Sunteți pe pagina 1din 12

Obesity, male infertility, and the

sperm epigenome
James R. Craig, M.D.,a,b Timothy G. Jenkins, Ph.D.,a Douglas T. Carrell, Ph.D., H.C.L.D.,a,c,d
and James M. Hotaling, M.D., M.S., F.E.C.S.M.a,b,c
a
Division of Urology, Department of Surgery, b Center for Men's Health and Reconstructive Surgery, and c Department of
Obstetrics and Gynecology, University of Utah; and d Department of Human Genetics, University of Utah School of
Medicine, Salt Lake City, Utah

Obesity is a growing epidemic and a common problem among reproductive-age men that can both cause and exacerbate male-factor
infertility by means of endocrine abnormalities, associated comorbidities, and direct effects on the delity and throughput of spermato-
genesis. Robust epidemiologic, clinical, genetic, epigenetic, and nonhuman animal data support these ndings. Recent works in the
burgeoning eld of epigenetics has demonstrated that paternal obesity can affect offspring metabolic and reproductive phenotypes
by means of epigenetic reprogramming of spermatogonial stem cells. Understanding the impact of this reprogramming is critical to
a comprehensive view of the impact of obesity on subsequent generations. Furthermore, and perhaps more importantly, conveying
the impact of these lifestyle changes on future progeny can serve as a powerful tool for obese men to modify their behavior. Reproduc-
tive urologists and endocrinologists must learn to assimilate these new ndings to better counsel men about the importance of paternal
preconception health, a topic recently being championed by the Centers for Disease Control and Prevention. (Fertil Steril 2017;107:
84859. 2017 by American Society for Reproductive Medicine.)
Key Words: Obesity, male infertility, sperm epigenetics, transgenerational inheritance
Discuss: You can discuss this article with its authors and with other ASRM members at https://www.fertstertdialog.com/users/
16110-fertility-and-sterility/posts/15227-23552

O
besity, dened as a body mass mellitus, sexual dysfunction, and sperm logic evidence was not available until
index (BMI) of >30 kg/m2, is epigenetic perturbations. In addition to a decade ago. In 2006, Sallmen et al.
a disease approaching the immediate effects that obesity has (11) described a group of 20,620 fam-
pandemic proportions, affecting more on the father, there is evidence that ilies in Iowa and North Carolina. They
than 1.9 billion adults over the age of negative effects may be transmitted to illustrated a dose-response relationship
18 years worldwide (1, 2) (Fig. 1). In the offspring via genetic and epigenetic between BMI and male infertility, with
the United States alone, the prevalence alterations of germ cell DNA (810). worsening male fertility for every 3-
of obese men who are of reproductive The objective of the present review is point increase in BMI >25 kg/m2,
age has tripled since the 1970s and to explore the epidemiology and patho- with an odds ratio (OR) of 1.12 (95%
currently affects 33.9% of the physiology of obesity-induced male condence interval [CI] 1.011.25).
population over the age of 20 years (3). infertility with an emphasis on the role These ndings were conrmed in later
The rise in obesity rates have paral- of epigenetics. studies across the globe, including in
leled reports of rising rates of Danish (12) and Norwegian (13) cohorts
poor sperm quality and male infertility that showed an association between
OBESITY AND MALE obesity and male infertility with
(4, 5). With the rate of male-related infer-
tility contributing to 45%50% of infer-
INFERTILITYTHE ORs of 1.53 (95% CI 1.321.77) and
tile couples (6, 7), there is an enlarging EPIDEMIOLOGIC 1.36 (95% CI 1.321.77), respectively
body of evidence linking male infertility ASSOCIATION (1219). A complete review of these
to obesity. Mechanisms by which obesity The negative effects of obesity on studies is presented in Table 1.
may affect spermatogenesis include semen parameters and androgen
thermal effects, hyperestrogenism, hypo- proles have been well established;
gonadotropic hypogonadism, diabetes however, population-based epidemio- OBESITY AND FERTILITY-
RELATED COMORBIDITIES
Received December 12, 2016; revised February 21, 2017; accepted February 25, 2017.
J.R.C. has nothing to disclose. T.G.J. has nothing to disclose. D.T.C. has nothing to disclose. J.M.H. has Obesity-related health decits include
nothing to disclose. increased risks of diabetes mellitus,
Reprint requests: James M. Hotaling, M.D., M.S., F.E.C.S.M., Department of Surgery (Urology), Univer-
sity of Utah, Suite 201, 675 Arapeen, Salt Lake City, UT 84108 (E-mail: jim.hotaling@hsc.utah.edu). cardiovascular disease, epigenetic
alterations, and certain malignancies
Fertility and Sterility Vol. 107, No. 4, April 2017 0015-0282/$36.00
Copyright 2017 American Society for Reproductive Medicine, Published by Elsevier Inc.
(2022). Obese men are also at greater
http://dx.doi.org/10.1016/j.fertnstert.2017.02.115 risk of developing hypogonadism,

848 VOL. 107 NO. 4 / APRIL 2017


Fertility and Sterility

FIGURE 1

Worldwide obesity incidence.


Craig. Obesity and sperm epigenetics. Fertil Steril 2017.

impaired spermatogenesis, and erectile dysfunction (2326). been shown to be required for normal spermatogenesis (40).
All of these factors are potential contributors to increased Thus, even a small decrease of the systemic testosterone
rates of male infertility in these patients. levels can reect a major reduction of the intratesticular
levels. GnRH is produced by the hypothalamus in a pulsatile
manner and stimulates LH and FSH. Normally, LH is
Obesity-Induced Endocrine Axis Derangements produced by the pituitary gland and acts to induce
Normal intratesticular testosterone levels are a prerequisite for steroidogenesis of testosterone by the Leydig cells. Once
normal spermatogenesis (27). Currently, our understanding of testosterone diffuses out of the Leydig cells, it is bound by
the hypothalamic-pituitary-gonadal (HPG) axis constitutes the proteins in circulation, mainly SHBG, and is then
core of our understanding of male reproduction. However, metabolized to estrogen by aromatase (41). FSH, while not
recent evidence has indicated that a number of neohor- strictly required for spermatogenesis in humans, does
mones, which include leptin (28, 29) and kisspeptin (30), augment Sertoli cell function, making it a core component
may also impact this axis. Furthermore, many of the of optimal testicular function (42).
comorbidities of obesity, such as diabetes (24) and sleep Hypogonadism in obesity can be mediated by both
apnea (3134), exacerbate these endocrine derangements. reduced pulse amplitude of the cyclical secretion of LH from
Below, we discuss in more detail the HPG axis, the effect of the pituitary as well as decreased response to LH by the
obesity on the HPG axis, and how these neohormones effect testis (25). Reductions in SHBG, FSH, and inhibin B and
these pathways. elevated E2, via increased aromatization of testosterone to
Testosterone levels measured within the testicle are found E2 peripherally, are also commonly seen (17, 4345). All of
to be 25125-fold greater than levels in serum (3539). The these changes result in reductions in the throughput and
physiologic need for elevated intratesticular levels is not delity of human spermatogenic function and, possibly,
completely understood; however, levels >70 nmol/L have alter the sperm epigenome (46).

VOL. 107 NO. 4 / APRIL 2017 849


850

VIEWS AND REVIEWS


TABLE 1

Epidemiologic studies on male infertility.


