Acessibilidade / Reportar erro

Paternal effects on fetal programming

Abstract

Paternal programming is the concept that the environmental signals from the sire’s experiences leading up to mating can alter semen and ultimately affect the phenotype of resulting offspring. Potential mechanisms carrying the paternal effects to offspring can be associated with epigenetic signatures (DNA methylation, histone modification and non-coding RNAs), oxidative stress, cytokines, and the seminal microbiome. Several opportunities exist for sperm/semen to be influenced during development; these opportunities are within the testicle, the epididymis, or accessory sex glands. Epigenetic signatures of sperm can be impacted during the pre-natal and pre-pubertal periods, during sexual maturity and with advancing sire age. Sperm are susceptible to alterations as dictated by their developmental stage at the time of the perturbation, and sperm and seminal plasma likely have both dependent and independent effects on offspring. Research using rodent models has revealed that many factors including over/under nutrition, dietary fat, protein, and ingredient composition (e.g., macro- or micronutrients), stress, exercise, and exposure to drugs, alcohol, and endocrine disruptors all elicit paternal programming responses that are evident in offspring phenotype. Research using livestock species has also revealed that sire age, fertility level, plane of nutrition, and heat stress can induce alterations in the epigenetic, oxidative stress, cytokine, and microbiome profiles of sperm and/or seminal plasma. In addition, recent findings in pigs, sheep, and cattle have indicated programming effects in blastocysts post-fertilization with some continuing into post-natal life of the offspring. Our research group is focused on understanding the effects of common management scenarios of plane of nutrition and growth rates in bulls and rams on mechanisms resulting in paternal programming and subsequent offspring outcomes. Understanding the implication of paternal programming is imperative as short-term feeding and management decisions have the potential to impact productivity and profitability of our herds for generations to come.

Keywords:
fetal programming; sire; epigenetics; offspring outcomes; paternal programming

Introduction

Developmental programming is the concept whereby environmental factors inflicted on gametes during their development and concepti during their gestation can have subsequent and long-term impacts on post-natal offspring development. Early evidence of developmental programming came from human epidemiological studies (Barker and Clark, 1997Barker DJ, Clark PM. Fetal undernutrition and disease in later life. Rev Reprod. 1997;2(2):105-12. http://dx.doi.org/10.1530/ror.0.0020105. PMid:9414472.
http://dx.doi.org/10.1530/ror.0.0020105...
) of pregnant women who experienced severe malnutrition during gestation due to the war-time associated food supply shortage, which is often referred to as the “Dutch Hunger Winter” or the “Dutch Famine”. Epidemiological analysis of this particular population of women revealed that, independent of weight at birth, children gestated during this famine had greater incidence of chronic degenerative diseases, altered structure and function of many organs, and increased behavioral abnormalities and mortality (Roseboom, 2019Roseboom TJ. Epidemiological evidence for the developmental origins of health and disease: effects of prenatal undernutrition in humans. J Endocrinol. 2019;242(1):T135-44. http://dx.doi.org/10.1530/JOE-18-0683. PMid:31207580.
http://dx.doi.org/10.1530/JOE-18-0683...
). In addition, male children with light birthweights had greater incidence of death from coronary disease (Godfrey and Barker, 2001Godfrey KM, Barker DJ. Fetal programming and adult health. Public Health Nutr. 2001;4(2B):611-24. http://dx.doi.org/10.1079/PHN2001145. PMid:11683554.
http://dx.doi.org/10.1079/PHN2001145...
). These studies also revealed programming effects that differed by stage of gestation during exposure and sex of the offspring. Subsequent research in livestock models has revealed that maternal nutrition can have impacts on major organ systems in the offspring including the musculoskeletal, digestive, reproductive, immune, endocrine, and central nervous systems (Wu et al., 2006Wu G, Bazer FW, Wallace JM, Spencer TE. Board-invited review: intrauterine growth retardation: implications for the animal sciences. J Anim Sci. 2006;84(9):2316-37. http://dx.doi.org/10.2527/jas.2006-156. PMid:16908634.
http://dx.doi.org/10.2527/jas.2006-156...
; Reynolds et al., 2010Reynolds LP, Borowicz PP, Caton JS, Vonnahme KA, Luther JS, Hammer CJ, Carlin KRM, Grazul-Bilska AT, Redmer DA. Developmental programming: the concept, large animal models, and the key role of uteroplacental vascular development. J Anim Sci. 2010;88(Suppl 13):E61-72. http://dx.doi.org/10.2527/jas.2009-2359. PMid:20023136.
http://dx.doi.org/10.2527/jas.2009-2359...
; Dahlen et al., 2021Dahlen CR, Borowicz PP, Ward AK, Caton JS, Czernik M, Palazzese L, Loi P, Reynolds LP. Programming of embryonic development. Int J Mol Sci. 2021;22(21):11668. http://dx.doi.org/10.3390/ijms222111668. PMid:34769097.
http://dx.doi.org/10.3390/ijms222111668...
; Hammer et al., 2023Hammer CJ, Caton JS, Dahlen CR, Ward AK, Borowicz PP, Reynolds LP. DOHaD: a menagerie of adaptations and perspectives: large animal models of developmental programming: sustenance, stress, and sex matter. Reproduction. 2023;165(6):F1-13. http://dx.doi.org/10.1530/REP-22-0453. PMid:36951791.
http://dx.doi.org/10.1530/REP-22-0453...
; Ibeagha-Awemu and Khatib, 2023Ibeagha-Awemu EM, Khatib H. Epigenetics of livestock health, production, and breeding. In: Tollefsbol TO, editor. Handbook of epigenetics. 3rd ed. Amsterdam: Academic Press; 2023. p. 569-610. http://dx.doi.org/10.1016/B978-0-323-91909-8.00041-4.
http://dx.doi.org/10.1016/B978-0-323-919...
; Zhao et al., 2023Zhao L, Liu X, Gomez NA, Gao Y, Son JS, Chae SA, Zhu MJ, Du M. Stage-specific nutritional management and developmental programming to optimize meat production. J Anim Sci Biotechnol. 2023;14(1):2. http://dx.doi.org/10.1186/s40104-022-00805-0. PMid:36597116.
http://dx.doi.org/10.1186/s40104-022-008...
).

Though extensive research has been conducted regarding the impacts of maternal environment during pregnancy on subsequent offspring outcomes, a growing body of work indicates that the experiences of a sire during spermatogenesis, sperm residence in the epididymis, and development of seminal fluids in the accessory sex glands may be implicated in offspring outcomes (Chan et al., 2020Chan JC, Morgan CP, Leu NA, Shetty A, Cisse YM, Nugent BM, Morrison KE, Jašarević E, Huang W, Kanyuch N, Rodgers AB, Bhanu NV, Berger DS, Garcia BA, Ament S, Kane M, Epperson CN, Bale TL. Reproductive tract extracellular vesicles are sufficient to transmit intergenerational stress and program neurodevelopment. Nat Commun. 2020;11(1):1499. http://dx.doi.org/10.1038/s41467-020-15305-w. PMid:32198406.
http://dx.doi.org/10.1038/s41467-020-153...
; Lismer and Kimmins, 2023Lismer A, Kimmins S. Emerging evidence that the mammalian sperm epigenome serves as a template for embryo development. Nat Commun. 2023;14(1):2142. http://dx.doi.org/10.1038/s41467-023-37820-2. PMid:37059740.
http://dx.doi.org/10.1038/s41467-023-378...
). This concept is termed as paternal programming and there are a variety of situations controlled by livestock herd managers that could result in paternal programming effects. The current review will focus on the proposed mechanisms of action of paternal programming and when and where these mechanisms are being initiated. The potential effect of timing of perturbations on these paternal programming associated mechanisms will also be discussed followed by a brief overview of research in non-livestock species and an update on current livestock research models of paternal programming.

Potential mechanisms of paternal programming

Epigenetic alterations, oxidative stress, immune responses, and the seminal microbiome are potential contributors to sire-borne effects on developmental programming. Sire environment can influence epigenetic marks of sperm including DNA methylation, histone modifications, and alterations of several types of coding (mRNA) and non-coding RNAs –miRNA, tRFs, piRNA, lncRNA, circRNA (Champroux et al., 2018Champroux A, Cocquet J, Henry-Berger J, Drevet JR, Kocer A. A decade of exploring the mammalian sperm epigenome: paternal epigenetic and transgenerational inheritance. Front Cell Dev Biol. 2018;6:50. http://dx.doi.org/10.3389/fcell.2018.00050. PMid:29868581.
http://dx.doi.org/10.3389/fcell.2018.000...
; Kretschmer and Gapp, 2022Kretschmer M, Gapp K. Deciphering the RNA universe in sperm in its role as a vertical information carrier. Environ Epigenet. 2022;8(1):dvac011. http://dx.doi.org/10.1093/eep/dvac011. PMid:35633894.
http://dx.doi.org/10.1093/eep/dvac011...
).Though extensive demethylation-remethlylation of the sire epigenome occurs in the embryo after fertilization, some sperm-carried epigenetic marks persist after demethylation, including those of imprinted genes, and can be present in the resultant blastocysts (Wu et al., 2020aWu C, Blondin P, Vigneault C, Labrecque R, Sirard M-A. Sperm miRNAs- potential mediators of bull age and early embryo development. BMC Genomics. 2020a;21(1):798. http://dx.doi.org/10.1186/s12864-020-07206-5. PMid:33198638.
http://dx.doi.org/10.1186/s12864-020-072...
). Mechanisms exist for sperm to transfer altered chromatin signatures, DNA methylation, and non-coding RNAs to the embryo, but it is unclear whether alterations in phenotype of offspring seen later in life are the result of an altered cascade of post-fertilization gene expression, or owing to the genes maintaining their pre-fertilization epigenetic marks and initiating cascades of gene expression later in life (Lismer and Kimmins, 2023Lismer A, Kimmins S. Emerging evidence that the mammalian sperm epigenome serves as a template for embryo development. Nat Commun. 2023;14(1):2142. http://dx.doi.org/10.1038/s41467-023-37820-2. PMid:37059740.
http://dx.doi.org/10.1038/s41467-023-378...
).

Oxidative stress in sperm can occur under conditions of heat stress, obesity, hypertension, insulin resistance, altered dietary ingredients or planes of nutrition, and psychological disorders (Ayad et al., 2022Ayad B, Omolaoye TS, Louw N, Ramsunder Y, Skosana BT, Oyeipo PI, Du Plessis SS. Oxidative stress and male infertility: evidence from a research perspective. Front Reprod Health. 2022;4:822257. http://dx.doi.org/10.3389/frph.2022.822257. PMid:36303652.
http://dx.doi.org/10.3389/frph.2022.8222...
), which likely comes from mitochondrial and enzymatic processes within sperm cells during these times of stress (Aitken, 2020Aitken RJ. Impact of oxidative stress on male and female germ cells: implications for fertility. Reproduction. 2020;159(4):R189-201. http://dx.doi.org/10.1530/REP-19-0452. PMid:31846434.
http://dx.doi.org/10.1530/REP-19-0452...
). Oxidative stress can subsequently lead to DNA damage in the sperm. The combination of oxidative stress and DNA damage has the potential to induce paternal programming effects on offspring resulting from damaged sperm that successfully fertilize an oocyte (Aitken, 2020Aitken RJ. Impact of oxidative stress on male and female germ cells: implications for fertility. Reproduction. 2020;159(4):R189-201. http://dx.doi.org/10.1530/REP-19-0452. PMid:31846434.
http://dx.doi.org/10.1530/REP-19-0452...
; Billah et al., 2022Billah MM, Khatiwada S, Morris MJ, Maloney CA. Effects of paternal overnutrition and interventions on future generations. Int J Obes. 2022;46(5):901-17. http://dx.doi.org/10.1038/s41366-021-01042-7. PMid:35022547.
http://dx.doi.org/10.1038/s41366-021-010...
).

