Вы находитесь на странице: 1из 10

Perspective

For reprint orders, please contact: reprints@futuremedicine.com

Transgenerational epigenetic inheritance:


an open discussion

Much controversy surrounds the idea of transgenerational epigenetics. Recent papers Corina Nagy1
argue that epigenetic marks acquired through experience are passed to offspring, & Gustavo Turecki*,1,2
but as in much of the field of epigenetics, there is lack of precision in the definitions
1
McGill Group for Suicide Studies,
Douglas Hospital University Institute,
and perhaps too much eagerness to translate animal research to humans. Here, we
6875 Lasalle boul, Montreal, QC, Canada
review operational definitions of transgenerational inheritance and the processes 2
Department of Psychiatry, McGill
of epigenetic programing during early development. Subsequently, based on this University, Montreal, QC, Canada
background, we critically examine some recent findings of studies investigating *Author for correspondence:
transgenerational inheritance. Finally, we discuss possible mechanisms that may explain gustavo.turecki@ mcgill.ca

transgenerational inheritance, including transmission of an epigenetic blueprint,


which may predispose offspring to specific epigenetic patterning. Taken together, we
conclude that presently, the evidence suggesting that acquired epigenetic marks are
passed to the subsequent generation remains limited.

Keywords: animal models • early development • histone modifications • human


• intergenerational • methylation • transgenerational

Conrad Waddington first used the term epi- germline transmission; epigenetic marks
genetics over half a century ago and defined might also be propagated forward through
it as ‘the branch of biology which studies the germline to offspring. This notion was
the causal interactions between genes and fuelled by both research studies in plants
their products which bring the phenotype and longitudinal epidemiological studies
into being.’ A more modern definition of in humans, which have suggested that the
epigenetics is: heritable chemical modifica- effects of environmental influences were
tions to DNA capable of influencing tran- observed in subsequent generations. Stud-
scriptional activity with no direct alteration ies in the plant Arabidopsis thaliana showed
to the DNA sequence itself. It should be that alterations in DNA methylation can be
noted that heritable in this definition refers transmitted through several generations [3] .
to ‘mitotic stability’ [1] or, in other words, to Famously in humans, epidemiological stud-
the fact that epigenetic information is passed ies examining the Dutch famine found that
to, and maintained, in the daughter cell upon those individuals who were exposed to the
mitotic division. Certainly, this type of epi- famine while in utero, had increased sus-
genetic heritability is of relevance to cancer ceptibility to diseases in adulthood such as,
research [2] . However, the concept of epigen- diabetes, cardiovascular disease and obesity,
etic heritability has broadened over the years. to name a few [4] . These findings begged the
Studies have progressively suggested that not question of lastingness, expressly, whether
only might epigenetic marks be inherited these predisposing factors, which seemingly
through mitotic processes, but also through stemmed from in utero exposure to famine,
meiosis, a concept akin to genetic inheri- could be transmitted to the next generation.
tance. That is, similar to the way we inher- Though there is no direct molecular link
part of
ited genetic traits from our parents through to these major health concerns, second-

10.2217/epi.15.46 © 2015 Future Medicine Ltd Epigenomics (2015) 7(5), 781–790 ISSN 1750-1911 781
Perspective  Nagy & Turecki

