Acessibilidade / Reportar erro

Obese fathers lead to an altered metabolism and obesity in their children in adulthood: review of experimental and human studies Please cite this article as: Ornellas F, Carapeto PV, Mandarim-de-Lacerda CA, Aguila MB. Obese fathers lead to an altered metabolism and obesity in their children in adulthood: review of experimental and human studies. J Pediatr (Rio J). 2017;93:551-9.

Abstract

Objective:

To discuss the recent literature on paternal obesity, focusing on the possible mechanisms of transmission of the phenotypes from the father to the children.

Sources:

A non-systematic review in the PubMed database found few publications in which paternal obesity was implicated in the adverse transmission of characteristics to offspring. Specific articles on epigenetics were also evaluated. As the subject is recent and still controversial, all articles were considered regardless of year of publication.

Summary of findings:

Studies in humans and animals have established that paternal obesity impairs their hormones, metabolism, and sperm function, which can be transmitted to their offspring. In humans, paternal obesity results in insulin resistance/type 2 diabetes and increased levels of cortisol in umbilical cord blood, which increases the risk factors for cardiovascular disease. Notably, there is an association between body fat in parents and the prevalence of obesity in their daughters. In animals, paternal obesity led to offspring alterations on glucose-insulin homeostasis, hepatic lipogenesis, hypothalamus/feeding behavior, kidney of the offspring; it also impairs the reproductive potential of male offspring with sperm oxidative stress and mitochondrial dysfunction. An explanation for these observations (human and animal) is epigenetics, considered the primary tool for the transmission of phenotypes from the father to offspring, such as DNA methylation, histone modifications, and non-coding RNA.

Conclusions:

Paternal obesity can induce programmed phenotypes in offspring through epigenetics. Therefore, it can be considered a public health problem, affecting the children's future life.

KEYWORDS
Paternal obesity; Programming; Obese child; Chronic diseases programming; Epigenetics

Resumo

Objetivo:

Discutir a literatura recente sobre obesidade paterna, focalizando os possíveis mecanismos de transmissão dos fenótipos do pai para os filhos.

Fontes:

Uma revisão não-sistemática no banco de dados PubMed encontrou poucas publicações com obesidade paterna implicada com a transmissão adversa das características à prole. Artigos específicos sobre epigenética também foram avaliados. Como o assunto é recente e ainda controverso, todos os trabalhos foram considerados independentemente do ano de publicação.

Resumo dos achados:

Estudos em seres humanos e animais estabeleceram que a obesidade do pai prejudica seus hormônios, metabolismo e função espermática, que pode ser transmitida à prole. Em humanos, a obesidade paterna resulta em resistência à insulina / diabetes tipo 2 e aumento do nível de cortisol no sangue do cordão umbilical, que aumenta os fatores de risco para doença cardiovascular. Notavelmente, existe associação entre a gordura corporal nos pais e a prevalência de obesidade em suas filhas. Em animais, pais obesos condicionam, na prole, a homeostase glicose-insulina, lipogênese hepática, hipotálamo / comportamento alimentar, rim, prejudicam o potencial reprodutivo da prole masculina com estresse oxidativo espermático e disfunção mitocondrial. Uma explicação para estas observações (humanos e animais) é a epigenética, considerada a ferramenta básica para a transmissão de fenótipos do pai à prole, como a metilação do DNA, modificações nas histonas, e RNA não codificante.

Conclusões:

A obesidade paterna pode induzir fenótipos programados na prole através da epigenética. Portanto, a obesidade paterna pode ser considerada um problema de saúde pública, afetando a vida futura das crianças.

PALAVRAS-CHAVE
Obesidade paterna; Programação; Criança obesa; Programação de doenças crônicas; Epigenética

Introduction

Obesity has been growing in a disorderly way, constituting a real epidemic described as "globesity," which represents a serious public health problem nowadays.11 Huang H, Yan Z, Chen Y, Liu F. A social contagious model of the obesity epidemic. Sci Rep. 2016;6:37961.

It is now known that the risk of developing obesity and metabolic syndrome (MS) in adulthood may be influenced by the initial period of life, especially through inadequate nutrition available to the fetus and newborn.22 Barker DJ. The fetal and infant origins of adult disease. BMJ. 1990;301:1111.,33 Barker DJ. The developmental origins of adult disease. J Am Coll Nutr. 2004;23:588S-95S. "Programming" is the process by which early life factors may influence the offspring's health in adulthood. Programming is considered an essential mechanism for the establishment of obesity and metabolic changes in the offspring.44 Gusmao-Correia ML, Volpato AM, Aguila MB, Mandarim-de-Lacerda CA. Developmental origins of health and disease: experimental and human evidence of fetal programming for metabolic syndrome. J Hum Hypertens. 2012;26:405-19.,55 Volpato AM, Schultz A, Magalhaes-da-Costa E, Correia ML, Aguila MB, Mandarim-de-Lacerda CA. Maternal high-fat diet programs for metabolic disturbances in offspring despite leptin sensitivity. Neuroendocrinology. 2012;96:272-84. Various models are used to understand the mechanisms associated with programming, in which the hormonal and metabolic environment during the prenatal or postnatal period is altered through changes in maternal nutritional status.66 Ornellas F, Souza-Mello V, Mandarim-de-Lacerda CA, Aguila MB. Combined parental obesity augments single-parent obesity effects on hypothalamus inflammation, leptin signaling (JAK/STAT), hyperphagia, and obesity in the adult mice offspring. Physiol Behav. 2016;153:47-55.

7 Bringhenti I, Ornellas F, Mandarim-de-Lacerda CA, Aguila MB. The insulin-signaling pathway of the pancreatic islet is impaired in adult mice offspring of mothers fed a high-fat diet. Nutrition. 2016;32:1138-43.
-88 Ornellas F, Mello VS, Mandarim-de-Lacerda CA, Aguila MB. Sexual dimorphism in fat distribution and metabolic profile in mice offspring from diet-induced obese mothers. Life Sci. 2013;93:454-63.

Most epidemiological and experimental studies have focused on the maternal influence on offspring's health. However, recent experiments with rodents have demonstrated that the paternal involvement affects glucose homeostasis and the lifetime of pancreatic islets in female offspring.99 Ng SF, Lin RC, Laybutt DR, Barres R, Owens JA, Morris MJ. Chronic high-fat diet in fathers programs beta-cell dysfunction in female rat offspring. Nature. 2010;467:963-6. Clinical and animal testing have challenged conventional ideas about metabolic programming, suggesting that something else might act in this process via paternal programming. Recent studies now indicate that paternal metabolic health at conception can also impact children's health, and that obese fathers are more likely to generate an obese child.1010 Li L, Law C, Lo Conte R, Power C. Intergenerational influences on childhood body mass index: the effect of parental body mass index trajectories. Am J Clin Nutr. 2009;89:551-7.

This review reports the recent findings and proposed mechanisms involved in paternal programming of the offspring.

