Skip to main content
Pediatric Investigation logoLink to Pediatric Investigation
. 2023 Feb 28;7(2):111–131. doi: 10.1002/ped4.12367

Developmental origins of health and disease: Impact of paternal nutrition and lifestyle

Qiaoyu Shi 1, Kemin Qi 1,
PMCID: PMC10262906  PMID: 37324600

ABSTRACT

Most epidemiological and experimental studies have focused on maternal influences on offspring's health. The impact of maternal undernutrition, overnutrition, hypoxia, and stress is linked to adverse offspring outcomes across a range of systems including cardiometabolic, respiratory, endocrine, and reproduction among others. During the past decade, it has become evident that paternal environmental factors are also linked to the development of diseases in offspring. In this article, we aim to outline the current understanding of the impact of male health and environmental exposure on offspring development, health, and disease and explore the mechanisms underlying the paternal programming of offspring health. The available evidence suggests that poor paternal pre‐conceptional nutrition and lifestyle, and advanced age can increase the risk of negative outcomes in offspring, via both direct (genetic/epigenetic) and indirect (maternal uterine environment) effects. Beginning at preconception, and during utero and the early life after birth, cells acquire an epigenetic memory of the early exposure which can be influential across the entire lifespan and program a child's health. Potentially not only mothers but also fathers should be advised that maintaining a healthy diet and lifestyle is important to improve offspring health as well as the parental health status. However, the evidence is mostly based on animal studies, and well‐designed human studies are urgently needed to verify findings from animal data.

Keywords: Epigenetics, Mental health, Metabolic outcomes, Offspring, Paternal exposure, Telomere length


Paternal exposure to environmental facts including unhealthy diets, malnutrition, and lifestyle during susceptible windows may affect offspring health, development, and chronic diseases, implicating indirect effects on the fetal component of the placenta, telomere length, sperm epigenetic, or seminal fluid protein mechanisms.

graphic file with name PED4-7-111-g002.jpg

INTRODUCTION

It is now well established that environmental stimuli in the early life stage impact offspring's health and development. Thirty years ago, Barker and colleagues first reported their findings on the association between in‐utero environment and cardiovascular disease in later life. 1 Subsequently, studies from population cohorts and animal models have demonstrated that the impact of maternal undernutrition, overnutrition, hypoxia, and stress is linked to adverse offspring outcomes. 2 As a result, Barker's fetal programming hypothesis is completed with the developmental origins of the health and disease concept, which posits that not only exposures to toxins and chemicals, use of drugs, infections, and other stimulations but also imbalanced nutritional status during pregnancy and postnatal early life, increase the risk of disease and dysfunction later in life, especially cardiovascular disease and metabolic comorbidities such as obesity and type‐2 diabetes (T2D). 3 , 4

During the past decade, a growing body of evidence suggests that male exposure to toxins and pharmaceutical drugs, chemical weapons and ionizing radiation, under‐/over‐nutrition, lifestyle factors such as exercise, alcohol, and tobacco consumption, trauma and stress situations, impact fetal development and adult offspring health and disease, beyond effects on sperm quality and infertility. 4 , 5 , 6 , 7 , 8 , 9 , 10 , 11 , 12 The evidence supports the involvement of paternal factors in the origin of health or disease in offspring, and a new concept of paternal origins of health and diseases has emerged, stressing that the impact of paternal environmental conditions could also influence the offspring's health and development. 11 Paternal lineage is responsible for more than just its genetically encoded information. The epigenetic marks are delivered by sperm to the zygote, with evidence pointing towards the involvement of DNA methylation, histone modifications, and non‐coding RNAs (ncRNAs), and further are transmissible through the germline to the next generation. 4 This review focuses on paternal diet and nutrition, lifestyle and related conditions and their potential effects on the health of future generations, and discusses the epigenetic mechanisms responsible for the transfer of information from one generation to the next.

EFFECTS OF PATERNAL DIET AND NUTRITION ON OFFSPRING

Historic studies from human cohorts and animals highlight the notion that paternal nutrition is important, with regard to health in future generations. 5 , 6 , 7 , 8 , 9 The Överkalix cohort study provided evidence for the transmission of effects from early nutritional exposures through the male germ line, showing that the longevity of grandchildren was determined by the grandparent's diet during pre‐puberty. 5 Subsequently, the impact on grandchild cardiovascular, diabetes mellitus and all‐cause mortality, and mental health is confirmed by the Överkalix cohort, 6 , 7 and others. 8 , 9 In a mouse model, a transgenerational effect on metabolic‐ and growth‐related parameters in the offspring was shown if fathers suffered from preconceptional food deprivation. 10 Thereafter, there is strong evidence for the influence of paternal‐specific nutrients on offspring's health and diseases (Table 1).

TABLE 1.

Effects of paternal nutrition and lifestyle on offspring health.

Paternal exposure Subjects Offspring outcomes Potential mechanisms References
Food supply
Human Survival shortening in male offspring with a surfeit of food for the paternal grandfather during the slow growth period (SGP). 5
Mortality risk ratio of grandsons and granddaughters linked to food access for paternal grandfather and grandmother respectively, with exposure during SGP. 6, 8
Low cardiovascular disease mortality with paternal and paternal grandmother famine, high diabetes mortality with a surfeit of food for paternal grandfather during SGP. 7
Higher mental health scores for the third generation, if their paternal grandfather (maternal grandmother) was exposed to famine during SGP. Evolutionary correction and possible epigenetic modifications of the gametes. 9
Mouse Decrease in serum glucose with paternal food deprivation. 10
Protein intake
Low Mouse Impaired cardiovascular and metabolic function. Modified DNA methylation, histone modifications, and RNA methylation in F1 testes; elevated TNF‐α, and reduced calcium signaling and metabolism in male offspring. 13, 16
Perturbed placental development, fetal growth, and skeletal development. Altered expression of Dnmt3b, Dnmt1, and Dnmt3L, and imprinted genes. 14, 15
Altered lipid metabolism in adipocytes. Altered genes are involved in lipogenesis and oxidation. 17
Altered cholesterol and lipid metabolism reprogramming of metabolic gene expression. Altered genes associated with lipid and cholesterol biosynthesis, DNA methylation, histone H3K9me2. 18, 175
Increased adiposity, glucose intolerance, nonalcoholic fatty liver disease, and altered gut bacterial profiles. Paternal sperm global hypomethylation is associated with reduced testicular expression of DNA methylation and folate‐cycle regulators. 19
Increased risk of breast cancer and tumors. Alterations in noncoding RNAs in mammary glands and tumors. 20
High Mouse Beneficial for body composition, insulin sensitivity, and gut microbiota; prevention of obesity and diabetes. β cell plasticity; altered expression of Dnmt1 and Dnmt3b intergenerationally. 21, 22
High Human and general population Increased risk for T2D, nonalcoholic fatty liver disease, and cardiovascular diseases. mTOR activation and reduced insulin sensitivity. 23, 24
High‐fat intake and/or obesity
Mouse Delayed development, reduced placental size and litter size, and disturbed embryo growth and development. Increase in spermatogonia and S‐phase population, decrease in Dnmt transcript and global DNA methylation levels, and histone H3K9me2 in germline 28, 29, 33, 34
Increased body weight; metabolic dysregulation; repaired insulin sensitivity and adverse effects on β‐cell development and function. Changes in adipokine promoter epigenetics in the offspring for multiple generations; sperm miRNA 193b, 204 and H3K9me2, altered Nr1h3 DNA methylation at paternal germ cells; altered Igf2/H19 DNA methylation paternal sperm and offspring liver; altered DNA methylation signature and expression of sncRNA, including miRNA and piRNA in paternal spermatozoa and offspring; enrichment in H3K4me1, H3K4me3, and H3K9/K27me3 in genes implicated in offspring metabolic, inflammatory and developmental processes. 32, 35, 38, 39, 40, 41, 173, 174, 180, 181
Increased risk of chronic kidney disease. Decreased lncRNA XR‐146683.1 expression and increased Cd300lf promoter methylation 45, 46
Human High birth weight, and childhood BMI z score; increased risk of LGA, or SGA, and macrosomia Altered DNA methylation in cord blood. 30, 31
High sugar intake
Mouse, Drosophila Disturbed liver metabolism and function; alterations of gut microbiota. Increased NFκB, SOCS3, JNK, TNF‐α, IL1‐β, and IL6 levels with upregulated expression of Srebp‐1c and Fas. 47, 48, 52
Obesity and abnormal energy metabolism. Altered sperm‐borne mRNAs; H3K9/K27me3‐dependent reprogramming of metabolic genes in germline and zygotic windows. 53, 54, 55
Abnormal adipokines, BP, and uric acid concentrations indicate elevated cardiovascular risk. 49
Decrease in reward‐seeking; anxiety‐like behavior; high body weight. 50, 51, 52
Methyl donor intake
Low folate intake Mouse Reduced pregnancy rate, post‐implantation loss, and increased birth defects. Altered imprinted gene methylation in paternal sperm and offspring; sperm histone H3 methylation or DNA methylation. 59, 60
Low intake of folate, methionine, and choline Rat, Mouse Increased risks of development and chronic diseases, depression‐like behavior, anxiety‐like behavior, and fear memory. Differential expression of memory‐related genes (Camk2α and PP1) and promoter methylation of the PP1 gene. 61, 62
Low intake of methionine, folate, betaine, choline Human Longer gestation and altered birth weight. Paternal global DNA hydroxymethylation; offspring global and IGF2 DNA methylation. 56, 57
High intake of methionine, betaine, choline, and vitamin B Mouse Deficits in learning and memory; increase in tumor volume and hepatic lipids. Altered spermatozoal miRNAs and lncRNAs, and offspring Kcnmb2 promoter methylation. 64, 65, 66
High vitamin B12 intake Human Associated with a higher risk of childhood brain tumors. 58
Physical activity
Exercise Mouse Decreased risk for obesity with higher glucose tolerance and insulin sensitivity; lower inflammation and oxidation; improved metabolic profile in the liver. Normalized abundance of X‐linked sperm microRNAs; induced expression of pancreatic microRNAs (let7d‐5p, 194‐5p); altered expression of metabolic genes in the testes and epididymis of the sires. 71, 72, 74, 182
Long‐term exercise Mouse Increased risk for obesity with glucose intolerance, and insulin insensitivity. Altered methylation patterns and micro‐RNA content in the sperm of fathers. 73
Climbing resistance exercise Mouse Muscle homeostasis maintenance; modulation on calcaneal tendon proteome and left ventricle proteome. Altered expression of genes relevant for key skeletal muscle remodeling pathways and inflammatory profiles; positive regulation of protein metabolism. 75, 76, 77
Treadmill exercise Mouse Improved spatial learning and memory capability. Increased expression of Bdnf. 78
Wheel running exercise or cage activity Mouse Modulation on placental weight, inflammation and nutrient transporter, and fetal weights. Decreased miRNA 193b expression and increased miRNA 204 in paternal sperm, with decreased expression of H3K9me2. 31
Active smoking
Human or mouse Increased body fat and weight, with a transgenerational effect across four generations. Altered paternal spermatozoal miRNAs. 6, 79, 80, 81, 90
Human High risk for childhood asthma; lower lung function. Altered lung and gonad DNA methylation and histone 3 and 4 acetylations, and PPARγ expression. 86, 88, 89
Poorer mental health (particularly hyperactivity/ADHD). 82
An increase in SGA, birth defects such as congenital heart defects, orofacial clefts, cancers, brain tumors, and acute lymphoblastic leukemia. 83
Prenatal exposure to maternal, but not paternal, tobacco smoking is associated with tobacco smoking and conduct disorder symptoms in offspring. 91, 92
Alcohol consumption
Mouse A prolonged period of fetal gestation and an increased incidence of intrauterine growth restriction; have deleterious effects on embryonic development. Altered expression of genes within the pro‐fibrotic TGF‐β signaling pathway and the LiverX/RetinoidX/FarnesoidX receptor pathways; alterations in DNA methylation, ncRNAs (tsRNA, piwiRNA), as well as damage in DNA integrity in the testicular germline and sperm. 93, 94, 95
Increase in activity and sensorimotor integration deficits, decreased balance, coordination, and short‐term motor learning; altered behavioral responses and responses to repetitive mild traumatic brain injury. Altered sensitivity to the GABAergic drug midazolam; modified expression of many genes and telomere length. 96, 97, 98
Human Higher risk of substance‐related disorders; increased risks of anxiety/depression and sleep problems in girls at age 4, and somatic complaints and rule‐breaking behaviors in boys at age 6; increased fetal birth defect risk; increased risk of total congenital heart diseases; shorter anogenital distance especially in boys. 100, 101, 102, 103, 104
Drug exposure
Human and mouse Negative impacts on future generations, ranging from behavioral to molecular and physiological changes. Regulation on the hypothalamic‐pituitary‐adrenal axis with increased corticosterone levels, as well as changes in dopaminergic signaling; changes in imprinted genes and microRNAs and genes related to glutamatergic and neurotrophic factor signaling. 12, 105, 106
Age
Increasing paternal age Human and mouse Birth defects include musculoskeletal anomalies; psychiatric disorders like autism, schizophrenia and bipolar disorders, and childhood cancer. Epigenetic changes, DNA mutations along with chromosomal aneuploidies in sperm and testes; longer telomere length. 83, 112, 113, 114, 115, 118, 119, 120
Increasing paternal age Human Risk factors for early‐onset type 1 diabetes and obesity. 110, 111