Insemination is followed by an immune response in the female reproductive tract (Marey et al., 2023Marey MA, Ma D, Yoshino H, Elesh IF, Zinnah MA, Fiorenza MF, Moriyasu S, Miyamoto A. Sperm induce proinflammatory responses in the uterus and peripheral blood immune cells of artificially inseminated cows. J Reprod Dev. 2023;69(2):95-102. http://dx.doi.org/10.1262/jrd.2022-124. PMid:36775285.
http://dx.doi.org/10.1262/jrd.2022-124...
). The uterus and oviducts of cattle have site-specific pro- and anti-inflammatory responses at different times after insemination (Marey et al., 2020Marey MA, Ezz MA, Akthar I, Yousef MS, Imakawa K, Shimada M, Miyamoto A. Sensing sperm via maternal immune system: a potential mechanism for controlling microenvironment for fertility in the cow. J Anim Sci. 2020;98(Suppl 1):S88-95. http://dx.doi.org/10.1093/jas/skaa147. PMid:32810249.
http://dx.doi.org/10.1093/jas/skaa147...
). These immune and inflammatory responses are likely to be essential for fertilization, implantation, and embryo growth and development (Fair, 2015Fair T. The contribution of the maternal immune system to the establishment of pregnancy in cattle. Front Immunol. 2015;6:7. http://dx.doi.org/10.3389/fimmu.2015.00007. PMid:25674085.
http://dx.doi.org/10.3389/fimmu.2015.000...
; Yockey and Iwasaki, 2018Yockey LJ, Iwasaki A. Interferons and proinflammatory cytokines in pregnancy and fetal development. Immunity. 2018;49(3):397-412. http://dx.doi.org/10.1016/j.immuni.2018.07.017. PMid:30231982.
http://dx.doi.org/10.1016/j.immuni.2018....
). In cultured bovine endometrial epithelial cells, seminal plasma originating from high and low fertility bulls resulted in differential release of cytokines (Nongbua et al., 2020Nongbua T, Guo Y, Ntallaris T, Rubér M, Rodriguez-Martinez H, Humblot P, Morrell JM. Bull seminal plasma stimulates in vitro production of TGF-β, IL-6 and IL-8 from bovine endometrial epithelial cells, depending on dose and bull fertility. J Reprod Immunol. 2020;142:103179. http://dx.doi.org/10.1016/j.jri.2020.103179. PMid:32717675.
http://dx.doi.org/10.1016/j.jri.2020.103...
). Cytokine profiles of bovine ejaculates can also be influenced by bull nutrition (Harrison et al., 2023Harrison TD, Chaney EM, Brandt KJ, Ault-Seay TB, Payton RR, Schneider LG, Strickland LG, Schrick FN, McLean KJ. The effects of nutritional level and body condition score on cytokines in seminal plasma of beef bulls. Front Anim Sci. 2023;3:1078960. http://dx.doi.org/10.3389/fanim.2022.1078960.
http://dx.doi.org/10.3389/fanim.2022.107...
). Therefore, the potential exists for a single female to mount differential immune responses to semen depending on the seminal cytokine profile at the time of mating.

Semen also carries with it a rich and diverse microbiota (Koziol et al., 2022Koziol JH, Sheets T, Wickware CL, Johnson TA. Composition and diversity of the seminal microbiota in bulls and its association with semen parameters. Theriogenology. 2022;182:17-25. http://dx.doi.org/10.1016/j.theriogenology.2022.01.029. PMid:35123307.
http://dx.doi.org/10.1016/j.theriogenolo...
; Webb et al., 2023Webb EM, Holman DB, Schmidt KN, Crouse MS, Dahlen CR, Cushman RA, Snider AP, McCarthy KL, Amat S. A longitudinal characterization of the seminal microbiota and antibiotic resistance in yearling beef bulls subjected to different rates of gain. Microbiol Spectr. 2023;11(2):e0518022. http://dx.doi.org/10.1128/spectrum.05180-22. PMid:36916922.
http://dx.doi.org/10.1128/spectrum.05180...
). Bacteria present in semen could be deposited into the oocyte at fertilization, or act indirectly via modulation of the female immune system (Schoenmakers et al., 2019Schoenmakers S, Steegers-Theunissen R, Faas M. The matter of the reproductive microbiome. Obstet Med. 2019;12(3):107-15. http://dx.doi.org/10.1177/1753495X18775899. PMid:31523266.
http://dx.doi.org/10.1177/1753495X187758...
; Luecke et al., 2022Luecke SM, Webb EM, Dahlen CR, Reynolds LP, Amat S. Seminal and vagino-uterine microbiome and their individual and interactive effects on cattle fertility. Front Microbiol. 2022;13:1029128. http://dx.doi.org/10.3389/fmicb.2022.1029128. PMid:36425035.
http://dx.doi.org/10.3389/fmicb.2022.102...
). A recent mouse-based study reported that fish oil feeding to male mice for one spermatogenic cycle before breeding resulted in a reduction in lung inflammation and expression of pulmonary pro-inflammatory cytokines in pups (Rumph et al., 2023Rumph JT, Stephens VR, Ameli S, Brown LK, Rayford KJ, Nde PN, Osteen KG, Bruner-Tran KL. A paternal fish oil diet preconception reduces lung inflammation in a toxicant-driven murine model of new bronchopulmonary dysplasia. Mar Drugs. 2023;21(3):161. http://dx.doi.org/10.3390/md21030161. PMid:36976210.
http://dx.doi.org/10.3390/md21030161...
). These authors also observed that offspring mice born from sires fed fish oil during spermatogenesis had distinct gut microbiota compared to that of offspring born from control sires, demonstrating the influence of paternal diet on offspring early life microbial colonization (Rumph et al., 2022Rumph JT, Stephens VR, Ameli S, Gaines PN, Osteen KG, Bruner-Tran KL, Nde PN. A paternal fish oil diet preconception modulates the gut microbiome and attenuates necrotizing enterocolitis in neonatal mice. Mar Drugs. 2022;20(6):390. http://dx.doi.org/10.3390/md20060390. PMid:35736193.
http://dx.doi.org/10.3390/md20060390...
).

Where are the effects being initiated?

It’s important to point out that some of the potential programming may originate in the testicle, in the epididymis, in the accessory sex glands during development of seminal plasma that is only added to sperm near the time of ejaculation. Sperm DNA methylation and histone modifications mainly take place within the testicle during early stages of sperm development, with sperm miRNA loading thought to occur in later portions of the tract. However, a portion of the miRNAs present in testicular parenchyma of rams and bulls are also present in segments of the epididymis (Guan et al., 2014Guan Y, Malecki IA, Hawken PAR, Linden MD, Martin GB. Under-nutrition reduces spermatogenic efficiency and sperm velocity, and increases sperm DNA damage in sexually mature male sheep. Anim Reprod Sci. 2014;149(3-4):163-72. http://dx.doi.org/10.1016/j.anireprosci.2014.07.014. PMid:25086661.
http://dx.doi.org/10.1016/j.anireprosci....
; Lima et al., 2021Lima AO, Afonso J, Edson J, Marcellin E, Palfreyman R, Porto-Neto LR, Reverter A, Fortes MRS. Network analyses predict small RNAs that might modulate gene expression in the testis and epididymis of Bos indicus bulls. Front Genet. 2021;12:610116. http://dx.doi.org/10.3389/fgene.2021.610116. PMid:33995471.
http://dx.doi.org/10.3389/fgene.2021.610...
; Wu et al., 2021Wu C, Wang C, Zhai B, Zhao Y, Zhao Z, Yuan Z, Zhang M, Tian K, Fu X. Study of microRNA expression profile in different regions of ram epididymis. Reprod Domest Anim. 2021;56(9):1209-19. http://dx.doi.org/10.1111/rda.13978. PMid:34169586.
http://dx.doi.org/10.1111/rda.13978...
), indicating that the testicle could have been a loading site for a portion of sperm miRNAs. To further support this concept, an evaluation of non-coding RNAs in bull sperm starting in the testicular parenchyma and continuing throughout the regions of the epididymis revealed dynamic transitions in sperm miRNA content as the sperm moved through the reproductive tract (Sellem et al., 2021Sellem E, Marthey S, Rau A, Jouneau L, Bonnet A, Le Danvic C, Guyonnet B, Kiefer H, Jammes H, Schibler L. Dynamics of cattle sperm sncRNAs during maturation, from testis to ejaculated sperm. Epigenetics Chromatin. 2021;14(1):24. http://dx.doi.org/10.1186/s13072-021-00397-5. PMid:34030709.
http://dx.doi.org/10.1186/s13072-021-003...
). Sperm within the testicular parenchyma contained small amounts of miRNAs, and this amount increased substantially as the sperm moved through the approximately 14 day journey in the epididymis (Sellem et al., 2021Sellem E, Marthey S, Rau A, Jouneau L, Bonnet A, Le Danvic C, Guyonnet B, Kiefer H, Jammes H, Schibler L. Dynamics of cattle sperm sncRNAs during maturation, from testis to ejaculated sperm. Epigenetics Chromatin. 2021;14(1):24. http://dx.doi.org/10.1186/s13072-021-00397-5. PMid:34030709.
http://dx.doi.org/10.1186/s13072-021-003...
).

Extracellular vesicles are released in the epididymis (i.e. epididymosomes) and contain miRNAs which are subsequently loaded into sperm. Once the sncRNA are transferred from the epididymosomes to sperm, they can be delivered to the oocyte at the time of fertilization to regulate maternal RNAs in the cytoplasm of the oocyte, or into the nucleus of the cell to affect the transcriptome of the early embryo (Hur et al., 2017Hur SS, Cropley JE, Suter CM. Paternal epigenetic programming: evolving metabolic disease risk. J Mol Endocrinol. 2017;58(3):R159-68. http://dx.doi.org/10.1530/JME-16-0236. PMid:28100703.
http://dx.doi.org/10.1530/JME-16-0236...
). As proof of concept that epididymosomes are responsible for a portion of offspring programming, several investigators have isolated and injected miRNA from stressed males into unstressed zygotes, which resulted in offspring with a stressed phenotype (Chan et al., 2020Chan JC, Morgan CP, Leu NA, Shetty A, Cisse YM, Nugent BM, Morrison KE, Jašarević E, Huang W, Kanyuch N, Rodgers AB, Bhanu NV, Berger DS, Garcia BA, Ament S, Kane M, Epperson CN, Bale TL. Reproductive tract extracellular vesicles are sufficient to transmit intergenerational stress and program neurodevelopment. Nat Commun. 2020;11(1):1499. http://dx.doi.org/10.1038/s41467-020-15305-w. PMid:32198406.
http://dx.doi.org/10.1038/s41467-020-153...
; Duffy et al., 2021Duffy KA, Bale TL, Epperson CN. Germ cell drivers: transmission of preconception stress across generations. Front Hum Neurosci. 2021;15:642762. http://dx.doi.org/10.3389/fnhum.2021.642762. PMid:34322003.
http://dx.doi.org/10.3389/fnhum.2021.642...
; Wang et al., 2021Wang Y, Chen Z-P, Hu H, Lei J, Zhou Z, Yao B, Chen L, Liang G, Zhan S, Zhu X, Jin F, Ma R, Zhang J, Liang H, Xing M, Chen XR, Zhang CY, Zhu JN, Chen X. Sperm microRNAs confer depression susceptibility to offspring. Sci Adv. 2021;7(7):eabd7605. http://dx.doi.org/10.1126/sciadv.abd7605. PMid:33568480.
http://dx.doi.org/10.1126/sciadv.abd7605...
).