Study n Location Design Variables studied Association with male BMI
Studies on association between BMI and male infertility
Sallmen et al. 1,329 United States Secondary analysis of data from the Infertility (dened as not conceiving Adjusted for female BMI, male and female ages,
2006 (11) Agricultural Health Study on couples a pregnancy after R12 mo of smoking status, alcohol use, and exposure to
with a certied pesticide applicator unprotected intercourse) solvents and pesticides:
and spouse 1. OR for infertility 1.12 (95% CI 1.011.25).
2. Positive dose-effect relationship, with maximal
effect in the BMI 3243 kg/m2 group and a
plateau beyond that.
Ramlau-Hansen et al. 47,835 Denmark Prospective study: Danish Birth Cohort of Subfecundity (time to pregnancy) Adjusted for the partner's BMI:
2007 (12) couples from 1996 to 2002 1. OR 1.18 (95% CI 1.101.27) in overweight men.
2. OR 1.53 (95% CI 1.321.77) in obese men.
Nguyen et al. 26,303 Norway Retrospective study: Norwegian Mother Subfecundity (time to pregnancy) Adjusted for partner's BMI, coital frequency, and
2007 (13) and Child Cohort Study on pregnancies the ages and smoking habits of both partners:
from 1999 to 2005 1. OR 1.20 (95% CI 1.041.38) in overweight men.
2. OR 1.36 (95% CI 1.131.63) in obese men, with
a plateauing effect over a BMI of 32 kg/m2.
Stewart et al. 225 Australia Cross-sectional study. Initiated by WHO on 1. Time to pregnancy 1. Obesity was signicantly related to reduced total
2009 (14) male partners of women who conceived 2. Semen analysis sperm count (mean 324 vs. 231 million;
naturally 3. Hormone prole P< .05).
4. Physical exam to rule out local 2. Obese men (BMI >30 kg/m2) had signicantly
causes for infertility lower T, SHBG, and inhibin but not FSH.
Studies on association between BMI and sperm parameters  hormone prole
Jensen et al. 1,558 Denmark Prospective study on military recruits 1. Semen analysis 1. BMI >25 kg/m2 had a reduction in sperm
2004 (15) from 1996 to 1998 2. Hormone prole concentration of 21.6% (95% CI
3. Physical exam to rule out local 4.0%39.4%) and reduced total sperm count of
causes for infertility and to 23.9% (95% CI 4.7%43.2%). Nonsignicant
measure testis size reduction in sperm count. Testis size, semen
volume, and percentage of motile spermatozoa
were not affected by high BMI.
2. Decreased serum T, SHBG, and inhibin B. E2
demonstrated a positive relationship.
Kort et al. 520 United States Prospective study on normal Semen parameters, including sperm 1. Signicant decrease in sperm motility: normal
2006 (16) healthy men chromatin integrity (DNA BMI 18.6  106 cells; overweight 3.6 
fragmentation index [DFI]) 106 cells; obese 0.7  106 cells.
2. Signicant increase in DFI: normal BMI 19.9%
(1.96%); overweight 25.8% (2.23%); obese
27.0% (3.16%).
Craig. Obesity and sperm epigenetics. Fertil Steril 2017.
VOL. 107 NO. 4 / APRIL 2017
VOL. 107 NO. 4 / APRIL 2017

TABLE 1

Continued.
Study n Location Design Variables studied Association with male BMI
Aggerholm et al. 1,989 Europe Prospective study on men aged 1869 y. Data from 1. Semen analysis 1. Overweight group had slightly lower sperm
2008 (17) ve previous population-based environmental 2. Hormone prole concentration (56  106 per mL; 95% CI
studies of semen quality were combined 5260) compared with normal-BMI group
into one database. (59  106 per mL; 95% CI 5563) and lower
total sperm count (154  106; 95% CI
142168) compared with normal-BMI group
(168  106; 95% CI 157180). Obese men had
higher sperm density (68  106 per mL; 95% CI
5878) and total sperm count (190  106;
95% CI 161223) than the reference group of
normal-weight men. None of these differences
were statistically signicant.
2. Decreased T and inhibin (25%32% lower) in
obese men. E2 concentration 6% higher in
obese men.
Martini et al. 794 Argentina Prospective study on male partners of infertile 1. Semen analysis 1. Decreased sperm motility in the high-BMI group.
2010 (18) couples from 2006 to 2007 2. Seminal hormone and Percentage of sperm motility: normal BMI
biochemical prole 51.4% (95% CI 50.2%52.6%); overweight
50.2% (95% CI 49.2%51.2%); obese 46.6%
(95% CI 44.9%48.3%).
2. No association between BMI and sperm
concentration.
3. No signicant differences were detected in
seminal T levels between groups.
Bieniek et al. 4,440 North America Prospective multicenter study from 2002 to 2014 1. Semen analysis 1. BMI had weak but signicant negative
2016 (19) 2. Hormone prole correlations with ejaculate volume (r 0.04),
sperm concentration (r 0.08), motility
(r 0.07), and morphology (r 0.04).
2. Rates of azoospermia and oligospermia were
also more prevalent among obese (12.7% and
31.7%, respectively) compared with normal-
weight (9.8% and 24.5%) men.
3. Testosterone had a signicant negative
correlation, with BMI. E2 demonstrated a
positive relationship.
4. Neither FSH nor LH demonstrated signicant
correlations with BMI.
Note: BMI body mass index; CI condence interval; OR odds ratio; WHO World Health Organization.

Fertility and Sterility


Craig. Obesity and sperm epigenetics. Fertil Steril 2017.
851
VIEWS AND REVIEWS

FIGURE 2

Obesity and male infertility: mechanisms. Obesity operates through multiple pathways to alter male reproductive potential. It creates epigenetic
changes, some of which can inuence subsequent generations. In addition, it alters the male androgenic axis, inuences a host of other
neohormones, and raises insulin levels. Finally, it has been linked to erectile dysfunction and causes stress, inammation and sleep apnea, all
of which can further reduce male fertility. Kp kisspeptin.
Craig. Obesity and sperm epigenetics. Fertil Steril 2017.

In addition to perturbations by these standard levels of E2 and estrone down-regulate the release of kisspep-
pathways, obesity is associated with dysregulation of a number tin from KISS neurons and decrease the activity of the HPG
of neohormones which act to further disrupt the male axis (48, 49). In addition to aromatization of testosterone,
endocrine axis (2830). Under normal physiologic control, adipocytes also produce a hormone called leptin, a critical
adipocytes release leptin, increasing release of kisspeptin factor in regulating energy homeostasis (50). Mutations in
from KISS neurons. This acts to increase the release of GnRH the leptin gene or leptin receptor leads to increased food
from the hypothalamus, elevating FSH and LH secretion from intake and decreased energy expenditure. However, in obese
the anterior pituitary, which in turn increases testicular individuals without mutations in the leptin gene or receptor,
testosterone biosynthesis. Obesity induces changes via leptin levels are chronically elevated, leading to leptin
multiple pathways in the HPG axis that lead to resistance (51). This resistance is seen centrally in the
hypogonadotropic hyperestrogenic hypoandrogenism (Fig. 2). hypothalamus, where leptin is unable to stimulate the HPG
Obese men have increased adipocyte-related aromatiza- axis in these leptin-resistant men, leading to decreased testos-
tion of testosterone into E2 and estrone (47). These increased terone levels (28, 29).