Dietary protein

The protein content of the paternal diet has been demonstrated to influence offspring in many aspects mostly by animal studies. Paternal low protein diet (LPD) increases female‐to‐male ratio and fetal weight, with reduced placental weight and junctional zone area, and perturbed fetal skeletal development whereas dietary methyl‐donor supplementation alters placental morphology and gene expression differentially to that observed with the LPD alone. 13 , 14 , 15 Also, paternal LPD impairs adult offspring's cardiovascular function with relative hypotension and elevated heart rate, by modifying renin‐angiotensin system activity and calcium signaling. 13 , 16 Studies on offspring metabolism indicate that pups derived from fathers with the LPD are more prone to increased adiposity and body weight, impaired glucose tolerance, perturbed hepatic gene expression, symptomatic of nonalcoholic fatty liver disease, altered lipid metabolism and gut bacterial profiles. 17 , 18 , 19 Furthermore, daughters of LPD fathers have higher rates of mammary cancer, with the AMP‐activated protein kinase pathway suppressed and consequent mammalian target of rapamycin signaling activated. 20 Conversely, in animal models, consumption of the high protein diet (HPD) by fathers improves body composition, insulin sensitivity, β cell mass and plasticity, circulating satiety hormones, and cecal short‐chain fatty acids with altered gut microbial composition in the offspring. 21 , 22 Although few studies have been reported on the association of paternal HPD with offspring health in general populations, subjects with an HPD resulted in higher branched‐chain amino acid concentration may activate the mammalian target of rapamycin (mTOR) complex 1 and reduce insulin‐stimulated glucose uptake, and thus leading to an increased risk for T2D, nonalcoholic fatty liver disease, and possibly cardiovascular diseases. 23 , 24 , 25 Further studies demonstrate that a higher intake of animal protein instead of plant protein is associated with insulin resistance and risk of prediabetes and T2D, 25 and that compared to a conventional diabetes diet, an isocaloric carbohydrate‐reduced high‐protein diet reduces HbA1c and hepatic fat content in individuals with T2D. 26 A hypercaloric (50% higher) high protein/high sucrose and low‐fat diet increases ectopic fat accumulation in the liver and muscle, and reduces energy expenditure, although its effect is smaller compared to an isocaloric low protein/ high sucrose and high‐fat diet. 27 Therefore, the relationship between protein intake and health is complex and influenced by overall diet composition, total energy intake, and even body weight. The habitual high‐protein intake with no reductions in carbohydrate and energy intake may contribute to the increased disease risk in themselves, 23 and offspring when it occurs in paternal preconception.

High‐fat intake

Besides the effects on subfertility in subsequent generations, 28 paternal obesity induced by a high‐fat diet (HFD) has significant negative effects on blastocyst attachment, growth, and implantation due to increased spermatogonia and S‐phase population, and DNA hypomethylation, resulting in delayed development, reduced placental size and litter size, and disturbed embryo growth and development. 29 , 30 , 31 , 32 , 33 Both in human and animal models, paternal HFD may impact offspring's postnatal obesity and metabolic outcomes, but the results are inconsistent. 34 , 35 , 36 , 37 The whole‐body and skeletal muscle insulin sensitivity in offspring early life is enhanced by paternal HFD feeding, 34 but metabolic syndrome‐like phenomena are induced, including weight and fat gain, glucose intolerance, hypertriglyceridemia, abnormal adipocytokine levels, and hypertension in adulthood of offspring, via endoplasmic reticulum stress and elevated expression of phosphoenolpyruvate carboxykinase. 35 , 38 , 39 Furthermore, programming by paternal HFD has demonstrated adverse effects on β‐cell development and function, and numerous adipose metabolic pathways in offspring, 40 , 41 and an additive effect on offspring for two generations. 35 However, in a rat study with parental HFD food intake at mating/conception, maternal but not paternal HFD predisposes for offspring metabolic health (obesity, hyperleptinemia, hyperglycemia, insulin resistance) over generations. To note, this inconsistent result does not exclude the possibility that paternal HFD exposure occurs at earlier ages, that is, during critical developmental stages (gametogenesis/adolescence), for a longer time period, and/or the severity of obesity‐induced. 42 Further investigation on the brain has demonstrated that male exposure to the HFD may contribute to stable behavioral variation among females in courtship, maternal care, and anxiety‐like behavior, 43 as well as cognitive impairments in the offspring due to altered methylation and expression of the BDNF gene. 44 Moreover, paternal HFD‐induced obesity is associated with renal triglyceride accumulation and histological changes in tubules, accompanied by upregulated expression of acetyl‐CoA acetyltransferase 1, and epigenetic alterations of renal mRNA expression of genes (Enpp6, Tmem144, Cd300lf, and Actr3b) in offspring, implying a higher risk of chronic kidney disease. 45 , 46

High sugar intake

The consumption of a high‐sugar diet (HSD) or high‐sugar/fat diet by fathers is associated with adverse effects on liver metabolism and function, and alterations of gut microbiota in adult male offspring. 47 , 48 Consumption of a fructose‐rich diet by the father, the mother, or both negatively affects blood pressure, adipokines, and uric acid concentrations of mature offspring, which correspond to markers of elevated cardiovascular risk in progeny. 49 Le et al. found that paternal sucrose self‐administration experience results in a decrease in reward‐seeking and induces anxiety‐like behavior in the offspring of rats. 50 Also, paternal HFD and HSD cause significant phenotypic changes in the offspring, including higher body weight, hypothalamic inflammation, altered energy homeostasis, gut microbiota, stress reactivity, and social behavior. 51 , 52 , 53 In a Drosophila model, paternal sugar‐induced intergenerational metabolic reprogramming (lipid metabolism, immune response, obesity, etc.) is attributable to modification of sperm and offspring chromatin state, and histone methylation associated gene transcription, and this similar pathway may be involved in obesity susceptibility and phenotype variation in mice and humans. 54 , 55

Vitamin or methyl donor in diet

There are emerging shreds of evidence that paternal vitamin or methyl donor (methionine, folate, betaine, choline) intake impacts the offspring's health and disease onset. Human studies demonstrate that an increase in paternal folate intake is associated with longer gestation, 56 and that methionine/betaine intake is negative, but choline intake is positively associated with birth weight. 57 In a case‐control study, paternal preconception high intake of folate, B6 or B12 is associated with an increased risk of childhood brain tumors. 58 In rodent models, paternal folate deficiency or low intake decreases sperm counts 59 and alters the sperm and placenta epigenome and genes’ expression implicated in the development of chronic diseases such as cancer, diabetes, autism, and schizophrenia, thus linking to adverse pregnancy outcomes, such as a reduced pregnancy rate, post‐implantation loss, increased birth defects, increased postnatal‐preweaning mortality and risks of developmental and chronic diseases. 59 , 60 Offspring of paternal methyl donor (folate) deficient diet shows greater depression‐like behavior, anxiety‐like behavior, and fear memory, accompanied by differences in the expression of memory‐related genes and gene promoter methylation in the hippocampus. 61 , 62 Paternal over intake of folic acid and methionine also results in deficits in hippocampus‐dependent learning and memory, impaired hippocampal synaptic plasticity and theta oscillations, and impaired metabolic functions in the offspring. 63 , 64 , 65 Meanwhile, a stepwise increase in tumor volume and hepatic triglycerides and cholesterol in female offspring has been found with increasing paternal B vitamin intake prior to mating. 66 Studies on other vitamins display that paternal consumption of vitamin E can improve offspring's negative parameters resulting from feeding fathers with trans‐fatty acids. 67

To summarize, the above‐mentioned imbalanced nutrient intake, encompassing high‐fat/sugar intake, lower intake of fiber, vitamins, minerals, and so forth in populations including preconception fathers, which are resulted from refined grains, processed and red meat, desserts, and drinks in the prevailing western dietary pattern, may exert a negative impact on offspring health outcomes.

EFFECTS OF PATERNAL LIFESTYLE ON OFFSPRING

A growing body of literature has provided strong evidence for a clustered and co‐occurring unhealthy pattern such as smoking, alcohol, physical inactivity, and poor diet in adult populations, particularly males and those with greater social disadvantage. 68 Beyond its contribution to the development of chronic diseases such as cardiovascular diseases and cancer in adults themselves, this clustered behavioral pattern may adversely affect sperm, which modulates both the genotype and phenotype of progeny in intergenerational or transgenerational inheritance.

Physical activity

Paternal exercise training, mostly in animal models, has been shown to impact various physiological systems, benefit cognitive development, and combat the development of metabolic dysfunction in offspring. 69 , 70 Increases in risk of T2D and metabolic impairments in offspring when the father consumes an HFD, can be normalized when the father also exercises during preconception. This protection may occur by increasing insulin signaling potential within offspring skeletal muscle, normalizing adiposity, attenuating fibrosis, inflammatory profile, and redox status, and restoring pancreatic islet cell morphology and ncRNA expression in sperm. 70 , 71 , 72 However, Murashov et al. reported that offspring from fathers subjected to long‐term exercise are more susceptible to low energy expenditure and increased risk for obesity in mice, potentially via microRNA (miRNA)‐induced modification of sperm. 73 Effects on other systems demonstrate that paternal exercise training can improve the metabolic profile in the liver of the progeny, thereby ameliorating the negative effects of obesity. 74 Meanwhile, key skeletal muscle remodeling pathways and inflammatory profiles are altered in offspring from exercised fathers, with positive regulation of proteins essential for the homeostasis maintenance of tendon integrity. 75 , 76 Moreover, paternal regular training demonstrates to be an important intervention capable of inducing beneficial effects on the left ventricle proteome and associated homeostasis maintenance in offspring. 77 As well, paternal treadmill exercise improves the spatial learning and memory capability of male pups, which is accompanied by increased expression of BDNF and Reelin. 78 In addition, in a mouse model of paternal HFD, the reduction in placental and fetal tissue weights, up‐regulation in nutrient transporter gene expression, and inflammatory gene expression, are reversed by paternal exercise in a sex‐specific manner, which may reduce offspring T2D risk. 31

Smoking

The Avon Longitudinal Study of Parents and Children cohort study first reported an association between prepubertal (<11 years) start of father's smoking and raised body mass index in adolescence in their sons, 6 , 79 with a transgenerational effect across four generations. 80 In the Respiratory Health in Northern Europe, Spain, and Australia study, fathers’ smoking onset before conception of the offspring (onset ≥15 years) is associated with higher body mass index in the offspring's adulthood. 81 Systematic reviews and meta‐analyses demonstrate that paternal smoking during pregnancy is related to greater odds of attention deficit hyperactivity disorder, 82 an increase in small for gestational age, birth defects such as congenital heart defects and orofacial clefts, cancers, brain tumors, and acute lymphoblastic leukemia in offspring. 83 Recently, to rule out maternal passive smoke, several studies have indicated that just preconception paternal smoking can affect offspring's health and development. For example, it has been reported that only preconception smoking exposure or both pre‐ and post‐conception exposure is significantly associated with offspring overweight/obesity in boys. 84 Preconception paternal smoking may be associated with congenital heart diseases, limb abnormalities, and neural tube defects in the offspring, and infants whose fathers stopped or decreased smoking during pregnancy are at lower risk of these defects. 85 A hospital‐based birth retrospective observational birth cohort study showed that either only paternal smoking or only maternal smoking increases the risk of childhood asthma, and paternal cessation of smoking during pregnancy reduces the risk of asthma regardless of maternal smoking. 86 Another study reported that the risk of childhood asthma is highest if the father starts smoking before age 15 years, even if he stopped more than 5 years before conception. 87 Additionally, not only fathers’ smoking in prepuberty but also paternal grandmothers’ smoking in pregnancy may cause lower lung function in three generations. 88 Mechanistic studies have shown that paternal smoking exposure regulates gonad DNA methylation and histone 3 and 4 acetylations, or spermatozoa miRNAs, which may become permanently programmed and transferred through the germline to subsequent generations, thus enhancing the risks of respiratory‐ and metabolic‐related diseases in offspring. 89 , 90 These data suggest that, unlike other factors, use of alcohol and caffeine or physical activity, which impact offspring outcomes via direct germline transmission, paternal smoking may affect intrauterine development by both direct pathways (germline transmission) with preconception smoking and indirect pathways (altering maternal environment and physiology) with post‐conception smoking and resulted in maternal passive smoke. However, an Australian birth cohort study has found insufficient evidence for an association between paternal smoking during pregnancy and the risk of tobacco smoking and conduct disorder symptoms in offspring. 91 As well, a meta‐analysis reveals that offspring exposed to maternal but not paternal prenatal tobacco smoking are at an increased risk of tobacco smoking/dependence. 92 These data suggest that paternal smoking affects offspring outcomes in a disease‐specific manner. Therefore, further differentiations between the impact on offspring outcomes of paternal smoke exposure in the preconception stage, and during and after pregnancy, remain to be a challenge.

