47
Transgenerational Epigenetic Inheritance: Prevalence, Mechanisms, and Implications for the Study of Heredity and Evolution Author(s): Eva Jablonka and Gal Raz Source: The Quarterly Review of Biology, Vol. 84, No. 2 (June 2009), pp. 131-176 Published by: The University of Chicago Press Stable URL: http://www.jstor.org/stable/10.1086/598822 . Accessed: 11/01/2011 00:48 Your use of the JSTOR archive indicates your acceptance of JSTOR's Terms and Conditions of Use, available at . http://www.jstor.org/page/info/about/policies/terms.jsp. JSTOR's Terms and Conditions of Use provides, in part, that unless you have obtained prior permission, you may not download an entire issue of a journal or multiple copies of articles, and you may use content in the JSTOR archive only for your personal, non-commercial use. Please contact the publisher regarding any further use of this work. Publisher contact information may be obtained at . http://www.jstor.org/action/showPublisher?publisherCode=ucpress. . Each copy of any part of a JSTOR transmission must contain the same copyright notice that appears on the screen or printed page of such transmission. JSTOR is a not-for-profit service that helps scholars, researchers, and students discover, use, and build upon a wide range of content in a trusted digital archive. We use information technology and tools to increase productivity and facilitate new forms of scholarship. For more information about JSTOR, please contact [email protected]. The University of Chicago Press is collaborating with JSTOR to digitize, preserve and extend access to The Quarterly Review of Biology. http://www.jstor.org

Transgenerational Epigenetic Inheritance: … EPIGENETIC INHERITANCE: PREVALENCE, MECHANISMS, AND IMPLICATIONS FOR THE STUDY OF HEREDITY AND EVOLUTION Eva Jablonka The Cohn Institute

  • Upload
    lebao

  • View
    232

  • Download
    3

Embed Size (px)

Citation preview

Transgenerational Epigenetic Inheritance: Prevalence, Mechanisms, and Implications for theStudy of Heredity and EvolutionAuthor(s): Eva Jablonka and Gal RazSource: The Quarterly Review of Biology, Vol. 84, No. 2 (June 2009), pp. 131-176Published by: The University of Chicago PressStable URL: http://www.jstor.org/stable/10.1086/598822 .Accessed: 11/01/2011 00:48

Your use of the JSTOR archive indicates your acceptance of JSTOR's Terms and Conditions of Use, available at .http://www.jstor.org/page/info/about/policies/terms.jsp. JSTOR's Terms and Conditions of Use provides, in part, that unlessyou have obtained prior permission, you may not download an entire issue of a journal or multiple copies of articles, and youmay use content in the JSTOR archive only for your personal, non-commercial use.

Please contact the publisher regarding any further use of this work. Publisher contact information may be obtained at .http://www.jstor.org/action/showPublisher?publisherCode=ucpress. .

Each copy of any part of a JSTOR transmission must contain the same copyright notice that appears on the screen or printedpage of such transmission.

JSTOR is a not-for-profit service that helps scholars, researchers, and students discover, use, and build upon a wide range ofcontent in a trusted digital archive. We use information technology and tools to increase productivity and facilitate new formsof scholarship. For more information about JSTOR, please contact [email protected].

The University of Chicago Press is collaborating with JSTOR to digitize, preserve and extend access to TheQuarterly Review of Biology.

http://www.jstor.org

TRANSGENERATIONAL EPIGENETIC INHERITANCE:PREVALENCE, MECHANISMS, AND IMPLICATIONS FOR THE

STUDY OF HEREDITY AND EVOLUTION

Eva JablonkaThe Cohn Institute for the History and Philosophy of Science and Ideas, Tel-Aviv University, Tel-Aviv

69978, Israel

e-mail: [email protected]

Gal RazThe Graduate School of Medicine, Tel-Aviv University, Tel-Aviv 69978, Israel

e-mail: [email protected]

keywordscell memory, epigenetics, induced heritable variations, Lamarckism,

microevolution, macroevolution

abstractThis review describes new developments in the study of transgenerational epigenetic inheritance, a

component of epigenetics. We start by examining the basic concepts of the field and the mechanismsthat underlie epigenetic inheritance. We present a comprehensive review of transgenerational cellularepigenetic inheritance among different taxa in the form of a table, and discuss the data containedtherein. The analysis of these data shows that epigenetic inheritance is ubiquitous and suggests linesof research that go beyond present approaches to the subject. We conclude by exploring some of theconsequences of epigenetic inheritance for the study of evolution, while also pointing to the importanceof recognizing and understanding epigenetic inheritance for practical and theoretical issues in biology.

Definitions: Epigenetics andEpigenetic Inheritance

EPIGENETICS has become one of thebuzz words of biology in recent years.

Following the success of genome projectsin defining what genomes are, the empha-sis has shifted to what they do, and there isrenewed interest in understanding the epi-genetic processes of development. Theterm “epigenetics,” however, has under-

gone many transformations since its origi-nal definition by Waddington (see Wad-dington 1968 for a discussion), reflectingthe changing foci of research in develop-mental molecular biology since the secondhalf of the 20th century (Jablonka andLamb 2002, 2007c; Haig 2004; Holliday2006). “Epigenetics” is therefore often em-ployed loosely and inconsistently, and issometimes used as a synonym for “epige-

The Quarterly Review of Biology, June 2009, Vol. 84, No. 2

Copyright © 2009 by The University of Chicago Press. All rights reserved.

0033-5770/2009/8402-0001$15.00

Volume 84, No. 2 June 2009THE QUARTERLY REVIEW OF BIOLOGY

131

netic inheritance.” To avoid misunder-standings, we define both terms as they areused in this review:

Epigenetics is the study of the processesthat underlie developmental plasticity andcanalization and that bring about persistentdevelopmental effects in both prokaryotesand eukaryotes. At the cellular level, theseare the processes involved in cell determina-tion and differentiation. At higher levels ofbiological organization, epigenetic mecha-nisms generate the context-dependent, self-sustaining interactions between groups ofcells that lead to physiological and morpho-logical persistence. The regulatory mecha-nisms that establish and maintain variant cel-lular and organismal states are known asepigenetic control mechanisms, or epigenetic con-trol systems (Nanney 1958).

Epigenetic inheritance is a componentof epigenetics. It occurs when phenotypicvariations that do not stem from variationsin DNA base sequences are transmitted tosubsequent generations of cells or organ-isms. Many of the discoveries about epige-netic inheritance between organisms arederived from studies in developmental bi-ology that look at inheritance in cell lin-eages within an organism. Cell heredity inmitotically dividing cells underlies the per-sistence of determined states in multicellu-lar organisms. That is, an individual’s kid-ney stem cells and skin stem cells generallybreed true, even though their DNA se-quences are identical and the developmen-tal stimuli that led to the different cellphenotypes are long gone. However, thesame cell heredity mechanisms that havebeen found in cell lineages during develop-ment are also observed when epigenetic in-heritance occurs between generations of in-dividuals. In single-celled organisms, such asbacteria and asexually reproducing protists,epigenetic inheritance leads to the clonalpersistence of induced and stochasticallygenerated phenotypic variations. In sexuallyreproducing organisms, heritable epigeneticvariations in germline cells can result in thetransmission of developmentally inducedand stochastically generated phenotypesfrom one generation of individuals to thenext through the gametes. Like “epigenet-

ics,” “epigenetic inheritance” is not alwaysconsistently employed. It is used in both abroad and a narrow sense.

Epigenetic inheritance in the broad sense is theinheritance of developmental variationsthat do not stem from differences in thesequence of DNA or from persistent induc-ing signals in the present environment. Aswell as cell-to-cell transmission of epige-netic variations in unicellular and multicel-lular organisms (as will be explained be-low), the definition covers body-to-body (orsoma-to-soma) information transference thatcan take place through developmental inter-actions between mother and offspring (e.g.,Weaver et al. 2004), through social learning(Avital and Jablonka 2000), and throughsymbolic communication (Richerson andBoyd 2005).

Cellular epigenetic inheritance is a narroweraspect of epigenetic inheritance as dis-cussed in the broad sense. It refers to epi-genetic transmission in sexual or asexualcell lineages, and the unit of this trans-mission is the cell. Following Holliday(1994, 2002, 2006), some biologists restrictthe term epigenetic inheritance solely tothe transmission of chromatin marks andRNAs (e.g., Wu and Morris 2001). How-ever, yeast geneticists use the term epige-netic inheritance to refer to the inheri-tance of protein conformations, such asprions (e.g., Uptain and Lindquist 2001),and the term is also used by biologistsstudying self-sustaining loops and chroma-tin inheritance in bacteria (e.g., Grandjeanet al. 1998; Laurent et al. 2005). We there-fore define cellular epigenetic inheritanceas the transmission from mother cell todaughter cell of variations that are not theresult of differences in DNA base sequenceand/or the present environment. Trans-mission can be through chromatin marks,through RNAs, through self-reconstructingthree-dimensional structures, and throughself-sustaining metabolic loops (Jablonkaet al. 1992; Jablonka and Lamb 1995, 2005,2007a). It occurs following cell division inprokaryotes, mitotic cell division in thesoma of eukaryotes, and sometimes follow-ing the meiotic divisions in the germline.The chromatin and RNA-mediated cellularinheritance systems seem to play an impor-tant role in inheritance through the germ-line in both females and males.

132 Volume 84THE QUARTERLY REVIEW OF BIOLOGY

In Figure 1, we illustrate the differencebetween the broad and narrow sense ofepigenetic inheritance by showing themain routes of between-generation trans-mission in a sexually reproducing multicel-lular organism. As our focus in this reviewis on cellular transgenerational epigenetic inher-itance, we are concentrating on the route ofbetween-generation transmission that in-volves a single-cell “bottleneck,” i.e., trans-mission through a gamete or a spore inmulticellular, sexually reproducing organ-isms, or through a single sexual or asexualcell in unicellular organisms. The environ-ment may induce epigenetic variation bydirectly affecting the germline or by affect-ing germ cells through the mediation ofthe soma, but, in either case, subsequenttransmission is through the germline. Al-though the direct soma-to-soma transmis-sion route of epigenetic variations is ofgreat importance for both developmentand evolution (Jablonka and Lamb 2005,2007a,b), its discussion is beyond the scopeof this review.

Mechanisms of Cellular EpigeneticInheritance (EISs)

Jablonka and Lamb (1989, 1995; Jablonka etal. 1992) suggested that the different mecha-nisms of epigenetic inheritance should beunderstood and studied within a shared evo-lutionary framework that incorporates thedevelopmental construction of heredity andthat acknowledges the Lamarckian aspects ofheredity and evolution. They called the pro-cesses and mechanisms that underlie cellularinheritance “epigenetic inheritance systems”(abbreviated to EISs by Maynard Smith[1990]). Cell heredity may occur when aninduced gene-product is diluted very slowlyby cell divisions, so that its concentrationremains above the threshold required for itsactivity for several cell generations (Zachari-oudakis et al. 2007), but such “memory” isshort-term. For more persistent memory andcell heredity, autocatalysis is necessary, andall the EISs we describe depend on mecha-nisms that enable self-perpetuation. Fourtypes of cellular EISs are recognized today:the EIS based on self-sustaining regulatoryloops, the EIS based on three-dimensional Fi

gu

re1.

Ro

ute

so

fT

ran

smis

sio

no

fE

pig

enet

icV

aria

tio

ns

ina

Mu

ltic

ellu

lar,

Sex

ual

lyR

epro

du

cin

gO

rgan

ism

Rou

tea

show

sth

ege

rmlin

e-to

-ger

mlin

etr

ansm

issi

onof

indu

ced

epig

enet

icva

riat

ion

s(e

.g.,

chro

mat

inm

arks

).A

vari

atio

nca

nbe

indu

ced

inth

ege

rmlin

ean

dca

nth

enbe

tran

smit

ted

from

one

gen

erat

ion

toth

en

ext,

orit

can

firs

tbe

indu

ced

inth

eso

ma,

then

affe

ctth

ege

rmlin

e,an

dth

erea

fter

bein

her

ited

thro

ugh

the

germ

line.

Rou

teb

show

sso

ma-

to-s

oma

tran

smis

sion

(for

exam

ple,

thro

ugh

the

tran

smis

sion

ofsy

mbi

onts

and

para

site

s,or

thro

ugh

the

self

-per

petu

atin

gef

fect

sof

mat

ern

albe

hav

ior,

soci

alle

arn

ing,

and

sym

bolic

com

mun

icat

ion

).A

broa

dvi

ewof

epig

enet

icin

her

itan

ceen

com

pass

esbo

thro

utes

aan

db,

wh

erea

sth

en

arro

wer

,ce

llula

rvi

ewin

clud

eson

lyro

ute

a—tr

ansm

issi

onth

roug

ha

sin

gle-

cell

“bot

tlen

eck,

”in

this

case

,a

gam

ete.

June 2009 133TRANSGENERATIONAL EPIGENETIC INHERITANCE

templating, the chromatin-marking EIS, andthe RNA-mediated EIS. Although these EISsare usually seen as very different types of de-velopmental mechanisms, all can contribute tobetween-generation epigenetic inheritance.Their dual nature as developmental mecha-nisms and as inheritance systems means thatthey can be studied from both perspectives.The developmental perspective is currentlythe dominant one, and the molecular basisof epigenetic control systems is one of themost intensely studied fields in biology today(e.g., see collections of articles in Nature Re-views Genetics 8(4) [Flintoft 2007], Nature447(7143 Insight) [Campbell 2007], and Cell128(4) [Marcus 2007], as well as Allis et al.2007). In this review, we focus on the lessdiscussed aspect of EISs and epigenetic in-heritance—the transmission of cellular epi-genetic states from one generation of organ-isms to the next. We therefore describe thefour EISs from a heredity-focused point ofview. All the examples to which we refer, aswell as many others, are presented here inTable 1 and in our expanded table (availableonline, with accompanying table references,at The Quarterly Review of Biology homepage,www.journals.uchicago.edu/QRB).

inheritance through self-sustainingloops

Self-sustaining feedback loops are metaboliccircuits through which different patternsof activity can be maintained, resulting inalternative heritable cell phenotypes. Thefirst experimental studies of such loopswere those involving the bistability of thelac operon of Escherichia coli (Novick andWeiner 1957), and this system has sincebeen thoroughly analyzed at both the mo-lecular and theoretical levels (Laurentet al. 2005). These studies show that, wheninducer concentrations are low, geneticallyidentical cells can generate two alternative,true-breeding, stable phenotypes.

Many other self-sustaining feedback loopsleading to alternative heritable phenotypeshave been described in bacteria and othertaxa (Dubnau and Losick 2006; Smits et al.2006; Malagnac and Silar 2003, 2006). Onewell-characterized example of positive regu-lation leading to alternative cell phenotypes

is found in the fungus Candida albicans,where an epigenetic switch underlies thetransition between white and opaquecells—two states that are heritable for manygenerations (Zordan et al. 2006). Self-sustaining loops need not be based on tran-scriptional regulation; they can also occur atthe post-translation stage, through proteinself-processing. An example is the enzymevacuolar protease B of Saccharomyces cerevisiae,whose active form is necessary for its ownconversion from an inactive precursor to anactive state. On glycerol media, where theprecursor is synthesized in high amounts,the self-processing of the enzyme is indefi-nitely self-sustaining (Roberts and Wickner2003). As noted by Wickner et al. (2004), itis likely that other protein-modifying pro-teins may also directly or indirectly affecttheir own modification and behave as self-sustaining, cell-transmissible loops.

structural inheritance:spatial templating

Structural inheritance refers to the inher-itance of alternative three-dimensional(3-D) structures through spatial templat-ing: a variant 3-D structure in a mother cellguides the formation of a similar structurefor a daughter cell, leading to the transmis-sion of the architectural variant (Nanney1968). The study of cellular structural in-heritance was initiated by investigations ofthe inheritance of cortical variations inciliates. Beisson and Sonneborn (1965)showed that an experimentally modifiedorganization of the cilia on Paramecium canbe inherited through many asexual andsexual generations. The inheritance of cor-tical variations induced by various physicaland chemical manipulations has also beendemonstrated for other ciliates (Nanney1985; Frankel 1989; Grimes and Aufder-heide 1991).