generation offspring was found to demonstrate the could be carried forward. In fact, some loci, includ-
same lower birth weights, as seen in first-generation ing repeated elements such as retrotransposons, do
affected individuals [4] . not undergo reprogramming during this phase [6,7] .
In contrast to the mitotic stability seen in somatic Interestingly, Molaro and colleagues [8] found a strik-
cells, transgenerational epigenetics requires germline ing difference in methylation of retrotransposon sub-
transmission of acquired epigenetic information across families when examining the sperm of humans and
generations. In cases of male exposure, the second chimps. Furthermore, when comparing these sperm
generation may be sufficient to assess transgenera- methylomes to embryonic stem cells, which they con-
tional effects. This is because exposure will affect the sider a mature germ cell having the product of the
F0 male as well as his germline (F1), therefore only two reprogramming events in mammal, they found
starting with the F2 generation can the effects of trans- distinct characteristic, suggesting that sperm and
generational epigenetic inheritance be considered. In somatic cells have different features, which deter-
females, however, studies require a third generation to mine DNA methylation patterns in each cell type.
test whether any of these DNA modifications are truly Furthermore, approximately 100 nonimprinted, non-
transmitted from one generation to the next. This is repetitive genes have been identified as maintaining
because one needs to rule out in utero exposure of the promoter methylation levels throughout a range of
second generation to an environmental effect associ- developmental stages, from mature gametes to blas-
ated with epigenetic changes. As an illustration, expos- tocysts  [9] . Nonmammalian organisms on the other
ing a gestating female, her fetus and thus the fetal hand, appear to not undergo whole genome demeth-
germline to a given environment, produces an F1 gen- ylation upon fertilization [10] . In a comprehensive
eration with an associated epigenetic change reflecting study using zebrafish, Jiang and colleagues [10] were
direct environmental effects through in utero exposure able to demonstrate that no global erasure occurs at
(Figure 1) . Prior to the F2 generation in males and the fertilization and by the midblastula stage the embryo’s
F3 generation in female, the inheritance detected in methylome is almost identical to the of sperm, while
the offspring can only be considered intergenerational, the oocyte’s methylome contribution lessens by the
in other words, parental effect due to direct expo- 16-cell state. This suggests that the early reprogram-
sure. This is important information to consider when ming mechanisms are different between mammalian
reviewing some of the recent publications in this field. and non-mammalian organisms.
As mentioned above, both the mammalian mater-
Early developmental mechanisms: nal and paternal genomes undergo demethylation and
epigenetic reset they also experience active chromatin remodeling. This
Early epigenetic reprogramming has been extensively is particularly evident for the paternal genome. The
studied in rodents. Though the same processes of early process of spermatogenesis in humans requires that
developmental epigenetic erasure occur in humans, anywhere between 85 and 96% of histones be replaced
the definitive characteristics and timeline of these by protamines, resulting in a tenfold compaction of
events have still to be established [5] . Work in mice the DNA [12,13] . It is believed that this process protects
has demonstrated that maternal and paternal cells the paternal genome from physical and chemical dam-
not only undergo a global erasure of DNA methyla- age. Protamines, like histones, carry post-translational
tion but the chromatin also undergoes active remod- modifications such as phosphorylation. The role of
eling. Global erasure occurs at two points. First, as these modifications remains unknown, although they
the primordial germ cells (PGC) or gamete precursors are believed to act similarly to histone modifications
develop and become part of the embryo, the epigenetic altering the availability of the DNA sequencing to
landscape is cleared to allow for cellular totipotency the cellular transcriptional machinery [12] . Thus, the
and the development of future generations through majority of paternally derived histone marks are lost
PGCs. This stage is followed by gametogenesis when during this process while the histones not replaced by
the genomes undergo de novo methylation, a process protamines are thought to belong to genes expressed
that occurs later in the maternal genome. The second early in development [13] . Interestingly, the repressive
wave of demethylation occurs after fertilization as mark, histone H3 lysine 27 trimethylation, is retained
the gametes fuse to form the zygote (Figure 2) . Here, on some genes in the human and mouse spermatozoa.
however, imprinted genes escape the second round of This is arguably another possible avenue for inheri-
demethylation ( Figure 2, dotted lines), to carry for- tance; conversely, it could be a methodological arti-
ward parent-specific monoallelic expression in somatic fact, and simply be the result of a histone mark being
cells. If epigenetic marks were to be maintained across re-established so quickly that its period of absence is
generations, it is presumably at this point that they not detected. Gradually, studies are demonstrating

782 Epigenomics (2015) 7(5) future science group


Transgenerational epigenetic inheritance: an open discussion  Perspective

Female Male

F0

F1 F0

F2 F1

F2 F1

F3 and subsequent generations F2 and subsequent generations

Figure 1. The difference between intergenerational and transgenerational epigenetic inheritance in females and
males. F0 represents maternal or paternal exposure to environmental factors, which directly impacts the fetus (F1)
and its already formed germline (F2) in females or the germline in males (F1). This demonstrates the importance
of studying the F2 generation in males and the F3 generation in females to avoid parental effects, in other words,
intergenerational epigenetics. Studying the F2 (in males) and F3 (in females) and/or their subsequent generations
could provide information on transgenerational epigenetics as they would be free of direct environmental
exposure and parental effects.
For color images please see online www.futuremedicine.com/doi/full/10.2217/epi.15.46

the impressive dynamics of chromatin, for example, takes the form of nucleosome, whereas following the
the protein involved in heterochromatin formation histone to protamine exchange during spermiogen-
HP1 binds for just a few minutes [14] and the wrap- esis, the paternal genome incorporates histones from
ping and unwrapping of nucleosomes, a process which the maternal genome [18] . Importantly, epigenetic pro-
erases histone modifications, is so rapid that the turn- cesses in early mammalian development result in non-
over is faster than a complete cell cycle [15] . Thus, the canonical forms of histones in both the maternal and
highly dynamic nature of histone marks makes them paternal genomes.
less likely to act as a mechanism of transgenerational Finally, there is interesting epigenetic asymmetry
epigenetics. Equally, canonical forms of histones in between parental genomes, which remains poorly
the maternal genome, such as H2A, are replaced with understood. For example, de novo methylation in
H2A.Z during early embryonic development [16] . It gametes, and demethylation after fertilization occurs
is thought that H2A.Z is responsible for establishing more rapidly in paternal genome, denoted respectively
heterchromatin in early development [17] , furthermore, by the blue and pink line graph in Figure 2. In males,
its absence in the early embryonic development results during protamine removal, the paternal genome is
in death shortly after implantation [16] . The maternal repackaged into new histones, as well as into mater-
genome has also been shown to transfer components nally derived histones, whereas maternal chromatin
of a repressive complex, PRC1, to the paternal genome maintains histone methylation throughout early cleav-
after fertilization [18] , resulting in direct silencing in age  [19] . The timing of methylation could be directly
the zygote. At gamete fusion, the maternal genome related to the presence or absence of histone marks