Alcohol exposure

Paternal alcohol exposure (PAE) is found to cause histological and epigenetic changes, as well as damage in DNA integrity in the testicular germline and sperm, which give rise to deleterious effects on embryonic development and to testicular and spermatic changes in the offspring. 93 , 94 , 95 Besides, PAE can induce several developmental aberrations and alters learning and locomotor activity, affective behavior, and sensitivity to the rewarding properties of alcohol in offspring. 12 Notably, the PAE effects are in a sex‐specific manner with a negative impact on offspring neocortical development, causing deficits in activity and sensorimotor integration deficits and decreased balance, coordination, and short‐term motor learning postnatally. 96 Female and male offspring display unique responses to repetitive mild traumatic brain injury and paternal experiences (exercise, caffeine, and alcohol), showing male instead of female offspring are affected by paternal alcohol treatment. 98 Also, reduced sensitivity to stress‐induced hyperthermia is induced by PAE, selectively in male offspring. 97 Additionally, PAE negatively affects fetal growth in the male offspring to a greater extent than the females, and the growing deficits in males may persist into adult life with insulin hypersensitivity, increased markers of hepatic fibrosis, and alterations in immune signaling. 93 By contrast, the male offspring of alcohol‐exposed sires are found resistant to HFD‐induced obesity and improved glucose homeostasis via programmed increases in hepatic LXRα expression. 99 In humans, preconceptional paternal alcohol consumption increases offspring risks of substance‐related disorders, anxiety/depression, sleep problems, somatic complaints and rule‐breaking behaviors, fetal birth defects including total congenital heart diseases, and shorter anogenital distance. 100 , 101 , 102 , 103 , 104

Drug exposure

An extensive body of literature firmly establishes that preconception exposure to drugs of abuse has impacts on future generations, ranging from behavioral to molecular and physiological changes. 12 , 105 , 106 Human literature suggests that paternal exposure to the drug (alcohol, cocaine, opioids, cannabinoids, and nicotine) has inconsistent or even contradictory results on vulnerability to drug abuse in offspring, but a few similarities do arise. 105 Preconception exposure, particularly in the case of ethanol, cocaine, and opiates, decreases addiction to that particular drug in the offspring, but offspring of morphine‐exposed dams showed a higher propensity to take cocaine. Another area that revealed consistencies across studies involves hypothalamic‐pituitary‐adrenal axis regulation with increased corticosterone levels, as well as changes in dopaminergic signaling and reward circuitry in the brain. 106 Moreover, drug‐sired offspring show epigenetic‐mediated changes in imprinted genes and miRNAs and genes related to glutamatergic and neurotrophic factor signaling, which, in some instances, occur simultaneously in sire sperm and the brains of offspring, resulting in developmental and physiological abnormalities, deficits in cognitive and emotional domains and addiction‐like behaviors. 12

PATERNAL AGE

Since the correlation between increasing paternal age and genetic defects and disorders was first suggested in the late 1800s by Weinburg and strengthened in 1955 by Penrose, many other genetic disorders with single DNA errors have been considered to be associated with increasing paternal age, such as achondroplasia and osteogenesis imperfecta, as well as neurofibromatosis and Marfan syndrome. 107 , 108 Thereafter, a series of studies demonstrate an association of advanced paternal age (>50 years) at conception with infertility and adverse outcomes, which is less pronounced as compared to the effect of advanced maternal age showing increased risks of prolonged time to pregnancy, infertility, spontaneous abortions, and trisomy (>35 years), and risks of preterm births and stillbirths (>40 years). 109 More recently, an association with the risk of more common disorders, diabetes mellitus, 110 obesity, 111 birth defects including musculoskeletal anomalies, psychiatric disorders like autism, schizophrenia and bipolar disorders, and childhood cancer in offspring is increasing with advanced paternal age, particularly when they are 50 years or older. 83 , 112 , 113 , 114 , 115 , 116 , 117 In a large cohort study in Swedish, with every 10‐year increase in paternal age at the time of conception, the risk of an offspring having schizophrenia increased by 1.47 times, while offspring with younger fathers (<21 years) were also at a higher risk of schizophrenia and attention‐problems compared to the fathers aged 21–24 years at the time of conception. 118 , 119 Thus, paternal age shows an inverted U‐shape relation with neurodevelopment and psychiatric disorders, with the offspring of younger and older fathers at a disadvantage. 115 , 118 , 119 By contrast, Frans et al. found that an old grandmother's age increased the risk of schizophrenia in a grandchild but not the grandfather's age. 120 In addition, several large studies have evaluated the risk of leukemia and cancers in the offspring of fathers with advanced age, indicating an increased risk of acute lymphoblastic leukemia 116 , 117 but no protective effect on the risk of acute myeloid leukemia, 117 , 121 and decreased risk of cancers of the female genitalia as well as cancers of the respiratory and intrathoracic organs in offspring. 112 , 113 Although the current data implies that aging impairs sperm quality, which can affect fertility status and offspring outcomes, the results are conflicting. Given that most aged fathers are partnered with aged mothers, the complications of confounding factors, and the lack of systemic definitions and study design has contributed to the difficulty in interpreting data. Also, there is no clear definition of advanced paternal age in the current literature. 114 A survey on assistant reproduction technology (ART) in European countries indicates that the legal maximum age for sperm donation ranges from 35 years old in Hungary, Kazakhstan, Russia, and Slovakia to 55 years old in Slovenia, and that male maximum age for ART access is legally set in Portugal (60 years) and is recommended in Finland (60 years) and Sweden (56 years). 122 Therefore, advanced paternal age should be evaluated while accounting for normal or abnormal semen parameters, the age of the female partner, and the mode of conception.

MECHANISMS

The environment influences the health and well‐being of progeny by working through the germline to introduce spontaneous genetic mutations as well as a variety of epigenetic changes. In evolutionary terms, these changes create the phenotypic diversity that fuels the fires of natural selection. However, such variation may also generate a plethora of pathological disease states ranging from dominant genetic disorders to an increased risk of developing diseases or metabolic disorders in the next generation(s). 123 , 124 Unique maternal factors include altered structure/function of reproductive organs, alterations in the vaginal or gut microbiome, mitochondrial DNA (mtDNA) inheritance, placental function, or epigenetic phenotypes in female germ cells. By contrast, unique paternally mediated effects on offspring implicate indirect effects on the fetal component of the placenta, telomere length (TL), sperm epigenetic, or seminal fluid protein mechanisms. 125 , 126 Herein, we focus on genetic/epigenetic information and/or shorter TL that are associated with the transgenerational nature of phenotypic changes observed in offspring. 123

Telomere length

Mammalian telomeres are composed of non‐coding DNA repeats that form nucleoprotein complexes and are located at the extremities of chromosomes. They preserve genomic integrity by protecting chromosome ends from degradation as well as preventing end‐to‐end fusions. 127 TL inherited by the father, is heterogeneous within cells presenting a Gaussian distribution and undergoes progressive age‐dependent shortening in somatic cells while elongating in the male germline. 128 , 129 Several large cohort studies confirm that a strong positive association exists between paternal age at birth and participant leukocyte TL, with a weak inverse or no association with maternal age, 129 , 130 , 131 and that a 1‐year increase in father's age corresponds to a 0.26% increase in offspring TL at age 53. 132 A cross‐sectional study recruited subjects from 30 to 80 years of age shows that an increase of 17–22 bp is seen in children's TL per year of increasing paternal age. 130 , 133 Moreover, the association of paternal age at birth with longer TL in offspring is cumulative across multiple generations. 134 , 135

The possible mechanisms underlying the effect of paternal age at conception on offspring TL, include sperm telomere extension due to telomerase activity, and age‐dependent changes in the spermatogonia stem cell population, representing an intergenerational adaptation with increased maintenance effort to lower extrinsic mortality and delay reproduction. 136 , 137 While TL dynamics in the growing fetus/newborn are influenced by polymorphisms in the maternal mtDNA in the end, the paternal age at conception effect on the offspring TL may largely (and ironically) reflect variation in the mitochondrial genome that is derived from the offspring's paternal grandmother. 128 In addition, telomere biology varies considerably across species 138 , 139 and the effect of paternal age at conception on TL appears to vary considerably as well. 136 , 140 Thus, even strong experimental evidence of the biological mechanisms accounting for the paternal age at conception effect in one species should not be extrapolated uncritically to other species. 135

The length of telomeres becomes shortened after each chromosomal replication event 127 and occurs concurrently with aging in most proliferating tissues. 141 It has been found that short TL are implicated in instigating cell senescence, as well as genetic and epigenetic instability that increase the risk of cancer, 142 and further are transmitted to future generations through their gametes. 143 Numerous studies have shown that telomere shortening is associated with aging and aging‐related diseases, including AD, atherosclerosis, and chronic obstructive pulmonary disease. 144 Reduced TL in early childhood is independently associated with arterial wall thickness in later childhood, indicating a marker of vascular disease risk. 145 Despite consistency across studies on the association between paternal age and offspring TL, it is needed to further assess the impact of this association on offspring health and aging outcomes.

Epigenetics

Although DNA damage or mutations are often suggested as the biological background for father‐child effects from various environmental exposures, it is confirmed that the offspring's epigenetic profile and health status are influenced by paternal pre‐conceptional insults, such as exposures to endocrine disruptors or toxins, ionizing radiation, 146 and nutritional status. 18 , 147 , 148 , 149 Ng et al. first reported that the female offspring mice from fathers with an HFD has an early onset of impaired insulin secretion and glucose tolerance, with altered expression of several associated genes and DNA hypomethylation. 147 Meanwhile, Carone et al. found that male mice consuming an LPD from weaning until sexual maturity produce offspring with increased methylation at the key lipid transcription factor PPARα in the liver. 18 Offspring of pre‐diabetic fathers exhibit impaired glucose tolerance and insulin insensitivity, altered expression of genes involved in glucose metabolism, and differential DNA methylation patterns in the pancreatic islets. 150 Subsequently, in humans, the Newborn Epigenetics Study shows significant differences in DNA methylation at differentially methylated regions (DMRs) of several imprinted genes (IGF2, H19) if the father is obese. 148 , 149 DNA methylation marks are known to establish during gametogenesis, and deregulation of methylation at DMRs is related to chronic diseases or metabolic disorders in the offspring. 151 , 152 Since then, an increasing number of studies demonstrate that the information transferred from sire to progeny may occur through epigenetic marks in sperm, encompassing alterations in DNA methylation, histone modification, and/or small ncRNAs (Table 1). 123

DNA methylation

DNA methylation, a process by which a methyl group is added to a cytosine residue at cytosine‐guanine dinucleotides as well as other dinucleotide pairings, 153 is an important regulator of gene expression globally involved in gene regulation, and more specifically, in transposon silencing, genomic imprinting, maintenance of genome integrity or X chromosome inactivation. 154 , 155 During mammalian life, DNA methylation is a dynamic modification that is erased and reset during two major genome‐wide epigenetic reprogramming events. One takes place in primordial germ cells (PGCs) during gametogenesis with genome‐wide DNA demethylation, removal and resetting of parental imprints, histone modifications, and inactive‐X‐chromosome reactivation. The other one occurs in the preimplantation embryo when cells experience DNA demethylation, the removal, and resetting of parental imprints, and histone modifications. 154 , 155 , 156 Epidemiological studies and various animal models demonstrate that DNA methylation in sperm and offspring exhibits alterations when the father is exposed to environmental stress, such as diet, 156 mental stress, 157 , 158 and metabolic disorders. 159

Gene imprinting, a form of DNA methylation, plays an important role in modulating fetal‐placental growth and resource acquisition, postnatal growth and energy homeostasis, and transgenerational inheritance. 160 , 162 Imprinted genes together with transposable elements and some regions of the male pronuclei may escape the demethylation process during gametogenesis and preimplantation and pass through to descendants. 163 , 164 Many studies show that methylation alterations in DMRs of imprinted genes are found in exposed sires’ sperm and their offspring. 165 , 166 , 167 Abnormalities in imprinting genes caused by epigenetic aberrations, mutations, and deletions can result in loss of imprinting and imprinting disorders, such as complex syndromes of neurodevelopmental disabilities, cognitive problems, and metabolic diseases. 168

Histone modification

The role of histones in conveying epigenetic information in mature spermatozoa was doubted because a few studies have focused on these epigenetic marks. It is clear that most histone proteins are replaced by protamine during late spermatogenesis, facilitating a highly condensed state of the chromosomes, repressing transcriptional activity, and preventing DNA damage. 169 However, 2% and 5%–10% of the mouse and human histones persist in the sperm nucleus and remain unchanged, respectively. 170 , 171 , 172 The modifications present on these histone proteins in sperm provide one mechanism for epigenetic transmission, that relates to the selective establishment of specific DNA methylation patterns of developmental genes, including imprinted genes, miRNAs, and homeobox genes. 172 For example, paternal HFD or HSD induces H3K4me1 enrichment at transcription regulatory genes, and H3K4me3, H3K9/K27me3 in genes implicated in metabolic, inflammatory, and developmental processes in the offspring, which are associated with metabolic dysfunction and corresponded to genes enriched in embryos. 55 , 173 , 174 The LPD‐induced and ATF7‐dependent histone H3K9me2 in testicular germ cells play an important role in paternal diet‐induced metabolic reprogramming in offspring. 175