Interestingly, there is a body of work in rodents comparing vasectomized and intact males that demonstrates specific and independent effects of sperm and of seminal plasma (Morgan et al., 2020Morgan HL, Paganopoulou P, Akhtar S, Urquhart N, Philomin R, Dickinson Y, Watkins AJ. Paternal diet impairs F1 and F2 offspring vascular function through sperm and seminal plasma specific mechanisms in mice. J Physiol. 2020;598(4):699-715. http://dx.doi.org/10.1113/JP278270. PMid:31617219.
http://dx.doi.org/10.1113/JP278270...
; Morgan and Watkins, 2020Morgan HL, Watkins AJ. The influence of seminal plasma on offspring development and health. Semin Cell Dev Biol. 2020;97:131-7. http://dx.doi.org/10.1016/j.semcdb.2019.06.008. PMid:31254609.
http://dx.doi.org/10.1016/j.semcdb.2019....
). In these models, the signaling for altered phenotype is likely originating from components of seminal plasma excreted by accessory sex organs and might include cytokines, hormones, specific nutrients, or extracellular vesicles originating from the ampulla, seminal vesicles, or the prostate glands (Morgan and Watkins, 2020Morgan HL, Watkins AJ. The influence of seminal plasma on offspring development and health. Semin Cell Dev Biol. 2020;97:131-7. http://dx.doi.org/10.1016/j.semcdb.2019.06.008. PMid:31254609.
http://dx.doi.org/10.1016/j.semcdb.2019....
; Roca et al., 2022Roca J, Rodriguez-Martinez H, Padilla L, Lucas X, Barranco I. Extracellular vesicles in seminal fluid and effects on male reproduction. An overview in farm animals and pets. Anim Reprod Sci. 2022;246:106853. http://dx.doi.org/10.1016/j.anireprosci.2021.106853. PMid:34556398.
http://dx.doi.org/10.1016/j.anireprosci....
). An additional concept that cannot be discounted is the potential interaction of the sperm and seminal plasma components with the female reproductive tract in terms of the immune response and extracellular vesicles of uterine origin (Sharma, 2019Sharma U. Paternal contributions to offspring health: role of sperm small RNAs in intergenerational transmission of epigenetic information. Front Cell Dev Biol. 2019;7:215. http://dx.doi.org/10.3389/fcell.2019.00215. PMid:31681757.
http://dx.doi.org/10.3389/fcell.2019.002...
). Research in livestock species that evaluates the independent and combined effects of sperm and seminal plasma on the female immune response and subsequent offspring programming is warranted.

Timing of alterations

The timing of sperm or seminal plasma alterations can also be broken into several categories; time in terms of sire age, relative age of developing sperm, lag time from insult to observable effect, and persistence of effects in the epididymis or other regions of the male reproductive tract (Table 1). The effects of sire age can be broken down into further intervals, including the prenatal, pre-pubertal, peri-pubertal, reproductive maturity, and advanced reproductive age periods. Strong evidence in rodent models suggest that stimuli experienced even during the prenatal period can affect epigenetic marks in the offspring’s sperm that are produced during adulthood (Radford et al., 2014Radford EJ, Ito M, Shi H, Corish JA, Yamazawa K, Isganaitis E, Seisenberger S, Hore TA, Reik W, Erkek S, Peters AHFM, Patti ME, Ferguson-Smith AC. In utero undernourishment perturbs the adult sperm methylome and intergenerational metabolism. Science. 2014;345(6198):1255903. http://dx.doi.org/10.1126/science.1255903. PMid:25011554.
http://dx.doi.org/10.1126/science.125590...
). An additional report in sheep indicated that prenatal ewe nutrition can have an impact on offspring epigenetic marks in sperm at reproductive maturity (Toschi et al., 2020Toschi P, Capra E, Anzalone DA, Lazzari B, Turri F, Pizzi F, Scapolo PA, Stella A, Williams JL, Marsan PA, Loi P. Maternal peri-conceptional undernourishment perturbs offspring sperm methylome. Reproduction. 2020;159(5):513-23. http://dx.doi.org/10.1530/REP-19-0549. PMid:32103819.
http://dx.doi.org/10.1530/REP-19-0549...
). Sperm methylation patterns in post-pubertal bulls are influenced by their respective plane of nutritional management during the first 24 (Perrier et al., 2020Perrier JP, Kenny DA, Chaulot-Talmon A, Byrne CJ, Sellem E, Jouneau L, Aubert-Frambourg A, Schibler L, Jammes H, Lonergan P, Fair S, Kiefer H. Accelerating onset of puberty through modification of early life nutrition induces modest but persistent changes in bull sperm DNA methylation profiles post-puberty. Front Genet. 2020;11:945. http://dx.doi.org/10.3389/fgene.2020.00945. PMid:33005172.
http://dx.doi.org/10.3389/fgene.2020.009...
), or 32 (Johnson et al., 2022Johnson C, Kiefer H, Chaulot-Talmon A, Dance A, Sellem E, Jouneau L, Jammes H, Kastelic J, Thundathil J. Prepubertal nutritional modulation in the bull and its impact on sperm DNA methylation. Cell Tissue Res. 2022;389(3):587-601. http://dx.doi.org/10.1007/s00441-022-03659-0. PMid:35779136.
http://dx.doi.org/10.1007/s00441-022-036...
) weeks of age, which also encompasses the period of major Sertoli cell proliferation and the subsequent early increase in the number of germ cells (Negrin et al., unpublished). Perhaps spermatogonial stem cells are particularly susceptible to epigenetic alterations during this early period of rapid proliferation.

Table 1
Key concepts related to timing of epigenetic alterations.

Methylation patterns can be changed with advancing peri-pubertal age. Bulls increasing in age from 10 to 16 months had alterations in their sperm miRNA profiles and DNA methylation patterns (Lambert et al., 2018Lambert S, Blondin P, Vigneault C, Labrecque R, Dufort I, Sirard M-A. Spermatozoa DNA methylation patterns differ due to peripubertal age in bulls. Theriogenology. 2018;106:21-9. http://dx.doi.org/10.1016/j.theriogenology.2017.10.006. PMid:29031946.
http://dx.doi.org/10.1016/j.theriogenolo...
; Wu et al., 2020bWu C, Blondin P, Vigneault C, Labrecque R, Sirard M-A. The age of the bull influences the transcriptome and epigenome of blastocysts produced by IVF. Theriogenology. 2020b;144:122-31. http://dx.doi.org/10.1016/j.theriogenology.2019.12.020. PMid:31951983.
http://dx.doi.org/10.1016/j.theriogenolo...
). Once altered during the pre- and peri-pubertal period, some of these DNA methylation patterns persist into adulthood (Gross et al., 2020Gross N, Taylor T, Crenshaw T, Khatib H. The intergenerational impacts of paternal diet on DNA methylation and offspring phenotypes in sheep. Front Genet. 2020;11:597943. http://dx.doi.org/10.3389/fgene.2020.597943. PMid:33250925.
http://dx.doi.org/10.3389/fgene.2020.597...
). Epigenetic alterations continue to change throughout the reproductive lifespan of breeding males, but are especially susceptible to changes during the peri-pubertal and advanced paternal age periods (Ashapkin et al., 2023Ashapkin V, Suvorov A, Pilsner JR, Krawetz SA, Sergeyev O. Age-associated epigenetic changes in mammalian sperm: implications for offspring health and development. Hum Reprod Update. 2023;29(1):24-44. http://dx.doi.org/10.1093/humupd/dmac033. PMid:36066418.
http://dx.doi.org/10.1093/humupd/dmac033...
). The latter could be a mechanism linking advanced paternal age with increased development and health disorders of their children (Phillips et al., 2019Phillips N, Taylor L, Bachmann G. Maternal, infant and childhood risks associated with advanced paternal age: the need for comprehensive counseling for men. Maturitas. 2019;125:81-4. http://dx.doi.org/10.1016/j.maturitas.2019.03.020. PMid:31133222.
http://dx.doi.org/10.1016/j.maturitas.20...
).

The time required for spermatogenesis varies among species, with bulls completing the process in roughly 61 days (Staub and Johnson, 2018Staub C, Johnson L. Review: spermatogenesis in the bull. Animal. 2018;12(Suppl 1):s27-35. http://dx.doi.org/10.1017/S1751731118000435. PMid:29882505.
http://dx.doi.org/10.1017/S1751731118000...
) and rams every 47 days (Senger, 2012Senger P. Pathways to pregnancy and parturition. 3rd ed. Redmond: Current Conceptions, Inc., 2012.). A critical feature of anatomy to consider at this point is the blood/testis barrier, which comprises tight junctions between Sertoli cells and acts to protect developing sperm cells from blood-borne components of the host system (Xia et al., 2007Xia W, Mruk DD, Cheng CY. C-type natriuretic peptide regulates blood–testis barrier dynamics in adult rat testes. Proc Natl Acad Sci USA. 2007;104(10):3841-6. http://dx.doi.org/10.1073/pnas.0610100104. PMid:17360440.
http://dx.doi.org/10.1073/pnas.061010010...
). The structure of the seminiferous epithelium is such that the youngest developmental stages of sperm are present outside of the blood-testis barrier, and as they advance in developmental age they reposition closer to the lumen of the seminiferous tubule before being released during spermiation. Therefore, the age of developing sperm affects both the physical contact with components outside of the blood-testis barrier (with spermatogonia and sperm stem cells being outside of the blood testis barrier) and the specific type of epigenetic alterations that are able to occur given the dynamic changes in sperm DNA packaging at the different stages of development (Marcho et al., 2020Marcho C, Oluwayiose OA, Pilsner JR. The preconception environment and sperm epigenetics. Andrology. 2020;8(4):924-42. http://dx.doi.org/10.1111/andr.12753. PMid:31901222.
http://dx.doi.org/10.1111/andr.12753...
; Kiefer et al., 2021Kiefer H, Sellem E, Bonnet-Garnier A, Pannetier M, Costes V, Schibler L, Jammes H. The epigenome of male germ cells and the programming of phenotypes in cattle. Anim Front. 2021;11(6):28-38. http://dx.doi.org/10.1093/af/vfab062. PMid:34934527.
http://dx.doi.org/10.1093/af/vfab062...
).

Additionally, the nutrient-sensing mTOR pathway can contribute to the regulation of blood-testis barrier and affect sperm at several stages of development (Moreira et al., 2019Moreira BP, Oliveira PF, Alves MG. Molecular mechanisms controlled by mTOR in male reproductive system. Int J Mol Sci. 2019;20(7):1633. http://dx.doi.org/10.3390/ijms20071633. PMid:30986927.
http://dx.doi.org/10.3390/ijms20071633...
), providing a potential mechanism through which the sire’s nutrition can impact developing sperm within the testicle. Further evidence of stage of sperm development interacting with external stressors is found in models of bull heat stress recovery, where specific sperm morphological abnormalities can be found in ejaculates at specific times after heat stress (Table 2). This indicates that sperm of different developmental stages respond to stressors differently (Rahman et al., 2018Rahman MB, Schellander K, Luceño NL, Van Soom A. Heat stress responses in spermatozoa: mechanisms and consequences for cattle fertility. Theriogenology. 2018;113:102-12. http://dx.doi.org/10.1016/j.theriogenology.2018.02.012. PMid:29477908.
http://dx.doi.org/10.1016/j.theriogenolo...
; Garcia-Oliveros et al., 2022Garcia-Oliveros LN, Arruda RP, Batissaco L, Gonzaga VHG, Nogueira VJM, Florez-Rodriguez SA, Almeida FDS, Alves MBR, Pinto SCC, Nichi M, Losano JDA, Kawai GKV, Celeghini ECC. Chronological characterization of sperm morpho-functional damage and recovery after testicular heat stress in Nellore bulls. J Therm Biol. 2022;106:103237. http://dx.doi.org/10.1016/j.jtherbio.2022.103237. PMid:35636895.
http://dx.doi.org/10.1016/j.jtherbio.202...
).

Table 2
Impact of time after heat stress on abnormalities observed in sperm from Nellore bulls.* * Adapted from Garcia-Oliveros et al. (2022).