The propagation of prions is anotherform of structural inheritance. The funda-mental characteristic of a prion—a trans-missible protein—is that it has a conforma-tion that can initiate and sustain the re-production of a similar conformation innewly synthesized proteins. Prusiner (1998)suggested that infectious proteins are the

134 Volume 84THE QUARTERLY REVIEW OF BIOLOGY

causative agents of mammalian neurode-generative diseases such as kuru, scrapie,and mad cow disease and coined the termprion (proteineceous infectious particles)for such agents. He advanced and devel-oped the concept of an infectious proteinwhose propagation is based on the spatial-templating of the variant prion conforma-tion, which, in turn, converts a normally-shaped protein into its own shape. Theconcept was extended when Wickner (1994)characterized the [URE3] variant in yeastas a prion and explained its idiosyncraticbiochemical and hereditary characteristicsin terms of protein-based inheritance.Since then, more prions have been identi-fied in yeast and other fungi (Baxa et al.2006; Tuite and Cox 2006). Although someprions have deleterious effects, othersseem to have important biological func-tions (Wickner et al. 2004; Shorter andLindquist 2005; Benkemoun and Saupe2006).

One of the most interesting findingsabout prions is that a single protein canmisfold into several different conforma-tions that have specific growth dynamics,stabilities, pathologies, and cross-speciesinfectivity (Chien et al. 2004). Differentprions may interact, leading to the forma-tion of many different transmissible (cell-heritable and infectious) phenotypes. There-fore, unicellular organisms, which have thesame genotype and live in the same en-vironment, can exhibit heritably differ-ent morphologies and physiologies thatare the consequence of differently foldedidentical proteins. The differences resultfrom differences in the developmentalhistories of their ancestors.

Cavalier-Smith (2004) studied anotheraspect of spatial templating, an aspectassociated with membrane reproduction.The reproduction of most membranes, in-cluding the plasma membrane, the endo-plasmic reticulum, and the mitochondrialmembrane, requires the presence and tem-plating of pre-existing membrane struc-tures. Cavalier-Smith identified 18 types ofwhat he called “genetic membranes,” thestructures of which are cell-heritablethrough these guided processes, and he

argued that the information embedded inthis “membranome” is as essential for theconstruction of a cell as genomic informa-tion. He suggested that crucial events inthe evolution of cells and major groupswere associated with heritable changes inmembranomes.

the chromatin-marking EISChromatin, the stuff of chromosomes,

includes DNA and all the factors physicallyassociated with it: small chemical groupscovalently attached to DNA (e.g., methylgroups), bound histone and nonhistoneproteins, and associated RNA molecules.The organization of chromatin and chro-mosomes, their localization in the nucleus,and the dynamic interactions among thevarious components of chromatin deter-mine when, where, and to what extentgenes are transcribed, how DNA repair isorchestrated, how different chromosomaldomains are organized, and how chromo-somes, as units, behave during the vari-ous phases of the cell cycle. Patterns ofchromatin are reconstructed followingDNA transcription and replication, and, al-though the processes of reconstruction arenot well understood, there is evidence thatchromatin variations can be transmittedbetween generations of individuals. Thestudy of the chromatin-marking EIS istherefore crucial for the understanding ofboth development and heredity.

DNA methylation, the best-understoodsystem of chromatin inheritance, is an epi-genetic modification found in Eubacteria,Archeabacteria, and Eukaryota. It is in-volved in many important functions (forgeneral reviews see Casadesus and Low2006; Vanyushin 2006a,b), including de-fence against genomic parasites (Kidwelland Lisch 1997), regulation and mainte-nance of gene activity patterns (Barlow andBartolomei 2007; Li and Bird 2007), stabi-lization of chromosomal structure (Karpenand Hawley 2007), and DNA replicationand repair (Mortusewicz et al. 2005; Scher-melleh et al. 2007). In eukaryotes, methyl-ation usually occurs on the cytosines inCpG doublets and also in CpNpG tripletsin plants. Since CpG and CpNpG se-

June 2009 135TRANSGENERATIONAL EPIGENETIC INHERITANCE

quences are palindromic, the two strandsof the duplex are mirror images of eachother. Following replication, two hemi-methylated duplexes are formed, andthese hemimethylated regions are recog-nized by maintenance DNA methylases,which preferentially add methyls to non-methylated C in the new strand (Allshireand Selker 2007; Henderson and Jacobsen2007; Li and Bird 2007). The replication ofmethylation patterns is thus semi-conservative.The fidelity of transmission in cell lineagesranges from about 1% variation per cellgeneration up to the very high fidelity of10-6 variations per cell generation (Gene-reux et al. 2005; Richards 2006).

Variations in a DNA methylation patterncan also be inherited between generations,and examples of this include paramuta-tions in plants (interaction between allelesthat leads to a directed epigenetic herita-ble change in one of the interacting alleles[Brink 1973; Chandler 2007]); silencingof foreign duplicated sequences in fungi(Allshire and Selker 2007); variations intelomeric, centromeric, and rRNA regions(Karpen and Hawley 2007); and variationsin transgenes and endogenous genes. Spe-cific examples of all these types of trans-generational inheritance are presented inTable 1.

How a DNA methylation pattern affectsa cell’s or an organism’s phenotype de-pends on the way it interacts with the pro-tein components of the chromosome,which are also heritable. In eukaryotes, im-portant variations in chromatin are associ-ated with histones, the proteins that makeup the nucleosome core around whichDNA is wrapped. The dynamic nature ofhistones, their variability, and their associ-ation with every conceivable cellular func-tion (see Berger 2007; Groth et al. 2007;Blasco 2007; Morris and Moazed 2007)makes understanding the inheritance oftheir specific structure and organizationboth challenging and urgent. Several mod-els of this process have been constructed,and according to all of them, the nucleo-some variants and the post-transcriptionalmodifications (PTMs) of the parental nu-cleosomes are used as blueprints for the

restoration of the same nucleosome con-figuration. These reconstructions may beassisted by other chromatin componentssuch as DNA methylation patterns (Martinand Zhang 2007), RNAs (Grewal and Jia2007; Ringrose and Paro 2007), and by thelocation of the chromosomal domainswithin the nucleus (Misteli 2007).

In addition to DNA methylation patterns,as well as histone variants and their PTMs,chromatin has various enzymes that associatedynamically with the histones and DNAbound to them and that participate in theirregulatory and structural functions (Bantig-nies and Cavalli 2006). The patterns of asso-ciation of these nonhistone proteins withother chromatin components can also bereconstructed between generations of cellsand organisms (Ringrose and Paro 2004;Bantignies and Cavalli 2006; Schuettengru-ber et al. 2007). Recent data suggest thatRNA produced at regions that bind non-histone chromatin proteins may lead to theinheritance of the bound state throughRNA-DNA pairing (Grewal and Jia 2007;Ringrose and Paro 2007).

the RNA-mediated EISDuring the last decade, it has become

apparent that RNA is central to the regu-lation of cellular dynamics in eukaryotesand is also involved in cell and organismheredity. Gene silencing by small RNAs—RNA interference (RNAi)—has been foundin all eukaryote phyla from yeast to man,although a few species (e.g., buddingyeast) seem to have lost the capacity forthis. In all of the RNAi pathways, double-stranded RNA molecules (dsRNA), whichtrigger the process, are chopped intoshorter dsRNAs (usually between 21-30 nu-cleotides long) by the enzyme Dicer. Af-ter the original dsRNA is chopped, the re-sulting siRNA (small interfering RNA) isloaded onto a complex of proteins, onestrand of the duplex is removed, and theother strand—the guide strand—directs si-lencing. Silencing may occur through anyof the following mechanisms: (i) the siRNAis loaded onto an enzyme complex thatinterferes with the transcription or transla-tion of mRNAs with a homologous se-

136 Volume 84THE QUARTERLY REVIEW OF BIOLOGY

quence (Cullen 2004; Meister and Tuschl2004); (ii) the siRNA is loaded onto anenzyme complex that targets chromatin re-gions with DNA that is homologous to thesiRNA, and alters chromatin into a silentstate (Matzke and Birchler 2005; Ekwall2007; Huettel et al. 2007); or (iii) thesiRNA is loaded onto an enzymatic com-plex that degrades and/or excises the DNAsequences complementary to the siRNAs.The latter processes of RNA-regulatedDNA rearrangement are being intenselystudied in ciliates (Meyer and Chalker2007; Nowacki et al. 2008).

RNA can affect cell and organism hered-ity in at least three different, non-mutuallyexclusive ways. The first is the result of rep-lication of siRNA through RNA-dependentRNA polymerase. This replication is a two-stage process, the details of which differbetween taxa, and it leads to the amplifica-tion of siRNAs that act as repressors ofgene expression (Baulcombe 2007; Pak andFire 2007). These siRNAs are transmittedto daughter cells during cell division andcan migrate to other cells as well (e.g.,Palauqui et al. 1997; Himber et al. 2003).The second way in which RNAs can af-fect cell heredity is by guiding, targeting,and assisting in transmitting variations inchromatin structure that are reconstructedand reproduced in daughter cells throughthe chromatin-marking EIS (Matzke andBirchler 2005; Ekwall 2007; Huettel et al.2007). The third way is by targeting DNAbase sequences and guiding changes inthem that are then replicated by DNA poly-merases (Meyer and Chalker 2007; Nowackiet al. 2008). Heritable variations can be gen-erated and perpetuated through all threeroutes of RNA-mediated heredity, and theformation of a particular dsRNA may be af-fected by local conditions and may be devel-opmentally regulated. Once formed, suchdsRNA may have long-term hereditary ef-fects.

EISs and the germlineIt is obvious from the foregoing descrip-

tions that the different EISs are oftenmechanistically and functionally interre-lated; therefore, the division of EISs into

four categories is somewhat arbitrary andartificial. However, dividing EISs in thismanner highlights the variety and com-plexity of the cellular mechanisms of in-heritance. The details of how epigeneticvariations are transmitted through mitosisin asexual clonal lineages remain a puzzle,but in sexually reproducing organisms,especially multicellular ones, the puzzle iseven greater.

In sexually reproducing organisms, epi-genetic variations have to survive the com-plex process of meiosis in order to be trans-mitted to the next generation, and, inmulticellular organisms, they also have tosurvive gametogenesis and early embryo-genesis—two developmental stages that in-volve significant restructuring of both cellsand chromatin. Although there is as yet noevidence that prions and self-sustainingloops are transmitted between generationsthrough sperm and egg, there is evidencethat chromatin marks and RNAs can betransmitted in this manner, but it is notclear how this occurs. It seems likely thatsome footprints of chromatin marks re-main and lead to the reconstruction ofancestral states, or that some remnants ofancestral states (including some RNAs) areretained. Even in male vertebrates, where acomprehensive replacement of histones byprotamines takes place during gametogen-esis, the erasure of histone marks is notcomplete. In the mouse, for example,about 1% of the DNA remains wrappedaround histones, and two acetylated vari-ants of H4 are maintained through sper-miogenesis (Van der Heijden et al. 2006).Van der Heijden and his colleagues (2006)suggested that these histones, as well asmethylated cytosines in centromeric DNA,are associated with the transgenerationalmaintenance of the structure of centro-meric heterochromatin. Chong et al. (2007)reported that when male mice had muta-tions in genes involved in epigenetic pro-gramming—in a gene that encodes a chro-matin remodeler protein, and in anotherthat encodes DNA methyltransferase—there were phenotypic effects on theiroffspring, even when they did not inheritthe defective gene. Extensive epigenetic

June 2009 137TRANSGENERATIONAL EPIGENETIC INHERITANCE

(methylation) variation has been found inhuman germ cells (Flanagan et al. 2006),but whether and to what extent this varia-tion is passed between generations is notknown.

In addition to some chromatin marks,certain RNAs may be transmitted throughthe germline. Cells in the germline containsmall RNAs, known as Piwi-associated inter-fering RNAs (piRNAs), that are importantfor the proper maturation of germ cells(Kim 2006). One function of piRNAs maybe the detection and silencing—or dele-tion—of regions of unpaired DNA duringmeiosis. Unpaired regions are targets ofthe RNAi machinery; RNA transcribedfrom them guides enzymatic complexes tothe unpaired regions, which are then de-leted or heterochromatinized (Shiu et al.2001; Bean et al. 2004; Turner et al. 2005;Costa et al. 2006). Mammalian spermato-cytes are filled with piRNAs, and similarRNAs have been discovered in oocytes too.The abundance of piRNAs suggests that theymay be transmitted to the next generationand lead to transgenerational effects. Thetransmission of an epigenetic modificationin male mice (induced in heterozygotes for avariant Kit gene) is known to be mediatedthrough microRNAs with sequences partiallycomplementary to that of Kit RNA (Rassoul-zadegan et al. 2006; see supplementarymaterial for details, available online at TheQuarterly Review of Biology homepage, www.journals.uchicago.edu/QRB).

Transgenerational EpigeneticInheritance: Prevalence,

Distribution, and InductionThere is no recent review of cellular epi-

genetic inheritance between generationsthat encompasses all four types of EISs andtheir distributions across taxa. The onlycomprehensive survey was made by Jab-lonka and Lamb in 1995. Since then, manymore cases have been described, and ourunderstanding of the molecular mecha-nisms underlying epigenetic inheritancehas deepened and expanded. The table wepresent here (Table 1) brings togetherover one hundred cases of inherited epige-netic variations in bacteria, protists, fungi,

plants, and animals. We have included onlycases where there is convincing evidencefor epigenetic inheritance in the narrowcellular sense, either through a single asex-ual cell (as in bacteria, some protists, somefungi, and some plants) or through a sex-ually generated gamete or spore (most an-imals, plants, and fungi). In most (al-though not all) cases, molecular studieshave revealed the involvement of one ormore of the EISs, but a full molecular char-acterization through all the reproductivestages is not yet available for any organism.More details about the cases are given assupplementary material in an expanded ta-ble form (available online at The QuarterlyReview of Biology homepage, www.journals.uchicago.edu/QRB).

How common is epigenetic inheritance?This question is often raised, and the an-swer, based upon the data we have accu-mulated, is that it may be ubiquitous. Webelieve that epigenetic variants in every lo-cus in the eukaryotic genome can be inher-ited, but in what manner, for how long,and under what conditions has yet to bequalified. In other words, unlike the repli-cation of DNA variations, which is largelycontext insensitive, whether and for howlong a particular mark or cellular elementis transmitted between generations de-pends on genomic, developmental, and ex-ternal conditions. This does not mean thatconditions that allow epigenetic inheri-tance are particularly rare. In fungi, forexample, the widespread occurrence ofepigenetic inheritance is generally ac-knowledged. As long ago as 1949, Linde-gren, reviewing data on inheritance in Neu-rospora, said that two thirds of new variantsdo not show Mendelian segregation andare therefore discarded, and, in their re-cent review of protein inheritance in fungi,Benkemoun and Saupe (2006) comment-ed: “Many of us working with filamentousfungi know how often bizarre looking sectors orsegregates that defy Mendelism appear on ourplates. As a number of pioneering fungal genet-icists have done in the past, maybe we shouldhave a closer look before putting them in theautoclave” (p. 793). They suggested thatmany of these “anomalies” may be caused

138 Volume 84THE QUARTERLY REVIEW OF BIOLOGY

by some form of epigenetic inheritance. Inall organisms, chromatin inheritance cantheoretically occur at every locus in thegenome, and the double-stranded natureof DNA provides a theoretical possibility(when transcription occurs from bothstrands) for every DNA segment to formsmall dsRNA molecules that can lead toheritable silencing. The potential of mostproteins to form � sheets with spatialtemplating properties (Baxa et al. 2006)suggests that a prion-like transfer of con-formation between cells may occur morefrequently than previously thought.

cases included in the tableIn Table 1, we show only a small sample

of representative cases of epigenetic inher-itance in allopolyploid plant hybrids be-cause, in all cases that have been investi-gated, allopolyploidy is accompanied byextensive epigenetic changes, some ofwhich are inherited between generations.In most of the cases presented in the table,the molecular basis of the EIS has beenunravelled, but we included a few cases forwhich the evidence for cellular epigeneticinheritance seemed overwhelming, despitethe fact that molecular studies were lacking(e.g., inheritance of the star phenotype insilver foxes, and some cases of develop-mentally induced variability and inheri-tance in plants and fungi).