Noncoding RNA

It is known that regulatory ncRNAs are involved in transcriptional, posttranscriptional, and translational regulation for gene expression by interacting with histone modifications and the DNA methylation status of genes. 176 , 177 Two large subsets of ncRNAs are long ncRNAs and short ncRNAs (sncRNAs) like miRNAs, short interfering RNAs (siRNAs), and piwi‐interacting RNAs (piRNAs). 178 Recently, sncRNAs in sperm have seemed crucial to the transmission of paternal epigenetic information to offspring. 179 The alteration in the abundance of specific ncRNAs in sperm, induced by paternal environmental exposure including HFD and drugs, impacts the development of the embryo following fertilization and ultimately, behavioral and metabolic outcomes in the offspring. 106 , 179 , 180 , 181 , 182 When dysregulated ncRNAs in the sperm of male animals with metabolic diseases are injected into fertilized embryos sired by healthy parents, the offspring display behavioral and physiological changes comparable to those offspring born from sires with metabolic diseases. 183 , 184 In another study, paternal exposure to the stress hormone corticosterone altered miRNA‐98, miRNA‐144, and miRNA‐190b, which may interact with IGF2 and BDNF in the sires’ sperm, resulting in anxiety behavior increased in the offspring. 185 However, more studies should focus on the origin of RNA alteration, the target of RNA regulation, and their roles in preventing offspring diseases. 179

Effects from both sperm and seminal plasma

Besides the effects of sperm, maternal health, and post‐fertilization development during early pregnancy may be influenced by the seminal plasma, which contains bioactive signaling factors that encourage uterine remodeling by inducing a maternal immune response, including the clearance of excess sperm and microorganisms from the vagina and cervix, endometrial cytokine production, immune cell infiltration and activation in the reproductive tract. 186 Being different from sperm‐mediated embryo programming, the seminal plasma impacts offspring growth and metabolic and vascular health via the uterine environment. Several human and animal studies have observed that repeated exposure to a partner's seminal plasma leads to better fertilization, lower prevalence of preeclampsia and inflammation in gestation, and altered expression of genes involved in the cell cycle and growth, indicating that an increased duration of exposure to partner's semen may be beneficial for a woman's gestational wellbeing. 187 , 188 , 189 , 190 , 191 , 192 , 193 In contrast, less time exposure to a partner's semen or changing partner places a woman at an increased risk of developing preeclampsia. 194 , 195 Mice without seminal fluid have reduced fertility, impaired glucose tolerance, and exhibited obesity, placental hypertrophy, and hypertension in male offspring. 196

The novel data from animal studies reveal the impact of sub‐optimal paternal nutrition on offspring well‐being, programming offspring health through both sperm and seminal plasma‐specific mechanisms over successive generations. 197 In a mouse model, besides sperm contribution, seminal plasma‐specific pathways are also indicated to mediate the effects of paternal preconception LPD on offspring health programming, including growth, adiposity, glucose metabolism, liver metabolic function, and gut microbiota. 19 Likewise, either LPD sperm or seminal fluid at conception impairs adult offspring's vascular function in response to both vasoconstrictors and dilators with modified expression profiles of epigenetic regulators in adult F1 male testes. 23 Other components of semen, including exosomes and its containing sncRNAs, may be important for sperm maturation and associated with metabolic diseases in their offspring. 184 , 198

WINDOWS OF SUSCEPTIBILITY FOR PATERNAL INFLUENCE

Epigenetic programs are particularly dynamic in germ cells undergoing erasure, re‐establishment, and maintenance of patterns, and these events, potentially susceptible to prenatal and/or postnatal exposures, may have amplified long‐lasting effects. 199 Based on epigenetic alterations during the development of the paternal germ line, zygote, and embryo, four windows of susceptibility are defined during the father's lifespan where environmental effects can impact the epigenetic profile of his gametes, as follows: (i) during migration of PGCs to the genital ridge in early embryos, when genome‐wide epigenetic reprogramming by demethylation or methylation remaining occurs; (ii) before puberty, from PGC (or gonocytes) to spermatogonia, during which methylation profiles are largely established; (iii) during each reproductive cycle, from spermatogonium to spermatocyte and finally the spermatozoon, when DNA methylation is also fully established; (iv) in the zygote, when the acquired methylation marks need to withstand post‐zygotic epigenetic reprogramming at specific regions (e.g. imprinted genes), and the modifications present on histone proteins in sperm may relate to the selective establishment of specific DNA methylation patterns. 146 Disappointedly, current studies on the effects of internal and external factors are not enough to differentiate which stage of sperm development is influenced both in animals and humans, and this needs to be strengthened in the future. 4 , 11 , 146 Nonetheless, these data indicate that, from a father's perspective, pre‐puberty/puberty, adulthood, and the zygote phase are all valuable windows to start potential nutritional interventions and avoidance of environmental insult to maximize sperm epigenetic integrity and promote adequate fetal growth and development, thus preventing chronic diseases in adulthood.

CONCLUSION

In summary, based mostly on animal studies, paternal risk factors such as obesity, high‐fat, high‐sugar, or low‐protein diet, undernutrition, diabetes mellitus, hyperglycemia, advanced age, smoking as well as drug exposure may affect offspring health leading to adverse outcomes. These paternal factors regulate offspring development via both direct (genetic/epigenetic) and indirect (maternal uterine environment) effects. The mechanisms responsible for this inter‐and trans‐generational inheritance are beginning to be better understood, with evidence pointing towards the involvement of DNA methylation, histone modifications, and above all, ncRNAs. All these events can interfere with each other and are transmissible through the germline to the next generation without the intervention of genetic mechanisms. In addition, the seminal plasma, a complex fluid that interacts with the sperm and the female reproductive tract, can impact maternal gestational health, post‐fertilization, and postnatal development. Appropriate care during the closed circle of life including pre‐puberty/puberty, adulthood, and the zygote phase, specialized in medical, behavioral, and social health interventions, is very important to reduce the risk of negative environmental exposures, and consequently to improve offspring health as well as the parental health status (Figure 1). Owing to those findings from animal studies that cannot be directly related to human populations, where exposures, outcomes, physiology, and social contexts are very different, caution should be taken in extrapolating findings from animal models to humans. Nonetheless, preventive and educational approaches should include both mothers and fathers (to be), to reduce adverse health outcomes in their offspring caused by modifiable diet and lifestyle, and environmental risk factors. To attain this, well‐crafted human study designs are urgently needed and indispensable to verify findings from animal data.

FIGURE 1.

FIGURE 1

Effects of paternal environmental factors on offspring health. Paternal exposure to environmental factors including unhealthy diets, malnutrition, lifestyle, such as smoking as well as drug exposure, and advanced age, may affect offspring health, development, and chronic diseases. These unique paternally mediated effects on offspring implicate indirect effects on the fetal component of the placenta, telomere length, sperm epigenetic, or seminal fluid protein mechanisms. The epigenetic marks are delivered by sperm to the zygote, with evidence pointing towards the involvement of DNA methylation, histone modifications, and non‐coding RNAs, and further are transmissible through the germline to the next generation. Four windows of susceptibility during the father's lifetime where environmental effects can impact the epigenetic profile of his gametes are as follows: during migration of primordial germ cells (PGCs) to the genital ridge; before puberty; during each reproductive cycle and zygote formation. Appropriate care during the closed circle of life including pre‐puberty/puberty, adulthood, and the zygote phase, specialized in medical, behavioral, and social health interventions is very important to reduce the risk of negative environmental exposures, and consequently to improve offspring health as well as the parental health status.

CONFLICT OF INTEREST

The authors declare no conflict of interest.

Shi Q, Qi K. Developmental origins of health and disease: Impact of paternal nutrition and lifestyle. Pediatr Investig. 2023;7:111–131. 10.1002/ped4.12367