Human studies

Studies in humans analyzed the relationship between paternal lifestyle-related factors, environmental exposure factors, and offspring's health outcome in early and later life, suggesting that paternal effects may play a significant role in the pathogenesis of offspring chronic diseases in later life (e.g., insulin resistance and type 2 diabetes). More than 60% of all adults are classified as overweight or obese in most Westernized societies; as the prevalence of obesity increases, it is responsible for an ever-larger proportion of the overall burden of disease.99 Ng SF, Lin RC, Laybutt DR, Barres R, Owens JA, Morris MJ. Chronic high-fat diet in fathers programs beta-cell dysfunction in female rat offspring. Nature. 2010;467:963-6.,1111 Nguyen DM, El-Serag HB. The epidemiology of obesity. Gastroenterol Clin North Am. 2010;39:1-7.

There is clear evidence that paternal nutritional factors play a significant role in the health of offspring. For example, there is a correlation between paternal absolute and relative amounts of body fat and the same parameters in their daughters aged 4.8-8.9 years.1212 Chen YP, Xiao XM, Li J, Reichetzeder C, Wang ZN, Hocher B. Paternal body mass index (BMI) is associated with offspring intrauterine growth in a gender-dependent manner. PLoS ONE. 2012;7:e36329. Furthermore, paternal body mass index (BMI) might modulate the offspring phenotype in a sex-dependent manner. In the frames of a family cohort study (899 parent-offspring trios), paternal BMI was correlated with birth weight, biparietal diameter, head circumference, abdominal diameter, abdominal circumference, and thoracic diameter in male newborns only.1212 Chen YP, Xiao XM, Li J, Reichetzeder C, Wang ZN, Hocher B. Paternal body mass index (BMI) is associated with offspring intrauterine growth in a gender-dependent manner. PLoS ONE. 2012;7:e36329. Fathers1313 Kaati G, Bygren LO, Pembrey M, Sjostrom M. Transgenerational response to nutrition, early life circumstances and longevity. Eur J Hum Genet. 2007;15:784-90. or grandfathers1414 Bygren LO, Kaati G, Edvinsson S. Longevity determined by paternal ancestors' nutrition during their slow growth period. Acta Biotheor. 2001;49:53-9. exposed to either overfeeding or food restriction on the age period of 9-12 years predetermined reduced longevity of their male offspring. The second generation of offspring of these grandfathers had a four-fold risk of diabetes mortality.1515 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;10:682-8.

In Northern Sweden, the follow-up of three generations demonstrated that a grandfathers' surfeit of food is associated with reduced survivability1414 Bygren LO, Kaati G, Edvinsson S. Longevity determined by paternal ancestors' nutrition during their slow growth period. Acta Biotheor. 2001;49:53-9. and an increased risk of diabetes1515 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;10:682-8. in their grandchildren. Early onset of grandpaternal smoking is also related to increased grandson BMI.1616 Pembrey ME, Bygren LO, Kaati G, Edvinsson S, Northstone K, Sjostrom M, et al. Sex-specific, male-line transgenerational responses in humans. Eur J Hum Genet. 2006;14:159-66. This evidence indicates that paternal nutritional factors, not only before conception but also as early as in father's puberty, might affect the offspring in a sex-dependent manner.1717 Li J, Tsuprykov O, Yang X, Hocher B. Paternal programming of offspring cardiometabolic diseases in later life. J Hypertens. 2016;34:2111-26. Moreover, there is an interaction between parental genes and parental environmental factors that affect the phenotype of the offspring.1818 Hocher B. More than genes: the advanced fetal programming hypothesis. J Reprod Immunol. 2014;104-105:8-11. The gene-environment interaction becomes even more complicated, as it is also known that the socioeconomic status of an individual appears to have opposite effects on obesity in poor and rich countries.1919 Pampel FC, Denney JT, Obesity Krueger PM. SES, and economic development: a test of the reversal hypothesis. Soc Sci Med. 2012;74:1073-81.

In fathers, the caloric imbalance imposed by lifestyle choices, including high food consumption and low physical activity, are factors to be considered in programming studies. Epigenetic modifications can occur within the lifespan of numerous individuals within a population, and thus be transmitted immediately to a large number of offspring in the next generation, unlike genomic events that spread slowly through a population.2020 Slyvka Y, Zhang Y, Nowak FV. Epigenetic effects of paternal diet on offspring: emphasis on obesity. Endocrine. 2015;48:36-46. It is likely that changing circumstances within the individual or over several generations can recruit silent alleles back into the active genome and contribute to the reversibility of adaptive or acquired changes. A recent study in obese men showed changes in circulating microRNA (miRNA) that target VEGF (vascular endothelial growth factor), an adipocyte mitogen, which was reversible following weight loss.2121 Ortega FJ, Mercader JM, Catalan V, Moreno-Navarrete JM, Pueyo N, Sabater M, et al. Targeting the circulating microRNA signature of obesity. Clin Chem. 2013;59:781-92.

Paternal BMI during conception was associated with fetal development of the male offspring, but not of the female offspring. It was significantly correlated with birth weight and perinatal biparietal diameter, head circumference, abdominal diameter, abdominal circumference, and thoracic diameter measured in male offspring. There were no significant correlations between paternal BMI and these parameters in female offspring. Cord blood cortisol level was also associated with father's BMI in male offspring only. The authors concluded that a sex-specific transgenerational effect of paternal BMI on fetal cortisol secretion might represent a risk factor for cardiovascular disease in male offspring in later life.1212 Chen YP, Xiao XM, Li J, Reichetzeder C, Wang ZN, Hocher B. Paternal body mass index (BMI) is associated with offspring intrauterine growth in a gender-dependent manner. PLoS ONE. 2012;7:e36329. Furthermore, increased paternal BMI is associated with decreased blastocyst development and live birth rates after in vitro fertilization.2222 Bakos HW, Henshaw RC, Mitchell M, Lane M. Paternal body mass index is associated with decreased blastocyst development and reduced live birth rates following assisted reproductive technology. Fertil Steril. 2011;95:1700-4. In obese fathers (BMI > 25 kg/m2), a higher reactive oxygen species was detected in sperm, as well as increased seminal fluid neopterin (a marker of reproductive tract macrophage activation), decreased sperm counts and serum testosterone, and increased serum estradiol.2323 Tunc O, Bakos HW, Tremellen K. Impact of body mass index on seminal oxidative stress. Andrologia. 2011;43:121-8.

Animal studies

Animal models of male obesity are being used to assess the impact of paternal programming on offspring and to analyze the sperm function of the obese father. Animal models are important due to the difficulties in separating the effects of paternal genetic makeup from those of paternal environmental exposures on the offspring,1010 Li L, Law C, Lo Conte R, Power C. Intergenerational influences on childhood body mass index: the effect of parental body mass index trajectories. Am J Clin Nutr. 2009;89:551-7. as well as in clustering and interpreting data from human studies. A better understanding of the mechanisms of paternal programming can help in interventions to minimize adverse effects on the offspring.2424 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:E805-21.