As our main goal in this review is to showthat variations in epigenetic marks can beinherited for several generations, we didnot include classical cases of genomic im-printing in the table. With genomic im-printing, the epigenetic marks that are im-posed on parental chromosomes duringoogenesis differ from those imposed dur-ing spermatogenesis; therefore, in the off-spring, a gene’s expression pattern de-pends on whether it was inherited from thefather or from the mother (see Barlow andBartolomei 2007 for genomic imprintingin mammals, Alleman and Doctor 2000 forflowering plants, and for general reviews ofimprinting that include invertebrates, seeJablonka and Lamb 1995; Haig 2002). Bydefinition, the genomic imprints charac-terizing one sex are reversed when chro-

mosomes go through gametogenesis in theopposite sex in the next generation; thus,imprints are inherently reversible. How-ever, molecular studies of imprinting havebeen important in the history of epigeneticinheritance because they have led to a gen-eral recognition of the role of epigeneticcontrol mechanisms, such as changes inDNA methylation in critical regions, andhave opened the way to unraveling casesthat were similar to genomic imprintingbut that were not parent-sex specific (e.g.,the early studies by Hadchouel et al.1987 and Allen et al. 1990, as discussedin Jablonka and Lamb 1995). The onlyimprinting-related case included in the ta-ble is that of an imprint in a human grand-mother that was not erased in her son andthat subsequently led to Prader-Willi syn-drome in her grandchildren (Buiting et al.2003). This is a clear case of an epigeneticimprinting mark being transmitted as a re-sult of a fault in resetting. As yet, we are notaware of any studies showing that new vari-ations in genomic imprints are transmittedover several consecutive generations.

In most of the cases that we included inthe table, the epigenetic variations havebeen transmitted for more than two gen-erations, and we can rule out repeateddirect induction of the variation in eachgeneration. However, when the inducedparent carries an embryo, three genera-tions may be necessary to confirm epige-netic inheritance. For example, when a fe-male mammal is exposed to an inducingagent during pregnancy, direct inductionof the embryo’s germ cells has to be ex-cluded; therefore, three generations oftransmission (from F0 to F3) are requiredto establish that epigenetic inheritance,rather than direct induction of the embryoand its germline, has occurred (Jirtle andSkinner 2007). On the other hand, when amale mammal is exposed to an inducer,two generations of transmission (from F0

to F2) are sufficient to establish that therehas been epigenetic transmission throughthe germline rather than through directinduction. When the epigenetic effects(e.g., patterns of methylation) in the F1

and F2 generations are identical, and when

June 2009 139TRANSGENERATIONAL EPIGENETIC INHERITANCE

TA

BL

E1

Tra

nsge

nera

tiona

lep

igen

etic

inhe

rita

nce

inpr

okar

yote

san

deu

kary

otes

Tax

onT

rait

Loc

us/c

ellu

lar

syst

emSt

abili

tyIn

duci

ngco

ndit

ions

EIS

Ref

eren

ce

Bac

teri

aan

dth

eir

viru

ses

�ph

age

ofEs

cher

ichia

coli

Lys

ogen

ic/l

ytic

cycl

eC

lan

dC

roSt

able

Nut

riti

onal

stat

eof

the

hos

t,ph

age

den

sity

Self

-sus

tain

ing

loop

sPt

ash

ne

(200

5)

Bac

illus

subt

ilis

Inac

tiva

tion

ofch

rom

osom

eW

hol

ech

rom

osom

eSt

able

Poly

eth

ylen

glyc

ol(P

EG

)-in

duce

dfu

sion

Ch

rom

atin

-mar

kin

gG

ran

djea

net

al.

(199

8)

Lac

kof

cell

wal

lB

alan

cebe

twee

npe

ptid

ogly

can

syn

thes

isan

dde

stru

ctio

nSt

able

onag

arEx

peri

men

talr

emov

alof

cell

wal

lSt

ruct

ural

inh

erit

ance

Lan

dman

(199

1)

Spor

ulat

ion

Spo0

Aph

osph

orel

aySt

able

Nut

riti

onal

depr

ivat

ion

Self

-sus

tain

ing

loop

sV

een

ing

etal

.(2

005)

Nat

ural

com

pete

nce

(Kst

ate)

Com

Kac

tivi

ty10

%–2

0%in

lab

stra

ins,

1%in

the

wild

Stoc

has

tic,

elev

ated

byst

ress

Self

-sus

tain

ing

loop

sM

aam

aran

dD

ubna

u(2

005)

Esch

eric

hia

coli

Uti

lizat

ion

ofla

ctos

ea

Lac

oper

onac

tivi

tySt

able

unde

rco

ndi

tion

sof

low

indu

cer

con

cen

trat

ion

Stoc

hast

ic,g

row

thin

low

conc

entr

atio

nof

indu

cer

Self

-sus

tain

ing

loop

sC

ohn

and

Hor

ibat

a(1

959a

,b),

Lau

rent

etal

.(2

005)

,Nov

ick

and

Wei

ner

(195

7),O

zbud

aket

al.(

2004

)Fl

uffy

Agn

43Ph

ase

vari

atio

nPr

obab

lyox

idat

ive

stre

ssG

AT

Cm

eth

ylat

ion

Cas

ades

usan

dL

ow(2

006)

Pili

Pap

oper

on10

-4pe

rge

ner

atio

n,

10-3

reve

rsio

nC

han

ged

carb

onso

urce

,te

mpe

ratu

re,

and

spon

tan

eous

GA

TC

met

hyl

atio

nH

ern

day

etal

.(2

002)

Gro

wth

rate

(per

sist

erty

peII

)b

Prob

ably

man

yge

nes

10-6

per

gen

erat

ion

,10

-1pe

rge

ner

atio

nre

vers

ion

Spon

tan

eous

and

anti

biot

ictr

eatm

ent

Self

-sus

tain

ing

loop

sB

alab

anet

al.(

2004

),L

ewis

(200

7),N

.Bal

aban

(per

sona

lco

mm

unic

atio

n)R

esist

ance

toan

tibio

tics

(am

pici

llin,

tetr

acyc

line,

and

nalid

ixic

acid

)

Alt

ered

regu

lati

onof

lact

amas

ecr

ypti

cge

ne,

glut

amat

ege

ne,

and

deca

rbox

ylas

ege

ne;

poss

ible

invo

lvem

ent

ofD

NA

met

hyl

ase

gen

es

3%–2

0%su

rviv

al(d

epen

din

gon

con

cen

trat

ion

ofan

tibi

otic

)an

d50

%re

vers

ion

rate

Low

,and

succ

essi

vely

incr

ease

dco

ncen

trat

ions

ofan

tibio

tics

Poss

ibly

self

-sus

tain

ing

loop

san

d/o

rD

NA

met

hyl

atio

n

Ada

met

al.

(200

8)

Pseu

dom

onas

aeru

gino

saT

oxin

inje

ctio

nT

TSS

syst

emSt

able

Cel

lde

nsi

tySe

lf-s

usta

inin

glo

ops

Filo

pon

etal

.(2

006)

140 Volume 84THE QUARTERLY REVIEW OF BIOLOGY

Syne

coco

ccus

elon

gate

s(C

yan

obac

teri

a)

Cir

cadi

anrh

yth

mR

egul

ator

ylo

opin

volv

ing

key

Kai

Cpr

otei

nSe

vera

lda

ysL

igh

tan

dda

rkpu

lses

Self

-sus

tain

ing

loop

sK

ondo

and

Ish

iura

(200

0)

Pro

tist

sO

xytr

icha

trifa

llax

(Cili

ate)

Alt

erat

ion

ofge

ne

orde

r:ab

erra

nt

rear

ran

gem

ents

Gen

esth

atbe

com

eun

scra

mbl

edin

the

som

atic

mac

ronu

cleu

s(i

.e.,

mos

tge

nes)

Stab

leth

roug

has

exua

lre

prod

uctio

n;at

leas

t3ge

nera

tions

follo

win

gse

xual

repr

oduc

tion

Exp

erim

enta

lm

anip

ulat

ion

RN

A-m

edia

ted

rear

ran

gem

ent

Now

acki

etal

.(2

008)

Para

mec

ium

aure

lia(C

iliat

e)

Sero

type

sex

pres

sed

Cili

apr

otei

ns

Stab

leC

han

ges

inpH

,te

mpe

ratu

re,

food

supp

ly,

and

salin

ity

Self

-sus

tain

ing

loop

sL

andm

an(1

991)

Indu

ced

tole

ran

ceto

hea

t,sa

lt,

and

arse

nic

Not

spec

ified

Inh

erit

edfo

rm

any

gen

erat

ion

sbe

fore

fadi

ng

away

grad

ually

Exp

osur

eto

hig

hte

mpe

ratu

re,

hig

hsa

lt,

and

arse

nic

con

cen

trat

ion

s

Not

know

nJo

llos

(192

1);

revi

ewed

inJa

blon

kaet

al.

(199

2)

Para

mec

ium

tetr

aure

lia,

Styl

onch

iale

mna

e,T

etra

hym

ena

ther

mop

hila

(Cili

ates

)c

Var

ious

trai

tsre

late

dto

alte

rnat

ive

gen

etic

orga

niz

atio

npa

tter

ns

inth

em

acro

nuc

leus

Inpr

inci

pal,

any

DN

Ase

quen

cein

the

gen

ome

Stab

leth

roug

hm

acro

nuc

leus

repr

oduc

tion

,bo

thdu

rin

gas

exua

lan

dfo

llow

ing

sexu

alre

prod

ucti

on

Sequ

ence

com

pari

son

ofm

ater

nal

(old

)an

dzy

goti

c(n

ew)

nuc

leus

,ba

sed

onn

on-

expr

esse

dse

quen

ces,

lead

sto

the

inh

erit

ance

ofm

ater

nal

,n

ucle

us-

guid

edge

ne

orga

niz

atio

n

Inhe

rite

dD

NA

rear

rang

emen

t/ed

iting

/pr

ogra

mm

ing

ofth

em

acro

nucl

eus

med

iate

dby

smal

lR

NA

san

dch

rom

atin

mod

ifica

tions

Gar

nie

ret

al.

(200

4),

Jura

nek

etal

.(2

005)

,L

iuet

al.

(200

4),

Mey

eran

dC

hal

ker

(200

7),

Tav

ern

aet

al.

(200

2),

Yao

etal

.(2

003)

Cor

tica

lor

gan

izat

ion

Bas

albo

dyan

dco

rtex

prot

ein

sSt

able

inm

itos

isan

dso

met

imes

inm

eios

is

Exp

erim

enta

lm

anip

ulat

ion

,st

ress

Stru

ctur

alin

her

itan

ce(g

uide

das

sem

bly)

Gri

mes

and

Auf

derh

eide

(199

1)

Plas

mod

ium

falc

ipar

um(M

alar

iapa

rasi

te)

Tel

omer

ein

acti

vati

onT

elom

ere

sequ

ence

sSw

itch

ever

y�

15ge

ner

atio

ns

Spon

tan

eous

Ch

rom

atin

-mar

kin

gR

ober

tset

al.

(199

2) cont

inue

d

June 2009 141TRANSGENERATIONAL EPIGENETIC INHERITANCE

TA

BL

E1

Con

tinue

d

Tax

onT

rait

Loc

us/c

ellu

lar

syst

emSt

abili

tyIn

duci

ngco

ndit

ions

EIS

Ref

eren

ce

Tet

rahy

men

a(C

iliat

e)In

crea

sed

insu

linbi

ndin

gan

dpr

oduc

tion

follo

win

gex

posu

reto

diio

doty

rosin

e(T

2)

Not

spec

ified

Ver

yst

able

—hu

ndre

dsof

gene

ratio

nsIn

duct

ion

byin

sulin

ordi

iodo

tyro

sin

ePr

obab

lym

eth

ylat

ion

;tr

eatm

ent

wit

h5-

azac

ytid

ine

can

abol

ish

resp

onse

Csa

ba(2

008)

,Csa

baet

al.

(199

9),C

saba

and

Kov

acs

(199

0,19

95),

Csa

baet

al.

(198

2a,b

)

Volv

oxca

rter

iSi

lenc

ing

ofin

vitro

met

hyla

ted

tran

sgen

e

C-a

rstr

ansg

ene

Mor

eth

an10

0ge

ner

atio

ns

DN

Atr

ansf

orm

atio

nD

NA

met

hyl

atio

nB

abin

ger

etal

.(2

007)

Fung

iA

scob

olus

imm

erse

sT

ran

sgen

ein

acti

vati

onA

ny

dupl

icat

edtr

ansg

ene

Stab

lePr

e-m

eiot

ical

lyD

NA

met

hyl

atio

nM

artie

nsse

nan

dC

olot

(200

1),R

houn

imet

al.

(199

2)C

andi

daal

bica

nsd

Cel

lmor

phol

ogy,

abili

tyto

form

colo

nies

onva

riou

ssu

bstr

ates

,mat

ing

prop

ertie

s

Mas

ter

regu

lato

rW

OR

1pr

otei

nSw

itch

esev

ery

10,0

00ge

ner

atio

ns

Spon

tan

eous

,af

fect

edby

tem

pera

ture

Self

-sus

tain

ing

loop

Hua

ng

etal

.(2

006)

,M

alag

nac

and

Sila

r(2

003)

,Z

orda

net

al.

(200

6)

Cop

rinu

sci

nere

usM

eth

ylat

ion

patt

ern

Cen

trom

ere-

linke

dlo

cus

Stab

lew

hen

hig

hly

met

hyl

ated

Un

know

nC

hro

mat

in-m

arki

ng,

DN

Am

eth

ylat

ion

invo

lved

Zol

anan

dPu

kkila

(198

6)

Podo

spor

aan

seri

na(F

ilam

ento

usfu

ngu

s)

Cri

pple

dG

row

th(C

G)

C—

assu

med

tobe

atr

ansm

issi

ble

self-

sust

aini

ngca

scad

efa

ctor

invo

lvin

ga

Map

kina

sem

odul

e

Stab

lem

itot

icin

her

itan

ceT

rans

form

atio

nof

norm

alce

llsto

CG

cells

indu

ced

bycy

todu

ctio

n,pr

omot

ing

stat

iona

ryst

ate

and

grow

thon

med

ium

supp

lem

ente

dw

ithye

aste

xtra

ct

Self

-sus

tain

ing

loop

Kic

kaet

al(2

006)

,Mal

agna

can

dSi

lar

(200

6),S

ilar

etal

.(19

99)

[Het

-s*]

and

[Het

-s]va

rian

tsaf

fect

ing

vege

tativ

ein

com

patib

ility

het-s

Stab

le;

low

freq

uen

cy[H

et-s

]co

nve

rted

toin

acti

ve[H

et-

s*],

but

the

reve

rse

also

occu

rs,

albe

itat

low

freq

uen

cy

Spon

tan

eous

Stru

ctur

alin

her

itan

ce(p

rion

)M

adde

lein

etal

.(2

002)

142 Volume 84THE QUARTERLY REVIEW OF BIOLOGY

Sacc

haro

myc

esce

revi

siae

Enha

nced

resis

tanc

eto

star

vatio

non

dext

rose

-min

imal

plat

esan

dsu

ppre

ssio

nof

inab

ility

tosp

orul

ate

inm

utan

ts

[�]-

prio

nfo

rmof

the

PrB

vacu

olar

prot

ease

Inde

fin

ite

prop

agat

ion

Em

erge

sin

dele

tion

-mut

ants

;in

duct

ion

isin

crea

sed

asa

resu

ltof

PrB

over

-ex

pres

sion

Prio

n,

thro

ugh

self

-su

stai

nin

glo

opR

ober

tsan

dW

ickn

er(2

003)

Gro

wth

phen

otyp

eFL

Oge

nes

nea

rte

lom

ere

Swit

chev

ery

10to

15ge

ner

atio

ns

Spon

tan

eous

Ch

rom

atin

inh

erit

ance

Hal

me

etal

.(2

004)

Rea

dth

roug

h[P

S]St

able

Var

ious

Stru

ctur

alin

her

itan

ce(p

rion

)T

uite

and

Cox

(200

6)

Nit

roge

nca

tabo

licge

ne

expr

essi

on[U

RE

3]St

able

Spon

tan

eous

and

over

expr

essi

onSt

ruct

ural

inh

erit

ance

(pri

on)

Ben

kem

oun

and

Saup

e(2

006)

Asc

usfo

rmat

ion

[PIN

]�R

nq1

pSt

able

Spon

tan

eous

and

over

expr

essi

onof

Sup3

5St

ruct

ural

inh

erit

ance

(pri

on)

Ben

kem

oun

and

Saup

e(2

006)

Exp

ress

ion

ofex

peri

men

tally

mod

ified

GA

Ln

etw

ork

GA

Ln

etw

ork

Stab

leL

owin

duce

rco

nce

ntr

atio

ns

Self

-sus

tain

ing

loop

Aca

ret

al.