REFERENCES

  • 1. Barker DJ, Osmond C, Golding J, Kuh D, Wadsworth ME. Growth in utero, blood pressure in childhood and adult life, and mortality from cardiovascular disease. BMJ. 1989;298:564‐567. DOI: 10.1136/bmj.298.6673.564 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2. Hanson MA, Gluckman PD. Early developmental conditioning of later health and disease: physiology or pathophysiology. Physiol Rev. 2014;94:1027‐1076. DOI: 10.1152/physrev.00029.2013 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3. Gluckman PD, Hanson MA, Morton SM, Pinal CS. Life‐long echoes–a critical analysis of the developmental origins of adult disease model. Biol Neonate. 2005;87:127‐139. DOI: 10.1159/000082311 [DOI] [PubMed] [Google Scholar]
  • 4. Dimofski P, Meyre D, Dreumont N, Leininger‐Muller B. Consequences of paternal nutrition on offspring health and disease. Nutrients. 2021;13:2818. DOI: 10.3390/nu13082818 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5. Bygren LO, Kaati G, Edvinsson S. Longevity determined by paternal ancestors’ nutrition during their slow growth period. Acta Biotheor. 2001;49:53‐59. DOI: 10.1023/a:1010241825519 [DOI] [PubMed] [Google Scholar]
  • 6. Pembrey ME, Bygren LO, Kaati G, Edvinsson S, Northstone K, Sjöström M, et al. Sex‐specific, male‐line transgenerational responses in humans. Eur J Hum Genet. 2006;14:159‐166. DOI: 10.1038/sj.ejhg.5201538 [DOI] [PubMed] [Google Scholar]
  • 7. Kaati G, Bygren LO, Edvinsson S. Cardiovascular and diabetes mortality determined by nutrition during parents’ and grandparents’ slow growth period. Eur J Hum Genet. 2002;10:682‐688. DOI: 10.1038/sj.ejhg.5200859 [DOI] [PubMed] [Google Scholar]
  • 8. Vågerö D, Pinger PR, Aronsson V, van den Berg GJ. Paternal grandfather's access to food predicts all‐cause and cancer mortality in grandsons. Nat Commun. 2018;9:5124. DOI: 10.1038/s41467-018-07617-9 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9. van den Berg GJ, Pinger PR. Transgenerational effects of childhood conditions on third generation health and education outcomes. Econ Hum Biol. 2016;23:103‐120. DOI: 10.1016/j.ehb.2016.07.001 [DOI] [PubMed] [Google Scholar]
  • 10. Anderson LM, Riffle L, Wilson R, Travlos GS, Lubomirski MS, Alvord WG. Preconceptional fasting of fathers alters serum glucose in offspring of mice. Nutrition. 2006;22:327‐331. DOI: 10.1016/j.nut.2005.09.006 [DOI] [PubMed] [Google Scholar]
  • 11. Soubry A. POHaD: why we should study future fathers. Environ Epigenet. 2018;4:dvy007. DOI: 10.1093/eep/dvy007 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12. Nieto SJ, Kosten TA. Who's your daddy? Behavioral and epigenetic consequences of paternal drug exposure. Int J Dev Neurosci. 2019;78:109‐121. DOI: 10.1016/j.ijdevneu.2019.07.002 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13. Watkins AJ, Sinclair KD. Paternal low protein diet affects adult offspring cardiovascular and metabolic function in mice. Am J Physiol Heart Circ Physiol. 2014;306:H1444‐1452. DOI: 10.1152/ajpheart.00981.2013 [DOI] [PubMed] [Google Scholar]
  • 14. Morgan HL, Aljumah A, Rouillon C, Watkins AJ. Paternal low protein diet and the supplementation of methyl‐donors impact fetal growth and placental development in mice. Placenta. 2021;103:124‐133. DOI: 10.1016/j.placenta.2020.10.020 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15. Watkins AJ, Sirovica S, Stokes B, Isaacs M, Addison O, Martin RA. Paternal low protein diet programs preimplantation embryo gene expression, fetal growth and skeletal development in mice. Biochim Biophys Acta Mol Basis Dis. 2017;1863:1371‐1381. DOI: 10.1016/j.bbadis.2017.02.009 [DOI] [PubMed] [Google Scholar]
  • 16. Morgan HL, Paganopoulou P, Akhtar S, Urquhart N, Philomin R, Dickinson Y, et al. Paternal diet impairs F1 and F2 offspring vascular function through sperm and seminal plasma specific mechanisms in mice. J Physiol. 2020;598:699‐715. DOI: 10.1113/JP278270 [DOI] [PubMed] [Google Scholar]
  • 17. Ly NH, Maekawa T, Yoshida K, Liu Y, Muratani M, Ishii S. RNA‐sequencing analysis of paternal low‐protein diet‐induced gene expression change in mouse offspring adipocytes. G3 (Bethesda). 2019;9:2161‐2170. DOI: 10.1534/g3.119.400181 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18. Carone BR, Fauquier L, Habib N, Shea JM, Hart CE, Li R, et al. Paternally induced transgenerational environmental reprogramming of metabolic gene expression in mammals. Cell. 2010;143:1084‐1096. DOI: 10.1016/j.cell.2010.12.008 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19. Watkins AJ, Dias I, Tsuro H, Allen D, Emes RD, Moreton J, et al. Paternal diet programs offspring health through sperm‐ and seminal plasma‐specific pathways in mice. Proc Natl Acad Sci U S A. 2018;115:10064‐10069. DOI: 10.1073/pnas.1806333115 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20. da Cruz RS, Carney EJ, Clarke J, Cao H, Cruz MI, Benitez C, et al. Paternal malnutrition programs breast cancer risk and tumor metabolism in offspring. Breast Cancer Res. 2018;20:99. DOI: 10.1186/s13058-018-1034-7 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21. Chleilat F, Schick A, Deleemans JM, Ma K, Alukic E, Wong J, et al. Paternal high protein diet modulates body composition, insulin sensitivity, epigenetics, and gut microbiota intergenerationally in rats. FASEB J. 2021;35:e21847. DOI: 10.1096/fj.202100198RR [DOI] [PubMed] [Google Scholar]
  • 22. Gong P, Bailbé D, Bianchi L, Pommier G, Liu J, Tolu S, et al. Paternal high‐protein diet programs offspring insulin sensitivity in a sex‐specific manner. Biomolecules. 2021;11:751. DOI: 10.3390/biom11050751 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23. Fappi A, Mittendorfer B. Dietary protein intake and obesity‐associated cardiometabolic function. Curr Opin Clin Nutr Metab Care. 2020;23:380‐386. DOI: 10.1097/MCO.0000000000000689 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24. Ancu O, Mickute M, Guess ND, Hurren NM, Burd NA, Mackenzie RW. Does high dietary protein intake contribute to the increased risk of developing prediabetes and type 2 diabetes. Appl Physiol Nutr Metab. 2021;46:1‐9. DOI: 10.1139/apnm-2020-0396 [DOI] [PubMed] [Google Scholar]
  • 25. Chen Z, Franco OH, Lamballais S, Ikram MA, Schoufour JD, Muka T, et al. Associations of specific dietary protein with longitudinal insulin resistance, prediabetes and type 2 diabetes: the Rotterdam Study. Clin Nutr. 2020;39:242‐249. DOI: 10.1016/j.clnu.2019.01.021 [DOI] [PubMed] [Google Scholar]
  • 26. Skytte MJ, Samkani A, Petersen AD, Thomsen MN, Astrup A, Chabanova E, et al. A carbohydrate‐reduced high‐protein diet improves HbA(1c) and liver fat content in weight stable participants with type 2 diabetes: a randomised controlled trial. Diabetologia. 2019;62:2066‐2078. DOI: 10.1007/s00125-019-4956-4 [DOI] [PubMed] [Google Scholar]
  • 27. Surowska A, Jegatheesan P, Campos V, Marques AS, Egli L, Cros J, et al. Effects of dietary protein and fat content on intrahepatocellular and intramyocellular lipids during a 6‐Day hypercaloric, high sucrose diet: a randomized controlled trial in normal weight healthy subjects. Nutrients. 2019;11:209. DOI: 10.3390/nu11010209 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28. Fullston T, Palmer NO, Owens JA, Mitchell M, Bakos HW, 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. DOI: 10.1093/humrep/des030 [DOI] [PubMed] [Google Scholar]
  • 29. Binder NK, Hannan NJ, Gardner DK. Paternal diet‐induced obesity retards early mouse embryo development, mitochondrial activity and pregnancy health. PLoS One. 2012;7:e52304. DOI: 10.1371/journal.pone.0052304 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30. Guo T, Yang Y, Jia J, Deng Y, Wang Y, Zhang Y, et al. Preconception paternal/maternal body mass index and risk of small/large for gestational age infant in over 4.7 million Chinese women aged 20‐49 years: a population‐based cohort study in China. Br J Nutr. 2022;21:1‐28. DOI: 10.1017/S000711452200054X [DOI] [PubMed] [Google Scholar]
  • 31. Lin J, Gu W, Huang H, Effects of paternal obesity on fetal development and pregnancy complications: a prospective clinical cohort study. Front Endocrinol. 2022;13:826665. DOI: 10.3389/fendo.2022.826665 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32. Claycombe‐Larson KG, Bundy AN, Roemmich JN. Paternal high‐fat diet and exercise regulate sperm miRNA and histone methylation to modify placental inflammation, nutrient transporter mRNA expression and fetal weight in a sex‐dependent manner. J Nutr Biochem. 2020;81:108373. DOI: 10.1016/j.jnutbio.2020.108373 [DOI] [PubMed] [Google Scholar]
  • 33. Deshpande SS, Nemani H, Arumugam G, Ravichandran A, Balasinor NH. High‐fat diet‐induced and genetically inherited obesity differentially alters DNA methylation profile in the germline of adult male rats. Clin Epigenetics. 2020;12:179. DOI: 10.1186/s13148-020-00974-7 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34. Consitt LA, Saxena G, Slyvka Y, Clark BC, Friedlander M, Zhang Y, et al. Paternal high‐fat diet enhances offspring whole‐body insulin sensitivity and skeletal muscle insulin signaling early in life. Physiol Rep. 2018;6:e13583. DOI: 10.14814/phy2.13583 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35. Masuyama H, Mitsui T, Eguchi T, Tamada S, Hiramatsu Y. The effects of paternal high‐fat diet exposure on offspring metabolism with epigenetic changes in the mouse adiponectin and leptin gene promoters. Am J Physiol Endocrinol Metab. 2016;311:E236‐245. DOI: 10.1152/ajpendo.00095.2016 [DOI] [PubMed] [Google Scholar]
  • 36. Fullston T, McPherson NO, Owens JA, Kang WX, Sandeman LY, Lane M. Paternal obesity induces metabolic and sperm disturbances in male offspring that are exacerbated by their exposure to an “obesogenic” diet. Physiol Rep. 2015;3:e12336. DOI: 10.14814/phy2.12336 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37. Lecomte V, Maloney CA, Wang KW, Morris MJ. Effects of paternal obesity on growth and adiposity of male rat offspring. Am J Physiol Endocrinol Metab. 2017;312:E117‐E125. DOI: 10.1152/ajpendo.00262.2016 [DOI] [PubMed] [Google Scholar]
  • 38. Park JH, Yoo Y, Cho M, Lim J, Lindroth AM, Park YJ. Diet‐induced obesity leads to metabolic dysregulation in offspring via endoplasmic reticulum stress in a sex‐specific manner. Int J Obes (Lond). 2018;42:244‐251. DOI: 10.1038/ijo.2017.203 [DOI] [PubMed] [Google Scholar]
  • 39. Wu HY, Cheng Y, Jin LY, Zhou Y, Pang HY, Zhu H, et al. Paternal obesity impairs hepatic gluconeogenesis of offspring by altering Igf2/H19 DNA methylation. Mol Cell Endocrinol. 2021;529:111264. DOI: 10.1016/j.mce.2021.111264 [DOI] [PubMed] [Google Scholar]
  • 40. Ng SF, Lin RC, Maloney CA, Youngson NA, Owens JA, Morris MJ. Paternal high‐fat diet consumption induces common changes in the transcriptomes of retroperitoneal adipose and pancreatic islet tissues in female rat offspring. FASEB J. 2014;28:1830‐1841. DOI: 10.1096/fj.13-244046 [DOI] [PubMed] [Google Scholar]
  • 41. Cerf ME. Parental high‐fat programming of offspring development, health and β‐cells. Islets. 2011;3:118‐120. DOI: 10.4161/isl.3.3.15420 [DOI] [PubMed] [Google Scholar]
  • 42. Schellong K, Melchior K, Ziska T, Rancourt RC, Henrich W, Plagemann A. Maternal but not paternal high‐fat diet (HFD) exposure at conception predisposes for ‘Diabesity’ in offspring generations. Int J Environ Res Public Health. 2020;17. DOI: 10.3390/ijerph17124229 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43. Korgan AC, O'Leary E, King JL, Weaver I, Perrot TS. Effects of paternal high‐fat diet and rearing environment on maternal investment and development of defensive responses in the offspring. Psychoneuroendocrinology. 2018;91:20‐30. DOI: 10.1016/j.psyneuen.2018.02.010 [DOI] [PubMed] [Google Scholar]
  • 44. Zhou Y, Zhu H, Wu HY, Jin LY, Chen B, Pang HY, et al. Diet‐induced paternal obesity impairs cognitive function in offspring by mediating epigenetic modifications in spermatozoa. Obesity. 2018;26:1749‐1757. DOI: 10.1002/oby.22322 [DOI] [PubMed] [Google Scholar]
  • 45. Chowdhury SS, Lecomte V, Erlich JH, Maloney CA, Morris MJ. Paternal high fat diet in rats leads to renal accumulation of lipid and tubular changes in adult offspring. Nutrients. 2016;8:521. DOI: 10.3390/nu8090521 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46. Zhang X, Hasan AA, Wu H, Gaballa MMS, Zeng S, Liu L, et al. High‐fat, sucrose and salt‐rich diet during rat spermatogenesis lead to the development of chronic kidney disease in the female offspring of the F2 generation. FASEB J. 2022;36:e22259. DOI: 10.1096/fj.202101789RR [DOI] [PubMed] [Google Scholar]
  • 47. Carapeto PV, Ornellas F, Mandarim‐de‐Lacerda CA, Aguila MB. Liver metabolism in adult male mice offspring: consequences of a maternal, paternal or both maternal and paternal high‐fructose diet. J Dev Orig Health Dis. 2018;9:450‐459. DOI: 10.1017/S2040174418000235 [DOI] [PubMed] [Google Scholar]
  • 48. Zhang X, Dong Y, Sun G, Hasan AA, Tian M, Zeng S, et al. Paternal programming of liver function and lipid profile induced by a paternal pre‐conceptional unhealthy diet: potential association with altered gut microbiome composition. Kidney Blood Press Res. 2019;44:133‐148. DOI: 10.1159/000497487 [DOI] [PubMed] [Google Scholar]
  • 49. Ornellas F, Carapeto PV, Aguila MB, Mandarim‐de‐Lacerda CA. Sex‐linked changes and high cardiovascular risk markers in the mature progeny of father, mother, or both father and mother consuming a high‐fructose diet. Nutrition. 2020;71:110612. DOI: 10.1016/j.nut.2019.110612 [DOI] [PubMed] [Google Scholar]