Lag from insult to injury

The lag time from a perturbation until an effect manifestation likely depends on the site of the effect (e.g. whether the perturbation affected developing sperm, the miRNA payload in the epididymis, seminal plasma composition, the seminal microbiome, or come combination of these). When male mice were given a single dose of dexamethasone to induce acute stress, relatively few changes were observed in metabolism of offspring sired by sperm collected at 3 hours after dexamethasone administration. However, modest alterations were observed in the offspring sired by sperm collected at 7 days after administration, and many alterations in RNA payload and offspring metabolism were observed from matings with semen collected 14 days after dexamethasone administration (Gapp et al., 2021Gapp K, Parada GE, Gross F, Corcoba A, Kaur J, Grau E, Hemberg M, Bohacek J, Miska EA. Single paternal dexamethasone challenge programs offspring metabolism and reveals multiple candidates in RNA-mediated inheritance. iScience. 2021;24(8):102870. http://dx.doi.org/10.1016/j.isci.2021.102870. PMid:34386731.
http://dx.doi.org/10.1016/j.isci.2021.10...
). In a model of bull heat stress, the miRNA content of extracellular vesicles was altered at 7 days and sperm miRNA was altered at 21 days after heat stress (Alves et al., 2021Alves MBR, Arruda RP, Batissaco L, Garcia-Oliveros LN, Gonzaga VHG, Nogueira VJM, Almeida FDS, Pinto SCC, Andrade GM, Perecin F, Silveira JC, Celeghini ECC. Changes in miRNA levels of sperm and small extracellular vesicles of seminal plasma are associated with transient scrotal heat stress in bulls. Theriogenology. 2021;161:26-40. http://dx.doi.org/10.1016/j.theriogenology.2020.11.015. PMid:33278692.
http://dx.doi.org/10.1016/j.theriogenolo...
). These observations were supported by another study where some sperm morphological characteristics were impacted in the period immediately after heat stress and others requiring more time to be present in the ejaculate (Garcia-Oliveros et al., 2022Garcia-Oliveros LN, Arruda RP, Batissaco L, Gonzaga VHG, Nogueira VJM, Florez-Rodriguez SA, Almeida FDS, Alves MBR, Pinto SCC, Nichi M, Losano JDA, Kawai GKV, Celeghini ECC. Chronological characterization of sperm morpho-functional damage and recovery after testicular heat stress in Nellore bulls. J Therm Biol. 2022;106:103237. http://dx.doi.org/10.1016/j.jtherbio.2022.103237. PMid:35636895.
http://dx.doi.org/10.1016/j.jtherbio.202...
). Samples, however, were not analyzed to characterize the lag time required for altered epigenetic marks to be present in the ejaculate in a bovine model (Garcia-Oliveros et al., 2022Garcia-Oliveros LN, Arruda RP, Batissaco L, Gonzaga VHG, Nogueira VJM, Florez-Rodriguez SA, Almeida FDS, Alves MBR, Pinto SCC, Nichi M, Losano JDA, Kawai GKV, Celeghini ECC. Chronological characterization of sperm morpho-functional damage and recovery after testicular heat stress in Nellore bulls. J Therm Biol. 2022;106:103237. http://dx.doi.org/10.1016/j.jtherbio.2022.103237. PMid:35636895.
http://dx.doi.org/10.1016/j.jtherbio.202...
).

Persistence of post-insult effects

The concept of the lag from injury/stressor to effect is also related to the concept of length of time post-insult that the respective effects persist. In a model of a 4-week chronic stress in mice (induced by altering variables including 36 h of constant light, 1 h exposure to predator odor, 15 minute restraint, introducing novel objects or white noise overnight, multiple cage changed, and saturated bedding), untreated sperm were incubated with extracellular vesicles from stressed males at different time points after the stress, and then used to produce in vitro fertilization (IVF) embryos (Chan et al., 2020Chan JC, Morgan CP, Leu NA, Shetty A, Cisse YM, Nugent BM, Morrison KE, Jašarević E, Huang W, Kanyuch N, Rodgers AB, Bhanu NV, Berger DS, Garcia BA, Ament S, Kane M, Epperson CN, Bale TL. Reproductive tract extracellular vesicles are sufficient to transmit intergenerational stress and program neurodevelopment. Nat Commun. 2020;11(1):1499. http://dx.doi.org/10.1038/s41467-020-15305-w. PMid:32198406.
http://dx.doi.org/10.1038/s41467-020-153...
). Interestingly, the extracellular vesicles collected one week after the stress did not result in altered offspring hypothalamic-pituitary-adrenal (HPA) phenotype, but extracellular vesicles collected 12 weeks after the stress (nearly 2.5 times the duration of spermatogenesis in mice) resulted in an altered HPA phenotype. This observation indicates that effects can persist specifically in extracellular vesicles for some time after the perturbation (Chan et al., 2020Chan JC, Morgan CP, Leu NA, Shetty A, Cisse YM, Nugent BM, Morrison KE, Jašarević E, Huang W, Kanyuch N, Rodgers AB, Bhanu NV, Berger DS, Garcia BA, Ament S, Kane M, Epperson CN, Bale TL. Reproductive tract extracellular vesicles are sufficient to transmit intergenerational stress and program neurodevelopment. Nat Commun. 2020;11(1):1499. http://dx.doi.org/10.1038/s41467-020-15305-w. PMid:32198406.
http://dx.doi.org/10.1038/s41467-020-153...
). An evaluation of men who were self-reportedly recovering from stress showed large shifts in miRNA expression compared with men who reported being relatively stress-free (Chan et al., 2020Chan JC, Morgan CP, Leu NA, Shetty A, Cisse YM, Nugent BM, Morrison KE, Jašarević E, Huang W, Kanyuch N, Rodgers AB, Bhanu NV, Berger DS, Garcia BA, Ament S, Kane M, Epperson CN, Bale TL. Reproductive tract extracellular vesicles are sufficient to transmit intergenerational stress and program neurodevelopment. Nat Commun. 2020;11(1):1499. http://dx.doi.org/10.1038/s41467-020-15305-w. PMid:32198406.
http://dx.doi.org/10.1038/s41467-020-153...
). Thus, the persistence of altered RNA payload and potential paternal programming effects in livestock models should be a main target of future investigations.

Evidence of paternal programming in rodent models

An exhaustive review of rodent models of paternal programming is beyond the scope of this paper. However, it is important to note that many studies have clearly demonstrated that the environment experienced by sires during spermatogenesis impacts offspring phenotype. Paternal programming effects have been observed in offspring of sires managed in over/under nutrition models (Billah et al., 2022Billah MM, Khatiwada S, Morris MJ, Maloney CA. Effects of paternal overnutrition and interventions on future generations. Int J Obes. 2022;46(5):901-17. http://dx.doi.org/10.1038/s41366-021-01042-7. PMid:35022547.
http://dx.doi.org/10.1038/s41366-021-010...
), on high fat (Claycombe-Larson et al., 2020Claycombe-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. http://dx.doi.org/10.1016/j.jnutbio.2020.108373. PMid:32422425.
http://dx.doi.org/10.1016/j.jnutbio.2020...
) or low protein diets (Watkins et al., 2017Watkins 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(6):1371-81. http://dx.doi.org/10.1016/j.bbadis.2017.02.009. PMid:28189722.
http://dx.doi.org/10.1016/j.bbadis.2017....
). Similar findings were reported in cases of targeted supplementation (folic acid, B-12, methionine; components of the one-carbon metabolism pathway that induces epigenetic alterations; (Lambrot et al., 2013Lambrot R, Xu C, Saint-Phar S, Chountalos G, Cohen T, Paquet M, Suderman M, Hallett M, Kimmins S. Low paternal dietary folate alters the mouse sperm epigenome and is associated with negative pregnancy outcomes. Nat Commun. 2013;4(1):2889. http://dx.doi.org/10.1038/ncomms3889. PMid:24326934.
http://dx.doi.org/10.1038/ncomms3889...
; Bailey et al., 2020Bailey JL, Dalvai M, Lessard M, Herst PM, Charest PL, Navarro P. Beyond fertilisation: how the paternal environment influences future generations. Anim Reprod Sci. 2020;220:106503. http://dx.doi.org/10.1016/j.anireprosci.2020.106503. PMid:32536524.
http://dx.doi.org/10.1016/j.anireprosci....
), acute and chronic stress (Duffy et al., 2021Duffy KA, Bale TL, Epperson CN. Germ cell drivers: transmission of preconception stress across generations. Front Hum Neurosci. 2021;15:642762. http://dx.doi.org/10.3389/fnhum.2021.642762. PMid:34322003.
http://dx.doi.org/10.3389/fnhum.2021.642...
), exercise (Kusuyama et al., 2020Kusuyama J, Alves-Wagner AB, Makarewicz NS, Goodyear LJ. Effects of maternal and paternal exercise on offspring metabolism. Nat Metab. 2020;2(9):858-72. http://dx.doi.org/10.1038/s42255-020-00274-7. PMid:32929233.
http://dx.doi.org/10.1038/s42255-020-002...
), and exposure to drugs (Toussaint et al., 2023Toussaint AB, Ellis AS, Bongiovanni AR, Peterson DR, Bavley CC, Karbalaei R, Mayberry HL, Bhakta S, Dressler CC, Imperio CG, Maurer JJ, Schmidt HD, Chen C, Bland K, Liu-Chen LY, Wimmer ME. Paternal morphine exposure enhances morphine self-administration and induces region-specific neural adaptations in reward-related brain regions of male offspring. bioRxiv. In press 2023. PMid:36711571. in press), alcohol (Lee et al., 2013Lee HJ, Ryu J-S, Choi NY, Park YS, Kim YI, Han DW, Ko K, Shin CY, Hwang HS, Kang K-S, Ko K. Transgenerational effects of paternal alcohol exposure in mouse offspring. Anim Cells Syst. 2013;17(6):429-34. http://dx.doi.org/10.1080/19768354.2013.865675.
http://dx.doi.org/10.1080/19768354.2013....
), and endocrine disruptors (Liu et al., 2023Liu J, Shi J, Hernandez R, Li X, Konchadi P, Miyake Y, Chen Q, Zhou T, Zhou C. Paternal phthalate exposure-elicited offspring metabolic disorders are associated with altered sperm small RNAs in mice. Environ Int. 2023;172:107769. http://dx.doi.org/10.1016/j.envint.2023.107769. PMid:36709676.
http://dx.doi.org/10.1016/j.envint.2023....
).

The array of effects on offspring outcomes is model-dependent. Altered phenotype variables that have been reported include changes in appetite regulation; glucose and insulin metabolism; energy metabolism; growth and development of muscle, fat, and bone; reproductive efficiency; cardiovascular health; temperament; social and cognitive abilities; predator avoidance; drug and alcohol preference and dependency; and propensity for depression-associated behaviors [recently reviewed by Lismer and Kimmins (2023)Lismer A, Kimmins S. Emerging evidence that the mammalian sperm epigenome serves as a template for embryo development. Nat Commun. 2023;14(1):2142. http://dx.doi.org/10.1038/s41467-023-37820-2. PMid:37059740.
http://dx.doi.org/10.1038/s41467-023-378...
]. It’s also worth noting that several models have resulted in similar measurable responses in offspring (i.e. alterations in glucose/insulin metabolic responses) (Sharma, 2019Sharma U. Paternal contributions to offspring health: role of sperm small RNAs in intergenerational transmission of epigenetic information. Front Cell Dev Biol. 2019;7:215. http://dx.doi.org/10.3389/fcell.2019.00215. PMid:31681757.
http://dx.doi.org/10.3389/fcell.2019.002...
), begging the question whether different types of stressors in livestock sires could culminate in similar offspring outcomes. In addition, there are differences in the RNA landscape between rodents and livestock species, thus emphasizing the need for livestock-specific work (Chukrallah et al., 2021Chukrallah LG, Badrinath A, Seltzer K, Snyder EM. Of rodents and ruminants: a comparison of small noncoding RNA requirements in mouse and bovine reproduction. J Anim Sci. 2021;99(3):skaa388. http://dx.doi.org/10.1093/jas/skaa388. PMid:33677580.
http://dx.doi.org/10.1093/jas/skaa388...
).

Evidence of paternal programming in livestock models

The body of literature in livestock species is not nearly as extensive as in rodent models. Several models exists where management-related strategies have shown mechanisms of paternal programming in livestock similar to those reported in rodent models and will be reviewed below.