852 VOL. 107 NO. 4 / APRIL 2017


Fertility and Sterility

Comorbidities in Obesity and Their Impact on the Sperm Parameters


Androgenic Axis The impact of male obesity on the androgenic axis is well
Diabetes is associated with obesity and can negatively understood. However, unlike the impact of obesity on ovula-
affect the androgenic axis. The prevalence of diabetes in tion and the oocyte, the impact of obesity on sperm parameters
the obese population is signicant, with 3.4% of the is complex and likely multifactorial (16, 66). Although
American population affected by both conditions (52). derangements of the androgenic axis and epigenetic changes
The obese man with diabetes exhibits both central and in spermatogonial stem cells are thought to play a role, the
peripheral insulin resistance, leading to reduction of exact cause remains elusive.
SHBG synthesis by the liver. The reduction of SHBG Obesity drives changes in bulk seminal parameters and,
allows for a greater fraction of testosterone to remain perhaps more importantly, sperm DNA packaging and epige-
free, which magnies the negative feedback effect of E2 netics (46). Observational studies demonstrate that over-
through aromatization (52). Thus, obese men with diabetes weight (BMI >25 kg/m2) and obese (BMI >30 kg/m2) men
have further down-regulation of the androgenic axis. are at higher risk of oligozoospermia (count <20  106/mL)
Although data are conicting, generally fecundity rates (15, 66, 67), with an OR of 3.3 (95% CI 1.199.14) (66).
and semen quality have been shown to be decreased in Obesity affects not only count, but also total progressive
men with diabetes (53). motile count, with obese men being at greater risk for
Sleep apnea affects 4% of adult men, with two-thirds of total progressive motile counts <10  106, with an OR of
those men being obese. Sleep apnea, common in obese men, 3.4 (95% CI 1.1210.60) (66). DNA fragmentation index
can down-regulate testosterone. The nocturnal apneic events (DFI) in obese men ranges from 21.9% to 27% compared
characteristic of sleep apnea lead to sleep fragmentation. This, with 15%19.9% in men with normal body weight. Obese
in turn, decreases nocturnal LH production, further reducing men also have elevated risk of having signicantly elevated
circulating testosterone levels (31, 54). Furthermore, DFI, with an OR of 2.5 (95% CI 1.25.1) (16, 68).
nonhuman animal models demonstrate that intermittent Despite the associations between obesity and semen
hypoxia can affect gene expression, sperm motility, and parameters, it is clear that men with high levels of body fat
fertility (55). are, in general, still capable of siring offspring. This fact high-
Obesity-related accumulation of toxic substances, lights an important problem. Specically, the fact that sperm
particularly endocrine disruptors, is thought to disrupt are competent to fertilize an egg does not guarantee that these
both the male endocrine axis and perturb spermatogenesis sperm are entirely unaffected in obese men or that normal
(56, 57). This is compounded by the fat solubility of many embryonic development will progress uninhibited after fertil-
of these toxins, making obese men more susceptible to ization. In fact, clear data indicate that epigenetic patterns in
their effect (56, 57). sperm, described in more detail below, are affected in obese
Impairment of scrotal and testicular thermoregulatory men and include differences in sperm microRNA content
mechanisms by excess adipose tissue leads to impaired and DNA methylation variations (46, 69). These important
spermatogenesis. Obese men have increased lower abdom- markers in sperm, when altered, are thought to be
inal fat, thigh fat, and scrotal fat that function as an implicated in abnormal phenotypes in the offspring and
insulator for the testicles, leading to increased testicular perturbations in embryogenesis. Therefore, even if fertility
temperature. This negatively impacts both testosterone issues can be overcome in obese patients, additional
synthesis and spermatogenesis (58, 59). consideration should be given to potentially unt gametes
resulting from exposure to the testicular environments seen
in obese men.
Sexual Dysfunction
The ability to sustain an erection rigid enough for penetration MALE OBESITY AND ADVANCED
is a prerequisite for natural conception. Unfortunately, REPRODUCTIVE TECHNOLOGY OUTCOMES
impaired coitus due to erectile dysfunction is commonly Although the male contribution to assisted reproductive tech-
seen in infertile men (60). Compared with fertile control nology (ART) success has historically been minimized, it is
subjects, men with infertility have signicantly worse Sexual now well accepted that sperm have a signicant effect (70).
Health Inventory for Men scores, with 36% reporting a score Male obesity, similarly to female obesity, appears to affect clin-
of <22 (60). In addition, 79% of men who report having ical pregnancy, miscarriage, and live birth rates (71, 72).
erectile dysfunction are obese (61). The relationship between Campbell et al. have shown that obese men undergoing
obesity and erectile dysfunction is multifactorial, but is ART have a statistically signicant decrease in live birth
thought to stem from decreased testosterone levels and rate compared with normal-weight men: OR 0.65 (95% CI
elevated proinammatory factors (tumor necrosis factor a 0.440.97) (72). The odds of having a nonviable pregnancy
and interleukin 6) (62, 63). These inammatory substances are signicantly greater for couples with an obese male
lead to vascular endothelial dysfunction, resulting in partner compared with a normal-weight partner: OR 2.87
erectile dysfunction. Testosterone deciency can lead to (95% CI 1.346.13), with an absolute risk difference of 10%.
sexual dysfunction in a myriad of ways, mainly through Another study has reported lower pregnancy rate in males
decreased libido and lack of androgenic augmentation of undergoing IVF, but no signicant difference if ICSI was used
cavernosal smooth muscle function (64, 65). for ART therapy. In contrast, Thomsen et al. have reported no