  • 50. Le Q, Li Y, Hou W, Yan B, Yu X, Song H, et al. Binge‐Like sucrose self‐administration experience inhibits cocaine and sucrose seeking behavior in offspring. Front Behav Neurosci. 2017;11:184. DOI: 10.3389/fnbeh.2017.00184 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51. César H, Sertorio MN, de Souza EA, Jamar G, Santamarina A, Jucá A, et al. Parental high‐fat high‐sugar diet programming and hypothalamus adipose tissue axis in male Wistar rats. Eur J Nutr. 2022;61:523‐537. DOI: 10.1007/s00394-021-02690-1 [DOI] [PubMed] [Google Scholar]
  • 52. Bodden C, Pang TY, Feng Y, Mridha F, Kong G, Li S, et al. Intergenerational effects of a paternal Western diet during adolescence on offspring gut microbiota, stress reactivity, and social behavior. FASEB J. 2022;36:e21981. DOI: 10.1096/fj.202100920RR [DOI] [PubMed] [Google Scholar]
  • 53. Emborski C, Mikheyev AS. Ancestral diet transgenerationally influences offspring in a parent‐of‐origin and sex‐specific manner. Philos Trans R Soc Lond B Biol Sci. 2019;374:20180181. DOI: 10.1098/rstb.2018.0181 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54. Teltumbade M, Bhalla A, Sharma A. Paternal inheritance of diet induced metabolic traits correlates with germline regulation of diet induced coding gene expression. Genomics. 2020;112:567‐573. DOI: 10.1016/j.ygeno.2019.04.008 [DOI] [PubMed] [Google Scholar]
  • 55. Öst A, Lempradl A, Casas E, Weigert M, Tiko T, Deniz M, et al. Paternal diet defines offspring chromatin state and intergenerational obesity. Cell. 2014;159:1352‐1364. DOI: 10.1016/j.cell.2014.11.005 [DOI] [PubMed] [Google Scholar]
  • 56. Martín‐Calvo N, Mínguez‐Alarcón L, Gaskins AJ, Nassan FL, Williams PL, Souter I, et al. Paternal preconception folate intake in relation to gestational age at delivery and birthweight of newborns conceived through assisted reproduction. Reprod Biomed Online. 2019;39:835‐843. DOI: 10.1016/j.rbmo.2019.07.005 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57. Pauwels S, Truijen I, Ghosh M, Duca RC, Langie S, Bekaert B, et al. The effect of paternal methyl‐group donor intake on offspring DNA methylation and birth weight. J Dev Orig Health Dis. 2017;8:311‐321. DOI: 10.1017/S2040174417000046 [DOI] [PubMed] [Google Scholar]
  • 58. Greenop KR, Miller M, Bailey HD, Scott RJ, Attia J, Bower C, et al. Paternal dietary folate, B6 and B12 intake, and the risk of childhood brain tumors. Nutr Cancer. 2015;67:224‐230. DOI: 10.1080/01635581.2015.990571 [DOI] [PubMed] [Google Scholar]
  • 59. Ly L, Chan D, Aarabi M, Landry M, Behan NA, MacFarlane AJ, et al. Intergenerational impact of paternal lifetime exposures to both folic acid deficiency and supplementation on reproductive outcomes and imprinted gene methylation. Mol Hum Reprod. 2017;23:461‐477. DOI: 10.1093/molehr/gax029 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60. Lambrot R, Xu C, Saint‐Phar S, Chountalos G, Cohen T, Paquet M, et al. Low paternal dietary folate alters the mouse sperm epigenome and is associated with negative pregnancy outcomes. Nat Commun. 2013;4:2889. DOI: 10.1038/ncomms3889 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61. McCoy CR, Jackson NL, Brewer RL, Moughnyeh MM, Smith DL, Jr , Clinton SM. A paternal methyl donor depleted diet leads to increased anxiety‐ and depression‐like behavior in adult rat offspring. Biosci Rep. 2018;38:BSR20180730. DOI: 10.1042/BSR20180730 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62. Sahara Y, Matsuzawa D, Ishii D, Fuchida T, Goto T, Sutoh C, et al. Paternal methyl donor deficient diets during development affect male offspring behavior and memory‐related gene expression in mice. Dev Psychobiol. 2019;61:17‐28. DOI: 10.1002/dev.21801 [DOI] [PubMed] [Google Scholar]
  • 63. Wu S, Guo W, Li X, Liu Y, Li Y, Lei X, et al. Paternal chronic folate supplementation induced the transgenerational inheritance of acquired developmental and metabolic changes in chickens. Proc Biol Sci. 2019;286:20191653. DOI: 10.1098/rspb.2019.1653 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 64. Ryan DP, Henzel KS, Pearson BL, Siwek ME, Papazoglou A, Guo L, et al. A paternal methyl donor‐rich diet altered cognitive and neural functions in offspring mice. Mol Psychiatry. 2018;23:1345‐1355. DOI: 10.1038/mp.2017.53 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65. Hine C. A paternal diet for offspring success? Sci Transl Med. 2017;9:eaan2780. DOI: 10.1126/scitranslmed.aan2780 [DOI] [PubMed] [Google Scholar]
  • 66. Sabet JA, Park LK, Iyer LK, Tai AK, Koh GY, Pfalzer AC, et al. Paternal B vitamin intake is a determinant of growth, hepatic lipid metabolism and intestinal tumor volume in female Apc1638N mouse offspring. PLoS One. 2016;11:e0151579. DOI: 10.1371/journal.pone.0151579 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 67. Khamehiee N, Jazireian P, Ebrahimi B, Alizadeh A, Shahhoseini M. Paternal trans‐fatty acid and vitamin E diet affects rat offspring's semen quality and PPARs expression. Andrologia. 2021;53:e14082. DOI: 10.1111/and.14082 [DOI] [PubMed] [Google Scholar]
  • 68. Noble N, Paul C, Turon H, Oldmeadow C. Which modifiable health risk behaviours are related? A systematic review of the clustering of smoking, nutrition, alcohol and physical activity (‘SNAP’) health risk factors. Prev Med. 2015;81:16‐41. DOI: 10.1016/j.ypmed.2015.07.003 [DOI] [PubMed] [Google Scholar]
  • 69. Goli P, Yazdi M, Poursafa P, Kelishadi R. Intergenerational influence of paternal physical activity on the offspring's brain: a systematic review and meta‐analysis. Int J Dev Neurosci. 2021;81:10‐25. DOI: 10.1002/jdn.10081 [DOI] [PubMed] [Google Scholar]
  • 70. Kusuyama J, Alves‐Wagner AB, Makarewicz NS, Goodyear LJ. Effects of maternal and paternal exercise on offspring metabolism. Nat Metab. 2020;2:858‐872. DOI: 10.1038/s42255-020-00274-7 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 71. de Sousa Neto IV, Prestes J, Pereira GB, Almeida JA, Ramos GV, de Souza FHV, et al. Protective role of intergenerational paternal resistance training on fibrosis, inflammatory profile, and redox status in the adipose tissue of rat offspring fed with a high‐fat diet. Life Sci. 2022;295:120377. DOI: 10.1016/j.lfs.2022.120377 [DOI] [PubMed] [Google Scholar]
  • 72. McPherson NO, Owens JA, 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:E805‐821. DOI: 10.1152/ajpendo.00013.2015 [DOI] [PubMed] [Google Scholar]
  • 73. Murashov AK, Pak ES, Koury M, Ajmera A, Jeyakumar M, Parker M, et al. Paternal long‐term exercise programs offspring for low energy expenditure and increased risk for obesity in mice. FASEB J. 2016;30:775‐784. DOI: 10.1096/fj.15-274274 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 74. Batista RO, Budu A, Alves‐Silva T, Arakaki AM, Gregnani M, Rodrigues Húngaro TG, et al. Paternal exercise protects against liver steatosis in the male offspring of mice submitted to high fat diet. Life Sci. 2020;263:118583. DOI: 10.1016/j.lfs.2020.118583 [DOI] [PubMed] [Google Scholar]
  • 75. Salomão R, Neto IVS, Ramos GV, Tibana RA, Durigan JQ, Pereira GB, et al. Paternal resistance exercise modulates skeletal muscle remodeling pathways in fathers and male offspring submitted to a high‐fat diet. Front Physiol. 2021;12:706128. DOI: 10.3389/fphys.2021.706128 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 76. de Sousa Neto IV, Tibana RA, da Silva LGO, de Lira EM, do Prado GPG, de Almeida JA, et al. Paternal resistance training modulates calcaneal tendon proteome in the offspring exposed to high‐fat diet. Front Cell Dev Biol. 2020;8:380. DOI: 10.3389/fcell.2020.00380 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 77. de Sousa Neto IV, Tibana RA, Prestes J, de Oliveira da Silva LG, Almeida JA, Franco OL, et al. Paternal resistance training induced modifications in the left ventricle proteome independent of offspring diet. Oxid Med Cell Longev. 2020;2020:5603580. DOI: 10.1155/2020/5603580 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 78. Yin MM, Wang W, Sun J, Liu S, Liu XL, Niu YM, et al. Paternal treadmill exercise enhances spatial learning and memory related to hippocampus among male offspring. Behav Brain Res. 2013;253:297‐304. DOI: 10.1016/j.bbr.2013.07.040 [DOI] [PubMed] [Google Scholar]
  • 79. Northstone K, Golding J, Davey Smith G, Miller LL, Pembrey M. Prepubertal start of father's smoking and increased body fat in his sons: further characterisation of paternal transgenerational responses. Eur J Hum Genet. 2014;22:1382‐1386. DOI: 10.1038/ejhg.2014.31 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 80. Golding J, Gregory S, Northstone K, Pembrey M, Watkins S, Iles‐Caven Y, et al. Human transgenerational observations of regular smoking before puberty on fat mass in grandchildren and great‐grandchildren. Sci Rep. 2022;12:1139. DOI: 10.1038/s41598-021-04504-0 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 81. Knudsen GTM, Dharmage S, Janson C, Abramson MJ, Benediktsdóttir B, Malinovschi A, et al. Parents’ smoking onset before conception as related to body mass index and fat mass in adult offspring: findings from the RHINESSA generation study. PLoS One. 2020;15:e0235632. DOI: 10.1371/journal.pone.0235632 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 82. Easey KE, Sharp GC. The impact of paternal alcohol, tobacco, caffeine use and physical activity on offspring mental health: a systematic review and meta‐analysis. Reprod Health. 2021;18:214. DOI: 10.1186/s12978-021-01266-w [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 83. Oldereid NB, Wennerholm UB, Pinborg A, Loft A, Laivuori H, Petzold M, et al. The effect of paternal factors on perinatal and paediatric outcomes: a systematic review and meta‐analysis. Hum Reprod Update. 2018;24:320‐389. DOI: 10.1093/humupd/dmy005 [DOI] [PubMed] [Google Scholar]
  • 84. You Y, Liu R, Zhou H, Wu R, Lin R, Li B, et al. Effect of exposure to paternal smoking on overweight and obesity in children: findings from the children lifeway cohort in Shenzhen, Southern China. Obes Facts. 2022;15:609‐620. DOI: 10.1159/000525544 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 85. Zhou Q, Zhang S, Wang Q, Shen H, Zhang Y, Tian W, et al. Association between preconception paternal smoking and birth defects in offspring: evidence from the database of the National Free Preconception Health Examination Project in China. BJOG. 2020;127:1358‐1364. DOI: 10.1111/1471-0528.16277 [DOI] [PubMed] [Google Scholar]
  • 86. Harju M, Keski‐Nisula L, Georgiadis L, Heinonen S. Parental smoking and cessation during pregnancy and the risk of childhood asthma. BMC Public Health. 2016;16:428. DOI: 10.1186/s12889-016-3029-6 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 87. Svanes C, Koplin J, Skulstad SM, Johannessen A, Bertelsen RJ, Benediktsdottir B, et al. Father's environment before conception and asthma risk in his children: a multi‐generation analysis of the respiratory health in Northern Europe study. Int J Epidemiol. 2017;46:235‐245. DOI: 10.1093/ije/dyw151 [DOI] [PubMed] [Google Scholar]
  • 88. Accordini S, Calciano L, Johannessen A, Benediktsdóttir B, Bertelsen RJ, Bråbäck L, et al. Prenatal and prepubertal exposures to tobacco smoke in men may cause lower lung function in future offspring: a three‐generation study using a causal modelling approach. Eur Respir J. 2021;58:2002791. DOI: 10.1183/13993003.02791-2020 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 89. Rehan VK, Liu J, Naeem E, Tian J, Sakurai R, Kwong K, et al. Perinatal nicotine exposure induces asthma in second generation offspring. BMC Med. 2012;10:129. DOI: 10.1186/1741-7015-10-129 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 90. Hammer B, Kadalayil L, Boateng E, Buschmann D, Rezwan FI, Wolff M, et al. Preconceptional smoking alters spermatozoal miRNAs of murine fathers and affects offspring's body weight. Int J Obes (Lond). 2021;45:1623‐1627. DOI: 10.1038/s41366-021-00798-2 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 91. Duko B, Pereira G, Tait RJ, Newnham J, Betts K, Alati R. Prenatal tobacco exposure and the risk of conduct disorder symptoms in offspring at the age of 14 years: findings from the Raine Study. J Psychiatr Res. 2021;142:1‐8. DOI: 10.1016/j.jpsychires.2021.07.030 [DOI] [PubMed] [Google Scholar]
  • 92. Duko B, Pereira G, Tait RJ, Nyadanu SD, Betts K, Alati R. Prenatal tobacco exposure and the risk of tobacco smoking and dependence in offspring: a systematic review and meta‐analysis. Drug Alcohol Depend. 2021;227:108993. DOI: 10.1016/j.drugalcdep.2021.108993 [DOI] [PubMed] [Google Scholar]
  • 93. Chang RC, Wang H, Bedi Y, Golding MC. Preconception paternal alcohol exposure exerts sex‐specific effects on offspring growth and long‐term metabolic programming. Epigenetics Chromatin. 2019;12:9. DOI: 10.1186/s13072-019-0254-0 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 94. Cambiasso MY, Gotfryd L, Stinson MG, Birolo S, Salamone G, Romanato M, et al. Paternal alcohol consumption has intergenerational consequences in male offspring. J Assist Reprod Genet. 2022;39:441‐459. DOI: 10.1007/s10815-021-02373-0 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 95. Bedi Y, Chang RC, Gibbs R, Clement TM, Golding MC. Alterations in sperm‐inherited noncoding RNAs associate with late‐term fetal growth restriction induced by preconception paternal alcohol use. Reprod Toxicol. 2019;87:11‐20. DOI: 10.1016/j.reprotox.2019.04.006 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 96. Conner KE, Bottom RT, Huffman KJ. The impact of paternal alcohol consumption on offspring brain and behavioral development. Alcohol Clin Exp Res. 2020;44:125‐140. DOI: 10.1111/acer.14245 [DOI] [PubMed] [Google Scholar]