(200

5)

Indu

ctio

nof

GA

L1

and

GA

L7

byga

lact

ose

GA

Lge

nes

Up

to7

gen

erat

ion

sIn

duct

ion

byga

lact

ose

Slow

dilu

tion

ofab

unda

nt

regu

lato

ryG

AL

1pr

otei

n

Zach

ario

udak

iset

al.(

2007

)

An

ti-s

uppr

esso

rIS

P�?

Stab

leSp

onta

neo

usPr

obab

lyba

sed

onst

ruct

ural

inh

erit

ance

Ben

kem

oun

and

Saup

e(2

006)

,Kun

zan

dB

all

(197

7),T

allo

czy

etal

.(2

000)

,Vol

kov

etal

.(2

002)

Glu

cosa

min

ere

sist

ance

GR

?

Con

trol

ofki

ller

viru

sex

pres

sion

KI-

d?(s

uspe

cted

prio

ns)

Slow

grow

than

dad

diti

onal

requ

irem

ent

for

leuc

ine

Stru

ctur

alal

tera

tion

ofth

em

itoc

hon

drio

nM

ore

than

100

gen

erat

ion

sin

the

abse

nce

ofle

ucin

e

Spon

tan

eous

inh

erit

edlo

ssof

mit

och

ondr

ial

DN

A

Stru

ctur

alin

her

itan

ceL

ocks

hon

(200

2)

cont

inue

d

June 2009 143TRANSGENERATIONAL EPIGENETIC INHERITANCE

TA

BL

E1

Con

tinue

d

Tax

onT

rait

Loc

us/c

ellu

lar

syst

emSt

abili

tyIn

duci

ngco

ndit

ions

EIS

Ref

eren

ce

Schi

zosa

ccha

rom

yces

pom

beSu

rviv

alof

mut

ant

stra

inla

ckin

gth

ehc

dre

gion

-en

codi

ng,

hig

hly

con

serv

eddo

mai

nof

the

esse

nti

alch

aper

one

caln

exin

[cif

]C

alne

xin

inde

pend

ence

inhe

rite

dby

88.7

%of

spor

es

Mat

ing

cells

lack

ing

the

gene

codi

ngfo

rca

lnex

inw

ithca

lnex

in-

depe

nden

tce

lls

Stru

ctur

alin

her

itan

ce,

prob

ably

prio

nC

ollin

etal

.(2

004)

Mei

otic

telo

mer

ecl

uste

ring

and

chro

mat

inst

ruct

ure

Inte

ract

ion

ofte

lom

eric

and

subt

elom

eric

regi

ons

with

Taz

1St

able

Nor

mal

;var

iatio

nsh

own

byde

letin

gte

lom

ere

sequ

ence

sin

chro

mos

omes

Ch

rom

atin

-mar

kin

g,st

ruct

ural

inh

erit

ance

Sada

ieet

al.

(200

3)

Rep

orte

rtr

ansg

ene

sile

nci

ng

and

mat

ing

type

swit

chin

g

K�

::ura

4(t

ran

sgen

ein

sert

edat

K-r

egio

n)

At

leas

t30

mit

otic

gen

erat

ion

saf

ter

mei

osis

Tra

nsg

enic

ally

indu

ced

Ch

rom

atin

-org

aniz

ing

fact

ors

prob

ably

invo

lved

Gre

wal

and

Kla

r(1

996)

Pla

nts

Ant

irrhi

num

maj

us(S

napd

rago

n)

Flus

h/g

ran

ulat

edph

enot

ype

(par

amut

atio

n)

nive

alo

cus

Var

iega

ted

inF 1

,m

ore

stab

lein

F 2

Indu

ced

bycr

ossi

ng

Tra

nsp

osit

ion

sugg

este

d;ch

rom

atin

-mar

kin

gpl

ausi

ble

Kre

bber

set

al.

(198

7)

Ara

bido

psis

thal

iana

(Mou

seea

rcr

ess)

Rev

ersi

onof

dwar

fm

orph

olog

yC

pr1–

1ge

ne-

epia

llele

inte

ract

ing

wit

hba

lEf

fect

seen

inF 2

prog

eny

butn

otin

F 1

Indu

ced

bycr

ossi

ng

Un

know

nSt

okes

and

Ric

hard

s(2

002)

Size

,ros

ette

,and

petio

leab

norm

aliti

es;

activ

ityof

GFP

repo

rter

gene

Tra

nsg

enic

loci

E82

and

L91

inte

ract

ing

wit

hC

OP1

endo

gen

e

At

leas

t5

gen

erat

ion

sIn

duce

dby

cros

sin

gIn

volv

emen

tof

RN

A-

med

iate

dD

NA

met

hyl

atio

nsu

gges

ted

Qin

and

von

Arn

im(2

002)

Del

ayed

flow

erin

gfw

a(g

ene-

epia

llele

)V

ery

stab

leE

MS,

fast

neu

tron

trea

tmen

t,or

ddm

1in

duce

d(W

ildty

peD

DM

1re

quir

edto

mai

nta

inD

NA

met

hyl

atio

n)

DN

Am

eth

ylat

ion

,h

isto

ne

H3

met

hyl

atio

n(s

iRN

As

invo

lved

)

Kin

osh

ita

etal

.(2

007)

,L

ippm

anet

al.

(200

4),

Sopp

eet

al.

(200

0)

144 Volume 84THE QUARTERLY REVIEW OF BIOLOGY

Dw

arfis

m,c

onst

itutiv

eac

tivat

ion

ofde

fens

ere

spon

sepa

thw

ay

Bal

locu

sA

tle

ast

5ge

ner

atio

ns

ddm

1ef

fect

even

wh

ense

greg

ated

out

Prob

ably

DN

Am

eth

ylat

ion

Stok

eset

al.

(200

2)

Num

ber

ofre

prod

uctiv

eor

gans

SUP

At

leas

t2

gen

erat

ion

sV

ario

usm

utag

ens

DN

Am

eth

ylat

ion

Jaco

bsen

and

Mey

erow

itz

(199

7)Ex

pres

sion

leve

lsof

the

retr

otra

nspo

son

At2

g104

10

At2

g104

10ep

ialle

leA

tle

ast

8ge

ner

atio

ns

Spon

tan

eous

,fo

und

inn

atur

alpo

pula

tion

sD

NA

met

hyl

atio

nR

angw

ala

etal

.(2

006)

Incr

ease

dle

vels

ofho

mol

ogou

sre

com

bina

tion

inso

ma

Not

spec

ified

Atl

east

4ge

nera

tions

inca

seof

UV

-Cra

diat

ion;

atle

ast2

gene

ratio

nsin

case

ofin

trod

uced

flage

llin

Ult

ravi

olet

radi

atio

nor

intr

oduc

tion

offl

agel

lin

Not

spec

ified

Mol

inie

ret

al.

(200

6),

J.M

olin

ier

(per

son

alco

mm

unic

atio

n)

Los

sof

hyg

rom

ycin

resi

stan

ce(p

aram

utat

ion

)

hpt

tran

sgen

eN

oef

fect

inF 1

;F 2

affe

cted

Tra

nsg

ene-

indu

ced

Invo

lves

DN

Am

eth

ylat

ion

Sch

eid

etal

.(2

003)

Tra

nsc

ript

ion

alac

tivi

tyof

tran

spos

able

elem

ents

Var

ious

tran

spos

able

elem

ents

At

leas

t6

gen

erat

ion

sdd

m1

effe

ct,

even

afte

rse

greg

ated

out

DN

Am

eth

ylat

ion

,ch

rom

atin

mod

ifica

tion

s;R

NA

ipr

obab

lyin

volv

ed

Lipp

man

etal

.(20

03,2

004)

,V

.Col

ot(p

erso

nal

com

mun

icat

ion)

Blu

efl

uore

scen

ce,

tryp

toph

anan

dIA

Ade

fici

ency

,m

orph

olog

ical

abn

orm

alit

ies

PAI

(gen

e-ep

ialle

les)

At

leas

t6

gen

erat

ion

sT

-DN

Am

utag

enes

is,

cros

sin

g;va

riat

ion

foun

din

nat

ural

popu

lati

ons

DN

Am

ethy

latio

n;D

NA

/D

NA

pair

ing

issu

gges

ted

Ben

der

and

Fin

k(1

995)

,L

uff

etal

.(1

999)

Ara

bido

psis

Inte

rspe

cfic

hyb

rids

e

Man

ytr

aits

Man

ylo

ci,

both

codi

ng

and

non

codi

ng

Stab

ility

vari

esac

cord

ing

tolo

cus,

but

man

ylo

cish

owst

able

inh

erit

ance

Indu

ced

byh

ybri

diza

tion

follo

wed

bypo

lypl

oidi

zati

on

Ch

rom

atin

-mar

kin

g,D

NA

met

hyl

atio

n;

RN

Ai

syst

empr

obab

lyal

soin

volv

ed

Com

aiet

al.

(200

0),

Sch

eid

etal

.(2

003)

cont

inue

d

June 2009 145TRANSGENERATIONAL EPIGENETIC INHERITANCE

TA

BL

E1

Con

tinue

d

Tax

onT

rait

Loc

us/c

ellu

lar

syst

emSt

abili

tyIn

duci

ngco

ndit

ions

EIS

Ref

eren

ce

Beta

vulg

aris

(Sug

arbe

et)

Man

ytr

aits

inm

itotic

,ag

amos

perm

ic,a

ndin

bred

lines

incl

udin

g:sin

gle

orm

ultip

leflo

wer

initi

atio

n,se

lf-fe

rtili

ty,

poly

mor

phism

ofm

alic

enzy

me,

and

vari

atio

nin

num

ber

ofch

loro

plas

ts

Mm

,li,

Rf1

,an

dM

e1lo

ciV

aria

ble,

ofte

n2

orm

ore

gen

erat

ion

sM

ode

ofre

prod

ucti

on,

clim

atic

con

diti

ons,

dire

ctio

nof

cros

s

DN

Am

eth

ylat

ion

ispr

obab

lyin

volv

edL

evit

es(2

000)

,L

evit

esan

dM

alet

skii

(199

9),

Mal

etsk

ii(1

999)

Lin

aria

vulg

aris

(Com

mon

toad

flax

)

Flow

ersy

mm

etry

Lcy

c(g

ene-

epia

llele

)A

tle

ast

2ge

ner

atio

ns

Nat

ural

vari

atio

nin

popu

lati

ons

DN

Am

eth

ylat

ion

Cub

aset

al.

(199

9),

J.Pa

rker

(per

son

alco

mm

unic

atio

n)

Lin

um usita

tissi

mum

(Fla

x)

Plan

twei

ght,

heig

ht,

pero

xida

seiso

zym

epa

ttern

,see

dca

psul

ese

pta

hair

num

ber

r-D

NA

gen

esan

dre

peti

tive

sequ

ence

sSt

able

Fert

ilize

ran

dh

eat

regi

mes

Met

hyl

atio

nan

dD

NA

-re

patt

ern

ing

Cul

lis(2

005)

Flow

erin

gag

e,m

ain

stem

heig

htat

mat

urity

,and

num

ber

ofle

aves

Not

spec

ified

,bu

tep

imut

atio

ns

inat

leas

t3

inde

pen

den

t,n

onra

ndo

mlo

ciar

eas

sum

edto

bein

volv

ed

At

leas

t9

gen

erat

ion

s5-

azaC

trea

tmen

tD

NA

met

hyl

atio

n,

poss

ibly

asso

ciat

edch

rom

atin

rem

odel

ing

asw

ell

Fiel

des

and

Am

yot

(199

9),

Fiel

des

etal

.(2

005)

Lyc

oper

sico

nes

cule

ntum

(Tom

ato)

Inhi

bitio

nof

ripe

ning

and

deve

lopm

ento

fa

colo

rles

spe

rica

rp

KeS

PL-C

NR

(gen

eep

ialle

le)

Ver

yst

able

Spon

tan

eous

epim

utat

ion

DN

Am

eth

ylat

ion

Man

nin

get

al.

(200

6)

Col

orva

rieg

atio

n(P

aram

utat

ion)

Sulf

epia

llele

sV

ery

stab

leX

-ray

s;re

gen

erat

ion

Ch

rom

atin

inh

erit

ance

sugg

este

dH

agem

ann

(196

9,19

93),

Hag

eman

nan

dB

erg

(197

7),

Wis

man

etal

.(1

993)

,R

.H

agem

ann

(per

son

alco

mm

unic

atio

n)

146 Volume 84THE QUARTERLY REVIEW OF BIOLOGY

Mel

andr

ium

albu

m(W

hit

eca

mpi

on)

Bis

exua

lity

Dec

reas

ein

CG

met

hyl

atio

nin

man

ylo

ci2

succ

essi

vege

ner

atio

ns,

inh

erit

edth

roug

hth

em

ale

pare

nt

Tre

atm

ent

wit

h5-

azac

ytid

ine

DN

Am

eth

ylat

ion

Jan

ouse

ket

al.

(199

6)

Nico

tiana

taba

cum

(Tob

acco

)

Los

sof

kan

amyc

inre

sist

ance

;pa

ram

utat

ion

-like

effe

cts

Nos

pro

and

nos

At

leas

t2

gen

erat

ion

sT

ran

sgen

icsi

len

cin

gin

doub

lytr

ansf

orm

edpl

ants

DN

Am

eth

ylat

ion

Mat

zke

and

Mat

zke

(199

1),

Mat

zke

etal

.(19

89)

Loss

ofhy

grom

ycin

resis

tanc

eH

pttr

ansg

ene

2ge

ner

atio

ns

Tra

nsg

enic

sile

nci

ng

DN

Am

eth

ylat

ion

Park

etal

.(1

996)

Req

uire

men

tofl

eaf

cell

for

cyto

kini

nN

otsp

ecifi

edA

rise

sat

10-2

per

cell

gen

erat

ion

Subc

ultu

rin

gin

med

iaco

nta

inin

gsu

cces

sive

lylo

wer

con

cen

trat

ion

sof

cyto

kin

in

Un

know

n;

DN

Am

eth

ylat

ion

sugg

este

dM

ein

s(1

986,

1989

a,b)

,M

ein

san

dT

hom

as(2

003)

Ory

zasa

tiva

(Ric

e)C

pGm

ethy

latio

npa

ttern

ssh

owin

heri

ted

culti

var

spec

ifici

ty

Met

hyl

atio

nst

ate

ofcy

tosi

ne

inva

riou

sC

CG

Gsi

tes

acro

ssth

ege

nom

e

At

leas

t6

gen

erat

ion

sC

ross

ing

Nip

pon

bare

and

Kas

alat

hcu

ltiv

ars5

DN

Am

eth

ylat

ion

Ash

ikaw

a(2

001)

Var

ious

trai

tsM

eth

ylat

ion

stat

ein

vari

ous

CC

GG

site

sac

ross

the

gen

ome

At

leas

t2

gen

erat

ion

sIn

trog

ress

ion

and

selfi

ng5

DN

Am

eth

ylat

ion

Don

get

al.