VOL. 107 NO. 4 / APRIL 2017 853


VIEWS AND REVIEWS

decrease in semen quality or IVF outcome in obese men under- promoter regions to transcriptional machinery. This, in
going ART (73). turn, can inhibit gene expression. Nuclear proteins,
These studies still leave much unknown about the exact specically histones, can be post-translationally modied
impact of male obesity on ART outcomes. Answering these by means of methylation, acetylation, phosphorylation,
questions will require well controlled studies. For example, and others. These modications promote chromatin
the age and obesity of the female partner may well modulate compaction and control the access of transcription factors
the effects of sperm defects, both independently from and to DNA (86). In addition, multiple RNA species (mRNA,
depending on the male factors. For example, lower maternal microRNA [miRNA], Piwi-interacting RNA [piRNA], etc.)
age results in improved oocyte quality, which may indepen- can directly and indirectly affect a cell's expression prole.
dently improve outcomes but may also provide an improved Small regulatory RNAs are noncoding RNAs that regulate
opportunity for correction of some defects, such as sperm gene expression at the transcriptional or post-
epigenetic abnormalities, after fertilization (74, 75). transcriptional step, or via chromatin remodeling (87).
Because of the epigenome's role in transcription regula-
tion, epigenetic alterations can drive many abnormal
MECHANISMS BEHIND INFERTILITY IN THE fertility phenotypes, and these perturbations have been
OBESE MAN: HORMONAL, INFLAMMATION, implicated in multiple cases in humans. A study by Sonnack
GENETIC, EPIGENETIC et al. showed that men exhibiting qualitative or quantitative
The etiology of obesity-driven male infertility is multifacto- infertility have signicantly decreased levels of histone H4
rial, with many likely contributing factors, including acetylation associated with impaired spermatogenesis (88).
hormonal perturbations and potentially reversible epigenetic Furthermore, it has been established that human spermato-
alterations (Fig. 2). It is important to emphasize that each of zoa is composed of miRNAs (7%) piRNAs (17%), and
the mechanisms described as potential factors of altered repeat-associated small RNAs (65%) (89), and some of these
spermatogenesis may have isolated or interdependent actions have been implicated in the process of infertility. Addition-
on fertility. For example, epigenetic alterations in sperm, ally, obesity-associated comorbidities, including inamma-
affecting embryogenesis and health of offspring, may be tion, glucose intolerance, stress, and hypercholesterolemia,
induced by a number of factors, including endocrine alter- were good predictors for sperm miRNA abundance and
ations, thermal effects, and gene expression pathways result- offspring phenotypes (90). That study also showed that
ing from sleep apnea, stress, etc. Therefore, although isolated alteration in preconception diet or exercise interventions
studies of individual factors may point to important features in obese fathers resulted in normalization of miRNA proles.
of the syndrome, well designed systems analyses are crucial to Although the impact of these alterations on sperm function
understand the complete scope of the problem. is unclear, it is important to note that multiple studies have
shown associations between sperm DNA methylation alter-
ations and various semen analysis parameters. This is most
ADIPOSITY-INDUCED EPIGENETIC CHANGES pronounced with sperm motility, count, and viability (91).
IN SPERM Therefore, it is not unreasonable to assume that at least a
Epigenetic marks provide a mechanism by which environ- portion of these obesity-induced alterations have the poten-
mental factors can inuence our cells by leaving biochemical tial to affect normal sperm function and, in turn, fertility. In
imprints on our genome capable of gene regulation in the the context of obesity specically, multiple studies have
absence of mutations to DNA sequence. This modiable been performed. In brief, it has been shown that sperm
nature makes epigenetic signatures very interesting epigenetic patterns are altered in individuals with high
candidates to explain obesity-associated alterations to BMIs (46, 92). Importantly, these alterations do appear to
fertility (810, 7679). A complete review of these studies is be, at least to some degree, reversible.
presented in Table 2. Epigenetic signatures include three Although it appears from studies that perturbations to
major categories: DNA methylation, nuclear protein the sperm epigenome can drive alterations to normal
composition (with specic consideration of histone gamete function, it is important to consider the down-
modications and localization), and expression proles of stream implications. Because the sperm has two major
noncoding RNAs (80). In short, these marks drive (or, at a functional rolessafe delivery of the paternal DNA blue-
minimum, inuence) gene expression patterns in the cell. print to the oocyte and competence to contribute to
Plasticity is a hallmark of epigenetic marks in both somatic embryogenesis and offspring developmentit is important
tissues and the germ line. Epigenetic signatures can be to assess the impact of sperm epigenetic alterations on
readily altered by various modiers, including, but not both. Recent studies have shown that epigenetic marks in
limited to, diet, BMI, activity level, aging, and exposure to sperm are unique and are at regions important in embry-
various toxins and environmental agents (46, 81). onic development (93). Other studies have suggested that
The epigenome helps to ensure proper regulation of various individual miRNA species (most notably the
transcription patterns in all cell types by means of multiple mir34 C family) and collective RNA proles (RNA
mechanisms. DNA methylation has been shown in multiple elements) play a role in normal embryogenesis (94).
studies to be important in gene regulation (8285). Furthermore, in very recent studies, sperm DNA methyl-
Specically, when enriched at gene promoters, DNA ation patterns have been tightly linked to male infertility,
methylation has been shown to inhibit the accessibility of embryo quality, and fecundity (95, 96).

854 VOL. 107 NO. 4 / APRIL 2017


Fertility and Sterility

TABLE 2

Studies on paternal obesity and trans generational inheritance.


Study subjects and
Study Hypothesis tested methodology Results
Ng et al. 2010 (62) Paternal chronic high-fat diet Expression of pancreatic islet-cell Paternal HFD causes progressive
(HFD) causes beta-cell genes in female offspring of early onset of impaired
dysfunction in the female obese male rats insulin secretion and glucose
offspring. tolerance, but offspring had
normal adiposity.
Fullston et al. 2012 (63) Impact of diet-induced paternal Transgenerational assessment of HFD induces paternal initiation
obesity on the reproductive reproductive health by of subfertility in both male
health of two subsequent mating F0 and then F1 mice and female offspring of two
generations. to CD (control diet) mice generations of mice.
Fullston et al. 2013 (8) Impact of diet-induced paternal Transgenerational assessment of Diet-induced paternal obesity
obesity on offspring metabolic health by mating modulates sperm microRNA
metabolic, testicular F0 and then F1 mice to CD content and germ cell
transcriptome, and sperm mice methylation status,
microRNA prole. programming offspring
health and propagating
obesity and impaired
metabolic health to future
generations.
Soubry et al. 2013 (10) Association between obesity DNA from the umbilical cord of Paternal obesity is associated
and DNA methylation 79 newborns whose with insulin-like growth
proles in the offspring. mothers had been enrolled factor 2 hypomethylation.
in the Newborn Epigenetics
Study
McPherson et al. 2014 (9) Dietary and exercise regimes in Male mice were fed an HFD and Sperm parameters were
obese men improve the randomized to control or diet improved in the offspring of
reproductive health of and exercise regimen for obese male rats that were
subsequent generations. 9 wk, with analysis of allocated to diet  exercise.
offspring sperm health
Ng et al. 2014 (64) Paternal high-fat diet Transcriptome alterations in islet Paternal HFD consumption
consumption in rats alters cells of female offspring of triggers unique gene
the transcriptomes of obese male rats signatures consistent with
retroperitoneal adipose and premature aging and chronic
pancreatic islet tissues of degenerative disorders.
female offspring.
Masuyuma et al. 2016 (65) Paternal HFD-induced obesity Effects of paternal HFD over After paternal HFD exposure,
affects the metabolic status multiple generations with resumption of normal diet by
of offspring through examination of offspring subsequent generations
epigenetic changes in metabolic prole, including diminished and ultimately
adiponectin and leptin weight and fat gain, glucose abolished the impact of
genes. intolerance, paternal HFD on offspring
hypertriglyceridemia, metabolic prole and
abnormal adipocytokine adipocytokine promoter
levels, hypertension, epigenetic signatures initially
adiponectin and leptin gene seen in the offspring.
expression, and epigenetic
changes
Craig. Obesity and sperm epigenetics. Fertil Steril 2017.

TRANSGENERATIONAL AND a man's life (or even that were induced in utero) from the
INTERGENERATIONAL INHERITANCE OF father to the child only (typically from F0 to F1). Whereas
OBESITY the inheritance of an altered epigenetic state to the grand-
offspring and beyond (F2 or beyond) is termed transgenera-
Transgenerational and/or intergenerational inheritance tional inheritance. Based on epidemiologic evidence, many
occurs when induced epigenetic changes in the gametes are types of environmental challenges imposed on the parent,
passed on to the subsequent generation (97). This epigenetic such as hunger, specic diets, toxins, and trauma, have
inheritance can theoretically result in phenotypic alterations been found to inuence the development of the offspring
in the offspring, the grand-offspring, and beyond. When (98). Paternal age, subfertility, smoking, obesity, and
considering transmission of epigenetic signatures for the exposure to a range of environmental inuences, including
paternal gamete, it is important to understand the difference air pollution, radiofrequency electromagnetic radiation, and
between intergenerational and transgenerational inheritance xenobiotics, have also been implicated (99).
patterns. In effect, intergenerational epigenetic inheritance The strongest epidemiologic association of intergener-
includes any sperm epigenetic signatures altered throughout ational inheritance is recorded by the Dutch famine study,