  • 97. Eyolfson E, Bhatt D, Wang M, Lohman AW, Mychasiuk R. Paternal exposure to exercise and/or caffeine and alcohol modify offspring behavioral and pathophysiological recovery from repetitive mild traumatic brain injury in adolescence. Genes Brain Behav. 2021:egbb12736. DOI: 10.1111/gbb.12736 [DOI] [PubMed] [Google Scholar]
  • 98. Rathod RS, Ferguson C, Seth A, Baratta AM, Plasil SL, Homanics GE. Effects of paternal preconception vapor alcohol exposure paradigms on behavioral responses in offspring. Brain Sci. 2020;10:658. DOI: 10.3390/brainsci10090658 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 99. Chang RC, Thomas KN, Bedi YS, Golding MC. Programmed increases in LXRα induced by paternal alcohol use enhance offspring metabolic adaptation to high‐fat diet induced obesity. Mol Metab. 2019;30:161‐172. DOI: 10.1016/j.molmet.2019.09.016 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 100. Thor S, Hemmingsson T, Danielsson AK. Landberg J. Fathers' alcohol consumption and risk of substance‐related disorders in offspring. Drug Alcohol Depend. 2022;233:109354. DOI: 10.1016/j.drugalcdep.2022.109354 [DOI] [PubMed] [Google Scholar]
  • 101. Luan M, Zhang X, Fang G, Liang H, Yang F, Song X, et al. Preconceptional paternal alcohol consumption and the risk of child behavioral problems: a prospective cohort study. Sci Rep. 2022;12:1508. DOI: 10.1038/s41598-022-05611-2 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 102. Zhou Q, Song L, Chen J, Wang Q, Shen H, Zhang S, et al. Association of preconception paternal alcohol consumption with increased fetal birth defect risk. JAMA Pediatr. 2021;175:742‐743. DOI: 10.1001/jamapediatrics.2021.0291 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 103. Zhang S, Wang L, Yang T, Chen L, Zhao L, Wang T, et al. Parental alcohol consumption and the risk of congenital heart diseases in offspring: an updated systematic review and meta‐analysis. Eur J Prev Cardiol. 2020;27:410‐421. DOI: 10.1177/2047487319874530 [DOI] [PubMed] [Google Scholar]
  • 104. Xia R, Jin L, Li D, Liang H, Yang F, Chen J, et al. Association between paternal alcohol consumption before conception and anogenital distance of offspring. Alcohol Clin Exp Res. 2018;42:735‐742. DOI: 10.1111/acer.13595 [DOI] [PubMed] [Google Scholar]
  • 105. Goldberg LR, Gould TJ. Multigenerational and transgenerational effects of paternal exposure to drugs of abuse on behavioral and neural function. Eur J Neurosci. 2019;50:2453‐2466. DOI: 10.1111/ejn.14060 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 106. Baratta AM, Rathod RS, Plasil SL, Seth A, Homanics GE. Exposure to drugs of abuse induce effects that persist across generations. Int Rev Neurobiol. 2021;156:217‐277. DOI: 10.1016/bs.irn.2020.08.003 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 107. Weinberg W. Zur Verebung des Zwergwuchses. Arch Rass Gesamte Biol. 1912;9:710‐718. [Google Scholar]
  • 108. Penrose LS. Parental age and mutation. Lancet. 1955;269:312‐313. DOI: 10.1016/s0140-6736(55)92305-9 [DOI] [PubMed] [Google Scholar]
  • 109. Schmidt L, Sobotka T, Bentzen JG, Nyboe Andersen A. Demographic and medical consequences of the postponement of parenthood. Hum Reprod Update. 2012;18:29‐43. DOI: 10.1093/humupd/dmr040 [DOI] [PubMed] [Google Scholar]
  • 110. Lee HY, Lu CL, Chen HF, Su HF, Li CY. Perinatal and childhood risk factors for early‐onset type 1 diabetes: a population‐based case‐control study in Taiwan. Eur J Public Health. 2015;25:1024‐1029. DOI: 10.1093/eurpub/ckv059 [DOI] [PubMed] [Google Scholar]
  • 111. Eriksen W, Sundet JM, Tambs K. Paternal age at birth and the risk of obesity in young adulthood: a register‐based birth cohort study of Norwegian males. Am J Hum Biol. 2013;25:29‐34. DOI: 10.1002/ajhb.22333 [DOI] [PubMed] [Google Scholar]
  • 112. Halvaei I, Litzky J, Esfandiari N. Advanced paternal age: effects on sperm parameters, assisted reproduction outcomes and offspring health. Reprod Biol Endocrinol. 2020;18:110. DOI: 10.1186/s12958-020-00668-y [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 113. Sun Y, Li X, Jiang W, Fan Y, Ouyang Q, Shao W, et al. Advanced paternal age and risk of cancer in offspring. Aging. 2020;13:3712‐3725. DOI: 10.18632/aging.202333 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 114. Chan PTK, Robaire B. Advanced paternal age and future generations. Front Endocrinol. 2022;13:897101. DOI: 10.3389/fendo.2022.897101 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 115. Fico G, Oliva V, De Prisco M, Giménez‐Palomo A, Sagué‐Vilavella M, Gomes‐da‐Costa S, et al. The U‐shaped relationship between parental age and the risk of bipolar disorder in the offspring: a systematic review and meta‐analysis. Eur Neuropsychopharmacol. 2022;60:55‐75. DOI: 10.1016/j.euroneuro.2022.05.004 [DOI] [PubMed] [Google Scholar]
  • 116. Dockerty JD, Draper G, Vincent T, Rowan SD, Bunch KJ. Case‐control study of parental age, parity and socioeconomic level in relation to childhood cancers. Int J Epidemiol. 2001;30:1428‐1437. DOI: 10.1093/ije/30.6.1428 [DOI] [PubMed] [Google Scholar]
  • 117. Sergentanis TN, Thomopoulos TP, Gialamas SP, Karalexi MA, Biniaris‐Georgallis SI, Kontogeorgi E, et al. Risk for childhood leukemia associated with maternal and paternal age. Eur J Epidemiol. 2015;30:1229‐1261. DOI: 10.1007/s10654-015-0089-3 [DOI] [PubMed] [Google Scholar]
  • 118. Sipos A, Rasmussen F, Harrison G, Tynelius P, Lewis G, Leon DA, et al. Paternal age and schizophrenia: a population based cohort study. BMJ. 2004;329:1070. DOI: 10.1136/bmj.38243.672396.55 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 119. Veldkamp SAM, Zondervan‐Zwijnenburg MAJ, van Bergen E, Barzeva SA, Tamayo‐Martinez N, Becht AI, et al. Parental age in relation to offspring's neurodevelopment. J Clin Child Adolesc Psychol. 2021;50:632‐644. DOI: 10.1080/15374416.2020.1756298 [DOI] [PubMed] [Google Scholar]
  • 120. Frans EM, McGrath JJ, Sandin S, Lichtenstein P, Reichenberg A, Långström N, et al. Advanced paternal and grandpaternal age and schizophrenia: a three‐generation perspective. Schizophr Res. 2011;133:120‐124. DOI: 10.1016/j.schres.2011.09.027 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 121. Panagopoulou P, Skalkidou A, Marcotte E, Erdmann F, Ma X, Heck JE, et al. Parental age and the risk of childhood acute myeloid leukemia: results from the Childhood Leukemia International Consortium. Cancer Epidemiol. 2019;59:158‐165. DOI: 10.1016/j.canep.2019.01.022 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 122. Calhaz‐Jorge C, De Geyter CH, Kupka MS, Wyns C, Mocanu E, Motrenko T, et al. Survey on ART and IUI: legislation, regulation, funding and registries in European countries: the European IVF‐monitoring Consortium (EIM) for the European Society of Human Reproduction and Embryology (ESHRE). Hum Reprod Open. 2020;2020:hoz044. DOI: 10.1093/hropen/hoz044 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 123. Xavier MJ, Roman SD, Aitken RJ, Nixon B. Transgenerational inheritance: how impacts to the epigenetic and genetic information of parents affect offspring health. Hum Reprod Update. 2019;25:518‐540. DOI: 10.1093/humupd/dmz017 [DOI] [PubMed] [Google Scholar]
  • 124. Soubry A. Epigenetic inheritance and evolution: a paternal perspective on dietary influences. Prog Biophys Mol Biol. 2015;118:79‐85. DOI: 10.1016/j.pbiomolbio.2015.02.008 [DOI] [PubMed] [Google Scholar]
  • 125. Goyal D, Limesand SW, Goyal R. Epigenetic responses and the developmental origins of health and disease. J Endocrinol. 2019;242:T105‐T119. DOI: 10.1530/JOE-19-0009 [DOI] [PubMed] [Google Scholar]
  • 126. Sales VM, Ferguson‐Smith AC, Patti ME. Epigenetic mechanisms of transmission of metabolic disease across generations. Cell Metab. 2017;25:559‐571. DOI: 10.1016/j.cmet.2017.02.016 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 127. Blackburn EH. Telomeres and telomerase: their mechanisms of action and the effects of altering their functions. FEBS Lett. 2005;579:859‐862. DOI: 10.1016/j.febslet.2004.11.036 [DOI] [PubMed] [Google Scholar]
  • 128. Aviv A. The mitochondrial genome, paternal age and telomere length in humans. Philos Trans R Soc Lond B Biol Sci. 2018;373:20170210. DOI: 10.1098/rstb.2017.0210 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 129. Nordfjäll K, Larefalk A, Lindgren P, Holmberg D, Roos G. Telomere length and heredity: indications of paternal inheritance. Proc Natl Acad Sci U S A. 2005;102:16374‐16378. DOI: 10.1073/pnas.0501724102 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 130. De Meyer T, Rietzschel ER, De Buyzere ML, De Bacquer D, Van Criekinge W, De Backer GG, et al. Paternal age at birth is an important determinant of offspring telomere length. Hum Mol Genet. 2007;16:3097‐3102. DOI: 10.1093/hmg/ddm271 [DOI] [PubMed] [Google Scholar]
  • 131. Prescott J, Du M, Wong JY, Han J, De Vivo I. Paternal age at birth is associated with offspring leukocyte telomere length in the nurses' health study. Hum Reprod. 2012;27:3622‐3631. DOI: 10.1093/humrep/des314 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 132. Wulaningsih W, Hardy R, Wong A, Kuh D. Parental age and offspring leukocyte telomere length and attrition in midlife: evidence from the 1946 British birth cohort. Exp Gerontol. 2018;112:92‐96. DOI: 10.1016/j.exger.2018.09.008 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 133. Unryn BM, Cook LS, Riabowol KT. Paternal age is positively linked to telomere length of children. Aging Cell. 2005;4:97‐101. DOI: 10.1111/j.1474-9728.2005.00144.x [DOI] [PubMed] [Google Scholar]
  • 134. Eisenberg DT, Hayes MG, Kuzawa CW. Delayed paternal age of reproduction in humans is associated with longer telomeres across two generations of descendants. Proc Natl Acad Sci U S A. 2012;109:10251‐10256. DOI: 10.1073/pnas.1202092109 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 135. Eisenberg D, Lee NR, Rej PH, Hayes MG, Kuzawa CW. Older paternal ages and grandpaternal ages at conception predict longer telomeres in human descendants. Proc Biol Sci. 2019;286:20190800. DOI: 10.1098/rspb.2019.0800 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 136. Eisenberg DTA, Kuzawa CW. The paternal age at conception effect on offspring telomere length: mechanistic, comparative and adaptive perspectives. Philos Trans R Soc Lond B Biol Sci. 2018;373:20160442. DOI: 10.1098/rstb.2016.0442 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 137. Eisenberg DT. An evolutionary review of human telomere biology: the thrifty telomere hypothesis and notes on potential adaptive paternal effects. Am J Hum Biol. 2011;23:149‐167. DOI: 10.1002/ajhb.21127 [DOI] [PubMed] [Google Scholar]
  • 138. Seluanov A, Chen Z, Hine C, Sasahara TH, Ribeiro AA, Catania KC, et al. Telomerase activity coevolves with body mass not lifespan. Aging Cell. 2007;6:45‐52. DOI: 10.1111/j.1474-9726.2006.00262.x [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 139. Gomes NM, Ryder OA, Houck ML, Charter SJ, Walker W, Forsyth NR, et al. Comparative biology of mammalian telomeres: hypotheses on ancestral states and the roles of telomeres in longevity determination. Aging Cell. 2011;10:761‐768. DOI: 10.1111/j.1474-9726.2011.00718.x [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 140. Bauch C, Boonekamp JJ, Korsten P, Mulder E, Verhulst S. Epigenetic inheritance of telomere length in wild birds. PLoS Genet. 2019;15:e1007827. DOI: 10.1371/journal.pgen.1007827 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 141. Ishii A, Nakamura K, Kishimoto H, Honma N, Aida J, Sawabe M, et al. Telomere shortening with aging in the human pancreas. Exp Gerontol. 2006;41:882‐886. DOI: 10.1016/j.exger.2006.06.036 [DOI] [PubMed] [Google Scholar]
  • 142. Willeit P, Willeit J, Mayr A, Weger S, Oberhollenzer F, Brandstätter A, et al. Telomere length and risk of incident cancer and cancer mortality. JAMA. 2010;304:69‐75. DOI: 10.1001/jama.2010.897 [DOI] [PubMed] [Google Scholar]
  • 143. Roberts AR, Huang E, Jones L, Daxinger L, Chong S, Whitelaw E. Non‐telomeric epigenetic and genetic changes are associated with the inheritance of shorter telomeres in mice. Chromosoma. 2013;122:541‐554. DOI: 10.1007/s00412-013-0427-8 [DOI] [PubMed] [Google Scholar]
  • 144. Rossiello F, Jurk D, Passos JF, d'Adda di Fagagna F. Telomere dysfunction in ageing and age‐related diseases. Nat Cell Biol. 2022;24:135‐147. DOI: 10.1038/s41556-022-00842-x [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 145. Skilton MR, Nakhla S, Ayer JG, Harmer JA, Toelle BG, Leeder SR, et al. Telomere length in early childhood: early life risk factors and association with carotid intima‐media thickness in later childhood. Eur J Prev Cardiol. 2016;23:1086‐1092. DOI: 10.1177/2047487315607075 [DOI] [PubMed] [Google Scholar]
  • 146. Soubry A, Hoyo C, Jirtle RL, Murphy SK. A paternal environmental legacy: evidence for epigenetic inheritance through the male germ line. Bioessays. 2014;36:359‐371. DOI: 10.1002/bies.201300113 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 147. Ng SF, Lin RC, Laybutt DR, Barres R, Owens JA, Morris MJ. Chronic high‐fat diet in fathers programs β‐cell dysfunction in female rat offspring. Nature. 2010;467:963‐966. DOI: 10.1038/nature09491 [DOI] [PubMed] [Google Scholar]