(200

6)

Ker

nel

shap

ean

dti

ller

num

ber

S2,

S3,

and

vari

ous

unsp

ecifi

edm

eth

ylat

ion

site

s3–

6ge

nera

tions

ofse

lfing

Hig

h-p

ress

ure

trea

tmen

tgi

ven

tose

eds

DN

Am

eth

ylat

ion

Shen

etal

.(2

006)

Indu

ced

dwar

fism

;re

sist

ance

topa

thog

en

Gen

eral

chan

gein

DN

Am

ethy

latio

n;X

a21G

prom

oter

At

leas

t9

gen

erat

ion

sIn

duce

dby

5-az

aCD

NA

met

hyl

atio

nin

volv

edA

kim

oto

etal

.(2

007)

,Sa

no

etal

.(1

990)

Petu

nia

hybr

ida

(Pet

unia

)W

hit

e-fl

ower

ing

An3

epia

llele

and

the

dTph

1tr

ansp

oson

At

leas

t3

gen

erat

ion

sof

self

-fe

rtili

zati

on

Indu

ced

byh

eter

ozyg

osit

yU

nkn

own

;ep

igen

etic

inte

ract

ion

betw

een

atle

ast

3dT

ph1

copi

es

Van

Hou

wel

inge

net

al.

(199

9)

Tri

ticum

aest

ivum

(Wh

eat)

Cyt

osin

em

eth

ylat

ion

Glu

ten

inge

ne

At

leas

t2

gen

erat

ion

sIn

bree

din

gC

hro

mat

in-m

arki

ng,

DN

Am

eth

ylat

ion

Flav

ell

and

O’D

ell

(199

0)

Lon

ger

prod

ucti

vesp

ikes

,la

rger

seed

s,an

dot

her

quan

tita

tive

trai

ts

Hl1

and

pclo

ci57

gen

erat

ion

sN

icot

inic

acid

Un

know

nB

ogda

nov

a(2

003)

cont

inue

d

June 2009 147TRANSGENERATIONAL EPIGENETIC INHERITANCE

TA

BL

E1

Con

tinue

d

Tax

onT

rait

Loc

us/c

ellu

lar

syst

emSt

abili

tyIn

duci

ngco

ndit

ions

EIS

Ref

eren

ce

Trit

icum

inte

rspe

cific

synt

hetic

hybr

idse

Man

ytr

aits

�13

%of

the

gen

ome

Stab

ility

depe

nds

onlo

cus;

man

yve

ryst

able

Hyb

ridi

zati

onan

dpo

lypl

oidi

zati

onC

hrom

atin

and

DN

Am

ethy

latio

n;R

NA

isy

stem

prob

ably

invo

lved

Lev

yan

dFe

ldm

an(2

004)

Tri

tical

e(S

tabl

ew

heat

-rye

hybr

id)

Incr

ease

inst

atur

e,n

umbe

rof

tille

rs,

chan

ged

tim

eof

ripe

nin

g

Not

spec

ified

At

leas

t2

gen

erat

ion

s5-

azaC

trea

tmen

tC

hro

mat

in-m

arki

ng;

DN

Am

eth

ylat

ion

invo

lved

Hes

lop-

Har

riso

n(1

990)

Zea

may

s(M

aize

)R

educ

edpi

gmen

tati

on(p

aram

utat

ion

)

B1

(gen

e-ep

ialle

le)

Ver

yst

able

Para

mut

agen

icB�

epia

llele

arise

ssp

onta

neou

slyfr

omB-

Ial

lele

ata

freq

uenc

yof

�1%

to10

%

Chr

omat

inin

heri

tanc

e;siR

NA

-dir

ecte

dch

rom

atin

mod

ifica

tion

isin

volv

ed

Alle

man

etal

.(20

06),

Cha

ndle

r(2

007)

,C

hand

ler

etal

.(20

00),

Coe

(196

6),S

tam

etal

.(2

002)

Red

uced

pigm

enta

tion

(par

amut

atio

n)

r1(c

ompl

exlo

ci)

Var

iabl

eSp

onta

neo

us,

and

expo

sure

tova

ried

ligh

tdu

rati

ons

and

tem

pera

ture

DN

Am

eth

ylat

ion

Ch

andl

eret

al.

(200

0),

Mik

ula

(199

5),W

alke

ran

dPa

nav

as(2

001)

Red

uced

,lig

ht-

depe

nden

tpi

gmen

tatio

n(p

aram

utat

ion)

pl(g

ene-

epia

llele

)V

arie

sfr

omm

etas

tabl

eto

very

stab

lein

her

itan

ce

Spon

tan

eous

Ch

rom

atin

-mar

kin

gsu

gges

ted

Ch

andl

eret

al.

(200

0),

Hol

lick

and

Ch

andl

er(1

998)

,H

ollic

ket

al.

(199

5,20

00)

Red

uced

peri

carp

colo

rbu

tda

rkpi

gmen

tati

onat

the

poin

tof

silk

atta

chm

ent

(par

amut

atio

n)

p1(g

ene-

epia

llele

)A

tle

ast

5ge

ner

atio

ns

Spon

tan

eous

DN

Am

eth

ylat

ion

Coc

ciol

one

etal

.(20

01),

Das

and

Mes

sing

(199

4),

Raj

ande

epet

al.(

2007

),Si

dore

nko

and

Pete

rson

(200

1),S

ekho

net

al.

(200

7)Sp

otti

ng

ofke

rnel

sM

uDR

tran

spos

able

elem

ent

Sile

ncin

gef

fect

ofth

eM

uklo

cus

onM

uDR

was

mai

ntai

ned

over

atle

ast4

gene

ratio

ns,e

ven

whe

nM

ukha

dse

greg

ated

away

Cro

ssin

gle

adin

gto

silen

cing

ofth

ere

gula

tory

tran

spos

onM

uDR

DN

Am

eth

ylat

ion

;si

RN

As

are

invo

lved

inm

ain

tain

ing

her

itab

lem

eth

ylat

ion

stat

esin

Mu1

and

MuD

Rel

emen

ts

Lis

chet

al.

(200

2),

Slot

kin

etal

.(2

003)

,Sl

otki

net

al.

(200

5)

148 Volume 84THE QUARTERLY REVIEW OF BIOLOGY

Ani

mal

sC

aeno

rhab

ditis

eleg

ans

(Nem

atod

e)

Smal

lan

ddu

mpy

appe

aran

ceR

NA

iof

ceh-

13O

ver

40ge

ner

atio

ns

Feed

ing

wit

hba

cter

iaex

pres

sin

gds

RN

Ata

rget

ing

ceh-

13

Ch

rom

atin

rem

odel

ing;

RN

Ai-m

edia

ted

Vas

ten

hou

wet

al.

(200

6),

N.

Vas

ten

hou

w(p

erso

nal

com

mun

icat

ion

)Si

len

cin

gof

gree

nfl

uore

scen

tpr

otei

n(G

FP)

RN

Ai

ofgf

ptr

ansg

ene

At

leas

t40

gen

erat

ion

sFe

edin

gw

ith

bact

eria

expr

essi

ng

dsR

NA

targ

etin

ggf

p

Ch

rom

atin

rem

odel

ing;

RN

Ai-m

edia

ted

Vas

ten

hou

wet

al.

(200

6),

N.

Vas

ten

hou

w(p

erso

nal

com

mun

icat

ion

)V

ario

usef

fect

s,n

otre

port

edR

NA

iof

13ge

nes

At

leas

t10

gen

erat

ion

sFe

edin

gw

ith

bact

eria

expr

essi

ng

dsR

NA

targ

etin

gth

e13

gen

es

Ch

rom

atin

rem

odel

ing;

RN

Ai-m

edia

ted

Vas

ten

hou

wet

al.

(200

6),

N.

Vas

ten

hou

w(p

erso

nal

com

mun

icat

ion

)D

aphn

iapu

lex

(Wat

erfl

ea)

Exp

ress

ion

ofG

6PD

San

dN

vari

ants

G6P

Dlo

cus

orit

sre

gula

tor

Spon

tan

eous

reve

rsio

nra

tebe

twee

nth

e2

form

sw

as1

in10

and

1in

2.

Spon

tan

eous

and

gluc

ose

indu

ced;

pres

ence

ofS

form

rela

ted

tost

ress

ful

con

diti

ons

Not

know

nR

uvin

sky

etal

.(1

983

a,b,

1986

)

Dia

phan

osom

ace

lebe

nsis

(Cla

doce

ran

)

Tim

ing

ofre

prod

ucti

onan

dn

umbe

rof

offs

prin

g

Not

spec

ified

2ge

ner

atio

ns

Indu

ced

byth

en

atur

ales

trog

enE

2N

otkn

own

;pa

ralle

lef

fect

onin

duce

dpa

ren

tsan

dth

eir

offs

prin

g

Mar

cial

and

Hag

iwar

a(2

007)

Dro

soph

ilam

elan

ogas

ter

(Fru

itfl

y)

Mod

ifyi

ng

abili

tyof

Ych

rom

osom

eIm

prin

tor

gen

ein

tera

ctio

n11

gen

erat

ion

sT

ran

sien

tef

fect

ofim

prin

tor

gen

eC

hro

mat

inm

arks

Dor

net

al.

(199

3)

Ect

opic

outg

row

thin

eyes

Kr

(KrIf

-1al

lele

),vt

d3(T

rxG

mut

atio

n)

At

leas

t13

gen

erat

ion

sG

elda

nam

ycin

trea

tmen

tgi

ven

toK

rIf-1

stra

in,

ortr

ansi

ent

pres

ence

ofT

rxG

mut

atio

nvt

d3

Ch

rom

atin

-mar

kin

gin

volv

edR

uden

etal

.(2

003)

,So

llars

etal

.(2

003)

Eye

-col

orT

ran

sgen

icFa

b-7

flan

kin

gla

cZan

dm

ini-w

hite

repo

rter

tran

sgen

es

At

leas

t4

gen

erat

ion

sT

ran

sien

tpr

esen

ceof

GA

L-4

-pro

tein

Prob

ably

chro

mat

inin

her

itan

ceC

aval

lian

dPa

ro(1

998,

1999

)

cont

inue

d

June 2009 149TRANSGENERATIONAL EPIGENETIC INHERITANCE

TA

BL

E1

Con

tinue

d

Tax

onT

rait

Loc

us/c

ellu

lar

syst

emSt

abili

tyIn

duci

ngco

ndit

ions

EIS

Ref

eren

ce

Eye

-col

or(d

ueto

de-

repr

essi

onof

min

i-w

hite

repo

rter

gen

ecl

oned

dow

nst

ream

totr

ansg

enic

Fab-

7)

Act

ivat

ion

stat

eof

endo

gen

ous

and

tran

sgen

icFa

b-7

elem

ents

con

tain

ing

CM

M

At

leas

t4

gen

erat

ion

s(m

ore

than

4ye

ars)

Hig

hte

mpe

ratu

rePr

obab

lych

rom

atin

inh

erit

ance

Ban

tign

ies

etal

.(2

003)

,F.

Ban

tign

ies

(per

son

alco

mm

unic

atio

n)

Supp

ress

ion

ofw

ing

defo

rmat

ion

sD

e-re

pres

sion

ofsd

Not

spec

ified

(sta

bilit

ylo

wer

and

har

der

tode

tect

)

Hig

hte

mpe

ratu

rePr

obab

lych

rom

atin

inh

erit

ance

Susc

epti

bilit

yto

tum

orig

enes

isPr

obab

lyse

vera

llo

ci,

incl

udin

gh

erit

able

epig

enet

icva

riat

ion

inth

eftz

prom

oter

Incr

ease

dtu

mor

igen

icity

(2ge

nera

tions

);m

odifi

edftz

met

hyla

tion

(atl

east

1ge

nera

tion)

Cro

ssin

gw

ith

hopT

um-l

and

Kr1

mut

ants

DN

Am

eth

ylat

ion

,ch

rom

atin

inh

erit

ance

Xin

get

al.

(200

7)

Ephe

stia

kueh

niel

la(M

oth

)

Rev

ersi

onof

shor

tene

dan

tenn

aean

das

soci

ated

mat

ing

disa

dvan

tage

Supp

ress

orof

sa(s

aWT)

Up

to5

gen

erat

ion

s;in

com

plet

ely

inh

erit

edfr

omm

oth

erbu

tal

mos

tfu

llyin

her

ited

from

fath

er

Exp

osur

eof

larv

aan

dpu

pato

lith

ium

ion

s,al

tern

ate

elec

tric

alfi

eld,

or25

°Cat

late

5th

inst

arla

rval

and

pupa

lph

ases

Prob

ably

chro

mat

inin

her

itan

cePa

velk

aan

dK

onde

lova

(200

1)

Hom

osa

pien

s(H

uman

)C

ardi

ovas

cula

rm

orta

lity

and

diab

etes

susc

epti

bilit

y

Impr

inte

dta

nde

mre

peat

upst

ream

ofIN

S-IG

F2-H

19re

gion

At

leas

t2

gen

erat

ion

sFo

odav

aila

bilit

ydu

rin

gch

ildh

ood

grow

thpe

riod

Poss

ibly

met

hyl

atio

n;

tran

smit

ted

thro

ugh

mal

ege

rmlin

e

Kaa

tiet

al.

(200

2,20

07),

Pem

brey

(200

2)

An

gelm

anan

dPr

ader

-W

illi

syn

drom

es15

q11-

q13

Inhe

rite

dfr

ompa

tern

algr

andm

othe

r(n

oim

prin

tera

sure

inth

efa

ther

)

Spon

tan

eous

DN

Am

eth

ylat

ion

Bui

tin

get

al.

(200

3),

Zog

hbi

and

Bea

udet

(200

7)

Mus

mus

culu

s(M

ouse

)Pr

obab

ility

ofde

velo

pin

gye

llow

coat

colo

ran

dob

esit

y,as

wel

las

susc

epti

bilit

yto

diab

etes

and

can

cer

Avy

(gen

e-ep

ialle

le)

and

prob

ably

oth

erlo

ciM

etas

tabl

e(a

tle

ast

2ge

ner

atio

ns

ofag

outi

epig

enot

ype)

;cu

mul

ativ

e,th

ree-

gen

erat

ion

effe

cton

obes

ity

Spon

tan

eous

;af

fect

edby

diet

Ch

rom

atin

-mar

kin

g,in

clud

ing

DN

Am

eth

ylat

ion

Ble

wit

tet

al.

(200

6),

Cro

pley

etal

.(2

006)

,M

orga

net

al.

(199

9),

Wat

erla

nd

etal

.(2

007)

,W

olff

etal

.(1

998)

,W

ater

lan

det

al.