VOL. 107 NO. 4 / APRIL 2017 855


VIEWS AND REVIEWS

which, among other items, assessed the outcomes of severe Pharmacotherapy


caloric restriction and poor nutrition in the offspring of in- The goal of medical management of infertility in the obese
dividuals exposed to famine conditions. One particular man is to optimize androgenic hormonal pathways. Typical
analysis assessed a cohort of 300,000 young men born manifestations of obesity include low testosterone, high E2,
around the time of Dutch famine of 1944. Their mothers or a combination of both (17, 4345). Clomiphene citrate, a
had been exposed to caloric rations of <1,000 calories selective estrogen receptor modulator, is used to augment
per day during their pregnancies. Their offspring exhibited endogenous testosterone production via elevation of LH and
a signicant increase in glucose intolerance, coronary spermatogenesis via elevation of FSH (110). Aromatase
heart disease, increased stress responsiveness and inhibitors, such as anastrozole or letrozole, are used to
obesity. The association was more signicant when the block the conversion of testosterone to E2 by aromatase,
mother took the very low calorie diet during early gesta- resulting in restoration of the optimal testosterone to E2
tion (100, 101). Another well known epidemiologic study, ratio of >10:1 (111, 112).
in the United Kingdom by Barker et al., established an
association between intrauterine growth restriction and
subsequent cardiovascular morbidity and mortality in the Gastric Bypass Surgeries
offspring (102). The American Society for Metabolic and Bariatric Surgery
Obesity has been shown to directly drive transgenera- recommends bariatric surgery for a person with BMI
tional effects in successive generations. Studies with the use R40 kg/m2, >100 pounds overweight, or with BMI
of nonhuman animal models have found altered metabolic R35 kg/m2 and at least two obesity-related comorbidities
processing in offspring sired by obese fathers (103). Further (113). In obese men who undergo bariatric surgery, gastric
studies in mice have shown similar patterns of offspring bypass and banding operations are proven to be highly suc-
metabolic perturbations but have also identied specic cessful in treating morbid obesity, reducing obesity-related
epigenetic alterations in the sperm (8). In humans, an comorbidities, correcting hormonal balance, and improving
intriguing recent study found alterations in DNA methylation sexual function (114116). Azoospermic men undergoing
at the insulin-like growth factor 2 gene in newborns sired by gastric bypass demonstrate increased serum testosterone
obese fathers (10). Taken together, these data strongly suggest and sperm count after bariatric surgeryinduced weight loss
that obesity is capable of affecting sperm function, embryo- (117), but studies about sperm motility, morphology, and
genesis, and even offspring health. DNA fragmentation show mixed results (114). Sudden
severe weight loss associated with bariatric surgery may
STRATEGIES TO COUNTER OBESITY result in a negative effect on spermatogenesis and initially
result in oligoasthenozoospermia (118). The exact impact of
Although the full scope of treatment of obesity as it relates preconception paternal extreme weight loss on offspring
to male infertility is the beyond the scope of this review, we remains unknown, but much can be inferred from
think that it is important to highlight a few of the basic steps nonhuman animal studies demonstrating improved
that can be taken to improve the reproductive prole of epigenetic proles in metabolic genes. For example, a
these men. Perhaps the most important point to remember comparison of children born before and after maternal
is to council these men that their lifestyle choices now gastrointestinal bypass surgery found that genes were
have consequences for the entire life course trajectory of differentially methylated in these two groups. The children
their progeny. born after gastric bypass surgery had markedly improved
epigenetic proles of receptor signaling in insulin, leptin,
Diet and Exercise and glucoregulatory pathways that were seen up to 18 years
after conception (119).
Traditionally, men seek health care at lower rates than
women and are less likely to have a primary care provider
(Summary Health Statistics: National Health Interview Sur- CONCLUSION
vey, 2014). When men do seek health care, it is typically for Male obesity is a signicant problem that has grown to
a specic health concern, such as infertility (104). Therefore, epidemic proportions. Obese men are more likely to be infer-
an infertility evaluation may represent a unique opportunity tile. Although the cause is not exactly known, it is likely a
to improve a man's health, and, by extension, the health of combination of obesity-induced endocrine derangements
his progeny. Despite seeking health care at lower rates than and its deleterious effects on spermatogenesis. Recent epige-
women, men are actually slightly more likely to lose netic evidence suggests that the metabolic memory of a
weight through diet, exercise, and behavioral modication man's obesity at the time of conception may be directly
(105, 106). Infertility and the burgeoning recognition of transmitted to his offspring through his sperm and thus
the effect of paternal lifestyle on offspring health provides may have major consequences for their somatic health.
a powerful motivator for many men to lose weight. Once Improved understanding of intergenerational inheritance
achieved, weight loss has been shown to improve and epigenetics is crucial to both understanding these mech-
testosterone levels, SHBG, erectile dysfunction, sperm anisms and using them as a powerful tool for preconception
motility, sperm mitochondrial function, and epigenetic behavioral change in men. Just as it is now well established
prole (9, 107109). that morbidly obese women have higher chances of