future science group www.futuremedicine.com 783


Perspective  Nagy & Turecki

like H3K9me2, which are intimately related to DNA Escaping intergenerational reprogramming
methylation, the absence of this mark may promote In light of early developmental reprogramming, the
DNA demethylation [5] . For instance, PGCs show sex question researchers must address is whether certain
differences in the imprinted Igf2r gene, in both the epigenomic transcriptional drivers, in the form of
timing of demethylation in males and de novo methyl- DNA methylation or histone modifications, escape
ation in females [6] . This asymmetry is likely an expla- this prototypical intergenerational reprogramming.
nation of why some traits are only propagated through The classic experiment in the agouti viable yellow
one parental line, examples of which are discussed in mouse (Avy) was the first study to report intergenera-
later sections. tional inheritance in mammals. If a repeat element

De novo
E10.5–13.5
methylation DNA demethylation DNA methylation

Gametes Zygote Blastocyst Post-implantation

PGC Imprints
(gonads) maintained
Epiblast
Epiblast

PGC
DNA demethylation precursor

Methylation cycle
Male Imprinting
Methylation

Female

PGC in Gamatogenesis Fertilization Cell division


embryo
E10.5–13.5

F1 F2

Figure 2. Very early embryonic development corresponds to epigenetic programing. Primordial germ cells in the
embryo undergo global DNA methylation erasure, or ‘reprogramming’ from their epiblast state (red arrow). This
first wave of demethylation is also denoted in the methylation cycle graph depicting both the male and female
genomes devoid of DNA methylation, including imprinted genes. Gametes are then de novo methylated at
different rates, with maternal methylation marks being established later (graph pink line) than paternal marks. A
second round of ‘reprogramming’ occurs upon fusion of the gametes (sperm and oocyte) producing totipotent or
pluripotent cell states. At this point, demethylation occurs more rapidly in the paternal genome (graph blue line),
moreover, imprinted genes escape erasure (graph dotted lines) maintaining their methylation marks. Genome-
wide remethylation occurs in both parental genomes at implantation (green arrow). The timeline denoted in this
schematic refer to event in the mouse life cycle. The timeline in humans is not yet full defined, though the events
are considered to occur in a similar manner.
PGC: Primordial germ cell.
Adapted with permission from [11] .