(200

8)

150 Volume 84THE QUARTERLY REVIEW OF BIOLOGY

Red

uced

body

wei

ght,

redu

ced

leve

lof

prot

ein

sin

volv

edin

sexu

alre

cogn

itio

n,

and

poss

ibly

hig

her

mor

talit

ybe

twee

nbi

rth

and

wea

nin

g

Not

spec

ified

,bu

tep

imut

atio

nis

con

nec

ted

toM

ajor

Uri

nar

yPr

otei

n(M

UP)

and

Olf

acto

ryM

arke

rPr

otei

n(O

MP)

gen

es

Prel

imin

ary

resu

lts

sugg

esti

ng

tran

smis

sion

ofth

etr

aits

toB

2

(th

ese

con

dge

ner

atio

n)

Indu

ced

bytr

ansf

erof

mou

sepr

onuc

lei

atth

eon

e-ce

llst

age

toeg

gsof

adi

ffer

ent

gen

otyp

e;tr

aits

are

tran

smit

ted

tom

ost

ofth

eof

fspr

ing

thro

ugh

mal

ege

rmlin

e

DN

Am

eth

ylat

ion

assu

med

tobe

invo

lved

Roe

mer

etal

.(1

997)

Prob

abili

tyof

kin

ked

tail

shap

eA

xin-

fuse

d(g

ene-

epia

llele

)an

dIA

Ptr

ansp

osab

leel

emen

tSp

onta

neou

sra

teof

inac

tivat

ion

6%;r

ate

ofre

activ

atio

n1%

Spon

tan

eous

;in

flue

nce

dby

diet

.In

ject

ion

ofh

ydro

cort

ison

edu

rin

gsp

erm

ioge

nes

isre

duce

spe

net

ran

ce

Ch

rom

atin

-mar

kin

gin

clud

ing

DN

Am

eth

ylat

ion

Bel

yaev

etal

(198

1a,1

983)

,R

akya

net

al.(

2003

),W

ater

land

etal

.(20

06),

D.M

artin

(per

sona

lco

mm

unic

atio

n)W

hit

e-sp

otte

dta

ilan

dfe

etK

it(p

aram

utat

ion

)2

gen

erat

ion

sof

outb

red

cros

sin

g;6

gen

erat

ion

sof

inbr

edcr

ossi

ng

betw

een

para

mut

ants

Tra

nsi

ent

pres

ence

ofK

ittm1A

lfm

utat

ion

RN

Ain

her

itan

ce;

RN

Ai

invo

lved

Ras

soul

zade

gan

etal

.(2

006)

,M

.R

asso

ulza

dega

n(p

erso

nal

com

mun

icat

ion

)

Rep

ress

ion

ofre

com

bin

atio

nof

the

Lox

Pel

emen

tan

dco

nco

mit

ant

met

hyl

atio

n

Tra

nsg

enic

Lox

Pan

dsu

rrou

ndi

ng

chro

mos

omal

sequ

ence

s

Met

hyl

ated

stat

em

ain

tain

edfo

rat

leas

t3

gen

erat

ion

s

Tra

nsi

ent

pres

ence

ofSy

cp1-

Cre

;ex

posu

reof

wild

type

tore

com

bin

ase

acti

vity

DN

Am

eth

ylat

ion

Ras

soul

zade

gan

etal

.(2

002)

Gen

ome

stab

ility

Man

yA

tle

ast

3ge

ner

atio

ns

Irra

diat

ion

Ch

rom

atin

met

hyl

atio

nB

arbe

ret

al.

(200

2),

Dub

rova

(200

3)G

luco

sein

tole

ranc

eN

otsp

ecifi

ed2

gen

erat

ion

s,so

me

effe

ctin

the

3rd

gen

erat

ion

Bet

eln

utin

gest

ion

Not

know

nB

ouch

eret

al.

(199

4)

Ten

denc

yto

deve

lop

tum

ors

Ele

vate

dex

pres

sion

ofge

ne

codi

ng

for

LF

(an

estr

ogen

-re

spon

sive

prot

ein

)an

dC

-fos

App

aren

tin

the

F 1an

dF 2

gen

erat

ion

sIn

duce

dby

diet

hyl

stilb

estr

oldu

rin

gF 0

preg

nan

cy

DN

Am

eth

ylat

ion

prob

ably

invo

lved

New

bold

etal

(200

6)

Car

diac

hype

rtro

phy

Th

em

icro

RN

Am

iR-1

At

leas

t3

gen

erat

ion

sM

icro

inje

ctio

nof

miR

-1in

tofe

rtili

zed

eggs

RN

Ain

her

itan

ceW

agn

eret

al.

(200

8) cont

inue

d

June 2009 151TRANSGENERATIONAL EPIGENETIC INHERITANCE

TA

BL

E1

Con

tinue

dT

axon

Tra

itL

ocus

/cel

lula

rsy

stem

Stab

ility

Indu

cing

cond

itio

nsE

ISR

efer

ence

Myz

uspe

rsica

e(P

each

pota

toap

hid)

Loss

ofin

sect

icid

ere

sista

nce

Prob

ably

ampl

ified

resi

stan

cege

nes

Stab

lein

her

itan

ceof

lost

resi

stan

cein

clon

esth

ath

ave

ampl

ified

DN

A

Indu

ced

byD

NA

ampl

ifica

tion

DN

Am

eth

ylat

ion

invo

lved

Fiel

det

al.

(198

9)

Rat

tus

norv

egic

us(R

at)

Mod

ified

sero

toni

nco

nten

tin

imm

une

cells

g

Not

spec

ified

2ge

ner

atio

ns

Intr

amus

cula

rad

min

istr

atio

nof

�-e

ndo

rph

indu

rin

g19

thda

yof

preg

nan

cy

Not

spec

ified

Csa

baet

al.

(200

5)

Incr

ease

dex

pres

sion

ofge

nes

codi

ngfo

rm

etab

olic

fact

ors

Prom

oter

sof

PPA

R�

and

GR

inliv

er;

incr

ease

dex

pres

sion

ofot

her

RN

As

At

leas

t2

gen

erat

ion

sPr

otei

n-r

estr

icte

ddi

etdu

rin

gpr

egn

ancy

DN

Am

eth

ylat

ion

Bur

dge

etal

.(2

007)

,G

.B

urdg

e(p

erso

nal

com

mun

icat

ion

)D

ecre

ased

sper

mat

ogen

icca

paci

ty;e

leva

ted

inci

denc

eof

tum

or,

pros

tate

,and

kidn

eydi

seas

es,s

erum

chol

este

roll

evel

s,an

dim

mun

esy

stem

abno

rmal

ities

;pr

emat

ure

agin

gan

dm

ale

mat

ing

disa

dvan

tage

Met

hyl

atio

nst

ate

of15

diff

eren

tD

NA

sequ

ence

s.R

educ

edex

pres

sion

ofan

kyri

n28

,N

cstn

,R

ab12

,L

rrn6

aan

dN

CA

M1

foun

din

vin

cloz

olin

grou

pas

wel

las

incr

ease

dex

pres

sion

ofFa

dd,

Pbm

1b,

snR

P1c

and

Was

pip

Atl

east

3ge

nera

tions

;tr

ansm

issio

nth

roug

hth

em

ale

germ

line

Vin

cloz

olin

orm

eth

oxyc

hlo

rtr

eatm

ent

duri

ng

gest

atio

nD

NA

met

hyl

atio

nA

nway

etal

.(20

05,2

006a ,

2006

b ),C

hang

etal

.(2

006)

,Cre

ws

etal

.(20

07)

Alt

ered

gluc

ose

hom

eost

asis

Not

repo

rted

F 0–F

3ge

ner

atio

ns

Low

-pro

tein

diet

inF 0

,fr

omda

y1

ofpr

egn

ancy

thro

ugh

lact

atio

n

Not

spec

ified

,D

NA

met

hyl

atio

npr

obab

lyin

volv

edin

F 1an

imal

s

Ben

yshe

ket

al.(

2006

),D

.C.B

enys

hek

(per

sona

lco

mm

unic

atio

n)Vu

lpes

vulp

es(F

ox)

Pieb

ald

spot

tin

gA

ctiv

atio

nst

ate

ofSt

arge

ne

Star

(sem

idom

inan

tal

lele

)ac

tivat

edin

�1%

ofdo

mes

ticat

edan

imal

s;in

heri

ted

for

mor

eth

an2

gene

ratio

ns

Spon

tan

eous

inta

med

foxe

sra

ised

infu

rfa

rms;

hor

mon

alst

ress

sugg

este

df

Poss

ibly

her

itab

lech

rom

atin

mod

ifica

tion

Bel

yaev

etal

.(1

981b

),T

rut

etal

.(2

004)

Not

e:T

he

spec

ies

inea

chca

tego

ryar

eor

dere

dal

phab

etic

ally

.R

efer

ence

sto

the

tabl

ear

eav

aila

ble

onlin

eat

The

Qua

rter

lyR

evie

wof

Bio

logy

hom

epag

e,w

ww

.jour

nal

s.uc

hic

ago.

edu/

QR

B.

a Sim

ilar

syst

ems

hav

ebe

ende

scri

bed

for

arab

inos

e-ut

iliza

tion

inE.

coli

(Kh

lebn

ikov

etal

.20

00)

and

the

lact

ose

oper

onin

Salm

onel

laen

teri

caty

phim

uriu

m(T

olke

r-N

iels

enet

al.

1998

).b P

roba

bly

occu

rsin

man

ypa

thog

ens

(Lew

is20

07).

c It

seem

sth

atal

lcili

ates

show

cort

ical

inh

erit

ance

and

guid

edas

sem

bly

ofco

rtic

alst

ruct

ures

(Fra

nke

l198

9).C

iliat

esh

ave

asi

len

tm

icro

nuc

leus

and

anac

tive

mac

ron

ucle

us,f

rom

wh

ich

non

codi

ng

sequ

ence

sar

eex

cise

dan

dco

din

gse

quen

ces

are

ampl

ified

.Fol

low

ing

con

juga

tion

orau

toga

my,

an

ew(z

ygot

ic)

mac

ron

ucle

usis

form

edfr

omth

efu

sed

mei

otic

prod

uct

ofth

em

icro

nuc

leus

,an

dth

eol

d,m

ater

nal

mac

ron

ucle

usde

gen

erat

es.

Th

eco

mpl

expr

oces

ses,

guid

edby

the

mat

ern

aln

ucle

us,

that

lead

toth

ein

her

itan

ceof

patt

ern

sof

chro

mos

omal

rear

ran

gem

ents

inth

ezy

goti

cn

ucle

usse

emto

bech

arac

teri

stic

ofal

lci

liate

spec

ies

(Mey

eran

dC

hal

ker

2007

).d S

imila

rph

enom

ena

are

foun

din

C.

galb

orat

a,C

.tr

opic

alis

,C

.pa

rpsi

losi

s,an

din

the

basi

omyc

etes

fun

gus

Cry

stoc

occu

sne

ofor

man

s.e I

nte

rspe

cifi

cpl

ant

hyb

rids

,an

dso

met

imes

hyb

rids

betw

een

cult

ivar

s,di

spla

yep

igen

etic

vari

atio

ns

that

are

her

itab

leac

ross

gen

erat

ion

s.W

hen

hyb

ridi

zati

onis

follo

wed

bypo

lypl

oidi

za-

tion

,th

isse

ems

tobe

an

orm

alan

din

vari

ant

gen

omic

resp

onse

.In

The

Bio

logi

calJ

ourn

alof

the

Lin

nean

Soci

ety

82(4

)(A

llen

2004

),m

any

exam

ples

ofep

igen

omic

effe

cts

ofh

ybri

diza

tion

inpl

ants

,in

clud

ing

mai

ze,

wh

eat,

rice

,co

tton

,an

dsu

nfl

ower

,ar

ere

view

ed.

f Th

ere

are

sim

ilar,

olde

rst

udie

sof

tran

sgen

erat

ion

alef

fect

sfo

llow

ing

adm

inis

trat

ion

ofh

orm

ones

and

drug

sin

ava

riet

yof

mam

mal

s.T

hes

est

udie

s,w

hic

hdi

dn

otin

clud

em

olec

ular

data

,w

ere

revi

ewed

byC

ampb

ell

and

Perk

ins

(198

8).

152 Volume 84THE QUARTERLY REVIEW OF BIOLOGY

molecular data show that the gametes ofthe F1 generation have acquired alteredepigenetic marks, the transmission of epi-genetic variations rather than the directinduction of such variations is a reasonableassumption. Based on this, we includedsome cases in our table in which only twogenerations of transmission following in-duction were reported. For instance, weincluded a case reported by Burdge et al.(2007) in which protein restriction duringa rat grandmother’s pregnancy led to iden-tical somatic and epigenetic (methylation)variations in her F1 and F2 descendants, aswell as a case reported by Newbold et al.(2006) that showed that F0 female rats ex-posed to diethylstilbestrol had F1 and F2

offspring with an increased susceptibility totumors associated with persistent changesin the DNA methylation and expression ofan oncogene (see Ruden et al. 2005 for anepigenetic model explaining these data).The case of germline inherited epigeneticvariants (and their corresponding pheno-types) that were acquired stochastically inthe Avy locus of the mouse was includedsince transgenerational epigenetic effectson coat color and obesity were reported.However, this is a complex case that seemsto involve epigenetic modifications in theAvy locus as well as in other loci in the ge-nome (Blewitt et al. 2006; Cropley et al.2006; Waterland et al. 2007, 2008). An-other case we included was that reportedby Csaba et al. (2005) in which the modi-fied serotonin content induced in the im-mune cells of mice treated with endomor-phin during pregnancy is subsequentlytransmitted to their grandchildren. We didnot include the many cases of single gen-eration inheritance, even though epige-netic inheritance seems quite plausible insome of them, as, for example, when apredisposition to cancer in humans is re-lated to an epimutation (an epigenetic he-reditary abnormality in gene expression)in a mismatch-repair gene, and this epimu-tation is transmitted from mother to son(Hitchins et al. 2007). We also excluded asimilar case in mice which showed that tu-mor risk was increased following chro-mium III chloride exposure and that this

risk was then transmitted from father toson (Shiao et al. 2005). Other cases ofsingle-generation inheritance, such as pro-tection against type I diabetes in humans(where a paramutation-like process hasbeen reported) (Bennett et al. 1997), hy-drostatic pressure-induced alterations inDNA methylation in Japonica rice (Long etal. 2006), and parallel alterations in geneexpression profiles in White Leghornchickens and their offspring followingstress in the parental generation (Lindqvistet al. 2007), were also excluded, althoughepigenetic inheritance may well have oc-curred and may be revealed when subse-quent generations are studied. However,within the limitations imposed by the re-search designs of the studies we reviewedand the qualifications we have mentioned,we believe that the table provides a fairlyexhaustive overview of the recognized casesof cellular transgenerational epigeneticinheritance that have been described inEnglish-language journals, although, inev-itably, we likely missed some cases.

taxonomic distribution and inducingconditions

The data in the table probably representthe tip of a very large iceberg. What ismissing from the table is important, be-cause the absences point to gaps that needto be filled. For instance, there is no infor-mation about epigenetic inheritance in thekingdom Archea, and most phyla are notrepresented. There are also few data di-rectly addressing epigenetic inheritance inviruses, although it may plausibly be as-sumed that viruses exploit and use the epi-genetic adaptations adopted by their hosts.Data on epigenetic inheritance in chloro-plasts and mitochondria are also veryscant. It is worth noting that the organismsthat show the greatest evidence for epige-netic inheritance are the classical modelorganisms of genetics—E. coli, yeast, Arabi-dopsis, maize, rice, Caenorhabditis, Drosoph-ila, the mouse, and the rat. However, asystematic investigation of epigenetic in-heritance in different conditions is not yetavailable for any of these model organisms.It is also worth noting that all the model

June 2009 153TRANSGENERATIONAL EPIGENETIC INHERITANCE

animals studied belong to taxa in whichthe segregation between germline andsoma occurs early, and, therefore, epige-netic inheritance may be more limitedthan in the non-represented animal taxawhere segregation occurs late if it occurs atall (Buss 1987; Jablonka and Lamb 1995).Although the non-systematic way in whichthe data were collected precludes generalconclusions, it seems as if epigenetic inher-itance in multicellular organisms is mostcommon in plants and fungi. This is prob-ably in part due to the lack of segregationbetween soma and germline in thesegroups that enables developmentally in-duced epigenetic variations occurring insomatic cells to be transferred to the ga-metes when these somatic cells assumegermline functions. However, there aretwo, additional considerations that may berelevant to the difference between animals,on the one hand, and plants and fungi, onthe other. First, the lack of nervous system-directed mobility and activity in plants andfungi means that they cannot adapt tochanging conditions behaviorally; if theconditions experienced by offspring arelikely to be similar to those of their par-ents, then inheriting epigenetic adapta-tions from them is an alternative adaptivestrategy to behavior and is likely to be pos-itively selected in plants and fungi (Jab-lonka et al. 1995; Jablonka and Lamb 1995;Lachmann and Jablonka 1996). Second,mobility and CNS-dependent flexible learn-ing in animals may often limit the predict-ability of the environment in descendentgenerations; therefore, wide-ranging stableepigenetic cellular inheritance throughthe germline may be selected against. Ingeneral, it seems that the difference be-tween the life strategies of plants and ani-mals may account for the observation thatepigenetic inheritance in multicellular or-ganisms is more common in plants andfungi than among animals.