856 VOL. 107 NO. 4 / APRIL 2017


Fertility and Sterility

complications during ART and of higher-risk pregnancies, it 20. Twig G, Yaniv G, Levine H, Leiba A, Goldberger N, Derazne E, et al. Body-
may become established that obese men father children who mass index in 2.3 million adolescents and cardiovascular death in adult-
hood. N Engl J Med 2016;374:243040.
are also at higher risk of metabolic derangements. It is prob-
21. Lauby-Secretan B, Scoccianti C, Loomis D, Grosse Y, Bianchini F, Straif K,
able that we may, one day in the not too distant future, et al. Body fatness and cancerviewpoint of the IARC Working Group.
establish protocols focusing on male weight loss before N Engl J Med 2016;375:7948.
conception to improve offspring health. 22. Ozanne SE. Epigenetic signatures of obesity. N Engl J Med 2015;372:9734.
23. Lamm S, Chidakel A, Bansal R. Obesity and Hypogonadism. Urol Clin North
Am 2016;43:23945.
REFERENCES 24. Dandona P, Dhindsa S. Update: hypogonadotropic hypogonadism in type
1. American Medical Association. Recognition of obesity as a disease. 2 diabetes and obesity. J Clin Endocrinol Metab 2011;96:264351.
H-440.842. In: Public Health Policy. Annual Meeting, 2013. Available at: 25. Vermeulen A, Kaufman JM, Deslypere JP, Thomas G. Attenuated luteinizing
https://searchpf.ama-assn.org/SearchML/searchDetails.action?uri=%2FA hormone (LH) pulse amplitude but normal LH pulse frequency, and its
MADoc%2FHOD.xml-0-3858.xml#. Last accesed March 12, 2017. relation to plasma androgens in hypogonadism of obese men. J Clin
2. World Health Organization. Obesity and overweight. Fact Sheet. 2016. Endocrinol Metab 1993;76:11406.
Available at: http://www.who.int/mediacentre/factsheets/fs311/en/. Last 26. MacDonald AA, Herbison GP, Showell M, Farquhar CM. The impact of
accessed March 12, 2017. body mass index on semen parameters and reproductive hormones in
3. National Center for Health Statistics. Health, United States, 2008, with human males: a systematic review with meta-analysis. Hum Reprod Update
chartbook. Hyattsville, MD: National Center for Health Statistics; 2009. 2010;16:293311.
4. Katib A. Mechanisms linking obesity to male infertility. Cent Eur J Urol 27. OShaughnessy PJ, Verhoeven G, de Gendt K, Monteiro A, Abel MH. Direct
2015;68:7985. action through the Sertoli cells is essential for androgen stimulation of
5. Swan SH, Elkin EP, Fenster L. The question of declining sperm density spermatogenesis. Endocrinology 2010;151:23438.
revisited: an analysis of 101 studies published 19341996. Environ Health 28. Munzberg H, Myers MG Jr. Molecular and anatomical determinants of
Perspect 2000;108:9616. central leptin resistance. Nat Neurosci 2005;8:56670.
6. Rowe PJ, World Health Organization. WHO manual for the standardized 29. Farooqi IS, ORahilly S. Leptin: a pivotal regulator of human energy
investigation, diagnosis, and management of the infertile male. Cambridge, homeostasis. Am J Clin Nutr 2009;89:980S4S.
UK: Cambridge University Press; 2000. 30. Clarke H, Dhillo WS, Jayasena CN. Comprehensive review on kisspeptin and
7. Lamb DJ, Lipshultz LI. Male infertility: recent advances and a look toward its role in reproductive disorders. Endocrinol Metab (Seoul) 2015;30:12441.
the future. Curr Opin Urol 2000;10:35962. 31. Luboshitzky R, Lavie L, Shen-Orr Z, Herer P. Altered luteinizing hormone
8. Fullston T, Ohlsson Teague EM, Palmer NO, DeBlasio MJ, Mitchell M, and testosterone secretion in middle-aged obese men with obstructive
Corbett M, et al. Paternal obesity initiates metabolic disturbances in two sleep apnea. Obes Res 2005;13:7806.
generations of mice with incomplete penetrance to the F2 generation 32. Axelsson J, Ingre M, Akerstedt T, Holmback U. Effects of acutely displaced
and alters the transcriptional prole of testis and sperm microRNA content. sleep on testosterone. J Clin Endocrinol Metab 2005;90:45305.
FASEB J 2013;27:422643. 33. Boyar RM, Rosenfeld RS, Kapen S, Finkelstein JW, Roffwarg HP,
9. McPherson NO, Fullston T, Bakos HW, Setchell BP, Lane M. Obese fathers Weitzman ED, et al. Human puberty. Simultaneous augmented secretion
metabolic state, adiposity, and reproductive capacity indicate sons of luteinizing hormone and testosterone during sleep. J Clin Invest 1974;
reproductive health. Fertil Steril 2014;101:86573. 54:60918.
10. Soubry A, Schildkraut JM, Murtha A, Wang F, Huang Z, Bernal A, et al. 34. Schiavi RC, White D, Mandeli J. Pituitary-gonadal function during sleep in
Paternal obesity is associated with IGF2 hypomethylation in newborns: healthy aging men. Psychoneuroendocrinology 1992;17:599609.
results from a Newborn Epigenetics Study (NEST) cohort. BMC Med 35. Comhaire FH, Vermeulen A. Testosterone concentration in the uids of
2013;11:29. seminiferous tubules, the interstitium and the rete testis of the rat. J
11. Sallmen M, Sandler DP, Hoppin JA, Blair A, Baird DD. Reduced fertility Endocrinol 1976;70:22935.
among overweight and obese men. Epidemiology 2006;17:5203. 36. Turner TT, Jones CE, Howards SS, Ewing LL, Zegeye B, Gunsalus GL. On the
12. Ramlau-Hansen CH, Thulstrup AM, Nohr EA, Bonde JP, Sorensen TI, androgen microenvironment of maturing spermatozoa. Endocrinology
Olsen J. Subfecundity in overweight and obese couples. Hum Reprod 1984;115:192532.
2007;22:16347. 37. Awoniyi CA, Santulli R, Sprando RL, Ewing LL, Zirkin BR. Restoration of
13. Nguyen RH, Wilcox AJ, Skjaerven R, Baird DD. Mens body mass index and advanced spermatogenic cells in the experimentally regressed rat testis:
infertility. Hum Reprod 2007;22:248893. quantitative relationship to testosterone concentration within the testis.
14. Stewart TM, Liu DY, Garrett C, Brown EH, Baker HW. Recruitment bias in Endocrinology 1989;124:121723.
studies of semen and other factors affecting pregnancy rates in fertile men. 38. Maddocks S, Hargreave TB, Reddie K, Fraser HM, Kerr JB, Sharpe RM. Intrates-
Hum Reprod 2009;24:24018. ticular hormone levels and the route of secretion of hormones from the testis
15. Jensen TK, Andersson AM, Jorgensen N, Andersen AG, Carlsen E, of the rat, guinea pig, monkey and human. Int J Androl 1993;16:2728.
Petersen JH, et al. Body mass index in relation to semen quality and repro- 39. Jarow JP, Chen H, Rosner TW, Trentacoste S, Zirkin BR. Assessment of the
ductive hormones among 1,558 Danish men. Fertil Steril 2004;82:86370. androgen environment within the human testis: minimally invasive method
16. Kort HI, Massey JB, Elsner CW, Mitchell-Leef D, Shapiro DB, Witt MA, et al. to obtain intratesticular uid. J Androl 2001;22:6405.
Impact of body mass index values on sperm quantity and quality. J Androl 40. Zirkin BR, Santulli R, Awoniyi CA, Ewing LL. Maintenance of advanced sper-
2006;27:4502. matogenic cells in the adult rat testis: quantitative relationship to testos-
17. Aggerholm AS, Thulstrup AM, Toft G, Ramlau-Hansen CH, Bonde JP. Is terone concentration within the testis. Endocrinology 1989;124:30439.
overweight a risk factor for reduced semen quality and altered serum sex 41. Kim HH, Schlegel PN. Endocrine manipulation in male infertility. Urol Clin
hormone prole? Fertil Steril 2008;90:61926. North Am 2008;35:30318.
18. Martini AC, Tissera A, Estofan D, Molina RI, Mangeaud A, de Cuneo MF, 42. Levallet J, Pakarinen P, Huhtaniemi IT. Follicle-stimulating hormone ligand
et al. Overweight and seminal quality: a study of 794 patients. Fertil Steril and receptor mutations, and gonadal dysfunction. Arch Med Res 1999;30:
2010;94:173943. 48694.
19. Bieniek JM, Kashanian JA, Deibert CM, Grober ED, Lo KC, Brannigan RE, 43. Ramlau-Hansen CH, Hansen M, Jensen CR, Olsen J, Bonde JP,
et al. Inuence of increasing body mass index on semen and reproductive Thulstrup AM. Semen quality and reproductive hormones according to
hormonal parameters in a multi-institutional cohort of subfertile men. Fertil birthweight and body mass index in childhood and adult life: two decades
Steril 2016;106:10705. of follow-up. Fertil Steril 2010;94:6108.