784 Epigenomics (2015) 7(5) future science group


Transgenerational epigenetic inheritance: an open discussion  Perspective

called an intracisternal A particle (IAP), of which there tance of an epigenetic state [28–32] . In other words, in
are thousand of copies throughout the mouse genome, some cases, transmission of epigenetic marks can be
inserts itself upstream of the agouti locus, phenotypic secondary to inherited DNA sequence mutations.
variations can arise including altered coat color and
obesity [20] . DNA methylation at the promoter region Epigenetic inheritance: acquiring
of this IAP, which in turn regulates the agouti gene, knowledge
is inversely related to its transcriptional activity [20] . A number of recent studies have implied that epi-
As indicated above, repeat elements may escape repro- genetic alteration induced by environmental factors
gramming, which appears to be the case for this epial- affect the behavior of subsequent generations. Vas-
lele that is passed along the maternal but not paternal soler and colleagues [33] found decreased cocaine self-
germline  [20] . Interesting as this may be, this is not administration in the first generation offspring of
the result of a past environmental factor being propa- cocaine exposed male rats. The behavioral outcome
gated forward via epigenetic inheritance. Though the suggests an almost protective effect of fathers consum-
insertion of the IAP at the agouti locus may not be ing cocaine, which is not consistent with the clinical
entirely random [21] , it does not result from the effects literature  [34,35] . Molecularly, this is associated with
of environmental factors. However, once the IAP is increased Bdnf levels as well as increased H3 acetyla-
inserted and susceptible to methylation changes, envi- tion, an epigenetic mechanism potentially responsible
ronment can play a role. This has been demonstrated for this change. The authors of this intriguing study
by altering the food source of pregnant dams. Diets correctly suggested that more generations are required
high in methyl groups such as folic acid or betaine can to definitively conclude that cocaine exposure results
effectively ‘turn off ’ the Avy locus by methylating the in transgenerational epigenetic alterations [36] . Cer-
promoter region of the IAP [22,23] . Though it is tempt- tainly, cocaine itself can alter chromatin states [37] , but
ing to look at this as transgenerational inheritance, it it is possible that cocaine directly influenced the gam-
is rather the result of direct environmental exposure on etes of the parental generation, and thus the F1 prog-
the germline, and is thus the result of parental influ- eny. In a similar vein, a recent study in Caenorhabditis
ence, more appropriately termed, intergenerational elegans showed paternal transmission to the embryo of
inheritance. the repressive mark H3K27me3, which is under the
Early life environment is also known to alter meth- control of the Polycomb repression complex 2 [38] .
ylation states as classically demonstrated through rat Interestingly, histones containing this mark are not
maternal behavior paradigms [24] . Caution must be replaced by protamines during spermatogenesis in
taken when conducting transgenerational experi- mammals  [13] . This very interesting finding demon-
ments to control for both in utero and early life expe- strates mechanistic properties for the propagation of
riences to avoid confounding early-life environmental epigenetic marks through generations, but as pointed
effects with transgenerational inheritance. To avoid out previously, there is a need to demonstrate trans-
these potential confounding effects, both in utero and generational inheritance beyond the first generation of
in early life, studies in rodent models have focused progeny.
on paternal lineage as males are not involved in the A study examining early stress in mice addressed
in utero environment and are less likely to be involved the question of whether transgenerational inheritance
in the early-life postnatal environment. An interesting could be explained by the effect of parental environ-
example of this was found using outbred rats exposed ment on the gametes or through the transmission of
to the fungicide, vinclozolin in utero. As a result, males acquired epigenetic marks to the offspring, by observ-
exhibited diminished fertility over three to four gen- ing the transmission of depressive-like behaviors up to
erations of offspring, which was found to be transmit- the third generation [39,40] . The behavioral traits cose-
ted through the male germline [25] . Interestingly, these gregated with altered DNA methylation in the male
effects were not observed with another strain of rats. It germ line. The same genes were tested for methylation
is possible that this discrepancy results from method- levels in both the F2 sperm and the F2 female brains.
ological differences between studies but it also raises The methylation patterns in these cell types were simi-
the possibility that interaction with genetic variation lar, though certainly not identical. However, of note
might be responsible for the observed effect [26] . If this was a discrepancy in behavioral phenotype, where the
is the case, it represents an example of secondary epi- F1 and F3 males showed depressive-like behaviors but
mutations, whereby DNA sequence variants have an not the F2 males. The depressive-like phenotype was
effect on epigenetic marks (in cis or trans) [27] . Indeed only seen in the F2 females and the molecular changes
a number of studies have demonstrated that genetic- reported in this study were correlational, as there was
based alterations can be responsible for the inheri- no experiment in this study showing a causal link