The relative importance—and some-times even the very presence—of particu-lar EISs in different taxa varies. Buddingyeast seems to lack the RNAi system, soepigenetic inheritance based upon it is im-possible, although other types of RNA-

mediated inheritance cannot be ruled out.In some groups of animals, DNA methyl-ation appears to have been lost (Regev etal. 1998) and is not part of chromatinmarking in these organisms. Transgenera-tional structural inheritance and inheri-tance through self-sustaining loops seem tobe more common in unicellular organismsand in fungi where horizontal transfer ofinformation through hyphal interaction iscommon. It may be that the developmentof a germline involves such drastic alter-ations in cellular functions and structuresthat self-sustaining loops and many cellularstructures are destabilized and disrupted.The data presented in Table 1 lend sup-port to this conjecture, although their pau-city precludes decisive conclusions.

To understand why and when cellularepigenetic variants are inherited, we needto know the conditions that promote theirinduction and stability in cell lineages.Their developmental nature requires anapproach that is sensitive to context. How-ever, our knowledge of the chromatin-marking and RNA-mediated systems sug-gests that certain parts of the genome mayexhibit chromatin- and RNA-mediated epi-genetic inheritance more often than oth-ers. Repetitive DNA sequences (especiallyregions that code for RNAs that can formdsRNA or stem-loop structures), DNA re-gions where transcription is likely to startfrom both complementary strands, and re-peated chromosomal segments that pairectopically are all likely to exhibit RNA-mediated epigenetic inheritance under awide range of conditions. These and otherrepeated sequences that cooperatively bindprotein complexes and regions with CG dou-blets are all likely candidates for multigen-erational chromatin- and methylation-basedepigenetic inheritance.

The inducibility and transmissibility of epi-genetic variants depend on developmentalconditions. Conditions of stress seem to beparticularly important as inducers of herita-ble epigenetic variation, and lead to changesin epigenetic and genetic organization thatare targeted to specific genomic sequences.We mentioned earlier that the genomicstresses of allopolyploidization and, to a

154 Volume 84THE QUARTERLY REVIEW OF BIOLOGY

lesser extent, autopolyploidization leadto epigenetic and genetic re-patterning(Grant-Downton and Dickinson 2005,2006; Rapp and Wendel 2005). A well-known epigenetic phenomenon associatedwith hybridization is nucleolar dominance,or the expression in the hybrid of therRNA gene complex (NOR) from only oneparent. The preferential silencing of oneNOR is a large-scale gene-silencing phe-nomenon associated with heritable DNAmethylation and repressive histone modifi-cations (Pikaard 2000, 2003). However, theepigenetic changes occurring in hybridsare not restricted to rRNA genes. An ex-ample we present in the table is that of thegenome-wide changes that occur in syn-thetic wheat hybrids that were formed inorder to simulate the evolution of domes-tic wheat. Levy and Feldman (2004) re-viewed evidence showing that, in these hy-brids, 13% of the genome undergoessignificant methylation changes, whilechanges also occur in genome organiza-tion (e.g., rearrangements and eliminationof some sequences). The methylationchanges affect both low copy numbers andrepetitive DNA sequences, and are associ-ated with heritable transcriptional silenc-ing. In addition, the activation of retroele-ments leads to heritable alterations in geneexpression at other loci, thus resulting inmajor changes in the profile of gene ex-pression. The changes in the epigeneticstate of the genome are region and chro-mosome specific: they are targeted to par-ticular genomic sequences and reoccur,with localized variations, upon repeatedformation of the same type of al-lopolyploid. In species of the cordgrassSpartina, genome-wide epigenetic and ge-netic changes were observed in two re-cently formed, morphologically different,natural hybrids (and an allopolyploid), inwhich 30% of the parental methylation pat-terns were altered, in addition to similarstructural changes in the DNA sequences ofthese two independently formed and genet-ically similar hybrids (Salmon et al. 2005).Many similar effects of allopolyploidizationhave been reviewed in the extensive andgrowing literature on plant polyploidy (e.g.,

see The Biological Journal of the Linnean Society82(4) [Allen 2004]). The overall impressiongained from these studies is that heritableepigenetic changes accompany the firststages of allopolyploidization, and that thetypes of repetitive sequences in the parentalspecies, the amount of divergence betweenthem (especially with regard to elements thatmay be involved in epigenetic control), andthe direction of the cross all play importantroles in specifying the extent and nature ofepigenetic re-patterning.

Another form of genomic stress that maylead to heritable variation is that associatedwith a change in reproductive mode. Forexample, the transition from sexual toa-gametic reproduction in sugar beet leadsto heritable activation of some genes (Le-vites 2000). DNA damage also leads to her-itable epigenetic changes, and researchersare beginning to uncover some of the fac-tors that affect this response. FollowingDNA repair, the epigenetic structure of therepaired region is not fully reconstructedand carries with it a repair-specific chroma-tin signature that can be transmitted tosubsequent generations (Polo et al. 2006).Moreover, the loading of the histone vari-ant �-H2AX (which is associated with re-paired DNA segments) with cohesin leadsto sister-chromatid interactions that maycontribute to the radiation-induced ge-nome instability that arises and is inheritedfor several generations in the progeny ofdamaged cells (Little 2003). This may bethe basis for the heritable genomic insta-bility found in the offspring and grand-offspring of male mice that were exposedto irradiation (Dubrova 2003), as well asfor the increased level of recombinationseen for at least four generations afterUV-C irradiation of Arabidopsis plants (Mo-linier et al. 2006).

Some of the cases included in Table 1show that physiological stresses—for in-stance, nutritional stresses imposed duringsensitive periods in the development offlax (Cullis 2005)—can lead to both ge-netic and epigenetic re-patterning, andboth types of re-patterning seem to be cor-related and share a common mechanisticbasis. It seems likely that other cases in

June 2009 155TRANSGENERATIONAL EPIGENETIC INHERITANCE

which an environmental stressor has tar-geted effects on genome organization,such as the heat-induced changes in rRNA-encoding DNA repeats in Brassica (Watersand Schaal 1996), will also be found tobe associated with heritable epigeneticchanges in the genes or repeated elementsinvolved. In animals, alterations in hor-monal balance, especially those occurringover several generations, may also be fol-lowed by epigenetic changes. This may bethe basis for the pattern of inheritance ofwhite spotting seen in domesticated silverfoxes (as we will discuss later) (Trut et al.2004). However, stressful conditions maynot only affect the chromatin-marking andthe RNA-mediated EISs; if a new prion vari-ant can be generated in stressful condi-tions, it might cross previously existing spe-cies barriers and have novel effects in itsnew “host” species.

epigenetically-based similarities anddifferences between generations:

inducing epigenetic variationsin the germline

Many of the studies of multicellular,sexually reproducing organisms that wepresent in Table 1 show that, as a result ofan inducing stimulus or of changed condi-tions in the F0 (parent) generation, similarchromatin marks and similar phenotypesare reconstructed in subsequent genera-tions. However, the F0 generation itselfmay not show any phenotypic effects;changes in epigenetic marks and associ-ated somatic phenotypes may first appearin the F1 generation and may only then beinherited by subsequent generations. Forexample, in mice, the diethylstilbestrol-induced increase in the probability of de-veloping tumors appeared only in the F1

and F2 generations (Newbold et al. 2006).Understanding the inheritance of in-

duced variation in sexually reproducingmulticellular organisms is an importanttopic of research, not least because, in thepast, much of the debate about the impor-tance of acquired characters in evolutionrevolved around this issue (Delage andGoldsmith 1912). Traditionally, in multi-cellular organisms with a germline, three

types of induced heritable effects havebeen distinguished: direct induction, par-allel induction, and somatic induction.With direct induction, the germline is di-rectly affected without any effect on the F0

parent’s soma, while with parallel induc-tion, similar somatic phenotypic effects areapparent in both the induced ancestor andits descendants, but the induction events inthe somatic and germ lineages are inde-pendent. Finally, with somatic induction, achange is induced in the soma, and thissomatic effect causes a change in the germ-line that reconstructs the somatically-induced parental phenotype in the descen-dants (Fothergill 1952; Jablonka and Lamb1995). There is, however, a fourth possibil-ity: an induced effect on the soma of the F0

generation may cause changes in the germ-line, but the resulting somatic changes indescendants are dissimilar from the effecton the soma in the F0 generation. This is acase of parallel induction with nonparalleleffects. The different types of induced her-itable effects are represented schematicallyin Figure 2.

The literature shows that direct induc-tion, parallel induction, and parallel induc-tion with nonparallel or partially paralleleffects are common. In Table 1, manyof the cases of paramutation and of in-duced, heritable, transposable element ac-tivity can be classified as direct inductionor parallel induction, because the induc-ing conditions directly affect events in thegermline (and sometimes, in parallel, inthe soma as well). Changes in hormonedynamics that specifically target the germ-line will either not affect somatic charac-ters in the induced F0 generation, or willaffect them in a way that is unrelated to theeffect seen in the F1. For example, in onecase, vinclozolin, an androgen suppressor,induced testis disease in at least three gen-erations of males following its administra-tion to a pregnant female ancestor (Anwayet al. 2005, 2006a,b). This is an example ofdirect induction with differing effects inmales and females. Parallel induction isseen in the case in which nutritional andtemperature changes affected morphology

156 Volume 84THE QUARTERLY REVIEW OF BIOLOGY

in the F0 generation of flax, and similarphenotypic effects were found in subse-quent generations (Cullis 2005). Anothercase of parallel induction is that seen in themoth Ephestia kuehniella, where subjectingthe insect to modified temperature condi-tions, lithium ion treatment, or an alterna-tive electric field during the first half of itspupal development resulted in the sup-pression of a mutant short antennae in thetreated generation and five subsequentgenerations (Pavelka and Koudelova 2001).Also, glucose induction of the S form of theG6PD enzyme in Daphnia pulex may be illus-

trative of a further possible case of parallelinduction (Ruvinsky et al. 1983a,b).

The distinction between direct and par-allel induction is not always straightfor-ward, as epigenetic changes may occur inonly some of the somatic cells of the in-duced parent, thus resulting in a varie-gated somatic phenotype. This can be de-scribed as partial parallel induction. It canbe seen in some cases of paramutation inplants where, when the induction rate ishigh, sectors that are the result of paramu-tation in somatic tissues can be seen in theF0 generation (Chandler et al. 2000).

Figure 2. Inducing an Inherited Effect(A) Direct germline induction: An external stimulus induces a germline change from G0 to G1 with no effect on

the parental soma, which remains S0. The G1 state is inherited, and leads to the development of an S1 soma.(B) Parallel induction: An external stimulus induces a change in the parent’s soma from S0 to S1 and in itsgermline from G0 to G1. The G1 state is inherited and causes the development of an S1 soma in descendants.(C) Somatic induction: An external stimulus induces a change in the parent, altering its somatic phenotype fromS0 to S1. The effect is transmitted from the S1 soma to the germline, where G0 is changed into G1; G1 isconsequently inherited and results in the development of an S1 soma. (D) Parallel induction with nonparalleleffects: An external stimulus alters the soma from S0 to S2, and the germline from G0 to G1. The germlinemodification is inherited and leads to the development of S1 soma in subsequent generations. With all fourtypes of induction, S1 could have an effect on G1 in all descendants of the original induced parents (not shown).

June 2009 157TRANSGENERATIONAL EPIGENETIC INHERITANCE

Several cases in the table can be inter-preted as instances of somatic induction inwhich heritable variation is induced in thesoma and is seen as a somatic character,and the resulting effect is then transferredfrom the soma to the germline. The abilityof small RNAs to move from cell to cell mayfacilitate soma to germline informationtransmission and may form the basis ofsuch inheritance. The most impressive ex-ample of somatic induction via small RNAsmay be found in the case discussed by Vas-tenhouw et al. (2006), in which C. eleganswere fed bacteria with DNA sequences cod-ing for dsRNA, and the RNAs migratedfrom the somatic cells of the nematode to itsgerm cells, thus affecting subsequent gener-ations. Steele et al. (1998) suggested anotherroute of transmission that is initiated byRNA: the transfer of RNA transcripts fromthe immune cells of mammals to their germ-line, followed by reversed transcription andincorporation of the reverse transcribedDNA into the germline genome. Zhivotovsky(2002) has modeled the conditions thatcould lead to the evolution of such a system.

Another route of somatic induction is viainducing conditions that affect the secre-tion of hormones, which, in turn, affect thegermline. The effect of a hormone on thesomatic characters of the induced parent(F0) and on its descendants may be similar,but it is unlikely to be identical. This isbecause even if the same genes are affectedin the soma of the adult F0 animals as wellas in the F1 and subsequent generations ofoffspring, it is unlikely that the pattern ofactivity of these genes will be the same. Indescendants, the induced epigenetic changemay be expressed during embryonic as wellas adult stages, whereas, in the F0 animals,the change is induced in adults. Only inrare cases in which the pattern of timingand spatial expression is identical in the F0

and subsequent generations—and is lim-ited to the stage at which it was induced inthe F0—will identical somatic charactersoccur in parent and offspring generations.An additional complication is the sex ofthe F0 animals and their offspring, as sex-limited effects will obviously be transmittedin a sex-specific manner, and epigenetic

variations in an induced F0 parent maytherefore be different for descendants ofthe opposite sex. These considerations arerelevant to any kind of somatic induction,no matter the mechanism behind it, so,although partial similarity is likely, we ex-pect that somatic induction leading toidentical phenotypes in the F0 and subse-quent generations will be rare.

The foregoing discussion suggests thatthe traditional distinctions between direct,parallel, and somatic induction do not sat-isfactorily describe the possible interac-tions between the soma and germline. Thesimilarity between the somatic characteris-tics of an induced ancestor and its descen-dants, which the traditional classificationhighlights, is, of course, of interest, but wethink that the mode and mechanisms ofthe induction of germline variations ratherthan their effect (i.e., the similarity or lackof similarity between the F0 and the F1)should be the focus of study. An importantaspect of the problem is whether externalinducing conditions affect the germline di-rectly or whether their effect is mediatedthrough the soma. External environmentalconditions that are independent of the or-ganism’s activity and its development canhave important heritable epigenetic ef-fects, but when there are somatic mediat-ing signals (e.g., RNAs or hormones) thesesomatic signals may evolve to efficientlycommunicate information to the germ-line. We therefore expect that develop-mentally-mediated somatic effects mayhave adaptive consequences more oftenthan signals that act on the germline di-rectly, and that when there are suchsoma-mediated influences on the germ-line, the effects on the somatic cells ofthe F0 will often differ from those thatwill be seen in the next generations (Fig-ure 2D).

The possibility of a somatic effect on thegermline that is mediated by hormoneswas raised shortly after hormones were dis-covered. The Austrian Lamarckian zoolo-gist Paul Kammerer and the pioneer endo-crinologist Eugen Steinach (1920) foundthat exposing male rats to high tempera-tures led to morphological and physiolog-

158 Volume 84THE QUARTERLY REVIEW OF BIOLOGY

ical changes in their offspring and grand-offspring. They suggested that the presence ofhormone-secreting interstitial cells adjacentto germ cells in the gonads facilitated hor-monal interactions between them, andthey claimed that heat produced a changein hormone production in interstitial cells,thus affecting germline cells and carryinghereditary consequences (see Logan 2007for a discussion of Kammerer and Stein-ach’s work). Although the validity of thisclaim and its possible interpretation interms of epigenetic inheritance is at presentunclear, the possibility that there are hor-monal effects on epigenetic variation is nolonger considered heresy.