VOL. 107 NO. 4 / APRIL 2017 857


VIEWS AND REVIEWS

44. Paasch U, Grunewald S, Kratzsch J, Glander HJ. Obesity and age affect 67. Alshahrani S, Ahmed AF, Gabr AH, Abalhassan M, Ahmad G. The impact
male fertility potential. Fertil Steril 2010;94:2898901. of body mass index on semen parameters in infertile men. Andrologia
45. Chavarro JE, Toth TL, Wright DL, Meeker JD, Hauser R. Body mass index in 2016;48:11259.
relation to semen quality, sperm DNA integrity, and serum reproductive 68. Dupont C, Faure C, Sermondade N, Boubaya M, Eustache F, Clement P,
hormone levels among men attending an infertility clinic. Fertil Steril et al. Obesity leads to higher risk of sperm DNA damage in infertile
2010;93:222231. patients. Asian J Androl 2013;15:6225.
46. Soubry A, Guo L, Huang Z, Hoyo C, Romanus S, Price T, et al. Obesity- 69. Fullston T, Ohlsson-Teague EM, Print CG, Sandeman LY, Lane M. Sperm
related DNA methylation at imprinted genes in human sperm: results microRNA content is altered in a mouse model of male obesity, but the
from the TIEGER study. Clin Epigenetics 2016;8:51. same suite of microRNAs are not altered in offsprings sperm. PLoS One
47. Kley HK, Deselaers T, Peerenboom H, Kruskemper HL. Enhanced conver- 2016;11:e0166076.
sion of androstenedione to estrogens in obese males. J Clin Endocrinol 70. Lewis SE, Kumar K. The paternal genome and the health of the assisted
Metab 1980;51:112832. reproductive technology child. Asian J Androl 2015;17:61622.
48. Gautier A, Bonnet F, Dubois S, Massart C, Grosheny C, Bachelot A, et al. 71. Provost MP, Acharya KS, Acharya CR, Yeh JS, Steward RG, Eaton JL, et al. Preg-
Associations between visceral adipose tissue, inammation and sex steroid nancy outcomes decline with increasing body mass index: analysis of 239,127
concentrations in men. Clin Endocrinol (Oxf) 2013;78:3738. fresh autologous in vitro fertilization cycles from the 20082010 Society for
49. George JT, Millar RP, Anderson RA. Hypothesis: kisspeptin mediates male Assisted Reproductive Technology registry. Fertil Steril 2016;105:6639.
hypogonadism in obesity and type 2 diabetes. Neuroendocrinology 2010; 72. Campbell JM, Lane M, Owens JA, Bakos HW. Paternal obesity negatively
91:3027. affects male fertility and assisted reproduction outcomes: a systematic re-
50. Brennan AM, Mantzoros CS. Drug insight: the role of leptin in human view and meta-analysis. Reprod Biomed Online 2015;31:593604.
physiology and pathophysiologyemerging clinical applications. Nat Clin 73. Thomsen L, Humaidan P, Bungum L, Bungum M. The impact of male over-
Pract Endocrinol Metab 2006;2:31827. weight on semen quality and outcome of assisted reproduction. Asian J
51. Considine RV, Considine EL, Williams CJ, Nyce MR, Magosin SA, Bauer TL, Androl 2014;16:74954.
et al. Evidence against either a premature stop codon or the absence of 74. Jenkins TG, Carrell DT. The sperm epigenome and potential implications
obese gene mRNA in human obesity. J Clin Invest 1995;95:29868. for the developing embryo. Reproduction 2012;143:72734.
52. Mokdad AH, Ford ES, Bowman BA, Dietz WH, Vinicor F, Bales VS, et al. 75. Gannon JR, Emery BR, Jenkins TG, Carrell DT. The sperm epigenome:
Prevalence of obesity, diabetes, and obesity-related health risk factors, implications for the embryo. Adv Exp Med Biol 2014;791:5366.
2001. JAMA 2003;289:769. 76. Ng SF, Lin RC, Laybutt DR, Barres R, Owens JA, Morris MJ. Chronic high-fat
53. Eisenberg ML, Sundaram R, Maisog J, Buck Louis GM. Diabetes, medical diet in fathers programs beta-cell dysfunction in female rat offspring.
comorbidities and couple fecundity. Hum Reprod 2016;31:236976. Nature 2010;467:9636.
54. Luboshitzky R, Zabari Z, Shen-Orr Z, Herer P, Lavie P. Disruption of the 77. Fullston T, Palmer NO, Owens JA, Mitchell M, Bakos HW, Lane M. Diet-
nocturnal testosterone rhythm by sleep fragmentation in normal men. J induced paternal obesity in the absence of diabetes diminishes the
Clin Endocrinol Metab 2001;86:11349. reproductive health of two subsequent generations of mice. Hum Reprod
55. Torres M, Laguna-Barraza R, Dalmases M, Calle A, Pericuesta E, 2012;27:1391400.
Montserrat JM, et al. Male fertility is reduced by chronic intermittent 78. Ng SF, Lin RC, Maloney CA, Youngson NA, Owens JA, Morris MJ. Paternal
hypoxia mimicking sleep apnea in mice. Sleep 2014;37:175765. high-fat diet consumption induces common changes in the transcriptomes
56. Magnusdottir EV, Thorsteinsson T, Thorsteinsdottir S, Heimisdottir M, of retroperitoneal adipose and pancreatic islet tissues in female rat
Olafsdottir K. Persistent organochlorines, sedentary occupation, obesity offspring. FASEB J 2014;28:183041.
and human male subfertility. Hum Reprod 2005;20:20815. 79. Masuyama H, Mitsui T, Eguchi T, Tamada S, Hiramatsu Y. The effects of
57. Oliva A, Spira A, Multigner L. Contribution of environmental factors to the paternal high-fat diet exposure on offspring metabolism with epigenetic
risk of male infertility. Hum Reprod 2001;16:176876. changes in the mouse adiponectin and leptin gene promoters. Am J Physiol
58. Shak A, Olfat S. Lipectomy in the treatment of scrotal lipomatosis. Br J Endocrinol Metab 2016;311:E23645.
Urol 1981;53:5561. 80. Rajender S, Avery K, Agarwal A. Epigenetics, spermatogenesis and male
59. Jung A, Schill WB. [Male infertility. Current life style could be responsible infertility. Mutat Res 2011;727:6271.
for infertility]. MMW Fortschr Med 2000;142:313, German. 81. John S, Burgess R, Cheng-Ching E, Wisco D, Taqui A, Bain M, et al. Last
60. OBrien JH, Lazarou S, Deane L, Jarvi K, Zini A. Erectile dysfunction and resort: case of clot translocation in intra-arterial stroke therapy. J Neurointerv
andropause symptoms in infertile men. J Urol 2005;174:19324, discussion Surg 2014;6:e50.
1934. 82. Houshdaran S, Cortessis VK, Siegmund K, Yang A, Laird PW, Sokol RZ.
61. Feldman HA, Johannes CB, Derby CA, Kleinman KP, Mohr BA, Araujo AB, Widespread epigenetic abnormalities suggest a broad DNA methylation
et al. Erectile dysfunction and coronary risk factors: prospective results from erasure defect in abnormal human sperm. PLoS One 2007;2:e1289.
the Massachusetts male aging study. Prev Med 2000;30:32838. 83. Khazamipour N, Noruzinia M, Fatehmanesh P, Keyhanee M, Pujol P.
62. Burnett AL, Strong TD, Trock BJ, Jin L, Bivalacqua TJ, Musicki B. Serum MTHFR promoter hypermethylation in testicular biopsies of patients with
biomarker measurements of endothelial function and oxidative stress after nonobstructive azoospermia: the role of epigenetics in male infertility.
daily dosing of sildenal in type 2 diabetic men with erectile dysfunction. J Hum Reprod 2009;24:23614.
Urol 2009;181:24551. 84. Wu W, Shen O, Qin Y, Niu X, Lu C, Xia Y, et al. Idiopathic male infertility is
63. Arana Rosainz Mde J, Ojeda MO, Acosta JR, Elias-Calles LC, Gonzalez NO, strongly associated with aberrant promoter methylation of methylenete-
Herrera OT, et al. Imbalanced low-grade inammation and endothelial trahydrofolate reductase (MTHFR). PLoS One 2010;5:e13884.
activation in patients with type 2 diabetes mellitus and erectile dysfunction. 85. Hammoud SS, Purwar J, Pueger C, Cairns BR, Carrell DT. Alterations in
J Sex Med 2011;8:201730. sperm DNA methylation patterns at imprinted loci in two classes of infertility.
64. Kelleher S, Conway AJ, Handelsman DJ. Blood testosterone threshold for Fertil Steril 2010;94:172833.
androgen deciency symptoms. J Clin Endocrinol Metab 2004;89:38137. 86. Terashima M, Barbour S, Ren J, Yu W, Han Y, Muegge K. Effect of high fat
65. Becker AJ, Uckert S, Stief CG, Scheller F, Knapp WH, Hartmann U, et al. diet on paternal sperm histone distribution and male offspring liver gene
Cavernous and systemic testosterone plasma levels during different penile expression. Epigenetics 2015;10:86171.
conditions in healthy males and patients with erectile dysfunction. Urology 87. Mattick JS, Makunin IV. Small regulatory RNAs in mammals. Hum Mol
2001;58:43540. Genet 2005;14(Spec. No. 1):R12132.
66. Hammoud AO, Wilde N, Gibson M, Parks A, Carrell DT, Meikle AW. 88. Sonnack V, Failing K, Bergmann M, Steger K. Expression of hyperacetylated
Male obesity and alteration in sperm parameters. Fertil Steril 2008;90: histone H4 during normal and impaired human spermatogenesis.
22225. Andrologia 2002;34:38490.