Effects on mechanisms of action associated with paternal programming

Several authors have focused on discovering biomarkers for fertility-related epigenetic changes in bulls used for artificial insemination (AI), and have reported that sperm methylation (Kropp et al., 2017Kropp J, Carrillo JA, Namous H, Daniels A, Salih SM, Song J, Khatib H. Male fertility status is associated with DNA methylation signatures in sperm and transcriptomic profiles of bovine preimplantation embryos. BMC Genomics. 2017;18(1):280. http://dx.doi.org/10.1186/s12864-017-3673-y. PMid:28381255.
http://dx.doi.org/10.1186/s12864-017-367...
; Costes et al., 2022Costes V, Chaulot-Talmon A, Sellem E, Perrier JP, Aubert-Frambourg A, Jouneau L, Pontlevoy C, Hozé C, Fritz S, Boussaha M, Le Danvic C, Sanchez MP, Boichard D, Schibler L, Jammes H, Jaffrézic F, Kiefer H. Predicting male fertility from the sperm methylome: application to 120 bulls with hundreds of artificial insemination records. Clin Epigenetics. 2022;14(1):54. http://dx.doi.org/10.1186/s13148-022-01275-x. PMid:35477426.
http://dx.doi.org/10.1186/s13148-022-012...
), miRNA abundance (Werry et al., 2022Werry N, Russell SJ, Gillis DJ, Miller S, Hickey K, Larmer S, Lohuis M, Librach C, LaMarre J. Characteristics of miRNAs present in bovine sperm and associations with differences in fertility. Front Endocrinol. 2022;13:874371. http://dx.doi.org/10.3389/fendo.2022.874371. PMid:35663333.
http://dx.doi.org/10.3389/fendo.2022.874...
), and histone modifications (Kutchy et al., 2018Kutchy NA, Menezes ESB, Chiappetta A, Tan W, Wills RW, Kaya A, Topper E, Moura AA, Perkins AD, Memili E. Acetylation and methylation of sperm histone 3 lysine 27 (H3K27ac and H3K27me3) are associated with bull fertility. Andrologia. 2018;50(3):e12915. http://dx.doi.org/10.1111/and.12915. PMid:29057498.
http://dx.doi.org/10.1111/and.12915...
) differ among bulls of high or low fertility. In addition, seminal plasma from high and low fertility bulls affected the cytokine response of cultured cells differently (Nongbua et al., 2020Nongbua T, Guo Y, Ntallaris T, Rubér M, Rodriguez-Martinez H, Humblot P, Morrell JM. Bull seminal plasma stimulates in vitro production of TGF-β, IL-6 and IL-8 from bovine endometrial epithelial cells, depending on dose and bull fertility. J Reprod Immunol. 2020;142:103179. http://dx.doi.org/10.1016/j.jri.2020.103179. PMid:32717675.
http://dx.doi.org/10.1016/j.jri.2020.103...
). As previously mentioned, sperm methylation patterns (Lambert et al., 2018Lambert S, Blondin P, Vigneault C, Labrecque R, Dufort I, Sirard M-A. Spermatozoa DNA methylation patterns differ due to peripubertal age in bulls. Theriogenology. 2018;106:21-9. http://dx.doi.org/10.1016/j.theriogenology.2017.10.006. PMid:29031946.
http://dx.doi.org/10.1016/j.theriogenolo...
) and microRNA expression (Wu et al., 2020bWu C, Blondin P, Vigneault C, Labrecque R, Sirard M-A. The age of the bull influences the transcriptome and epigenome of blastocysts produced by IVF. Theriogenology. 2020b;144:122-31. http://dx.doi.org/10.1016/j.theriogenology.2019.12.020. PMid:31951983.
http://dx.doi.org/10.1016/j.theriogenolo...
) were influenced by age of bulls during the peri-pubertal period.

Plane of nutrition was reported to influence mechanisms of paternal programming, with underfed rams having differentially expressed miRNAs in testicular tissue (Guan et al., 2015Guan Y, Liang G, Hawken PAR, Malecki IA, Cozens G, Vercoe PE, Martin GB, Guan LL. Roles of small RNAs in the effects of nutrition on apoptosis and spermatogenesis in the adult testis. Sci Rep. 2015;5(1):10372. http://dx.doi.org/10.1038/srep10372. PMid:25996545.
http://dx.doi.org/10.1038/srep10372...
), increased DNA damage in sperm (Guan et al., 2014Guan Y, Malecki IA, Hawken PAR, Linden MD, Martin GB. Under-nutrition reduces spermatogenic efficiency and sperm velocity, and increases sperm DNA damage in sexually mature male sheep. Anim Reprod Sci. 2014;149(3-4):163-72. http://dx.doi.org/10.1016/j.anireprosci.2014.07.014. PMid:25086661.
http://dx.doi.org/10.1016/j.anireprosci....
), as well as altered miRNA, mRNA, and pre-mRNA splicing in sperm compared with overfed rams (Guan et al., 2017Guan Y, Liang G, Martin GB, Guan LL. Functional changes in mRNA expression and alternative pre-mRNA splicing associated with the effects of nutrition on apoptosis and spermatogenesis in the adult testis. BMC Genomics. 2017;18(1):64. http://dx.doi.org/10.1186/s12864-016-3385-8. PMid:28068922.
http://dx.doi.org/10.1186/s12864-016-338...
). In yearling and mature bulls experiencing body weight fluctuations, sperm methylation was altered (Moura et al., 2022Moura FH, Macias-Franco A, Pena-Bello CA, Archilia EC, Batalha IM, Silva AE, Moreira GM, Norris AB, Schütz LF, Fonseca MA. Sperm DNA 5-methyl cytosine and RNA N 6-methyladenosine methylation are differently affected during periods of body weight losses and body weight gain of young and mature breeding bulls. J Anim Sci. 2022;100(2):skab362. http://dx.doi.org/10.1093/jas/skab362. PMid:34902028.
http://dx.doi.org/10.1093/jas/skab362...
) and semen cytokine profiles were altered by plane of nutrition (Harrison et al., 2023Harrison TD, Chaney EM, Brandt KJ, Ault-Seay TB, Payton RR, Schneider LG, Strickland LG, Schrick FN, McLean KJ. The effects of nutritional level and body condition score on cytokines in seminal plasma of beef bulls. Front Anim Sci. 2023;3:1078960. http://dx.doi.org/10.3389/fanim.2022.1078960.
http://dx.doi.org/10.3389/fanim.2022.107...
). Heat stress in bulls induced oxidative stress, altered sperm characteristics, miRNA profile of sperm and extracellular vesicles (Alves et al., 2021Alves MBR, Arruda RP, Batissaco L, Garcia-Oliveros LN, Gonzaga VHG, Nogueira VJM, Almeida FDS, Pinto SCC, Andrade GM, Perecin F, Silveira JC, Celeghini ECC. Changes in miRNA levels of sperm and small extracellular vesicles of seminal plasma are associated with transient scrotal heat stress in bulls. Theriogenology. 2021;161:26-40. http://dx.doi.org/10.1016/j.theriogenology.2020.11.015. PMid:33278692.
http://dx.doi.org/10.1016/j.theriogenolo...
), and sperm chromatin protamination (Rahman et al., 2011Rahman MB, Vandaele L, Rijsselaere T, Maes D, Hoogewijs M, Frijters A, Noordman J, Granados A, Dernelle E, Shamsuddin M, Parrish JJ, Van Soom A. Scrotal insulation and its relationship to abnormal morphology, chromatin protamination and nuclear shape of spermatozoa in Holstein-Friesian and Belgian Blue bulls. Theriogenology. 2011;76(7):1246-57. http://dx.doi.org/10.1016/j.theriogenology.2011.05.031. PMid:21777969.
http://dx.doi.org/10.1016/j.theriogenolo...
). Though each of the models (and other similar models not referenced in the current review) provide a glimpse of their potential to elicit paternal programming responses, fewer livestock models have followed alterations in sperm through the processes of fertilization, embryo and fetal development, and parturition necessary to verify paternal programming responses.

Models demonstrating paternal programming effects

Specific examples do exist in swine, sheep, and bovine models in which paternal programming was evident (Table 3). A recent report examining the effects of boar housing revealed that boars housed in enriched crates during pre-breeding spermatogenesis sired more piglets than boars housed in crates without enrichment (Sabei et al., 2023Sabei L, Bernardino T, Parada Sarmiento M, Barbosa BS, Farias SS, Ghantous GF, Lima CG, Poletto R, Zanella AJ. Life experiences of boars can shape the survival, aggression, and nociception responses of their offspring. Front Anim Sci. 2023;4:1142628. http://dx.doi.org/10.3389/fanim.2023.1142628.
http://dx.doi.org/10.3389/fanim.2023.114...
). In addition, piglets sired by pen-raised boars had reduced incidence of skin lesions and an attenuated response to potentially painful stimulus (Sabei et al., 2023Sabei L, Bernardino T, Parada Sarmiento M, Barbosa BS, Farias SS, Ghantous GF, Lima CG, Poletto R, Zanella AJ. Life experiences of boars can shape the survival, aggression, and nociception responses of their offspring. Front Anim Sci. 2023;4:1142628. http://dx.doi.org/10.3389/fanim.2023.1142628.
http://dx.doi.org/10.3389/fanim.2023.114...
), indicating effects on stress/emotional response areas of the brain. Feeding methyl donors (methionine, cysteine, choline, betaine, vitamin B6, folate, and vitamin B12) to boars resulted in offspring that had differential gene expression and DNA methylation, as well as altered back fat (Braunschweig et al., 2012Braunschweig M, Jagannathan V, Gutzwiller A, Bee G. Investigations on transgenerational epigenetic response down the male line in F2 pigs. PLoS One. 2012;7(2):e30583. http://dx.doi.org/10.1371/journal.pone.0030583. PMid:22359544.
http://dx.doi.org/10.1371/journal.pone.0...
). Similarly, when sibling ram pairs were fed a diet with or without a rumen-protected methionine from weaning to puberty their post-pubertal sperm methylation was altered, and subsequent F1 offspring from rams that received methionine had reduced body weight at puberty and had smaller scrotal circumference than their counterparts from rams that did not receive methionine (Gross et al., 2020Gross N, Taylor T, Crenshaw T, Khatib H. The intergenerational impacts of paternal diet on DNA methylation and offspring phenotypes in sheep. Front Genet. 2020;11:597943. http://dx.doi.org/10.3389/fgene.2020.597943. PMid:33250925.
http://dx.doi.org/10.3389/fgene.2020.597...
).

Table 3
Summary of livestock models demonstrating paternal programming effects.

Following up on their observations of altered miRNA and DNA methylation in bulls of different ages, Sirard’s research group reported alterations in methylation patterns and gene expression in oocytes derived from IVF using semen collected from the same bulls at 10, 12, and 16 months of age (Wu et al., 2020aWu C, Blondin P, Vigneault C, Labrecque R, Sirard M-A. Sperm miRNAs- potential mediators of bull age and early embryo development. BMC Genomics. 2020a;21(1):798. http://dx.doi.org/10.1186/s12864-020-07206-5. PMid:33198638.
http://dx.doi.org/10.1186/s12864-020-072...
). Interestingly, pathway analysis of the resulting blastocysts indicate that metabolic pathways were affected, which warrants further post-natal evaluation. An additional evaluation of the high/low bull fertility model also revealed changes in mRNA abundance and methylation of resultant blastocysts (Kropp et al., 2017Kropp J, Carrillo JA, Namous H, Daniels A, Salih SM, Song J, Khatib H. Male fertility status is associated with DNA methylation signatures in sperm and transcriptomic profiles of bovine preimplantation embryos. BMC Genomics. 2017;18(1):280. http://dx.doi.org/10.1186/s12864-017-3673-y. PMid:28381255.
http://dx.doi.org/10.1186/s12864-017-367...
). Feeding omega-3 fatty acids in the form of fish meal or flax meal enhanced blastocyst rate and altered blastocyst gene expression compared with a saturated fatty acid control (Kalo et al., 2022Kalo D, Reches D, Netta N, Komsky-Elbaz A, Zeron Y, Moallem U, Roth Z. Carryover effects of feeding bulls with an omega-3-enriched-diet: from spermatozoa to developed embryos. PLoS One. 2022;17(3):e0265650. http://dx.doi.org/10.1371/journal.pone.0265650. PMid:35324945.
http://dx.doi.org/10.1371/journal.pone.0...
)

Seminal plasma has been targeted as having the potential to enhance fertility in cattle breeding systems (Bromfield, 2016Bromfield JJ. A role for seminal plasma in modulating pregnancy outcomes in domestic species. Reproduction. 2016;152(6):R223-32. http://dx.doi.org/10.1530/REP-16-0313. PMid:27601714.
http://dx.doi.org/10.1530/REP-16-0313...
). Though not always successful at increasing pregnancy rates, exposing heifers to seminal plasma from vasectomized bulls before transfer of IVF embryos resulted in enhanced embryo growth and altered conceptus gene expression in cattle receiving multiple IVF embryos (Mateo-Otero et al., 2020Mateo-Otero Y, Sánchez JM, Recuero S, Bagés-Arnal S, McDonald M, Kenny DA, Yeste M, Lonergan P, Fernandez-Fuertes B. Effect of exposure to seminal plasma through natural mating in cattle on conceptus length and gene expression. Front Cell Dev Biol. 2020;8:341. http://dx.doi.org/10.3389/fcell.2020.00341. PMid:32478076.
http://dx.doi.org/10.3389/fcell.2020.003...
). Likewise, increased birth weights of calves born from sex-sorted semen was reported (Ortiz et al., 2019Ortiz WG, Rizo JA, Carvalheira LR, Ahmed BMS, Estrada-Cortes E, Harstine BR, Bromfield JJ, Hansen PJ. Effects of intrauterine infusion of seminal plasma at artificial insemination on fertility of lactating Holstein cows. J Dairy Sci. 2019;102(7):6587-94. http://dx.doi.org/10.3168/jds.2019-16251. PMid:31103294.
http://dx.doi.org/10.3168/jds.2019-16251...
). Thus, seminal plasma-specific effects may be present in cattle, and are likely more important in natural mating where seminal plasma is concentrated in the ejaculate rather than in cases of AI where seminal plasma is diluted with semen extender in preparation for cryopreservation, or when adding an aliquot of seminal plasma at the time of AI. Specific components of bovine seminal plasma that elicit programming responses, however, are yet to be determined.

