical influence on sperm epigenetics. This includes changes in DNA methylation, histone modifications, enzyme activity, transcriptome, and sperm quality. These germline changes contribute to a reduced risk of breast cancer in their offspring by decreasing the incidence and tumor volume, delaying latency, and modulating genes associated with tumors. Upward arrows indicate an increase, and downward arrows indicate a decrease in the outcome. Created in https://BioRender.com (accessed on 11 September 2025).
Nutrients 17 03690 g005
Table 4. Impact of micronutrient deficiencies on sperm epigenetics and offspring health.
Table 4. Impact of micronutrient deficiencies on sperm epigenetics and offspring health.
MicronutrientRole in SpermDeficiency Effects on SpermOffspring/Health ConsequencesOrganismReferences
Folate (B9)One-carbon metabolism, DNA and histone methylation, nucleotide synthesisLow sperm count, increase DNA damageInfertility, congenital malformationsMice, rat[119]
Vitamin B6/B12DNA synthesis, cofactors in homocysteine metabolismChromosomal instability, hypomethylation Altered DNA methylationHuman, mice, rat [133,134]
Vitamin CTestosterone regulation, antioxidant protectionLow motility and sperm count, DNA oxidationProtects against smoking-related sperm DNA damageHuman, mice, rat[132]
Vitamin DVitamin D receptor in sperm/testis, calcium transferAltered morphology, low sperm countInfertility, interacts with epigenetic regulationHuman, mice, rat[132]
Vitamin EProtects DNA from ROS, antioxidantOxidative DNA damage Human, mice, rat[132]
Iron (Fe)Integral to Heme proteins and support DNA/RNA structure Impaired SpermatogenesisDevelopmental and metabolic riskRat[127]
IodineThyroid-dependent spermatogenesisTesticular atrophy with hypothyroxinemia, decrease motility and sperm count Rat, goat[127]
ZincChromatin stability, protamine cross-linking; antioxidant enzymes; transcriptional cofactorsOxidative damage, decrease motility/morphology, poor chromatin integrityOffspring cancer risk through germline DNA damageRat[134,135]
SeleniumSperm maturation, Selonoproteins (GPx-1 cofactor)ROS accumulation, reduces motilityAltered mammary development and increases breast cancer risk in daughtersMice[135]
MagnesiumGlutathione synthesisOxidative DNA damage, energy dysregulation Rat, goat[127]
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

Durojaye, T.J.; Ganguly, S.; Li, Y.; Tollefsbol, T.O. Nutrition-Based Paternal Influence on Gynecological Diseases in Female Offspring via Epigenetic Mechanisms. Nutrients 2025, 17, 3690. https://doi.org/10.3390/nu17233690

AMA Style

Durojaye TJ, Ganguly S, Li Y, Tollefsbol TO. Nutrition-Based Paternal Influence on Gynecological Diseases in Female Offspring via Epigenetic Mechanisms. Nutrients. 2025; 17(23):3690. https://doi.org/10.3390/nu17233690

Chicago/Turabian Style

Durojaye, Titilayomi J., Sebanti Ganguly, Yuanyuan Li, and Trygve O. Tollefsbol. 2025. "Nutrition-Based Paternal Influence on Gynecological Diseases in Female Offspring via Epigenetic Mechanisms" Nutrients 17, no. 23: 3690. https://doi.org/10.3390/nu17233690

APA Style

Durojaye, T. J., Ganguly, S., Li, Y., & Tollefsbol, T. O. (2025). Nutrition-Based Paternal Influence on Gynecological Diseases in Female Offspring via Epigenetic Mechanisms. Nutrients, 17(23), 3690. https://doi.org/10.3390/nu17233690

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