future science group www.futuremedicine.com 785


Perspective  Nagy & Turecki

between methylation changes and phenotype. Further- tions may act in a probabilistic manner. In other words,
more, the exposure of females to stressed males, even these changes are more or less likely to occur in broad
for a short period, could have had subtle effects on the genomic regions, resulting in altered behavior without
female behaviors and hormonal expression, which in producing a consistent molecular phenotype. A recent
turn may have affected maternal care. In vitro fertiliza- paper demonstrating that paternal sugar consumption
tion (IVF) of the sperm directly into control females in fruit flies influences the metabolic properties of the
has been used to avoid these potential confounding F1 generation, leading to an obesity phenotype, sup-
effects from fathers. When IVF was used in social ports this concept. Here, the authors demonstrate that
defeat paradigms, which is an animal model of depres- a network of genes is required for proper intergenera-
sion, the depressive-like phenotype that seemed to be tional metabolic reprogramming involving a number
propagated forward under normal mating conditions of changes to chromatin structure. No specific sites
was largely absent [40] . Together, these results suggest were identified but rather a pattern or signature of gene
that broad behavioral conditioning can be inherited. dysfunction was identified as conferring susceptibility
Although there is a possibility that these may cosegre- to obesity [44] .
gate with specific epigenetic profiles, additional work is
necessary to definitively make such conclusions. Epigenetic inheritance: from rodents to
Another study using the same early stress paradigm humans
in mice showed depressive-like behaviors associated The vast majority of studies examining transgenera-
with an upregulation of several miRNA in sperm tional inheritance have been conducted in animals
which appeared to affect serum and hippocampal other than humans. Though mice and rats provide
miRNAs levels in the subsequent generation [41] . convenient models for human disorders and behav-
Because small RNAs are highly present in sperm, they ioral traits, there are different physiological and bio-
have become candidate vectors for conveying trans- logical processes between humans and rodents. For
generational inheritance [42] . However, in this study, instance, a recent paper exploring the methylation
although the behavioral phenotype was detected in landscape of early human embryos showed features
the F3 generation, the miRNA levels of five candi- distinctive to humans, specifically levels of methyla-
date miRNAs in F2 sperm were unchanged and the tion and timing of genome-wide demethylation [45] ,
alterations seen in these candidate miRNAs from F2 while another study suggests the symmetry of epigen-
were no longer detectable in F3. The authors suggest etic reprogramming cycles may differ between spe-
that the initial change in miRNA levels resulting from cies  [6] . Moreover, many transcriptionally relevant
early stress may have been transferred to other epigen- epigenetic marks show considerable sequence diver-
etic marks, but give no evidence of this effect. Another gence between mice and humans [46] . Furthermore,
study from the same group set out to demonstrate a information about epigenetic erasure during early
causal mechanism for the inherited stress responses in development comes primarily from mouse mod-
the offspring of stressed sires [43] . In this study, the els, and it is possible to be different in humans, but
authors were able to pharmacologically induce the remains, as yet, untested. Transgenerational effects
same behavioral traits seen in the stressed offspring, have been observed in humans through longitudinal
with corresponding alteration to gene expression levels studies [47–49] , whereas gene–environment interplay is
resulting from histone methyl- and acetyl-transferase suspected as a mediating factor in health outcome.
inhibitors, suggesting a causal role for mineral corti- Some researchers have attempted to approach epigen-
coid receptors in the altered stress response of pater- etic inheritance using in a systems biology approach,
nally stressed offspring. Here, however, the epigenetic whereby acquired behavioural and physiological
changes seen in the male germline differed from what changes, are considered through the framework of a
was found in the adult hippocampus of the offspring, “two hit” model. This approach recognizes both, the
specifically methylation changes where seen in the biology established in a previous generation and the
sperm whereas altered histone modifications were seen organism’s present environment, as involved in the
in the brains. Collectively, it appears that whatever development of these changes [50] . In a study explor-
environmentally induced epigenetic alteration is tak- ing vinclozolin-lineage rats presumed to be carry-
ing place in one generation may be propagated for- ing epigenetic alterations from previous generations,
ward, but as noted above, although possible, it is still the authors identified gene network and behavioural
premature to conclusively suggest a clear association alterations associated with the lineage specific rats
between one acquired epigenetic mark and transgener- when subjected to a stress paradigm [51] . Though this
ational behavioral phenotypes. An enticing possibility study does not answer the question of whether or not
is that epigenetic changes transmitted across genera- these genetic and behavioural changes would have

786 Epigenomics (2015) 7(5) future science group


Transgenerational epigenetic inheritance: an open discussion  Perspective

occurred in the absence of the induced environmental One should be particularly cautious when interpret-
stress (context) nor does it confirm the presence of ing the results of studies testing transgenerational epi-
epigenetically altered states, it nonetheless represents genetic inheritance in animals to the lay media or gen-
the attractive notion of dynamic interactions between eral public. Though likely not the intention, as pointed
context dependence and “predisposing factors”, in out previously [52] , animal studies have been vulgarized
this case, environment and inherited epigenetic state. and commonly anthropomorphized into a public mes-
Intuitively, most people can recognize that their envi- sage that overemphasizes the molecular impact of the
ronment plays a role in health and behavior, but we environment, including parents and grandparents onto
still lack conclusive evidence to indicate that such their children.
transgenerational effects are explained by acquired The field of epigenetics is fascinating and holds
epigenetic mechanisms inherited from one generation great potential in medicine, both to uncover dis-
to the next. ease biomarkers and therapeutic interventions. The
results suggesting that acquired epigenetic factors
Conclusion may be transmitted through generations and explain
The studies reviewed above suggest that there is some acquired phenotypic traits are promising and intrigu-
evidence that certain epigenetic marks escape erasure ing, but nevertheless, studies published thus far have
in early development. There is also evidence that cer- limitations and prevent us from making definitive
tain acquired behavioral phenotypes are transmitted conclusions.
through subsequent generations. Although promising,
the evidence suggesting that acquired epigenetic marks Future perspective
cosegregate with acquired behavioral phenotypes An important missing link in the study of transgen-
remains inconclusive and open to a number of potential erational epigenetic inheritance is a mechanism by
alternative interpretations, including methodological which gene-regulatory information is transferred
explanations. from somatic cells to germs cells. Efforts to uncover