Final considerations

Though human epidemiology and rodent models have established solid proof of paternal programming effects, the body of evidence in livestock species is limited. Potential mechanisms, magnitude, timing, and persistence of effects, and whether and to what extent specific paternal programming effects are present in livestock species all warrant further exploration. Our group is focused on common management scenarios of plane of nutrition and growth rates in bulls and rams and their effects on mechanisms responsible for paternal programming with the goal of understanding offspring outcomes. Once we demonstrate the impacts of common scenarios under direct control of our herd and flock managers, however, the work is likely just beginning as longer-term and interactive situations must be explored.

Though previously reported in rodent models (Aiken and Ozanne, 2014Aiken CE, Ozanne SE. Transgenerational developmental programming. Hum Reprod Update. 2014;20(1):63-75. http://dx.doi.org/10.1093/humupd/dmt043. PMid:24082037.
http://dx.doi.org/10.1093/humupd/dmt043...
), a recent report in sheep highlighted the potential for sire experiences during spermatogenesis to not only impact the F1 generation conceived during the initial breeding, but also to impact successive generations. In their model of feeding rumen protected methionine to prepuberal sheep, Kahtib’s lab revealed that paternal programming effects persisted until at least the F2 generation (Braz et al., 2022Braz CU, Taylor T, Namous H, Townsend J, Crenshaw T, Khatib H. Paternal diet induces transgenerational epigenetic inheritance of DNA methylation signatures and phenotypes in sheep model. PNAS Nexus. 2022;1(2):pgac040. http://dx.doi.org/10.1093/pnasnexus/pgac040. PMid:36713326.
http://dx.doi.org/10.1093/pnasnexus/pgac...
), with over 100 methylated cytosines and an altered scrotal circumference phenotype inherited by the F1 and F2 generations. Given the long generation interval of cattle, paternal programming effects would be affecting herds for years into the future. Another consideration paramount to the programming equation is the potential additive and/or interactive effects of paternal environment and experiences leading up to mating with those of the breeding females leading up to conception and during gestation on subsequent offspring outcomes. Taken together, as new information becomes available, our existing paradigms of the short-term nature of management decisions (e.g. how should we feed or manage our bulls today for outcomes in the current breeding season) may be shifted to include a long-term outlook on responses that could impact the productivity and profitability of our herds for generations to come.

  • #
    U.S. Department of Agriculture is an equal opportunity provider and employer.
  • Financial support: None.
  • How to cite: Dahlen CR, Amat S, Caton JS, Crouse MS, Diniz WJS, Reynolds LP. Paternal effects on fetal programming. Anim Reprod. 2023;20(2):e20230076. https://doi.org/10.1590/1984-3143-AR2023-0076