858 VOL. 107 NO. 4 / APRIL 2017


Fertility and Sterility

89. Krawetz SA, Kruger A, Lalancette C, Tagett R, Anton E, Draghici S, et al. A prevention and treatment of metabolic and endocrine diseases in adults:
survey of small RNAs in human sperm. Hum Reprod 2011;26:340112. cosponsored by the American Association of Clinical Endocrinologists/
90. McPherson NO, Owens JA, Fullston T, Lane M. Preconception diet or exer- the American College of Endocrinology and the Obesity Society. Endocr
cise intervention in obese fathers normalizes sperm microRNA prole and Pract 2013;19(Suppl 3):182.
metabolic syndrome in female offspring. Am J Physiol Endocrinol Metab 107. Kaukua J, Pekkarinen T, Sane T, Mustajoki P. Sex hormones and sexual
2015;308:E80521. function in obese men losing weight. Obes Res 2003;11:68994.
91. Jenkins TG, Aston KI, Hotaling JM, Shamsi MB, Simon L, Carrell DT. 108. Moran LJ, Brinkworth GD, Martin S, Wycherley TP, Stuckey B, Lutze J, et al.
Teratozoospermia and asthenozoospermia are associated with specic Long-term effects of a randomised controlled trial comparing high protein
epigenetic signatures. Andrology 2016;4:8439. or high carbohydrate weight loss diets on testosterone, SHBG, erectile and
92. Donkin I, Versteyhe S, Ingerslev LR, Qian K, Mechta M, Nordkap L, et al. urinary function in overweight and obese men. PLoS One 2016;11:
Obesity and bariatric surgery drive epigenetic variation of spermatozoa in e0161297.
humans. Cell Metab 2016;23:36978. 109. Collins CE, Jensen ME, Young MD, Callister R, Plotnikoff RC, Morgan PJ.
93. Hammoud SS, Nix DA, Zhang H, Purwar J, Carrell DT, Cairns BR. Distinctive Improvement in erectile function following weight loss in obese men:
chromatin in human sperm packages genes for embryo development. the SHED-IT randomized controlled trial. Obes Res Clin Pract 2013;7:
Nature 2009;460:4738. e4504.
94. Jodar M, Sendler E, Moskovtsev SI, Librach CL, Goodrich R, Swanson S, 110. Kim ED, McCullough A, Kaminetsky J. Oral enclomiphene citrate raises
et al. Absence of sperm RNA elements correlates with idiopathic male testosterone and preserves sperm counts in obese hypogonadal men, un-
infertility. Sci Transl Med 2015;7:295re6. like topical testosterone: restoration instead of replacement. BJU Int 2016;
95. Jenkins TG, Aston KI, Meyer TD, Hotaling JM, Shamsi MB, Johnstone EB, 117:67785.
et al. Decreased fecundity and sperm DNA methylation patterns. Fertil 111. Roth MY, Amory JK, Page ST. Treatment of male infertility secondary to
Steril 2016;105:517.e1-3. morbid obesity. Nat Clin Pract Endocrinol Metab 2008;4:4159.
96. Aston KI, Uren PJ, Jenkins TG, Horsager A, Cairns BR, Smith AD, et al. 112. Raman JD, Schlegel PN. Aromatase inhibitors for male infertility. J Urol
Aberrant sperm DNA methylation predicts male fertility status and embryo 2002;167:6249.
quality. Fertil Steril 2015;104:138897.e1-5. 113. American Society for Metabolic and Bariatric Surgery. Medical and
97. Bohacek J, Mansuy IM. Epigenetic inheritance of disease and disease risk. government groups support bariatric surgery. 2012. Available at: https://
Neuropsychopharmacology 2013;38:22036. asmbs.org/resources/bariatric-surgery-guidelines-and-recommendations.
98. Nilsson EE, Skinner MK. Environmentally induced epigenetic transgenerational Last accessed March 12, 2017.
inheritance of disease susceptibility. Transl Res 2015;165:127. 114. Reis LO, Dias FG. Male fertility, obesity, and bariatric surgery. Reprod Sci
99. Yuan TF, Li A, Sun X, Ouyang H, Campos C, Rocha NB, et al. Transgenera- 2012;19:77885.
tional inheritance of paternal neurobehavioral phenotypes: stress, 115. Hammoud A, Gibson M, Hunt SC, Adams TD, Carrell DT, Kolotkin RL,
addiction, ageing and metabolism. Mol Neurobiol 2016;53:636776. et al. Effect of Roux-en-Y gastric bypass surgery on the sex steroids
100. Smith CA. The effect of wartime starvation in Holland upon pregnancy and and quality of life in obese men. J Clin Endocrinol Metab 2009;94:
its product. Am J Obstet Gynecol 1947;53:599608. 132932.
101. Ravelli GP, Stein ZA, Susser MW. Obesity in young men after famine 116. Reis LO, Favaro WJ, Barreiro GC, de Oliveira LC, Chaim EA, Fregonesi A,
exposure in utero and early infancy. N Engl J Med 1976;295:34953. et al. Erectile dysfunction and hormonal imbalance in morbidly obese
102. Barker DJ, Osmond C. Infant mortality, childhood nutrition, and ischaemic male is reversed after gastric bypass surgery: a prospective randomized
heart disease in England and Wales. Lancet 1986;1:107781. controlled trial. Int J Androl 2010;33:73644.
103. de Castro Barbosa T, Ingerslev LR, Alm PS, Versteyhe S, Massart J, 117. el Bardisi H, Majzoub A, Arafa M, AlMalki A, al Said S, Khalafalla K,
Rasmussen M, et al. High-fat diet reprograms the epigenome of rat et al. Effect of bariatric surgery on semen parameters and sex hormone
spermatozoa and transgenerationally affects metabolism of the offspring. concentrations: a prospective study. Reprod Biomed Online 2016;33:
Mol Metab 2016;5:18497. 60611.
104. Tudiver F, Talbot Y. Why dont men seek help? Family physicians perspectives 118. Sermondade N, Massin N, Boitrelle F, Pfeffer J, Eustache F, Sifer C, et al.
on help-seeking behavior in men. J Fam Pract 1999;48:4752. Sperm parameters and male fertility after bariatric surgery: three case
105. McTiernan A, Sorensen B, Irwin ML, Morgan A, Yasui Y, Rudolph RE, et al. series. Reprod Biomed Online 2012;24:20610.
Exercise effect on weight and body fat in men and women. Obesity (Silver 119. Berglind D, Muller P, Willmer M, Sinha I, Tynelius P, Naslund E, et al. Differ-
Spring) 2007;15:1496512. ential methylation in inammation and type 2 diabetes genes in siblings
106. Gonzalez-Campoy JM, St. Jeor ST, Castorino K, Ebrahim A, Hurley D, born before and after maternal bariatric surgery. Obesity (Silver Spring)
Jovanovic L, et al. Clinical practice guidelines for healthy eating for the 2016;24:25061.

VOL. 107 NO. 4 / APRIL 2017 859

S-ar putea să vă placă și