Executive Summary
Early developmental mechanisms: epigenetic reset
• Epigenetic reprogramming occurs at two points during early development, after fertilization and in primordial
germ cells.
• Some genomic features, such as retrotransposons, as well as certain histone marks can carry forward
epigenetic marks in spite of these erasure mechanisms, supporting the possibility that epigenetic marks can be
maintained across generations.
Escaping intergenerational reprogramming
• Mice and rat models have been used to show that certain epigenetic marks can escape intergenerational
reprogramming, affecting the phenotype of future generations.
• Effects resulting from parental influences result in intergeneration inheritance, which is not to be confused
with transgenerational inheritance (effects that survive across generations in the absence of direct exposure).
• Secondary epimutations represent another factor that can influence possible transgenerational inheritance. In
these cases, epigenetic marks are secondary to inherited DNA sequence mutations.
Epigenetic inheritance: acquiring knowledge
• A number of studies including recent articles providing mechanistic insight show that epigenetic marks can be
inherited intergenerationally.
• Data for transgenerational inheritance on the other hand show certain inconsistencies, which may be
suggestive of probabilistic epigenetic changes capable of influencing phenotypic outcome in subsequent
generations.
Epigenetic inheritance: from rodents to humans
• It is difficult to provide concrete evidence that transgenerational epigenetics occurs in humans owing to the
lack of studies in humans and the physiological and biological differences between humans and rodents.
Conclusion
• There is evidence that epigenetic inheritance occurs intergenerationally by escaping erasure in early
development.
• However, evidence is lacking for the co-segregation of acquired epigenetic marks with acquired behavioral
traits transgenerationally.
• The field is very promising but more studies are required in order to provide definitive conclusions on the
topic of transgenerational inheritance, particularly in humans.

future science group www.futuremedicine.com 787


Perspective  Nagy & Turecki

these mechanisms have been made in nonmamma- phenotypic traits and acquired epigenetic marks, and
lian animals. For example, a number of studies in C. their co-segregation, through three generations at
elegans have implicated small RNAs in the process least. In addition to correlational evidence, studies
of inherited epigenetic marks [53–55] . It was recently should also investigate causative links between the
demonstrated in that dsRNA act as mobile elements molecular changes investigated and the phenotypes.
that mediating intertissue transfer of regulatory infor- Finally, although more difficult to conduct, studies
mation by entering the cytosol via a dsRNA-selective are also necessary in humans.
importer [56] . Equally, it has been speculated, notably
by a paper discussed here [41] and others [57,58] , that Financial & competing interests disclosure
small noncoding RNAs, like miRNA, mediate soma The authors have no relevant affiliations or financial in-
to germline transfer of regulatory information in volvement with any organization or entity with a financial
mammals, however, experimental evidence in mam- interest in or financial conflict with the subject matter or
mals is lacking. Tackling this question in mammals materials discussed in the manuscript. This includes employ-
and particularly in humans is an important next step ment, consultancies, honoraria, stock ownership or options,
in the research of transgenerational epigenetics. expert testimony, grants or patents received or pending, or
Moving forward in the field of transgenerational royalties.
epigenetics requires more precision in experimental No writing assistance was utilized in the production of
design. First, studies should investigate both acquired this manuscript.

References 11 Fresard L, Morisson M, Brun JM et al. Epigenetics and


Papers of special note have been highlighted as: phenotypic variability: some interesting insights from birds.
• of interest; •• of considerable interest Genet. Sel. Evol. 45, 16 (2013).
1 Skinner MK. Environmental epigenetic transgenerational 12 Brunner AM, Nanni P, Mansuy IM. Epigenetic marking
inheritance and somatic epigenetic mitotic stability. of sperm by post-translational modification of histones and
Epigenetics 6(7), 838–842 (2011). protamines. Epigenetics Chromatin 7(1), 2 (2014).
2 Dawson MA, Kouzarides T. Cancer epigenetics: from 13 Brykczynska U, Hisano M, Erkek S et al. Repressive and active
mechanism to therapy. Cell 150(1), 12–27 (2012). histone methylation mark distinct promoters in human and
mouse spermatozoa. Nat. Struct. Mol. Biol. 17(6), 679–687
3 Johannes F, Porcher E, Teixeira FK et al. Assessing the
(2010).
impact of transgenerational epigenetic variation on complex
traits. PLoS Genet. 5(6), e1000530 (2009). 14 Festenstein R, Pagakis SN, Hiragami K et al. Modulation of
heterochromatin protein 1 dynamics in primary mammalian
4 Painter RC, Osmond C, Gluckman P, Hanson M, Phillips
cells. Science 299(5607), 719–721 (2003).
DI, Roseboom TJ. Transgenerational effects of prenatal
exposure to the dutch famine on neonatal adiposity and 15 Deal RB, Henikoff S. Histone variants and modifications in
health in later life. BJOG 115(10), 1243–1249 (2008). plant gene regulation. Curr. Opin. Plant Biol. 14(2), 116–122
(2011).
5 Leitch HG, Tang WW, Surani MA. Primordial germ-cell
development and epigenetic reprogramming in mammals. 16 Faast R, Thonglairoam V, Schulz TC et al. Histone variant
Curr. Top. Dev. Biol. 104, 149–187 (2013). h2a.Z is required for early mammalian development. Curr.
Biol. 11(15), 1183–1187 (2001).
6 Hyldig SM, Croxall N, Contreras DA, Thomsen PD, Alberio
R. Epigenetic reprogramming in the porcine germ line. BMC 17 Banaszynski LA, Allis CD, Lewis PW. Histone variants in
Dev. Biol. 11, 11 (2011). metazoan development. Dev. Cell 19(5), 662–674 (2010).
7 Law JA, Jacobsen SE. Establishing, maintaining and 18 Puschendorf M, Terranova R, Boutsma E et al. Prc1
modifying DNA methylation patterns in plants and animals. and suv39h specify parental asymmetry at constitutive
Nat. Rev. Genet. 11(3), 204–220 (2010). heterochromatin in early mouse embryos. Nat. Genet. 40(4),
411–420 (2008).
8 Molaro A, Hodges E, Fang F et al. Sperm methylation
profiles reveal features of epigenetic inheritance and 19 Zhou LQ, Dean J. Reprogramming the genome to totipotency
evolution in primates. Cell 146(6), 1029–1041 (2011). in mouse embryos. Trends Cell. Biol. 25(2), 82–91 (2014).
9 Borgel J, Guibert S, Li Y et al. Targets and dynamics 20 Morgan HD, Sutherland HG, Martin DI, Whitelaw E.
of promoter DNA methylation during early mouse Epigenetic inheritance at the agouti locus in the mouse. Nat.
development. Nat. Genet. 42(12), 1093–1100 (2010). Genet. 23(3), 314–318 (1999).
10 Jiang L, Zhang J, Wang JJ et al. Sperm, but not oocyte, 21 Baillie JK, Barnett MW, Upton KR et al. Somatic
DNA methylome is inherited by zebrafish early embryos. Cell retrotransposition alters the genetic landscape of the human
153(4), 773–784 (2013). brain. Nature 479(7374), 534–537 (2011).
• In this study, the authors demonstrate that the timing and 22 Waterland RA, Jirtle RL. Transposable elements: targets for
inheritance of epigenetic marks in zebrafish are driven by early nutritional effects on epigenetic gene regulation. Mol.
the male germline. Cell. Biol. 23(15), 5293–5300 (2003).