A paternal programming (when obese fathers led to disturbance of glucose-insulin homeostasis in the female offspring) was initially described in animals.99 Ng SF, Lin RC, Laybutt DR, Barres R, Owens JA, Morris MJ. Chronic high-fat diet in fathers programs beta-cell dysfunction in female rat offspring. Nature. 2010;467:963-6. Obese fathers also program liver lipogenesis and beta-oxidation2525 Ornellas F, Souza-Mello V, Mandarim-de-Lacerda CA, Aguila MB. Programming of obesity and comorbidities in the progeny: lessons from a model of diet-induced obese parents. PLOS ONE. 2015;10:e0124737. and the hypothalamus of the offspring (hypothalamus inflammation was found in offspring, with an increase of interleukin [IL]-6 and tumor necrosis factor [TNF]-alpha expressions).66 Ornellas F, Souza-Mello V, Mandarim-de-Lacerda CA, Aguila MB. Combined parental obesity augments single-parent obesity effects on hypothalamus inflammation, leptin signaling (JAK/STAT), hyperphagia, and obesity in the adult mice offspring. Physiol Behav. 2016;153:47-55.

Obese fathers also altered the offspring's kidney, with tubular damage and loss of the tubular brush border, but not glomerular damage. The cholesterol acyltransferase-1 (Acat1) gene, involved in the input of fatty acid for beta-oxidation in the tubuli, was up-regulated in the offspring.2626 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.

Mammalian male germ cell development is susceptible to damage in different times and in specific diseases in offspring's later life: embryonic development, infancy, and prepubertal age, and preconception and spermatogenesis in adulthood.2727 Schagdarsurengin U, Steger K. Epigenetics in male reproduction: effect of paternal diet on sperm quality and offspring health. Nat Rev Urol. 2016;13:584-95.

28 Soubry A, Hoyo C, Jirtle RL, Murphy SK. A paternal environmental legacy: evidence for epigenetic inheritance through the male germ line. Bioessays. 2014;36:359-71.
-2929 Wu H, Hauser R, Krawetz SA, Pilsner JR. Environmental susceptibility of the sperm epigenome during windows of male germ cell development. Curr Environ Health Rep. 2015;2:356-66. Paternal obesity negatively impacts on the reproductive potential of the male offspring not only by altering function, quality, and molecular composition of sperm, but also by increasing sperm's oxidative stress, contributing to DNA damage and mitochondrial dysfunction.3030 Palmer NO, Bakos HW, Fullston T, Lane M. Impact of obesity on male fertility, sperm function and molecular composition. Spermatogenesis. 2012;2:253-63.

Many studies in animal model focus on the adverse factors influencing paternal exposure, from the prepubertal period to preconception. Therefore, is more plausible to consider that the epigenome undergoes reprogramming in pre-implantation embryos and in primordial germ cells.3131 Saitou M, Kagiwada S, Kurimoto K. Epigenetic reprogramming in mouse pre-implantation development and primordial germ cells. Development. 2012;139:15-31.

Environmental exposures in animal models over this period have been suggested to induce intergenerational and transgenerational effects through the sperm epigenome.3232 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;27:4226-43. Data demonstrate that numerous metabolic effects observed in the first generation are shown to persist in the second generation, with F1 females producing F2 males with increased adiposity. Paternal obesity modified the expression of various microRNAs, concomitant with alterations in sperm microRNA content and a reduction in global methylation of germ cell DNA.3232 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;27:4226-43.

From the moment the blastocyst is formed, the pre-implantation is over, and a new stage starts - the in utero development. After entering the gonads, primordial germ cells convert to gonocytes, and in mouse models decreasing decrease in DNA methylation levels is observed.3333 Seisenberger S, Andrews S, Krueger F, Arand J, Walter J, Santos F, et al. The dynamics of genome-wide DNA methylation reprogramming in mouse primordial germ cells. Mol Cell. 2012;48:849-62. The primary responsibilities in this timeframe encompass cell-specific gene expression, tissue differentiation, and tissue-specific epigenomes.2727 Schagdarsurengin U, Steger K. Epigenetics in male reproduction: effect of paternal diet on sperm quality and offspring health. Nat Rev Urol. 2016;13:584-95.

Obese mice fathers presented H3 retention and genomic imprints in the sperm, and differences in liver mRNA expression of several fat synthesis-related genes were observed in the offspring. Differences were observed in the liver expression of matallothionein-1 and -2 (Mt1 and Mt2), fatty acid synthase (Fasn), P450 cytochrome oxidoreductase,3434 Kilgore JA, Hoose SA, Gustafson TL, Porter W, Kladde MP. Single-molecule and population probing of chromatin structure using DNA methyltransferases. Methods. 2007;41:320-32. and acetyl-CoA carboxylase-α (Acaca) at the age of 24 weeks. Paternal obesity also increased histone H3 occupancy in the promoters of the genes responsible for the embryonic development and increased monomethylation of lysine 4 on histone H3 (H3K4me1) in genes responsible for embryogenesis regulation in spermatozoa. Altogether, findings suggest that dietary exposure can modulate histone composition at genes involved in the process of development.3535 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;10:861-71.

Epigenetics

The mechanisms explaining how the father may affect the development of the offspring are still under debate. Epigenetics is the primary tool for the transmission of paternal phenotypes to offspring, because the transient nutritional stimuli in critical stages of ontogenesis may influence the expression of various genes by changes in chromatin conformation and the accessibility of transcription factors.3636 Gallou-Kabani C, Junien C. Nutritional epigenomics of metabolic syndrome: new perspective against the epidemic. Diabetes. 2005;54:1899-906.

Therefore, the term epigenetic (Greek prefix epi- (ɛπί): over, outside of, around), proposed by Conrad Waddington, is described as a "process of development of the phenotype from the genotype."3737 Waddington C. The epigenotype. Endeavour. 1942;1:18-20. In other words, epigenetic is any transmissible and reversible modification in the expression of a gene without structural change in the sequence of DNA.3838 Heard E, Martienssen RA. Transgenerational epigenetic inheritance: myths and mechanisms. Cell. 2014;157:95-109. Unlike the genetic variation of the germline, which remains unchanged in all cells of the body, epigenetic modification is dynamic and varies among tissues in response to a range of environmental stimuli, including those that direct tissue differentiation during development and growth, and the serious risks that provoke an adaptive response of cells.3939 Estampador AC, Franks PW. Genetic and epigenetic catalysts in early-life programming of adult cardiometabolic disorders. Diabetes Metab Syndr Obes. 2014;7:575-86.

Some epigenetic marks during spermatogenesis may continue throughout the embryonic development. Environmental exposures (diet, lifestyle, and other exposures) that occur in male gametogenesis can cause irreversible epigenetic changes and phenotypic consequences expressed in the offspring.4040 Soubry A. Epigenetic inheritance and evolution: a paternal perspective on dietary influences. Prog Biophys Mol Biol. 2015;118:79-85. Epigenetic processes also modulate the effects through transcription regulation due to several processes, such as DNA methylation, histone alterations, and transcription of non-coding RNA (miRNA, for example).4141 Li N, Shen Q, Hua J. Epigenetic remodeling in male germline development. Stem Cells Int. 2016;2016:3152173.,4242 Zhao H, Zhao Y, Ren Y, Li M, Li T, Li R, et al. Epigenetic regulation of an adverse metabolic phenotype in polycystic ovary syndrome: the impact of the leukocyte methylation of PPARGC1A promoter. Fertil Steril. 2017;107:467-74.