  • 148. Soubry A, Murphy SK, Wang F, Huang Z, Vidal AC, Fuemmeler BF, et al. Newborns of obese parents have altered DNA methylation patterns at imprinted genes. Int J Obes (Lond). 2015;39:650‐657. DOI: 10.1038/ijo.2013.193 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 149. Soubry A, Schildkraut JM, Murtha A, Wang F, Huang Z, Bernal A, et al. Paternal obesity is associated with IGF2 hypomethylation in newborns: results from a Newborn Epigenetics Study (NEST) cohort. BMC Med. 2013;11:29. DOI: 10.1186/1741-7015-11-29 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 150. Wei Y, Yang CR, Wei YP, Zhao ZA, Hou Y, Schatten H, et al. Paternally induced transgenerational inheritance of susceptibility to diabetes in mammals. Proc Natl Acad Sci U S A. 2014;111:1873‐1878. DOI: 10.1073/pnas.1321195111 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 151. Jirtle RL, Skinner MK. Environmental epigenomics and disease susceptibility. Nat Rev Genet. 2007;8:253‐262. DOI: 10.1038/nrg2045 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 152. Murphy SK, Jirtle RL. Imprinting evolution and the price of silence. Bioessays. 2003;25:577‐588. DOI: 10.1002/bies.10277 [DOI] [PubMed] [Google Scholar]
  • 153. Varley KE, Gertz J, Bowling KM, Parker SL, Reddy TE, Pauli‐Behn F, et al. Dynamic DNA methylation across diverse human cell lines and tissues. Genome Res. 2013;23:555‐567. DOI: 10.1101/gr.147942.112 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 154. Hackett JA, Zylicz JJ, Surani MA. Parallel mechanisms of epigenetic reprogramming in the germline. Trends Genet. 2012;28:164‐174. DOI: 10.1016/j.tig.2012.01.005 [DOI] [PubMed] [Google Scholar]
  • 155. Messerschmidt DM, Knowles BB, Solter D. DNA methylation dynamics during epigenetic reprogramming in the germline and preimplantation embryos. Genes Dev. 2014;28:812‐828. DOI: 10.1101/gad.234294.113 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 156. Schagdarsurengin U, Steger K. Epigenetics in male reproduction: effect of paternal diet on sperm quality and offspring health. Nat Rev Urol. 2016;13:584‐595. DOI: 10.1038/nrurol.2016.157 [DOI] [PubMed] [Google Scholar]
  • 157. Chan JC, Nugent BM, Bale TL. Parental advisory: maternal and paternal stress can impact offspring neurodevelopment. Biol Psychiatry. 2018;83:886‐894. DOI: 10.1016/j.biopsych.2017.10.005 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 158. Zheng X, Li Z, Wang G, Wang H, Zhou Y, Zhao X, et al. 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. DOI: 10.1038/s41421-021-00343-5 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 159. Illum LRH, Bak ST, Lund S, Nielsen AL. DNA methylation in epigenetic inheritance of metabolic diseases through the male germ line. J Mol Endocrinol. 2018;60:R39‐R56. DOI: 10.1530/JME-17-0189 [DOI] [PubMed] [Google Scholar]
  • 160. Cassidy FC, Charalambous M. Genomic imprinting, growth and maternal‐fetal interactions. J Exp Biol. 2018;221:jeb164517. DOI: 10.1242/jeb.164517 [DOI] [PubMed] [Google Scholar]
  • 161. Thamban T, Agarwaal V, Khosla S. Role of genomic imprinting in mammalian development. J Biosci. 2020;45:20. [PubMed] [Google Scholar]
  • 162. Mitchell M, Strick R, Strissel PL, Dittrich R, McPherson NO, Lane M, et al. Gene expression and epigenetic aberrations in F1‐placentas fathered by obese males. Mol Reprod Dev. 2017;84:316‐328. DOI: 10.1002/mrd.22784 [DOI] [PubMed] [Google Scholar]
  • 163. Hackett JA, Sengupta R, Zylicz JJ, Murakami K, Lee C, Down TA, et al. Germline DNA demethylation dynamics and imprint erasure through 5‐hydroxymethylcytosine. Science. 2013;339:448‐452. DOI: 10.1126/science.1229277 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 164. Tang WW, Dietmann S, Irie N, Leitch HG, Floros VI, Bradshaw CR, et al. A Unique gene regulatory network resets the human germline epigenome for development. Cell. 2015;161:1453‐1467. DOI: 10.1016/j.cell.2015.04.053 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 165. Liang F, Diao L, Liu J, Jiang N, Zhang J, Wang H, et al. Paternal ethanol exposure and behavioral abnormities in offspring: associated alterations in imprinted gene methylation. Neuropharmacology. 2014;81:126‐133. DOI: 10.1016/j.neuropharm.2014.01.025 [DOI] [PubMed] [Google Scholar]
  • 166. Swinford‐Jackson SE, Fant B, Wimmer ME, Chan D, Knouse MC, Sarmiento M, et al. Cocaine‐induced changes in sperm Cdkn1a methylation are associated with cocaine resistance in male offspring. J Neurosci. 2022;42:2905‐2916. DOI: 10.1523/JNEUROSCI.3172-20.2022 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 167. Liu Y, Chen S, Pang D, Zhou J, Xu X, Yang S, et al. Effects of paternal exposure to cigarette smoke on sperm DNA methylation and long‐term metabolic syndrome in offspring. Epigenetics Chromatin. 2022;15:3. DOI: 10.1186/s13072-022-00437-8 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 168. Elhamamsy AR. Role of DNA methylation in imprinting disorders: an updated review. J Assist Reprod Genet. 2017;34:549‐562. DOI: 10.1007/s10815-017-0895-5 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 169. Jenkins TG, Carrell DT. The paternal epigenome and embryogenesis: poising mechanisms for development. Asian J Androl. 2011;13:76‐80. DOI: 10.1038/aja.2010.61 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 170. Bench GS, Friz AM, Corzett MH, Morse DH, Balhorn R. DNA and total protamine masses in individual sperm from fertile mammalian subjects. Cytometry. 1996;23:263‐271. DOI: 10.1002/(SICI)1097-0320(19960401)23:4<263::AID‐CYTO1>3.0.CO;2‐I [DOI] [PubMed] [Google Scholar]
  • 171. Gatewood JM, Cook GR, Balhorn R, Schmid CW, Bradbury EM. Isolation of four core histones from human sperm chromatin representing a minor subset of somatic histones. J Biol Chem. 1990;265:20662‐20666. [PubMed] [Google Scholar]
  • 172. Hammoud SS, Nix DA, Zhang H, Purwar J, Carrell DT, Cairns BR. Distinctive chromatin in human sperm packages genes for embryo development. Nature. 2009;460:473‐478. DOI: 10.1038/nature08162 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 173. Terashima M, Barbour S, Ren J, Yu W, Han Y, Muegge K. Effect of high fat diet on paternal sperm histone distribution and male offspring liver gene expression. Epigenetics. 2015;10:861‐871. DOI: 10.1080/15592294.2015.1075691 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 174. Pepin AS, Lafleur C, Lambrot R, Dumeaux V, Kimmins S. Sperm histone H3 lysine 4 tri‐methylation serves as a metabolic sensor of paternal obesity and is associated with the inheritance of metabolic dysfunction. Mol Metab. 2022;59:101463. DOI: 10.1016/j.molmet.2022.101463 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 175. Yoshida K, Maekawa T, Ly NH, Fujita SI, Muratani M, Ando M, et al. ATF7‐Dependent epigenetic changes are required for the intergenerational effect of a paternal low‐protein diet. Mol Cell. 2020;78:445‐458.e6. DOI: 10.1016/j.molcel.2020.02.028 [DOI] [PubMed] [Google Scholar]
  • 176. Saxena A, Carninci P. Long non‐coding RNA modifies chromatin: epigenetic silencing by long non‐coding RNAs. Bioessays. 2011;33:830‐839. DOI: 10.1002/bies.201100084 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 177. Neve B, Jonckheere N, Vincent A, Van Seuningen I. Epigenetic regulation by lncRNAs: an overview focused on UCA1 in colorectal cancer. Cancers. 2018;10:440. DOI: 10.3390/cancers10110440 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 178. Wei JW, Huang K, Yang C, Kang CS. Non‐coding RNAs as regulators in epigenetics (Review). Oncol Rep. 2017;37:3‐9. DOI: 10.3892/or.2016.5236 [DOI] [PubMed] [Google Scholar]
  • 179. Yin X, Anwar A, Wang Y, Hu H, Liang G, Zhang C. Paternal environmental exposure‐induced spermatozoal small noncoding RNA alteration meditates the intergenerational epigenetic inheritance of multiple diseases. Front Med. 2022;16:176‐184. DOI: 10.1007/s11684-021-0885-y [DOI] [PubMed] [Google Scholar]
  • 180. Crisóstomo L, Bourgery M, Rato L, Raposo JF, Batterham RL, Kotaja N, et al. Testicular “inherited metabolic memory” of ancestral high‐fat diet is associated with sperm sncRNA content. Biomedicines. 2022;10:909. DOI: 10.3390/biomedicines10040909 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 181. Fullston T, Ohlsson Teague EM, Palmer NO, DeBlasio MJ, Mitchell M, Corbett M, et al. Paternal obesity initiates metabolic disturbances in two generations of mice with incomplete penetrance to the F2 generation and alters the transcriptional profile of testis and sperm microRNA content. FASEB J. 2013;27:4226‐4243. DOI: 10.1096/fj.12-224048 [DOI] [PubMed] [Google Scholar]
  • 182. McPherson NO, Lane M, Sandeman L, Owens JA, Fullston T. An exercise‐only intervention in obese fathers restores glucose and insulin regulation in conjunction with the rescue of pancreatic islet cell morphology and MicroRNA expression in male offspring. Nutrients. 2017;9:122. DOI: 10.3390/nu9020122 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 183. Grandjean V, Fourré S, De Abreu DA, Derieppe MA, Remy JJ, Rassoulzadegan M. RNA‐mediated paternal heredity of diet‐induced obesity and metabolic disorders. Sci Rep. 2015;5:18193. DOI: 10.1038/srep18193 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 184. Chen Q, Yan M, Cao Z, Li X, Zhang Y, Shi J, et al. Sperm tsRNAs contribute to intergenerational inheritance of an acquired metabolic disorder. Science. 2016;351:397‐400. DOI: 10.1126/science.aad7977 [DOI] [PubMed] [Google Scholar]
  • 185. Short AK, Fennell KA, Perreau VM, Fox A, O'Bryan MK, Kim JH, et al. Elevated paternal glucocorticoid exposure alters the small noncoding RNA profile in sperm and modifies anxiety and depressive phenotypes in the offspring. Transl Psychiatry. 2016;6:e837. DOI: 10.1038/tp.2016.109 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 186. Schjenken JE, Robertson SA. The female response to seminal fluid. Physiol Rev. 2020;100:1077‐1117. DOI: 10.1152/physrev.00013.2018 [DOI] [PubMed] [Google Scholar]
  • 187. Klonoff‐Cohen HS, Savitz DA, Cefalo RC, McCann MF. An epidemiologic study of contraception and preeclampsia. JAMA. 1989;262:3143‐3147. [PubMed] [Google Scholar]
  • 188. Einarsson JI, Sangi‐Haghpeykar H, Gardner MO. Sperm exposure and development of preeclampsia. Am J Obstet Gynecol. 2003;188:1241‐1243. DOI: 10.1067/mob.2003.401 [DOI] [PubMed] [Google Scholar]
  • 189. Kho EM, McCowan LM, North RA, Roberts CT, Chan E, Black MA, et al. Duration of sexual relationship and its effect on preeclampsia and small for gestational age perinatal outcome. J Reprod Immunol. 2009;82:66‐73. DOI: 10.1016/j.jri.2009.04.011 [DOI] [PubMed] [Google Scholar]
  • 190. Sadat Z, Abedzadeh Kalahroudi M, Saberi F. The effect of short duration sperm exposure on development of preeclampsia in primigravid women. Iran Red Crescent Med J. 2012;14:20‐24. [PMC free article] [PubMed] [Google Scholar]
  • 191. Robertson SA, Sharkey DJ. Seminal fluid and fertility in women. Fertil Steril. 2016;106:511‐519. DOI: 10.1016/j.fertnstert.2016.07.1101 [DOI] [PubMed] [Google Scholar]
  • 192. Kenny LC, Kell DB. Immunological tolerance, pregnancy, and preeclampsia: the roles of semen microbes and the father. Front Med. 2017;4:239. DOI: 10.3389/fmed.2017.00239 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 193. Mateo‐Otero Y, Sánchez JM, Recuero S, Bagés‐Arnal S, McDonald M, Kenny DA, et al. Effect of exposure to seminal plasma through natural mating in cattle on conceptus length and gene expression. Front Cell Dev Biol. 2020;8:341. DOI: 10.3389/fcell.2020.00341 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 194. Skjaerven R, Wilcox AJ. Lie RT. The interval between pregnancies and the risk of preeclampsia. N Engl J Med. 2002;346:33‐38. DOI: 10.1056/NEJMoa011379 [DOI] [PubMed] [Google Scholar]
  • 195. Zhang J, Patel G. Partner change and perinatal outcomes: a systematic review. Paediatr Perinat Epidemiol. 2007;21:46‐57. DOI: 10.1111/j.1365-3016.2007.00837.x [DOI] [PubMed] [Google Scholar]
  • 196. Bromfield JJ, Schjenken JE, Chin PY, Care AS, Jasper MJ, Robertson SA. Maternal tract factors contribute to paternal seminal fluid impact on metabolic phenotype in offspring. Proc Natl Acad Sci U S A. 2014;111:2200‐2205. DOI: 10.1073/pnas.1305609111 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 197. Furse S, Watkins AJ, Williams HEL, Snowden SG, Chiarugi D, Koulman A. Paternal nutritional programming of lipid metabolism is propagated through sperm and seminal plasma. Metabolomics. 2022;18:13. DOI: 10.1007/s11306-022-01869-9 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 198. Vojtech L, Woo S, Hughes S, Levy C, Ballweber L, Sauteraud RP, et al. Exosomes in human semen carry a distinctive repertoire of small non‐coding RNAs with potential regulatory functions. Nucleic Acids Res. 2014;42:7290‐7304. DOI: 10.1093/nar/gku347 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 199. Ly L, Chan D, Trasler JM. Developmental windows of susceptibility for epigenetic inheritance through the male germline. Semin Cell Dev Biol. 2015;43:96‐105. DOI: 10.1016/j.semcdb.2015.07.006 [DOI] [PubMed] [Google Scholar]

Articles from Pediatric Investigation are provided here courtesy of Chinese Medical Association

RESOURCES