References

  • Aiken CE, Ozanne SE. Transgenerational developmental programming. Hum Reprod Update. 2014;20(1):63-75. http://dx.doi.org/10.1093/humupd/dmt043 PMid:24082037.
    » http://dx.doi.org/10.1093/humupd/dmt043
  • Aitken RJ. Impact of oxidative stress on male and female germ cells: implications for fertility. Reproduction. 2020;159(4):R189-201. http://dx.doi.org/10.1530/REP-19-0452 PMid:31846434.
    » http://dx.doi.org/10.1530/REP-19-0452
  • Alves MBR, Arruda RP, Batissaco L, Garcia-Oliveros LN, Gonzaga VHG, Nogueira VJM, Almeida FDS, Pinto SCC, Andrade GM, Perecin F, Silveira JC, Celeghini ECC. Changes in miRNA levels of sperm and small extracellular vesicles of seminal plasma are associated with transient scrotal heat stress in bulls. Theriogenology. 2021;161:26-40. http://dx.doi.org/10.1016/j.theriogenology.2020.11.015 PMid:33278692.
    » http://dx.doi.org/10.1016/j.theriogenology.2020.11.015
  • Ashapkin V, Suvorov A, Pilsner JR, Krawetz SA, Sergeyev O. Age-associated epigenetic changes in mammalian sperm: implications for offspring health and development. Hum Reprod Update. 2023;29(1):24-44. http://dx.doi.org/10.1093/humupd/dmac033 PMid:36066418.
    » http://dx.doi.org/10.1093/humupd/dmac033
  • Ayad B, Omolaoye TS, Louw N, Ramsunder Y, Skosana BT, Oyeipo PI, Du Plessis SS. Oxidative stress and male infertility: evidence from a research perspective. Front Reprod Health. 2022;4:822257. http://dx.doi.org/10.3389/frph.2022.822257 PMid:36303652.
    » http://dx.doi.org/10.3389/frph.2022.822257
  • Bailey JL, Dalvai M, Lessard M, Herst PM, Charest PL, Navarro P. Beyond fertilisation: how the paternal environment influences future generations. Anim Reprod Sci. 2020;220:106503. http://dx.doi.org/10.1016/j.anireprosci.2020.106503 PMid:32536524.
    » http://dx.doi.org/10.1016/j.anireprosci.2020.106503
  • Barker DJ, Clark PM. Fetal undernutrition and disease in later life. Rev Reprod. 1997;2(2):105-12. http://dx.doi.org/10.1530/ror.0.0020105 PMid:9414472.
    » http://dx.doi.org/10.1530/ror.0.0020105
  • Billah MM, Khatiwada S, Morris MJ, Maloney CA. Effects of paternal overnutrition and interventions on future generations. Int J Obes. 2022;46(5):901-17. http://dx.doi.org/10.1038/s41366-021-01042-7 PMid:35022547.
    » http://dx.doi.org/10.1038/s41366-021-01042-7
  • Braunschweig M, Jagannathan V, Gutzwiller A, Bee G. Investigations on transgenerational epigenetic response down the male line in F2 pigs. PLoS One. 2012;7(2):e30583. http://dx.doi.org/10.1371/journal.pone.0030583 PMid:22359544.
    » http://dx.doi.org/10.1371/journal.pone.0030583
  • Braz CU, Taylor T, Namous H, Townsend J, Crenshaw T, Khatib H. Paternal diet induces transgenerational epigenetic inheritance of DNA methylation signatures and phenotypes in sheep model. PNAS Nexus. 2022;1(2):pgac040. http://dx.doi.org/10.1093/pnasnexus/pgac040 PMid:36713326.
    » http://dx.doi.org/10.1093/pnasnexus/pgac040
  • Bromfield JJ. A role for seminal plasma in modulating pregnancy outcomes in domestic species. Reproduction. 2016;152(6):R223-32. http://dx.doi.org/10.1530/REP-16-0313 PMid:27601714.
    » http://dx.doi.org/10.1530/REP-16-0313
  • Champroux A, Cocquet J, Henry-Berger J, Drevet JR, Kocer A. A decade of exploring the mammalian sperm epigenome: paternal epigenetic and transgenerational inheritance. Front Cell Dev Biol. 2018;6:50. http://dx.doi.org/10.3389/fcell.2018.00050 PMid:29868581.
    » http://dx.doi.org/10.3389/fcell.2018.00050
  • Chan JC, Morgan CP, Leu NA, Shetty A, Cisse YM, Nugent BM, Morrison KE, Jašarević E, Huang W, Kanyuch N, Rodgers AB, Bhanu NV, Berger DS, Garcia BA, Ament S, Kane M, Epperson CN, Bale TL. Reproductive tract extracellular vesicles are sufficient to transmit intergenerational stress and program neurodevelopment. Nat Commun. 2020;11(1):1499. http://dx.doi.org/10.1038/s41467-020-15305-w PMid:32198406.
    » http://dx.doi.org/10.1038/s41467-020-15305-w
  • Chukrallah LG, Badrinath A, Seltzer K, Snyder EM. Of rodents and ruminants: a comparison of small noncoding RNA requirements in mouse and bovine reproduction. J Anim Sci. 2021;99(3):skaa388. http://dx.doi.org/10.1093/jas/skaa388 PMid:33677580.
    » http://dx.doi.org/10.1093/jas/skaa388
  • 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. http://dx.doi.org/10.1016/j.jnutbio.2020.108373 PMid:32422425.
    » http://dx.doi.org/10.1016/j.jnutbio.2020.108373
  • Costes V, Chaulot-Talmon A, Sellem E, Perrier JP, Aubert-Frambourg A, Jouneau L, Pontlevoy C, Hozé C, Fritz S, Boussaha M, Le Danvic C, Sanchez MP, Boichard D, Schibler L, Jammes H, Jaffrézic F, Kiefer H. Predicting male fertility from the sperm methylome: application to 120 bulls with hundreds of artificial insemination records. Clin Epigenetics. 2022;14(1):54. http://dx.doi.org/10.1186/s13148-022-01275-x PMid:35477426.
    » http://dx.doi.org/10.1186/s13148-022-01275-x
  • Dahlen CR, Borowicz PP, Ward AK, Caton JS, Czernik M, Palazzese L, Loi P, Reynolds LP. Programming of embryonic development. Int J Mol Sci. 2021;22(21):11668. http://dx.doi.org/10.3390/ijms222111668 PMid:34769097.
    » http://dx.doi.org/10.3390/ijms222111668
  • Duffy KA, Bale TL, Epperson CN. Germ cell drivers: transmission of preconception stress across generations. Front Hum Neurosci. 2021;15:642762. http://dx.doi.org/10.3389/fnhum.2021.642762 PMid:34322003.
    » http://dx.doi.org/10.3389/fnhum.2021.642762
  • Fair T. The contribution of the maternal immune system to the establishment of pregnancy in cattle. Front Immunol. 2015;6:7. http://dx.doi.org/10.3389/fimmu.2015.00007 PMid:25674085.
    » http://dx.doi.org/10.3389/fimmu.2015.00007
  • Gapp K, Parada GE, Gross F, Corcoba A, Kaur J, Grau E, Hemberg M, Bohacek J, Miska EA. Single paternal dexamethasone challenge programs offspring metabolism and reveals multiple candidates in RNA-mediated inheritance. iScience. 2021;24(8):102870. http://dx.doi.org/10.1016/j.isci.2021.102870 PMid:34386731.
    » http://dx.doi.org/10.1016/j.isci.2021.102870
  • Garcia-Oliveros LN, Arruda RP, Batissaco L, Gonzaga VHG, Nogueira VJM, Florez-Rodriguez SA, Almeida FDS, Alves MBR, Pinto SCC, Nichi M, Losano JDA, Kawai GKV, Celeghini ECC. Chronological characterization of sperm morpho-functional damage and recovery after testicular heat stress in Nellore bulls. J Therm Biol. 2022;106:103237. http://dx.doi.org/10.1016/j.jtherbio.2022.103237 PMid:35636895.
    » http://dx.doi.org/10.1016/j.jtherbio.2022.103237
  • Godfrey KM, Barker DJ. Fetal programming and adult health. Public Health Nutr. 2001;4(2B):611-24. http://dx.doi.org/10.1079/PHN2001145 PMid:11683554.
    » http://dx.doi.org/10.1079/PHN2001145
  • Gross N, Taylor T, Crenshaw T, Khatib H. The intergenerational impacts of paternal diet on DNA methylation and offspring phenotypes in sheep. Front Genet. 2020;11:597943. http://dx.doi.org/10.3389/fgene.2020.597943 PMid:33250925.
    » http://dx.doi.org/10.3389/fgene.2020.597943
  • Guan Y, Liang G, Hawken PAR, Malecki IA, Cozens G, Vercoe PE, Martin GB, Guan LL. Roles of small RNAs in the effects of nutrition on apoptosis and spermatogenesis in the adult testis. Sci Rep. 2015;5(1):10372. http://dx.doi.org/10.1038/srep10372 PMid:25996545.
    » http://dx.doi.org/10.1038/srep10372
  • Guan Y, Liang G, Martin GB, Guan LL. Functional changes in mRNA expression and alternative pre-mRNA splicing associated with the effects of nutrition on apoptosis and spermatogenesis in the adult testis. BMC Genomics. 2017;18(1):64. http://dx.doi.org/10.1186/s12864-016-3385-8 PMid:28068922.
    » http://dx.doi.org/10.1186/s12864-016-3385-8
  • Guan Y, Malecki IA, Hawken PAR, Linden MD, Martin GB. Under-nutrition reduces spermatogenic efficiency and sperm velocity, and increases sperm DNA damage in sexually mature male sheep. Anim Reprod Sci. 2014;149(3-4):163-72. http://dx.doi.org/10.1016/j.anireprosci.2014.07.014 PMid:25086661.
    » http://dx.doi.org/10.1016/j.anireprosci.2014.07.014
  • Hammer CJ, Caton JS, Dahlen CR, Ward AK, Borowicz PP, Reynolds LP. DOHaD: a menagerie of adaptations and perspectives: large animal models of developmental programming: sustenance, stress, and sex matter. Reproduction. 2023;165(6):F1-13. http://dx.doi.org/10.1530/REP-22-0453 PMid:36951791.
    » http://dx.doi.org/10.1530/REP-22-0453
  • Harrison TD, Chaney EM, Brandt KJ, Ault-Seay TB, Payton RR, Schneider LG, Strickland LG, Schrick FN, McLean KJ. The effects of nutritional level and body condition score on cytokines in seminal plasma of beef bulls. Front Anim Sci. 2023;3:1078960. http://dx.doi.org/10.3389/fanim.2022.1078960
    » http://dx.doi.org/10.3389/fanim.2022.1078960
  • Hur SS, Cropley JE, Suter CM. Paternal epigenetic programming: evolving metabolic disease risk. J Mol Endocrinol. 2017;58(3):R159-68. http://dx.doi.org/10.1530/JME-16-0236 PMid:28100703.
    » http://dx.doi.org/10.1530/JME-16-0236
  • Ibeagha-Awemu EM, Khatib H. Epigenetics of livestock health, production, and breeding. In: Tollefsbol TO, editor. Handbook of epigenetics. 3rd ed. Amsterdam: Academic Press; 2023. p. 569-610. http://dx.doi.org/10.1016/B978-0-323-91909-8.00041-4
    » http://dx.doi.org/10.1016/B978-0-323-91909-8.00041-4
  • Johnson C, Kiefer H, Chaulot-Talmon A, Dance A, Sellem E, Jouneau L, Jammes H, Kastelic J, Thundathil J. Prepubertal nutritional modulation in the bull and its impact on sperm DNA methylation. Cell Tissue Res. 2022;389(3):587-601. http://dx.doi.org/10.1007/s00441-022-03659-0 PMid:35779136.
    » http://dx.doi.org/10.1007/s00441-022-03659-0
  • Kalo D, Reches D, Netta N, Komsky-Elbaz A, Zeron Y, Moallem U, Roth Z. Carryover effects of feeding bulls with an omega-3-enriched-diet: from spermatozoa to developed embryos. PLoS One. 2022;17(3):e0265650. http://dx.doi.org/10.1371/journal.pone.0265650 PMid:35324945.
    » http://dx.doi.org/10.1371/journal.pone.0265650
  • Kiefer H, Sellem E, Bonnet-Garnier A, Pannetier M, Costes V, Schibler L, Jammes H. The epigenome of male germ cells and the programming of phenotypes in cattle. Anim Front. 2021;11(6):28-38. http://dx.doi.org/10.1093/af/vfab062 PMid:34934527.
    » http://dx.doi.org/10.1093/af/vfab062
  • Koziol JH, Sheets T, Wickware CL, Johnson TA. Composition and diversity of the seminal microbiota in bulls and its association with semen parameters. Theriogenology. 2022;182:17-25. http://dx.doi.org/10.1016/j.theriogenology.2022.01.029 PMid:35123307.
    » http://dx.doi.org/10.1016/j.theriogenology.2022.01.029
  • Kretschmer M, Gapp K. Deciphering the RNA universe in sperm in its role as a vertical information carrier. Environ Epigenet. 2022;8(1):dvac011. http://dx.doi.org/10.1093/eep/dvac011 PMid:35633894.
    » http://dx.doi.org/10.1093/eep/dvac011
  • Kropp J, Carrillo JA, Namous H, Daniels A, Salih SM, Song J, Khatib H. Male fertility status is associated with DNA methylation signatures in sperm and transcriptomic profiles of bovine preimplantation embryos. BMC Genomics. 2017;18(1):280. http://dx.doi.org/10.1186/s12864-017-3673-y PMid:28381255.
    » http://dx.doi.org/10.1186/s12864-017-3673-y
  • Kusuyama J, Alves-Wagner AB, Makarewicz NS, Goodyear LJ. Effects of maternal and paternal exercise on offspring metabolism. Nat Metab. 2020;2(9):858-72. http://dx.doi.org/10.1038/s42255-020-00274-7 PMid:32929233.
    » http://dx.doi.org/10.1038/s42255-020-00274-7
  • Kutchy NA, Menezes ESB, Chiappetta A, Tan W, Wills RW, Kaya A, Topper E, Moura AA, Perkins AD, Memili E. Acetylation and methylation of sperm histone 3 lysine 27 (H3K27ac and H3K27me3) are associated with bull fertility. Andrologia. 2018;50(3):e12915. http://dx.doi.org/10.1111/and.12915 PMid:29057498.
    » http://dx.doi.org/10.1111/and.12915
  • Lambert S, Blondin P, Vigneault C, Labrecque R, Dufort I, Sirard M-A. Spermatozoa DNA methylation patterns differ due to peripubertal age in bulls. Theriogenology. 2018;106:21-9. http://dx.doi.org/10.1016/j.theriogenology.2017.10.006 PMid:29031946.
    » http://dx.doi.org/10.1016/j.theriogenology.2017.10.006
  • Lambrot R, Xu C, Saint-Phar S, Chountalos G, Cohen T, Paquet M, Suderman M, Hallett M, Kimmins S. Low paternal dietary folate alters the mouse sperm epigenome and is associated with negative pregnancy outcomes. Nat Commun. 2013;4(1):2889. http://dx.doi.org/10.1038/ncomms3889 PMid:24326934.
    » http://dx.doi.org/10.1038/ncomms3889
  • Lee HJ, Ryu J-S, Choi NY, Park YS, Kim YI, Han DW, Ko K, Shin CY, Hwang HS, Kang K-S, Ko K. Transgenerational effects of paternal alcohol exposure in mouse offspring. Anim Cells Syst. 2013;17(6):429-34. http://dx.doi.org/10.1080/19768354.2013.865675
    » http://dx.doi.org/10.1080/19768354.2013.865675
  • Lima AO, Afonso J, Edson J, Marcellin E, Palfreyman R, Porto-Neto LR, Reverter A, Fortes MRS. Network analyses predict small RNAs that might modulate gene expression in the testis and epididymis of Bos indicus bulls. Front Genet. 2021;12:610116. http://dx.doi.org/10.3389/fgene.2021.610116 PMid:33995471.
    » http://dx.doi.org/10.3389/fgene.2021.610116
  • Lismer A, Kimmins S. Emerging evidence that the mammalian sperm epigenome serves as a template for embryo development. Nat Commun. 2023;14(1):2142. http://dx.doi.org/10.1038/s41467-023-37820-2 PMid:37059740.
    » http://dx.doi.org/10.1038/s41467-023-37820-2