Epigenetic modifications acting on the cell plasticity capacity prepare the individual for the extrauterine environment and may potentiate a survival advantage by regulating the differential genes encoding proteins involved in energy metabolism and adipogenesis.4343 Gao Q, Tang J, Chen J, Jiang L, Zhu X, Xu Z. Epigenetic code and potential epigenetic-based therapies against chronic diseases in developmental origins. Drug Discov Today. 2014;19:1744-50. However, in face of a deleterious metabolic condition such as obesity and related metabolic alterations, these modifications can be exacerbated or silenced, especially germ cells program, which constitute the species' perpetuation and phenotypic transmission.4444 Guerrero-Bosagna C, Skinner MK. Environmentally induced epigenetic transgenerational inheritance of male infertility. Curr Opin Genet Dev. 2014;26:79-88.

Recent hypotheses consider that some paternal dietary patterns reside in spermatozoa bearing epigenetic information.2727 Schagdarsurengin U, Steger K. Epigenetics in male reproduction: effect of paternal diet on sperm quality and offspring health. Nat Rev Urol. 2016;13:584-95. The sperm "epigenome" was traditionally considered insignificant, since it was postulated that its DNA methylation profile was erased immediately after fertilization. However, in recent years, there has been an increase in the number of reported cases of apparent epigenetic inheritance through the male germline, suggesting that this epigenome may transmit information between generations.2828 Soubry A, Hoyo C, Jirtle RL, Murphy SK. A paternal environmental legacy: evidence for epigenetic inheritance through the male germ line. Bioessays. 2014;36:359-71.,4040 Soubry A. Epigenetic inheritance and evolution: a paternal perspective on dietary influences. Prog Biophys Mol Biol. 2015;118:79-85.

The development of sperm- and spermatid-derived frog embryos allow the observation that the sperm not only transfers DNA, but also contributes to the epigenetic information required for proper embryonic gene expression, being the key for epigenetic information.4545 Teperek M, Simeone A, Gaggioli V, Miyamoto K, Allen GE, Erkek S, et al. Sperm is epigenetically programmed to regulate gene transcription in embryos. Genome Res. 2016;26:1034-46.

Methylation

DNA methylation is one of the chemical reactions that occur most frequently in eukaryotes such as plants, fungi, invertebrates, and vertebrates.3434 Kilgore JA, Hoose SA, Gustafson TL, Porter W, Kladde MP. Single-molecule and population probing of chromatin structure using DNA methyltransferases. Methods. 2007;41:320-32. This chemical modification is characterized by the addition of a methyl group at the C5 position of the cytosine ring, catalyzed by the DNA methyltransferases, leading to the formation of 5-methylcytosine.4646 Caiafa P, Zampieri M. DNA methylation and chromatin structure: the puzzling CpG islands. J Cell Biochem. 2005;94:257-65.,4747 Fan H, Zhao ZJ, Cheng J, Su XW, Wu QX, Shan YF. Overexpression of DNA methyltransferase 1 and its biological significance in primary hepatocellular carcinoma. World J Gastroenterol. 2009;15:2020-6. The frequency of 5-methylcytidine is less than 1% of the total number of nucleotides in the genome.4848 Yen CY, Huang HW, Shu CW, Hou MF, Yuan SS, Wang HR, et al. DNA methylation, histone acetylation and methylation of epigenetic modifications as a therapeutic approach for cancers. Cancer Lett. 2016;373:185-92. This methylation process acts in normal embryonic development, inactivation of the X chromosome, genetic regulation, genomic press, and chromatin modifications.4949 Bird A. DNA methylation patterns and epigenetic memory. Genes Dev. 2002;16:6-21.

Most DNA methylation occurs in regions called CpG islands, which corresponds to genomic regions of more than 1000 base pairs in length and with many GC dinucleotides; about 55% of these islands are in the promoter regions of approximately 40% of mammalian genes. These CpG (cytosine-phosphate-guanine) islands are kept non-methylated, except in imprinting genes or when located on the inactive X chromosome.5050 Deng X, Ma W, Ramani V, Hill A, Yang F, Ay F, et al. Bipartite structure of the inactive mouse X chromosome. Genome Biol. 2015;16:152.,5151 Kota SK, Roy Chowdhury D, Rao LK, Padmalatha V, Singh L, Bhadra U. Uncoupling of X-linked gene silencing from XIST binding by DICER1 and chromatin modulation on human inactive X chromosome. Chromosoma. 2015;124:249-62. Therefore, CpG islands located in the promoter region of housekeeping and developmental regulator genes with dense cytosine and guanine distribution are resistant to DNA methylation.5252 Xu W, Wang F, Yu Z, Xin F. Epigenetics and cellular metabolism. Genet Epigenet. 2016;8:43-51. The CpG islands, which allow the binding of proteins and enzymes, initiate the cascade of transcription. In contrast, methylated CpG islands are related to transcriptional silencing.5353 Egger G, Liang G, Aparicio A, Jones PA. Epigenetics in human disease and prospects for epigenetic therapy. Nature. 2004;429:457-63.

Hypermethylation of promoter regions rich in dinucleotides has a significant role in the loss of gene expression.5454 Luczak MW, Jagodzinski PP. The role of DNA methylation in cancer development. Folia Histochem Cytobiol. 2006;44:143-54. Typically, hypomethylation of DNA triggers an increase in gene expression, while hypermethylation decreases the expression of target genes.5555 Liddle RA, Jirtle RL. Epigenetic silencing of genes in human colon cancer. Gastroenterology. 2006;131:960-2. Furthermore, transcription factors do not recognize and bind to transcription initiation sites due to the modification of cytosine into 5-methylcytidine. This is the case of adipocyte protein 2 (AP-2), cMYC/murine homolog of max (cMYC/MYN), cyclic adenosine monophosphate response element binding protein (CREB), E2 factor (E2F), and nuclear factor-κB (NF-KB). However, these binding sites may be occupied by other proteins such as methyl-CpG binding protein 2 (MeCP-2) and methyl-CpG-binding domain protein (MBD)1, MBD2, MBD3, and MBD4, which bind to methylated cytosines and stimulate chromatin condensation, inactivating the gene.4646 Caiafa P, Zampieri M. DNA methylation and chromatin structure: the puzzling CpG islands. J Cell Biochem. 2005;94:257-65.,5656 Weyrich A, Lenz D, Jeschek M, Chung TH, Rubensam K, Goritz F, et al. Paternal intergenerational epigenetic response to heat exposure in male Wild guinea pigs. Mol Ecol. 2016;25:1729-40.

The enzymes responsible for the addition of a methyl group to cytosine molecules belong to the family of DNA methyltransferases (DNMTs), including DNMT-1, DNMT3A, DNMT3B and its isoforms, and DNMT3L.4747 Fan H, Zhao ZJ, Cheng J, Su XW, Wu QX, Shan YF. Overexpression of DNA methyltransferase 1 and its biological significance in primary hepatocellular carcinoma. World J Gastroenterol. 2009;15:2020-6.,5757 Sarabi MM, Naghibalhossaini F. Association of DNA methyltransferases expression with global and gene-specific DNA methylation in colorectal cancer cells. Cell Biochem Funct. 2015;33:427-33. DNMT1 is primarily responsible for maintaining DNA methylation patterns during mitosis. Furthermore, DNMT (differentially methylated regions)-s3 is responsible for de novo methylation of newly synthesized DNA molecules and is most important during the early stages of embryonic development; it could be the process involved in paternal programming for phenotypic transmission.5858 Dolinoy DC, Weidman JR, Jirtle RL. Epigenetic gene regulation: linking early developmental environment to adult disease. Reprod Toxicol. 2007;23:297-307.,5959 Moss TJ, Wallrath LL. Connections between epigenetic gene silencing and human disease. Mutat Res. 2007;618:163-74. A schematic view of the main paternal epigenetic modifications can be seen in Fig. 1.