788 Epigenomics (2015) 7(5) future science group


Transgenerational epigenetic inheritance: an open discussion  Perspective

• Evidences that transposable elements can escape early 39 Franklin TB, Russig H, Weiss IC et al. Epigenetic
developmental reprogramming. transmission of the impact of early stress across generations.
23 Wolff GL, Kodell RL, Moore SR, Cooney CA. Maternal Biol. Psychiatry 68(5), 408–415 (2010).
epigenetics and methyl supplements affect agouti gene 40 Dietz DM, Laplant Q, Watts EL et al. Paternal transmission
expression in avy/a mice. FASEB J. 12(11), 949–957 (1998). of stress-induced pathologies. Biol. Psychiatry 70(5), 408–414
24 Weaver IC, Cervoni N, Champagne FA et al. Epigenetic (2011).
programming by maternal behavior. Nat. Neurosci. 7(8), 41 Gapp K, Jawaid A, Sarkies P et al. Implication of sperm rnas
847–854 (2004). in transgenerational inheritance of the effects of early trauma
in mice. Nat. Neurosci. 17(5), 667–669 (2014).
• Demonstrates how environment can have a relatively long-
lasting effect on behavior through epigenetic alterations. 42 Kawano M, Kawaji H, Grandjean V, Kiani J, Rassoulzadegan
M. Novel small noncoding rnas in mouse spermatozoa,
25 Anway MD, Cupp AS, Uzumcu M, Skinner MK.
zygotes and early embryos. PLoS ONE 7(9), e44542 (2012).
Epigenetic transgenerational actions of endocrine disruptors
and male fertility. Science 308(5727), 1466–1469 (2005). 43 Gapp K, Soldado-Magraner S, Alvarez-Sanchez M et al.
Early life stress in fathers improves behavioural flexibility in
26 Schneider S, Kaufmann W, Buesen R, Van Ravenzwaay B.
their offspring. Nat. Commun. 5, 5466 (2014).
Vinclozolin – the lack of a transgenerational effect after oral
maternal exposure during organogenesis. Reprod. Toxicol. •• Provides evidence in mammals that certain histone marks
25(3), 352–360 (2008). can be mantained through developmental reprogramming.
27 Oey H, Whitelaw E. On the meaning of the word 44 Ost A, Lempradl A, Casas E et al. Paternal diet defines
‘epimutation’. Trends Genet. 30(12), 519–520 (2014). offspring chromatin state and intergenerational obesity. Cell
28 Bell JT, Pai AA, Pickrell JK et al. DNA methylation 159(6), 1352–1364 (2014).
patterns associate with genetic and gene expression variation 45 Guo H, Zhu P, Yan L et al. The DNA methylation landscape
in hapmap cell lines. Genome Biol. 12(1), R10 (2011). of human early embryos. Nature 511(7511), 606–610 (2014).
29 Gertz J, Varley KE, Reddy TE et al. Analysis of DNA 46 Yue F, Cheng Y, Breschi A et al. A comparative encyclopedia
methylation in a three-generation family reveals widespread of DNA elements in the mouse genome. Nature 515(7527),
genetic influence on epigenetic regulation. PLoS Genet. 355–364 (2014).
7(8), e1002228 (2011). 47 Bygren LO, Kaati G, Edvinsson S. Longevity determined by
30 Gutierrez-Arcelus M, Ongen H, Lappalainen T et al. paternal ancestors’ nutrition during their slow growth period.
Tissue-specific effects of genetic and epigenetic variation on Acta Biotheor. 49(1), 53–59 (2001).
gene regulation and splicing. PLoS Genet. 11(1), e1004958 48 Kaati G, Bygren LO, Edvinsson S. Cardiovascular and
(2015). diabetes mortality determined by nutrition during parents’