  • Liu J, Shi J, Hernandez R, Li X, Konchadi P, Miyake Y, Chen Q, Zhou T, Zhou C. Paternal phthalate exposure-elicited offspring metabolic disorders are associated with altered sperm small RNAs in mice. Environ Int. 2023;172:107769. http://dx.doi.org/10.1016/j.envint.2023.107769 PMid:36709676.
    » http://dx.doi.org/10.1016/j.envint.2023.107769
  • Luecke SM, Webb EM, Dahlen CR, Reynolds LP, Amat S. Seminal and vagino-uterine microbiome and their individual and interactive effects on cattle fertility. Front Microbiol. 2022;13:1029128. http://dx.doi.org/10.3389/fmicb.2022.1029128 PMid:36425035.
    » http://dx.doi.org/10.3389/fmicb.2022.1029128
  • Marcho C, Oluwayiose OA, Pilsner JR. The preconception environment and sperm epigenetics. Andrology. 2020;8(4):924-42. http://dx.doi.org/10.1111/andr.12753 PMid:31901222.
    » http://dx.doi.org/10.1111/andr.12753
  • Marey MA, Ezz MA, Akthar I, Yousef MS, Imakawa K, Shimada M, Miyamoto A. Sensing sperm via maternal immune system: a potential mechanism for controlling microenvironment for fertility in the cow. J Anim Sci. 2020;98(Suppl 1):S88-95. http://dx.doi.org/10.1093/jas/skaa147 PMid:32810249.
    » http://dx.doi.org/10.1093/jas/skaa147
  • Marey MA, Ma D, Yoshino H, Elesh IF, Zinnah MA, Fiorenza MF, Moriyasu S, Miyamoto A. Sperm induce proinflammatory responses in the uterus and peripheral blood immune cells of artificially inseminated cows. J Reprod Dev. 2023;69(2):95-102. http://dx.doi.org/10.1262/jrd.2022-124 PMid:36775285.
    » http://dx.doi.org/10.1262/jrd.2022-124
  • Mateo-Otero Y, Sánchez JM, Recuero S, Bagés-Arnal S, McDonald M, Kenny DA, Yeste M, Lonergan P, Fernandez-Fuertes B. Effect of exposure to seminal plasma through natural mating in cattle on conceptus length and gene expression. Front Cell Dev Biol. 2020;8:341. http://dx.doi.org/10.3389/fcell.2020.00341 PMid:32478076.
    » http://dx.doi.org/10.3389/fcell.2020.00341
  • Moreira BP, Oliveira PF, Alves MG. Molecular mechanisms controlled by mTOR in male reproductive system. Int J Mol Sci. 2019;20(7):1633. http://dx.doi.org/10.3390/ijms20071633 PMid:30986927.
    » http://dx.doi.org/10.3390/ijms20071633
  • Morgan HL, Paganopoulou P, Akhtar S, Urquhart N, Philomin R, Dickinson Y, Watkins AJ. Paternal diet impairs F1 and F2 offspring vascular function through sperm and seminal plasma specific mechanisms in mice. J Physiol. 2020;598(4):699-715. http://dx.doi.org/10.1113/JP278270 PMid:31617219.
    » http://dx.doi.org/10.1113/JP278270
  • Morgan HL, Watkins AJ. The influence of seminal plasma on offspring development and health. Semin Cell Dev Biol. 2020;97:131-7. http://dx.doi.org/10.1016/j.semcdb.2019.06.008 PMid:31254609.
    » http://dx.doi.org/10.1016/j.semcdb.2019.06.008
  • Moura FH, Macias-Franco A, Pena-Bello CA, Archilia EC, Batalha IM, Silva AE, Moreira GM, Norris AB, Schütz LF, Fonseca MA. Sperm DNA 5-methyl cytosine and RNA N 6-methyladenosine methylation are differently affected during periods of body weight losses and body weight gain of young and mature breeding bulls. J Anim Sci. 2022;100(2):skab362. http://dx.doi.org/10.1093/jas/skab362 PMid:34902028.
    » http://dx.doi.org/10.1093/jas/skab362
  • Nongbua T, Guo Y, Ntallaris T, Rubér M, Rodriguez-Martinez H, Humblot P, Morrell JM. Bull seminal plasma stimulates in vitro production of TGF-β, IL-6 and IL-8 from bovine endometrial epithelial cells, depending on dose and bull fertility. J Reprod Immunol. 2020;142:103179. http://dx.doi.org/10.1016/j.jri.2020.103179 PMid:32717675.
    » http://dx.doi.org/10.1016/j.jri.2020.103179
  • Ortiz WG, Rizo JA, Carvalheira LR, Ahmed BMS, Estrada-Cortes E, Harstine BR, Bromfield JJ, Hansen PJ. Effects of intrauterine infusion of seminal plasma at artificial insemination on fertility of lactating Holstein cows. J Dairy Sci. 2019;102(7):6587-94. http://dx.doi.org/10.3168/jds.2019-16251 PMid:31103294.
    » http://dx.doi.org/10.3168/jds.2019-16251
  • Perrier JP, Kenny DA, Chaulot-Talmon A, Byrne CJ, Sellem E, Jouneau L, Aubert-Frambourg A, Schibler L, Jammes H, Lonergan P, Fair S, Kiefer H. Accelerating onset of puberty through modification of early life nutrition induces modest but persistent changes in bull sperm DNA methylation profiles post-puberty. Front Genet. 2020;11:945. http://dx.doi.org/10.3389/fgene.2020.00945 PMid:33005172.
    » http://dx.doi.org/10.3389/fgene.2020.00945
  • Phillips N, Taylor L, Bachmann G. Maternal, infant and childhood risks associated with advanced paternal age: the need for comprehensive counseling for men. Maturitas. 2019;125:81-4. http://dx.doi.org/10.1016/j.maturitas.2019.03.020 PMid:31133222.
    » http://dx.doi.org/10.1016/j.maturitas.2019.03.020
  • Radford EJ, Ito M, Shi H, Corish JA, Yamazawa K, Isganaitis E, Seisenberger S, Hore TA, Reik W, Erkek S, Peters AHFM, Patti ME, Ferguson-Smith AC. In utero undernourishment perturbs the adult sperm methylome and intergenerational metabolism. Science. 2014;345(6198):1255903. http://dx.doi.org/10.1126/science.1255903 PMid:25011554.
    » http://dx.doi.org/10.1126/science.1255903
  • Rahman MB, Schellander K, Luceño NL, Van Soom A. Heat stress responses in spermatozoa: mechanisms and consequences for cattle fertility. Theriogenology. 2018;113:102-12. http://dx.doi.org/10.1016/j.theriogenology.2018.02.012 PMid:29477908.
    » http://dx.doi.org/10.1016/j.theriogenology.2018.02.012
  • Rahman MB, Vandaele L, Rijsselaere T, Maes D, Hoogewijs M, Frijters A, Noordman J, Granados A, Dernelle E, Shamsuddin M, Parrish JJ, Van Soom A. Scrotal insulation and its relationship to abnormal morphology, chromatin protamination and nuclear shape of spermatozoa in Holstein-Friesian and Belgian Blue bulls. Theriogenology. 2011;76(7):1246-57. http://dx.doi.org/10.1016/j.theriogenology.2011.05.031 PMid:21777969.
    » http://dx.doi.org/10.1016/j.theriogenology.2011.05.031
  • Reynolds LP, Borowicz PP, Caton JS, Vonnahme KA, Luther JS, Hammer CJ, Carlin KRM, Grazul-Bilska AT, Redmer DA. Developmental programming: the concept, large animal models, and the key role of uteroplacental vascular development. J Anim Sci. 2010;88(Suppl 13):E61-72. http://dx.doi.org/10.2527/jas.2009-2359 PMid:20023136.
    » http://dx.doi.org/10.2527/jas.2009-2359
  • Roca J, Rodriguez-Martinez H, Padilla L, Lucas X, Barranco I. Extracellular vesicles in seminal fluid and effects on male reproduction. An overview in farm animals and pets. Anim Reprod Sci. 2022;246:106853. http://dx.doi.org/10.1016/j.anireprosci.2021.106853 PMid:34556398.
    » http://dx.doi.org/10.1016/j.anireprosci.2021.106853
  • Roseboom TJ. Epidemiological evidence for the developmental origins of health and disease: effects of prenatal undernutrition in humans. J Endocrinol. 2019;242(1):T135-44. http://dx.doi.org/10.1530/JOE-18-0683 PMid:31207580.
    » http://dx.doi.org/10.1530/JOE-18-0683
  • Rumph JT, Stephens VR, Ameli S, Brown LK, Rayford KJ, Nde PN, Osteen KG, Bruner-Tran KL. A paternal fish oil diet preconception reduces lung inflammation in a toxicant-driven murine model of new bronchopulmonary dysplasia. Mar Drugs. 2023;21(3):161. http://dx.doi.org/10.3390/md21030161 PMid:36976210.
    » http://dx.doi.org/10.3390/md21030161
  • Rumph JT, Stephens VR, Ameli S, Gaines PN, Osteen KG, Bruner-Tran KL, Nde PN. A paternal fish oil diet preconception modulates the gut microbiome and attenuates necrotizing enterocolitis in neonatal mice. Mar Drugs. 2022;20(6):390. http://dx.doi.org/10.3390/md20060390 PMid:35736193.
    » http://dx.doi.org/10.3390/md20060390
  • Sabei L, Bernardino T, Parada Sarmiento M, Barbosa BS, Farias SS, Ghantous GF, Lima CG, Poletto R, Zanella AJ. Life experiences of boars can shape the survival, aggression, and nociception responses of their offspring. Front Anim Sci. 2023;4:1142628. http://dx.doi.org/10.3389/fanim.2023.1142628
    » http://dx.doi.org/10.3389/fanim.2023.1142628
  • Schoenmakers S, Steegers-Theunissen R, Faas M. The matter of the reproductive microbiome. Obstet Med. 2019;12(3):107-15. http://dx.doi.org/10.1177/1753495X18775899 PMid:31523266.
    » http://dx.doi.org/10.1177/1753495X18775899
  • Sellem E, Marthey S, Rau A, Jouneau L, Bonnet A, Le Danvic C, Guyonnet B, Kiefer H, Jammes H, Schibler L. Dynamics of cattle sperm sncRNAs during maturation, from testis to ejaculated sperm. Epigenetics Chromatin. 2021;14(1):24. http://dx.doi.org/10.1186/s13072-021-00397-5 PMid:34030709.
    » http://dx.doi.org/10.1186/s13072-021-00397-5
  • Senger P. Pathways to pregnancy and parturition. 3rd ed. Redmond: Current Conceptions, Inc., 2012.
  • Sharma U. Paternal contributions to offspring health: role of sperm small RNAs in intergenerational transmission of epigenetic information. Front Cell Dev Biol. 2019;7:215. http://dx.doi.org/10.3389/fcell.2019.00215 PMid:31681757.
    » http://dx.doi.org/10.3389/fcell.2019.00215
  • Staub C, Johnson L. Review: spermatogenesis in the bull. Animal. 2018;12(Suppl 1):s27-35. http://dx.doi.org/10.1017/S1751731118000435 PMid:29882505.
    » http://dx.doi.org/10.1017/S1751731118000435
  • Toschi P, Capra E, Anzalone DA, Lazzari B, Turri F, Pizzi F, Scapolo PA, Stella A, Williams JL, Marsan PA, Loi P. Maternal peri-conceptional undernourishment perturbs offspring sperm methylome. Reproduction. 2020;159(5):513-23. http://dx.doi.org/10.1530/REP-19-0549 PMid:32103819.
    » http://dx.doi.org/10.1530/REP-19-0549
  • Toussaint AB, Ellis AS, Bongiovanni AR, Peterson DR, Bavley CC, Karbalaei R, Mayberry HL, Bhakta S, Dressler CC, Imperio CG, Maurer JJ, Schmidt HD, Chen C, Bland K, Liu-Chen LY, Wimmer ME. Paternal morphine exposure enhances morphine self-administration and induces region-specific neural adaptations in reward-related brain regions of male offspring. bioRxiv. In press 2023. PMid:36711571.
  • Wang Y, Chen Z-P, Hu H, Lei J, Zhou Z, Yao B, Chen L, Liang G, Zhan S, Zhu X, Jin F, Ma R, Zhang J, Liang H, Xing M, Chen XR, Zhang CY, Zhu JN, Chen X. Sperm microRNAs confer depression susceptibility to offspring. Sci Adv. 2021;7(7):eabd7605. http://dx.doi.org/10.1126/sciadv.abd7605 PMid:33568480.
    » http://dx.doi.org/10.1126/sciadv.abd7605
  • 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(6):1371-81. http://dx.doi.org/10.1016/j.bbadis.2017.02.009 PMid:28189722.
    » http://dx.doi.org/10.1016/j.bbadis.2017.02.009
  • Webb EM, Holman DB, Schmidt KN, Crouse MS, Dahlen CR, Cushman RA, Snider AP, McCarthy KL, Amat S. A longitudinal characterization of the seminal microbiota and antibiotic resistance in yearling beef bulls subjected to different rates of gain. Microbiol Spectr. 2023;11(2):e0518022. http://dx.doi.org/10.1128/spectrum.05180-22 PMid:36916922.
    » http://dx.doi.org/10.1128/spectrum.05180-22
  • Werry N, Russell SJ, Gillis DJ, Miller S, Hickey K, Larmer S, Lohuis M, Librach C, LaMarre J. Characteristics of miRNAs present in bovine sperm and associations with differences in fertility. Front Endocrinol. 2022;13:874371. http://dx.doi.org/10.3389/fendo.2022.874371 PMid:35663333.
    » http://dx.doi.org/10.3389/fendo.2022.874371
  • Wu C, Blondin P, Vigneault C, Labrecque R, Sirard M-A. Sperm miRNAs- potential mediators of bull age and early embryo development. BMC Genomics. 2020a;21(1):798. http://dx.doi.org/10.1186/s12864-020-07206-5 PMid:33198638.
    » http://dx.doi.org/10.1186/s12864-020-07206-5
  • Wu C, Blondin P, Vigneault C, Labrecque R, Sirard M-A. The age of the bull influences the transcriptome and epigenome of blastocysts produced by IVF. Theriogenology. 2020b;144:122-31. http://dx.doi.org/10.1016/j.theriogenology.2019.12.020 PMid:31951983.
    » http://dx.doi.org/10.1016/j.theriogenology.2019.12.020
  • Wu C, Wang C, Zhai B, Zhao Y, Zhao Z, Yuan Z, Zhang M, Tian K, Fu X. Study of microRNA expression profile in different regions of ram epididymis. Reprod Domest Anim. 2021;56(9):1209-19. http://dx.doi.org/10.1111/rda.13978 PMid:34169586.
    » http://dx.doi.org/10.1111/rda.13978
  • Wu G, Bazer FW, Wallace JM, Spencer TE. Board-invited review: intrauterine growth retardation: implications for the animal sciences. J Anim Sci. 2006;84(9):2316-37. http://dx.doi.org/10.2527/jas.2006-156 PMid:16908634.
    » http://dx.doi.org/10.2527/jas.2006-156
  • Xia W, Mruk DD, Cheng CY. C-type natriuretic peptide regulates blood–testis barrier dynamics in adult rat testes. Proc Natl Acad Sci USA. 2007;104(10):3841-6. http://dx.doi.org/10.1073/pnas.0610100104 PMid:17360440.
    » http://dx.doi.org/10.1073/pnas.0610100104
  • Yockey LJ, Iwasaki A. Interferons and proinflammatory cytokines in pregnancy and fetal development. Immunity. 2018;49(3):397-412. http://dx.doi.org/10.1016/j.immuni.2018.07.017 PMid:30231982.
    » http://dx.doi.org/10.1016/j.immuni.2018.07.017
  • Zhao L, Liu X, Gomez NA, Gao Y, Son JS, Chae SA, Zhu MJ, Du M. Stage-specific nutritional management and developmental programming to optimize meat production. J Anim Sci Biotechnol. 2023;14(1):2. http://dx.doi.org/10.1186/s40104-022-00805-0 PMid:36597116.
    » http://dx.doi.org/10.1186/s40104-022-00805-0

Publication Dates

  • Publication in this collection
    28 Aug 2023
  • Date of issue
    2023

History

  • Received
    26 May 2023
  • Accepted
    18 July 2023
Colégio Brasileiro de Reprodução Animal Coronel José dias Bicalho, 1224, CEP: , 31275-050, Belo Horizonte, MG - Brasil, Tel.: 55-31-3491 7122 - Belo Horizonte - MG - Brazil
E-mail: animreprod.journal@gmail.com