Figure 1
Epigenetics of paternal programming on the offspring. Paternal metabolic changes related to obesity may result in modifications to genetic information contained in the spermatozoa. DNA methylation in specific promoter regions most often prevents gene transcription by inactivating the gene in question. As for histone modifications, hypermethylation (depending on histone and amino acid) favors the condensation of chromatin, making difficult access to regulatory proteins that promote transcription. However, non-methylated histones ensure decondensed chromatin, supporting gene transcription. In contrast, histone acetylation opens the chromatin, allowing coupling of the transcriptional machinery.

In a contrary process, DNA demethylation is also an important epigenetic component of gene transcription and epigenetic programming, which occurs through several enzymatic reactions mediating the oxidation of 5-methylcytosine to 5-hydroxymethylcytosine.6060 Scourzic L, Mouly E, Bernard OA. TET proteins and the control of cytosine demethylation in cancer. Genome Med. 2015;7:9. The presence of 5-hydroxymethylcytosine in promoter regions results in increased transcription, indicating a role in long- and short-term regulation of gene expression.6161 Ficz G, Branco MR, Seisenberger S, Santos F, Krueger F, Hore TA, et al. Dynamic regulation of 5-hydroxymethylcytosine in mouse ES cells and during differentiation. Nature. 2011;473:398-402.

Histone modifications

Histone modifications are other biological actions that regulate gene expression. Histones are basic proteins found in eukaryotic cell nuclei, which help in the packaging of DNA into nucleosomes, the binding blocks of chromatin.6262 Sarkar DK. Male germline transmits fetal alcohol epigenetic marks for multiple generations: a review. Addict Biol. 2016;21:23-34. Chromatin should be remodeled, modifying the accessibility of the DNA transcription machinery to control the transcription process.6363 Clapier CR, Cairns BR. The biology of chromatin remodeling complexes. Annu Rev Biochem. 2009;78:273-304. Events such as acetylation, methylation, phosphorylation and ubiquitination, often occur in the tail of the histones that extend from the center of the nucleosomes.6464 Howell PM, Liu S, Ren S, Behlen C, Fodstad O, Riker AI. Epigenetics in human melanoma. Cancer Control. 2009;16:200-18. Other chemical modifications also alter histones, such as histone lysine glucosamine acylation (GlcNAcylation), butyrylation, malonylation, and crotonylation.6565 Jenuwein T, Allis CD. Translating the histone code. Science. 2001;293:1074-80.,6666 Gelato KA, Fischle W. Role of histone modifications in defining chromatin structure and function. Biol Chem. 2008;389:353-63.

Histones 3 and 4 (H3 and H4) are commonly studied, and acetylation is the main epigenetic modification considered; it occurs on the lysine and arginine and neutralizes the positive charge of basic residues. The histone acetylase enzymes add acetyl groups to the histone lysine residues, and it is believed that the acetylated histones have a reduced affinity between DNA and histones, leaving chromatin in a relaxed (euchromatin) and transcriptionally active state.6666 Gelato KA, Fischle W. Role of histone modifications in defining chromatin structure and function. Biol Chem. 2008;389:353-63.

In contrast, histone deacetylase removes the acetyl groups, which are more condensed, and prevents gene expression.6767 Ausio J, Levin DB, De Amorim GV, Bakker S, Macleod PM. Syndromes of disordered chromatin remodeling. Clin Genet. 2003;64:83-95. These chemical modifications alter the interaction between DNA and histones, changing the degree of chromatin folding and the gene activity.6868 Deobagkar DD, Chandra HS. The inactive X chromosome in the human female is enriched in 5-methylcytosine to an unusual degree and appears to contain more of this modified nucleotide than the remainder of the genome. J Genet. 2003;82:13-6. Therefore, heterochromatin is related with hypoacetylation for H3 and H4, and di- or trimethylation of the ninth lysine residue on H3 (H3K9me2 or H3K9me3).6969 Arney KL, Fisher AG. Epigenetic aspects of differentiation. J Cell Sci. 2004;117:4355-63.

Non-coding RNA (microRNA)

In addition to DNA methylation and histone modification, sperm RNA can be an epigenetic regulator. Spermatozoa contain an array of both mRNAs7070 Carrell DT. Contributions of spermatozoa to embryogenesis: assays to evaluate their genetic and epigenetic fitness. Reprod Biomed Online. 2008;16:474-84. and noncoding RNAs, including miRNA.7171 Yan W, Morozumi K, Zhang J, Ro S, Park C, Yanagimachi R. Birth of mice after intracytoplasmic injection of single purified sperm nuclei and detection of messenger RNAs and microRNAs in the sperm nuclei. Biol Reprod. 2008;78:896-902. Most of this RNA is delivered to the oocyte. However, the role of miRNA in early preimplantation embryos is still under debate.

Evidence of the direct biological effect of miRNAs in the pre-implantation period is supported by observations in mice with a chromosomal lesion in the Dicer gene. Loss of enzymatic processing of Dicer miRNA in oocytes leads to early lethality in the development process, where the zygotes cannot survive the division in the first place.7272 Bernstein E, Kim SY, Carmell MA, Murchison EP, Alcorn H, Li MZ, et al. Dicer is essential for mouse development. Nat Genet. 2003;35:215-7. Therefore, this suggests the interference of miRNA in zygote development.

miRNA is a major part of the group of such small non-coding. These components have approximately 21nt-long RNA molecules that repress their target mRNA.7373 Bhat SS, Jarmolowski A, Szweykowska-Kulinska Z. MicroRNA biogenesis: epigenetic modifications as another layer of complexity in the microRNA expression regulation. Acta Biochim Pol. 2016;63:717-23. In males, they are observed in the nucleus of spermatozoa, keeping the DNA connected to histones during spermiogenesis and in early embryonic development.7474 Hamatani T. Human spermatozoal RNAs. Fertil Steril. 2012;97:275-81. miRNA regulates various biological functions, influencing the epigenetic inactivation of genes and the protection of the DNA against viruses and transposons.7575 Luteijn MJ, Ketting RF. PIWI-interacting RNAs: from generation to transgenerational epigenetics. Nat Rev Genet. 2013;14:523-34.

In a general way, miRNA in animals is mostly located within the introns of protein coding or non-coding RNA genes,7676 Kim VN, Han J, Siomi MC. Biogenesis of small RNAs in animals. Nat Rev Mol Cell Biol. 2009;10:126-39. being produced by RNA polymerase II transcription. In humans, miRNA appears to be synthesized by RNA polymerase III.7777 Borchert GM, Lanier W, Davidson BL. RNA polymerase III transcribes human microRNAs. Nat Struct Mol Biol. 2006;13:1097-101. In recent years, over- or under-expression of miRNA has been associated with disease development, but the mechanisms and actions are still ambiguous.7878 Li Y, Kowdley KV. MicroRNAs in common human diseases. Genomics Proteomics Bioinform. 2012;10:246-53.