31 Zhang D, Cheng L, Badner JA et al. Genetic control of and grandparents’ slow growth period. Eur. J. Hum. Genet.
individual differences in gene-specific methylation in 10(11), 682–688 (2002).
human brain. Am. J. Hum. Genet. 86(3), 411–419 (2010). 49 Pembrey ME, Bygren LO, Kaati G et al. Sex-specific, male-
32 Cini G, Carnevali I, Quaia M et al. Concomitant mutation line transgenerational responses in humans. Eur. J. Hum.
and epimutation of the mlh1 gene in a lynch syndrome Genet. 14(2), 159–166 (2006).
family. Carcinogenesis 36(4), 452–458 (2015). 50 Crews D, Gore AC. Transgenerational epigenetics – current
33 Vassoler FM, White SL, Schmidt HD, Sadri-Vakili G, controversiesand debates. In: Transgenerational Epigenetics.
Pierce RC. Epigenetic inheritance of a cocaine-resistance Tollefsbol T (Ed.).  Elsevier, MA, USA, 371–387 (2014).
phenotype. Nat. Neurosci. 16(1), 42–47 (2013). 51 Crews D, Gillette R, Scarpino SV, Manikkam M, Savenkova
34 Johnson JL, Leff M. Children of substance abusers: MI, Skinner MK. Epigenetic transgenerational inheritance of
overview of research findings. Pediatrics 103(5 Pt. 2), altered stress responses. Proc. Natl Acad. Sci. USA. 109(23),
1085–1099 (1999). 9143–9148 (2012).
35 Delaney-Black V, Chiodo LM, Hannigan JH et al. Prenatal 52 Juengst ET, Fishman JR, McGowan ML, Settersten RA Jr.
and postnatal cocaine exposure predict teen cocaine use. Serving epigenetics before its time. Trends Genet. 30(10),
Neurotoxicol. Teratol. 33(1), 110–119 (2011). 427–429 (2014).
36 Vassoler FM, Sadri-Vakili G. Mechanisms of 53 Buckley BA, Burkhart KB, Gu SG et al. A nuclear argonaute
transgenerational inheritance of addictive-like behaviors. promotes multigenerational epigenetic inheritance and
Neuroscience 264, 198–206 (2014). germline immortality. Nature 489(7416), 447–451 (2012).
37 Maze I, Feng J, Wilkinson MB, Sun H, Shen L, Nestler 54 Ashe A, Sapetschnig A, Weick EM et al. Pirnas can trigger a
EJ. Cocaine dynamically regulates heterochromatin and multigenerational epigenetic memory in the germline of C.
repetitive element unsilencing in nucleus accumbens. elegans. Cell 150(1), 88–99 (2012).
Proc. Natl Acad. Sci. USA 108(7), 3035–3040 (2011). 55 Burton NO, Burkhart KB, Kennedy S. Nuclear rnai
38 Gaydos LJ, Wang W, Strome S. Gene repression. H3K27me maintains heritable gene silencing in Caenorhabditis elegans.
and PRC2 transmit a memory of repression across Proc. Natl Acad. Sci. USA 108(49), 19683–19688 (2011).
generations and during development. Science 345(6203), 56 Devanapally S, Ravikumar S, Jose AM. Double-stranded
1515–1518 (2014). RNA made in C. elegans neurons can enter the germline and

future science group www.futuremedicine.com 789


Perspective  Nagy & Turecki

cause transgenerational gene silencing. Proc. Natl Acad. Sci. 58 Sharma A. Novel transcriptome data analysis implicates
USA 112(7), 2133–2138 (2015). circulating microRNAs in epigenetic inheritance in
57 Sharma A. Bioinformatic analysis revealing association of mammals. Gene 538(2), 366–372 (2014).
exosomal mrnas and proteins in epigenetic inheritance.
J. Theor. Biol. 357, 143–149 (2014).

790 Epigenomics (2015) 7(5) future science group

Вам также может понравиться