Epigenetics in paternal programming

Several studies in animals and humans have demonstrated the influence of paternal diet on their offspring's phenotype through the epigenome.

Male mice fed a high-fat diet generated female offspring with impaired glucose-insulin homeostasis, associated with an altered expression in one of the 642 pancreatic islet genes and in the hypomethylated interleukin 13 receptor alpha 2 (IL13ra2) gene.99 Ng SF, Lin RC, Laybutt DR, Barres R, Owens JA, Morris MJ. Chronic high-fat diet in fathers programs beta-cell dysfunction in female rat offspring. Nature. 2010;467:963-6. These alterations were recently observed in the transcriptomes of retroperitoneal adipose and pancreatic islet tissues in female offspring. In retroperitoneal adipose tissue, 5108 genes were differentially expressed due to paternal high-fat diet, whose functions are related to mitochondrial and cellular response to stress, telomerase signaling, cell death and survival, cell cycle, cellular growth and proliferation, and cancer.7979 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;28:1830-41.

In a mice model of paternal low-protein diet, the offspring showed an elevated methylation in peroxisome proliferator-activated receptor (PPAR)-alpha in the liver, a gene involved in the formation of the first enzymes for the oxidation of lipids in mitochondria, being an essential lipid regulator.8080 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;143:1084-96. Also, paternal insulin resistance has altered the methylation status of various insulin-signaling genes in the offspring, increasing the offspring's susceptibility to diabetes through gametic epigenetic alterations. Such includes the genes phosphoinositide-3-kinase regulatory subunit 1 (Pik3r1), phosphoinositide-3-kinase catalytic alpha polypeptide (Pik3ca), Ptpn1 protein tyrosine phosphatase, non-receptor type 1 (Ptpn1), and Pik3ca in sperm.8181 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;111:1873-8. In mice models, paternal high-fat diet led to an increase in transfer RNA (tRNA) in sperm as an inherited epigenetic key influenced by paternal diet and related to metabolic impairment in the offspring, leading to glucose intolerance and insulin resistance.8282 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.

Paternal obesity initiates metabolic disturbances in two generations of mice, altering the transcriptional profile of testis and sperm miRNA content. The differential content of canonical miRNAs in the sperm suggested dysregulation in spermatogenesis, embryo development, and metabolic function. Furthermore, paternal high-fat diet affected the metabolic status of offspring through epigenetic changes in the adiponectin and leptin genes for two generations.8383 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:E236-45. Interestingly, an increase in miR-29 (microRNA 29) has been associated with a decrease in methylation of repeat elements in the male germline.3232 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;27:4226-43. Moreover, 13 sperm-borne miRNA were modulated by paternal high-fat diet and transferred off this altered miRNA payload to the embryo at fertilization, changing its growth trajectory and affecting adult offspring phenotype.8484 Fullston T, Ohlsson-Teague EM, Print CG, Sandeman LY, Lane M. Sperm microRNA content is altered in a mouse model of male obesity, but the same suite of microRNAs are not altered in offspring's sperm. PLOS ONE. 2016;11:e0166076.

In newborns, an association was observed between preconception obesity and DNA methylation profiles in the offspring, particularly in the differentially methylated region (DMR) of the imprinted insulin-like growth factor 2 (IGF2) gene. The hypomethylation at the IGF2 DMR in the offspring was associated with paternal obesity.8585 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. It is conceivable that additional or novel epigenetic regulators (such as prions) are present in sperm, that sperm quality is affected by diet, and that environment may direct the genetic changes. Although it is important to emphasize that inbred mouse strains were used in this study.8080 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;143:1084-96. Furthermore, γ-radiation-induced DNA damage in the sperm has been shown to be heritable, with offspring similarly exhibiting sperm DNA damage.8686 Adiga SK, Upadhya D, Kalthur G, Bola Sadashiva SR, Kumar P. Transgenerational changes in somatic and germ line genetic integrity of first-generation offspring derived from the DNA-damaged sperm. Fertil Steril. 2010;93:2486-90.

Epigenetic modification is a continual process, and some changes may be reversible.2020 Slyvka Y, Zhang Y, Nowak FV. Epigenetic effects of paternal diet on offspring: emphasis on obesity. Endocrine. 2015;48:36-46. Combined, these data reveal the impact of paternal programming (in particular through nutritional manipulations) on the future life of the offspring, influenced mainly by phenotype transmission via the epigenetic process.

Final considerations

Data from epidemiological and animal studies provide evidence that paternal feeding and paternal health conditions can program following generations. Thus, the mother is not the only responsible for offspring's health. The father shares the responsibility in providing sperm-specific epigenetic stamp to the oocyte, affecting the embryo developmental trajectory and health of adult offspring. Although the role of paternal influence may be clearly identified, the current knowledge on reprogramming, alteration, and possible prevention of paternal effects remains limited. To date, human studies are not progressing in the same manner as animal studies, so there is little information about the mechanism and contribution of paternal programming in the child's health, as men are widely used as controls in studies with women. Therefore, considering that paternal obesity can also be a public health problem, it is important to improve epidemiological studies to assess the exact role of paternal health on the sperm and on the health of the offspring to propose further interventions.

  • Please cite this article as: Ornellas F, Carapeto PV, Mandarim-de-Lacerda CA, Aguila MB. Obese fathers lead to an altered metabolism and obesity in their children in adulthood: review of experimental and human studies. J Pediatr (Rio J). 2017;93:551-9.

Acknowledgements

This laboratory (www.lmmc.uerj.br) is sponsored by Conselho Nacional de Desenvolvimento Científico e Tecnológico (CNPq, grant #302.154/2011-6 to CAML, and #306.077/2013-2 to MBA), and Fundação Carlos Chagas Filho de Amparo à Pesquisa do Rio de Janeiro (FAPERJ, grant #102.944/2011 to CAML, #103.062/2011 to MBA).

References

  • 1
    Huang H, Yan Z, Chen Y, Liu F. A social contagious model of the obesity epidemic. Sci Rep. 2016;6:37961.
  • 2
    Barker DJ. The fetal and infant origins of adult disease. BMJ. 1990;301:1111.
  • 3
    Barker DJ. The developmental origins of adult disease. J Am Coll Nutr. 2004;23:588S-95S.
  • 4
    Gusmao-Correia ML, Volpato AM, Aguila MB, Mandarim-de-Lacerda CA. Developmental origins of health and disease: experimental and human evidence of fetal programming for metabolic syndrome. J Hum Hypertens. 2012;26:405-19.
  • 5
    Volpato AM, Schultz A, Magalhaes-da-Costa E, Correia ML, Aguila MB, Mandarim-de-Lacerda CA. Maternal high-fat diet programs for metabolic disturbances in offspring despite leptin sensitivity. Neuroendocrinology. 2012;96:272-84.
  • 6
    Ornellas F, Souza-Mello V, Mandarim-de-Lacerda CA, Aguila MB. Combined parental obesity augments single-parent obesity effects on hypothalamus inflammation, leptin signaling (JAK/STAT), hyperphagia, and obesity in the adult mice offspring. Physiol Behav. 2016;153:47-55.
  • 7
    Bringhenti I, Ornellas F, Mandarim-de-Lacerda CA, Aguila MB. The insulin-signaling pathway of the pancreatic islet is impaired in adult mice offspring of mothers fed a high-fat diet. Nutrition. 2016;32:1138-43.
  • 8
    Ornellas F, Mello VS, Mandarim-de-Lacerda CA, Aguila MB. Sexual dimorphism in fat distribution and metabolic profile in mice offspring from diet-induced obese mothers. Life Sci. 2013;93:454-63.
  • 9
    Ng SF, Lin RC, Laybutt DR, Barres R, Owens JA, Morris MJ. Chronic high-fat diet in fathers programs beta-cell dysfunction in female rat offspring. Nature. 2010;467:963-6.
  • 10
    Li L, Law C, Lo Conte R, Power C. Intergenerational influences on childhood body mass index: the effect of parental body mass index trajectories. Am J Clin Nutr. 2009;89:551-7.
  • 11
    Nguyen DM, El-Serag HB. The epidemiology of obesity. Gastroenterol Clin North Am. 2010;39:1-7.
  • 12
    Chen YP, Xiao XM, Li J, Reichetzeder C, Wang ZN, Hocher B. Paternal body mass index (BMI) is associated with offspring intrauterine growth in a gender-dependent manner. PLoS ONE. 2012;7:e36329.
  • 13
    Kaati G, Bygren LO, Pembrey M, Sjostrom M. Transgenerational response to nutrition, early life circumstances and longevity. Eur J Hum Genet. 2007;15:784-90.
  • 14
    Bygren LO, Kaati G, Edvinsson S. Longevity determined by paternal ancestors' nutrition during their slow growth period. Acta Biotheor. 2001;49:53-9.
  • 15
    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;10:682-8.
  • 16
    Pembrey ME, Bygren LO, Kaati G, Edvinsson S, Northstone K, Sjostrom M, et al. Sex-specific, male-line transgenerational responses in humans. Eur J Hum Genet. 2006;14:159-66.
  • 17
    Li J, Tsuprykov O, Yang X, Hocher B. Paternal programming of offspring cardiometabolic diseases in later life. J Hypertens. 2016;34:2111-26.
  • 18
    Hocher B. More than genes: the advanced fetal programming hypothesis. J Reprod Immunol. 2014;104-105:8-11.
  • 19
    Pampel FC, Denney JT, Obesity Krueger PM. SES, and economic development: a test of the reversal hypothesis. Soc Sci Med. 2012;74:1073-81.
  • 20
    Slyvka Y, Zhang Y, Nowak FV. Epigenetic effects of paternal diet on offspring: emphasis on obesity. Endocrine. 2015;48:36-46.
  • 21
    Ortega FJ, Mercader JM, Catalan V, Moreno-Navarrete JM, Pueyo N, Sabater M, et al. Targeting the circulating microRNA signature of obesity. Clin Chem. 2013;59:781-92.
  • 22
    Bakos HW, Henshaw RC, Mitchell M, Lane M. Paternal body mass index is associated with decreased blastocyst development and reduced live birth rates following assisted reproductive technology. Fertil Steril. 2011;95:1700-4.
  • 23
    Tunc O, Bakos HW, Tremellen K. Impact of body mass index on seminal oxidative stress. Andrologia. 2011;43:121-8.
  • 24
    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:E805-21.
  • 25
    Ornellas F, Souza-Mello V, Mandarim-de-Lacerda CA, Aguila MB. Programming of obesity and comorbidities in the progeny: lessons from a model of diet-induced obese parents. PLOS ONE. 2015;10:e0124737.
  • 26
    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.
  • 27
    Schagdarsurengin U, Steger K. Epigenetics in male reproduction: effect of paternal diet on sperm quality and offspring health. Nat Rev Urol. 2016;13:584-95.
  • 28
    Soubry A, Hoyo C, Jirtle RL, Murphy SK. A paternal environmental legacy: evidence for epigenetic inheritance through the male germ line. Bioessays. 2014;36:359-71.
  • 29
    Wu H, Hauser R, Krawetz SA, Pilsner JR. Environmental susceptibility of the sperm epigenome during windows of male germ cell development. Curr Environ Health Rep. 2015;2:356-66.
  • 30
    Palmer NO, Bakos HW, Fullston T, Lane M. Impact of obesity on male fertility, sperm function and molecular composition. Spermatogenesis. 2012;2:253-63.
  • 31
    Saitou M, Kagiwada S, Kurimoto K. Epigenetic reprogramming in mouse pre-implantation development and primordial germ cells. Development. 2012;139:15-31.
  • 32
    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;27:4226-43.
  • 33
    Seisenberger S, Andrews S, Krueger F, Arand J, Walter J, Santos F, et al. The dynamics of genome-wide DNA methylation reprogramming in mouse primordial germ cells. Mol Cell. 2012;48:849-62.
  • 34
    Kilgore JA, Hoose SA, Gustafson TL, Porter W, Kladde MP. Single-molecule and population probing of chromatin structure using DNA methyltransferases. Methods. 2007;41:320-32.
  • 35
    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;10:861-71.
  • 36
    Gallou-Kabani C, Junien C. Nutritional epigenomics of metabolic syndrome: new perspective against the epidemic. Diabetes. 2005;54:1899-906.
  • 37
    Waddington C. The epigenotype. Endeavour. 1942;1:18-20.
  • 38
    Heard E, Martienssen RA. Transgenerational epigenetic inheritance: myths and mechanisms. Cell. 2014;157:95-109.
  • 39
    Estampador AC, Franks PW. Genetic and epigenetic catalysts in early-life programming of adult cardiometabolic disorders. Diabetes Metab Syndr Obes. 2014;7:575-86.
  • 40
    Soubry A. Epigenetic inheritance and evolution: a paternal perspective on dietary influences. Prog Biophys Mol Biol. 2015;118:79-85.
  • 41
    Li N, Shen Q, Hua J. Epigenetic remodeling in male germline development. Stem Cells Int. 2016;2016:3152173.
  • 42
    Zhao H, Zhao Y, Ren Y, Li M, Li T, Li R, et al. Epigenetic regulation of an adverse metabolic phenotype in polycystic ovary syndrome: the impact of the leukocyte methylation of PPARGC1A promoter. Fertil Steril. 2017;107:467-74.
  • 43
    Gao Q, Tang J, Chen J, Jiang L, Zhu X, Xu Z. Epigenetic code and potential epigenetic-based therapies against chronic diseases in developmental origins. Drug Discov Today. 2014;19:1744-50.
  • 44
    Guerrero-Bosagna C, Skinner MK. Environmentally induced epigenetic transgenerational inheritance of male infertility. Curr Opin Genet Dev. 2014;26:79-88.
  • 45
    Teperek M, Simeone A, Gaggioli V, Miyamoto K, Allen GE, Erkek S, et al. Sperm is epigenetically programmed to regulate gene transcription in embryos. Genome Res. 2016;26:1034-46.
  • 46
    Caiafa P, Zampieri M. DNA methylation and chromatin structure: the puzzling CpG islands. J Cell Biochem. 2005;94:257-65.
  • 47
    Fan H, Zhao ZJ, Cheng J, Su XW, Wu QX, Shan YF. Overexpression of DNA methyltransferase 1 and its biological significance in primary hepatocellular carcinoma. World J Gastroenterol. 2009;15:2020-6.
  • 48
    Yen CY, Huang HW, Shu CW, Hou MF, Yuan SS, Wang HR, et al. DNA methylation, histone acetylation and methylation of epigenetic modifications as a therapeutic approach for cancers. Cancer Lett. 2016;373:185-92.
  • 49
    Bird A. DNA methylation patterns and epigenetic memory. Genes Dev. 2002;16:6-21.
  • 50
    Deng X, Ma W, Ramani V, Hill A, Yang F, Ay F, et al. Bipartite structure of the inactive mouse X chromosome. Genome Biol. 2015;16:152.
  • 51
    Kota SK, Roy Chowdhury D, Rao LK, Padmalatha V, Singh L, Bhadra U. Uncoupling of X-linked gene silencing from XIST binding by DICER1 and chromatin modulation on human inactive X chromosome. Chromosoma. 2015;124:249-62.
  • 52
    Xu W, Wang F, Yu Z, Xin F. Epigenetics and cellular metabolism. Genet Epigenet. 2016;8:43-51.
  • 53
    Egger G, Liang G, Aparicio A, Jones PA. Epigenetics in human disease and prospects for epigenetic therapy. Nature. 2004;429:457-63.
  • 54
    Luczak MW, Jagodzinski PP. The role of DNA methylation in cancer development. Folia Histochem Cytobiol. 2006;44:143-54.
  • 55
    Liddle RA, Jirtle RL. Epigenetic silencing of genes in human colon cancer. Gastroenterology. 2006;131:960-2.
  • 56
    Weyrich A, Lenz D, Jeschek M, Chung TH, Rubensam K, Goritz F, et al. Paternal intergenerational epigenetic response to heat exposure in male Wild guinea pigs. Mol Ecol. 2016;25:1729-40.
  • 57
    Sarabi MM, Naghibalhossaini F. Association of DNA methyltransferases expression with global and gene-specific DNA methylation in colorectal cancer cells. Cell Biochem Funct. 2015;33:427-33.
  • 58
    Dolinoy DC, Weidman JR, Jirtle RL. Epigenetic gene regulation: linking early developmental environment to adult disease. Reprod Toxicol. 2007;23:297-307.
  • 59
    Moss TJ, Wallrath LL. Connections between epigenetic gene silencing and human disease. Mutat Res. 2007;618:163-74.
  • 60
    Scourzic L, Mouly E, Bernard OA. TET proteins and the control of cytosine demethylation in cancer. Genome Med. 2015;7:9.
  • 61
    Ficz G, Branco MR, Seisenberger S, Santos F, Krueger F, Hore TA, et al. Dynamic regulation of 5-hydroxymethylcytosine in mouse ES cells and during differentiation. Nature. 2011;473:398-402.
  • 62
    Sarkar DK. Male germline transmits fetal alcohol epigenetic marks for multiple generations: a review. Addict Biol. 2016;21:23-34.
  • 63
    Clapier CR, Cairns BR. The biology of chromatin remodeling complexes. Annu Rev Biochem. 2009;78:273-304.
  • 64
    Howell PM, Liu S, Ren S, Behlen C, Fodstad O, Riker AI. Epigenetics in human melanoma. Cancer Control. 2009;16:200-18.
  • 65
    Jenuwein T, Allis CD. Translating the histone code. Science. 2001;293:1074-80.
  • 66
    Gelato KA, Fischle W. Role of histone modifications in defining chromatin structure and function. Biol Chem. 2008;389:353-63.
  • 67
    Ausio J, Levin DB, De Amorim GV, Bakker S, Macleod PM. Syndromes of disordered chromatin remodeling. Clin Genet. 2003;64:83-95.
  • 68
    Deobagkar DD, Chandra HS. The inactive X chromosome in the human female is enriched in 5-methylcytosine to an unusual degree and appears to contain more of this modified nucleotide than the remainder of the genome. J Genet. 2003;82:13-6.
  • 69
    Arney KL, Fisher AG. Epigenetic aspects of differentiation. J Cell Sci. 2004;117:4355-63.
  • 70
    Carrell DT. Contributions of spermatozoa to embryogenesis: assays to evaluate their genetic and epigenetic fitness. Reprod Biomed Online. 2008;16:474-84.
  • 71
    Yan W, Morozumi K, Zhang J, Ro S, Park C, Yanagimachi R. Birth of mice after intracytoplasmic injection of single purified sperm nuclei and detection of messenger RNAs and microRNAs in the sperm nuclei. Biol Reprod. 2008;78:896-902.
  • 72
    Bernstein E, Kim SY, Carmell MA, Murchison EP, Alcorn H, Li MZ, et al. Dicer is essential for mouse development. Nat Genet. 2003;35:215-7.
  • 73
    Bhat SS, Jarmolowski A, Szweykowska-Kulinska Z. MicroRNA biogenesis: epigenetic modifications as another layer of complexity in the microRNA expression regulation. Acta Biochim Pol. 2016;63:717-23.
  • 74
    Hamatani T. Human spermatozoal RNAs. Fertil Steril. 2012;97:275-81.
  • 75
    Luteijn MJ, Ketting RF. PIWI-interacting RNAs: from generation to transgenerational epigenetics. Nat Rev Genet. 2013;14:523-34.
  • 76
    Kim VN, Han J, Siomi MC. Biogenesis of small RNAs in animals. Nat Rev Mol Cell Biol. 2009;10:126-39.
  • 77
    Borchert GM, Lanier W, Davidson BL. RNA polymerase III transcribes human microRNAs. Nat Struct Mol Biol. 2006;13:1097-101.
  • 78
    Li Y, Kowdley KV. MicroRNAs in common human diseases. Genomics Proteomics Bioinform. 2012;10:246-53.
  • 79
    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;28:1830-41.
  • 80
    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;143:1084-96.
  • 81
    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;111:1873-8.
  • 82
    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.
  • 83
    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:E236-45.
  • 84
    Fullston T, Ohlsson-Teague EM, Print CG, Sandeman LY, Lane M. Sperm microRNA content is altered in a mouse model of male obesity, but the same suite of microRNAs are not altered in offspring's sperm. PLOS ONE. 2016;11:e0166076.
  • 85
    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.
  • 86
    Adiga SK, Upadhya D, Kalthur G, Bola Sadashiva SR, Kumar P. Transgenerational changes in somatic and germ line genetic integrity of first-generation offspring derived from the DNA-damaged sperm. Fertil Steril. 2010;93:2486-90.

Publication Dates

  • Publication in this collection
    Nov-Dec 2017

History

  • Received
    29 Dec 2016
  • Accepted
    18 Jan 2017
Sociedade Brasileira de Pediatria Av. Carlos Gomes, 328 cj. 304, 90480-000 Porto Alegre RS Brazil, Tel.: +55 51 3328-9520 - Porto Alegre - RS - Brazil
E-mail: jped@jped.com.br