The experimental work of Vanyushin etal. (2006a) has shown that methylation pat-terns in the rat genome are controlled byhydrocortisone dynamics, and that phyto-hormones of different classes cause a de-cline in global DNA methylation and therepression of de novo methylation in plants.Moreover, the evidence reviewed by Nazand Sellamuthu (2006) suggests that, de-spite doubts about some of the reportedinformation, there are 8 hormone recep-tors and 16 cytokine/growth receptors inmature ejaculated sperm, thereby allowingfor the possibility that hormones couldexert their effects on male gametes.Hormone and neurotransmitter recep-tors have also been found in oocytes and infemale germline cells. For example, theoestrogen receptor (Wu et al. 1992), sero-tonin receptor 5-HTID (Vesela et al. 2003),Notch1 and Notch2 receptors (Cormier etal. 2004), and the �2-andrenoceptor (Ci-kos et al. 2005) have all been detected inoocytes, and the GH receptor has beendetected in fertilized eggs (Pantaleon et al.1997; Kolle et al. 2001). It is interesting tonote that the changes in expression ofsome of the genes coding for these recep-tors coincide temporally with early waves ofepigenetic re-programming during devel-opment. The presence of hormone recep-tors in gametes, and the modulation ofreceptors’ synthesis during sensitive devel-opmental periods when hormonal changesoccur, suggest that induced variations inhormonal conditions may affect the epige-

netic state of genes within germline cells,and these, in turn, can be transmitted tothe next generation.

The involvement of hormones in the in-duction of heritable epigenetic variationsis no longer a mere speculation: several ofthe mammalian examples presented in Ta-ble 1 suggest that changes in hormonalstimuli induce heritable epigenetic changes.For example, the penetrance of the fusedphenotype is altered in the progeny ofmouse parents treated with hydrocortisone(Belyaev et al. 1983). In silver foxes se-lected for tame behavior, hormonal effectsin the serotonin system that controls ag-gression seem to be involved in the herita-ble activation of the star gene that leads towhite spotting (Belyaev et al. 1981a,b; Trutet al. 2004; Popova 2006). The best inves-tigated case of hormonally-mediated ef-fects on epigenetic marks is that of thetransgenerational effect of the estrogenicandrogen disruptors vinclozolin and me-thoxychlor on testes development in malerats (Anway et al. 2005, 2006a,b; Chang etal. 2006; Crews et al. 2007). With vinclozo-lin, 15 different DNA sequences isolatedfrom sperm were shown to have alteredmethylation patterns, and these patternswere transmitted to the F1-F3 generationsof offspring of treated F0 females. Cru-cially, vinclozolin was effective only whenadministered between 8 and 15 days postcoitum, and had no effect when adminis-tered later, between 15 and 20 days. Thissensitive developmental period coincideswith the epigenetic remethylation phase inthe male (Hajkova et al. 2002), thus sug-gesting that the hormonal effect of andro-gens is developmentally specific (limited tothis period of epigenetic reprogramming)and is not a general toxic effect. If so,modifications in these methylation pat-terns in the soma of F0 vinclozolin-treatedfemales are not expected. In plants, whereno epigenetic reprogramming phase simi-lar to that in mammals is apparent, and thegermline is continuously produced duringdevelopment (Matzke and Scheid 2007),changes in hormonal stimulations duringall phases of somatic development are

June 2009 159TRANSGENERATIONAL EPIGENETIC INHERITANCE

likely to affect epigenetic variations in thegermline.

epigenetic recall and otherdirectional changes in heritable

epigenetic marksWe defined cellular epigenetic inheri-

tance as the transmission from mother cellto daughter cell of variations that are notthe result of differences in DNA sequence,or of persistent inducing signals in thecell’s environment. The examples pre-sented in Table 1 support our assertionthat when epigenetic marks are inherited,the same pattern of marks is more or lessfaithfully reconstructed across generations.If a particular mark—for example, a pat-tern of 5 methylated cytosine sites—is in-duced at a particular locus in the germline,this pattern is then reconstructed (with acertain error rate) in the descendants andhas similar phenotypic effects (Figure 3A).Stable epigenetic inheritance is at oneextreme pole, and total reset to a singledefault state—that of the uninduced par-ents—is at the other pole of developmen-tally influenced inheritance. However, thedevelopmental nature of epigenetic inher-itance and our knowledge of the construc-tion of chromatin marks suggest that weconsider other possibilities; the examplesin the table, therefore, represent only avery small fraction of the types of epige-netic hereditary phenomena that probablyexist.

The first possibility is that of partial re-construction—an intermediate between thetwo extreme poles of complete reset andfaithful reconstruction. For example, ofthe 5 induced methylation sites, only 3 arereconstructed in the offspring, and, in theabsence of the inducing stimulus, if thephenotype of the offspring is the same asthat of an uninduced individual, this wouldnot be seen as a case of epigenetic inheri-tance, even if the threshold for the devel-opmental response was lowered or thespeed of reaction was enhanced in descen-dants (see Figure 3B). The situation is sim-ilar to that found with neural memory,when the original stimulus that leads to theinitial learnt response is required to trigger

the response again, but because memorytraces remain, there is recall—facilitatedreconstruction of the learnt response uponre-induction. We suggest that the inheri-tance of some epigenetic memory tracesmay lead to epigenetic recall—a facilitatedresponse in descendants that requires aninducer. The inherited, partial epigeneticpatterns that facilitate a response arecalled epigenetic engrams. (Engram is a termthat was invented by Richard Semon in1904, to mean, roughly, “memory trace”;see Schacter 2001.) In order to recognizeepigenetic recall, the kinetics of inducedresponses in the F1 and subsequent gener-ations need to be studied. None of theexamples in Table 1 fulfils this require-ment, and, to the best of our knowledge,this kind of investigation—searching forepigenetic engrams and for facilitated butstill inducer-dependent responses—has notbeen part of the research program of epi-genetics. Such responses, however, arelikely to be common, as the mechanismsfor them are all in place, and a systemenabling the reconstruction of epigeneticengrams that allow recall would be selec-tively advantageous in many conditions,just as, in spite of the different timescale,the evolution of neural sensitization hasbeen favored. Agrawal et al. (1999) studiedinduced defenses against predators in wildradish (Raphanus raphanistrum) and the wa-ter flea (Daphnia cucullata), and showedthat induction in the parental generationmade offspring better adapted to preda-tors than the offspring of uninduced par-ents. They suggested that the persistent pa-rental effect in radish plants may be eithera direct maternally-induced effect or theresult of more rapid induction of plantdefenses in the offspring of damagedmothers. If the latter proves correct, it willrepresent a case of epigenetic recall, un-derlain by as yet uncharacterized epige-netic engrams. (For an extended discus-sion of the possibility of learning in cellsand non-neural organisms, see Ginsburgand Jablonka 2008.)

In addition to epigenetic recall based onpartial reconstruction, we must also con-sider the possibility that although induced

160 Volume 84THE QUARTERLY REVIEW OF BIOLOGY

Figure 3. Types of Transgenerationally Inherited Epigenetic EffectsAn inactive gene (gray rectangle) and its corresponding phenotype are depicted at the top of the figure. (A)

Epigenetic copying and corresponding phenotypic inheritance: An epigenetic mark consisting of 5 � sites is inducedin the parent and affects the marking of the gene and the phenotype of the induced individual. (The “�” signsindicate an altered methylation or histone modification site, and the inducer is indicated by a curved arrow.)The epigenetic mark is reliably transmitted through the germline, thus leading to a modified heritablemorphology in the uninduced progeny (heritable site-states are indicated by “�” signs within the rectangle, andthe straight black arrows indicate transitions between generations). (B) Epigenetic recall: Partial inheritance ofthe epigenetic pattern (represented by 3 internalized “�” signs) that was established in the induced parentdoes not lead to modified morphology in progeny. However, the amount of inducer needed to re-establish thefull epigenetic pattern (5 � pattern) and the corresponding induced phenotype is much smaller than in theparent (the smaller curved arrow indicates low level stimulation). (C) Reactive but dissimilar effects of inheritedepigenetic patterns: (i) antagonistic - The parental epigenetic mark (5 � signs) is inherited faithfully, but, in amismatched postnatal environment (triangle around progeny), it leads to a different phenotype in theprogeny. (ii) accumulative - Following recurrent induction in each generation, epigenetically modified and“internalized” sites accumulate, and result in correspondingly more extreme phenotypes. When the epigeneticpattern reaches a certain configuration (5 � internalized sites), it is inherited even in the absence of theinducer, and this is a form of epigenetic assimilation. (iii) lingering-fading - Following induction, the mark andits corresponding morphology are established, but fade away gradually in subsequent generations in a nonin-ducing environment.

June 2009 161TRANSGENERATIONAL EPIGENETIC INHERITANCE

marks in one generation may be faithfullyinherited, they might lead to a non-matchingyet predictable phenotype in the subse-quent generation if the environments ofparent and progeny are drastically differ-ent (Figure 3Ci). The offspring’s responsecould be interpreted as a misfired predic-tive response—the consequences of a strat-egy that evolved when the parents’ and theoffspring’s conditions matched. The ef-fects of such mismatches and their medicalsignificance have been discussed by Gluck-man and Hanson (2005; Gluckman et al.2007).

Another possibility worth consideringis that of directional changes in heritableepigenetic marks over the course of gener-ations. For instance, if inducing condi-tions persist for several generations, epige-netic marks may accumulate (Jablonka andLamb 2005). This could lead to a moreextreme phenotype (Figure 3Cii) and, pos-sibly, to a greater fidelity of transmission.Inducing conditions might endure due tothe persistence of external environmen-tal factors (e.g., there is multi-generationalexposure to a chemical), continual trans-mission through one sex (i.e., for severalgenerations a particular epiallele is trans-mitted only through females, or onlythrough males), or continuous transmis-sion through old parents, thus leading tothe Lansing effect (see Lamb 1994). Theopposite (Figure 3Ciii) may also occur;that is, when induction in the parental gen-eration is followed by non-inducing condi-tions in the subsequent offspring genera-tions, the induced epigenetic variationsmay linger and gradually fade away, withsome marks being lost in each generation.This might be the basis of the “lingering”modifications described by Jollos (1921),who found that following exposure to higharsenic or salt concentrations, or to highheat, paramecia showed heritable pheno-types that slowly faded over many genera-tions (Jablonka et al. 1992). We suggestthat the study of epigenetic engrams, andthe study of the kinetics of epigeneticmemory changes in different conditions,will lead to an expansion of the research

agenda of epigenetics (Ginsburg and Jab-lonka 2008).

Implications: Evolutionary,Practical, and Theoretical

Given that epigenetic variations are of-ten less stable than genetic variations, whatevolutionary significance do they hold? Weargue that a view of heredity that incorpo-rates the transmission of epigenetic infor-mation through cellular EISs presents chal-lenges and opportunities to applied andtheoretical research in evolutionary biol-ogy. Since, with few exceptions, the in-corporation of epigenetic inheritance andepigenetic control mechanisms into evolu-tionary models and empirical studies is stillrare, our discussion is, inevitably, some-what speculative.

implications for the study ofevolution

Heritable epigenetic variations and epi-genetic control mechanisms are relevantfor the empirical and theoretical study ofevolution because they affect both the pro-cesses of adaptation and of divergence(Jablonka and Lamb 1995, 2005, 2006a,2007b). Five types of effects are character-ized: (i) evolutionary change occurringthrough selection of epigenetic variants,without involvement of genetic variation;(ii) evolutionary change in which an initialepigenetic modification guides the selec-tion of correlated genetic variations; (iii)evolutionary change stemming from thedirect effects of epigenetic variations andepigenetic control mechanisms on the gen-eration of local and systemic epigenomicvariations; (iv) evolutionary change resultingfrom the constraints and affordances thatepigenetic inheritance imposes on develop-ment; and (v) evolutionary change that leadsto new modes of epigenetic inheritance.

Evolution through Selection ofEpigenetic Variants

Adaptation can occur through the selec-tion of heritable epialleles, without any ge-netic change. This may be of particularimportance when populations are small

162 Volume 84THE QUARTERLY REVIEW OF BIOLOGY

and lack genetic variability (e.g., in situa-tions of intense inbreeding following isola-tion or following changes in reproduc-tive strategies). As the examples listed inTable 1 indicate, epigenetic variants areoften induced when environmental condi-tions change, so several individuals in thepopulation may acquire similar modifica-tions at the same time. This means that ad-aptation through the inheritance of newlyinduced epigenetic variants may be veryrapid (Jablonka and Lamb 1995, 2005; Kus-sell and Leibler 2005; Richards 2006; Boss-dorf et al. 2008), thus leading to the accu-mulation of epigenetic variations. Several ofthe epigenetic variations presented in thetable are beneficial for their carriers, such asincreased epigenetically heritable antibioticresistance in bacteria (Adam et al. 2008) andthe switch between morphotypes in Candidaalbicans (Zordan et al. 2006). Other cases,such as increased mutability as a result ofradiation in mice (Dubrova 2003), increasedrecombination rate in plants (Molinier et al.2006), and alterations in flowering time,color, and flower morphology (see table forseveral specific examples and references) arelikely to be adaptive under some conditions;therefore, positive selection of such variantsis plausible.

In order to assess the role of epigeneticvariation in microevolution, it is importantto evaluate the extent and heritability ofepigenetic variations in natural popula-tions (Yi et al. 2004). In a programmaticpaper that outlines the framework for eco-logical epigenetics, Bossdorf et al. (2008)present some of the fundamental researchquestions that need to be asked about epi-genetic variations in natural populations,regarding the extent and structure of epi-genetic variation, its correlation with phe-notypic variation, its inducibility, and itseffects on fitness.

The mechanisms of epigenetic controlmay play an interesting role in structuringepigenetic variation because they can coor-dinate patterns of gene expression. Zuck-erkandl and Cavalli (2007) believe that re-peated sequences in “junk DNA” might becarriers of epigenetic marks, and thatmarks on these sequences can be commu-

nicated to other regions in the genome.They suggested that an altered mark couldtherefore result in coordinated hereditarychanges in the expression of several differ-ent genes simultaneously, hence accelerat-ing adaptive evolution.

Coordinated hereditary epigenetic changesmay have been involved in the process of do-mestication. For instance, forty-six genera-tions of selection for tameness in silver foxesby Belyaev and his research group in No-vosibirsk resulted in a complex of heritablechanges. The foxes became dog-like in theirbehavior and displayed skeletal, hormonal,and spotting changes, as well as altered tailand ear posture, altered vocalizations, andan increased number of supernumerarychromosomes (Belyaev et al. 1981a,b; Trut etal. 2004). Analysis of the pattern of inheri-tance of white spotting revealed that spottingbehaved like a dominant or semi-dominanttrait, but the rate of appearance and disap-pearance of the character was far too highfor new mutations to be a likely explanation.These reversible changes could not be ex-plained as an effect of inbreeding either,because the coefficient of inbreeding wasonly 0.03 (Trut et al. 2004). A probable ex-planation is that the stress of domesticationand selection for tameness targeted geneswith large effects in the neuro-hormonal sys-tem (Trut et al. 2004; Popova 2006) and mayhave heritably reactivated some of them(Belyaev 1981a,b). This epigenetic interpre-tation, in terms of new epimutations ratherthan new mutations, explains the high rateof appearance and disappearance of somephenotypes, and support for this comesfrom the fact that at least two of the genes(Agouti and C-kit) that seem to be involved inthe changes are known to have heritable epi-genetic variants in mice (Trut et al. 2004).The induction and selection of epigeneticvariations may also have been important inthe domestication of plants: ecological andgenomic stress conditions caused by mov-ing plants to new conditions and crossingdivergent strains induce many epigeneticvariations, and selection of such variationsprobably played a part in domestic plantevolution.

June 2009 163TRANSGENERATIONAL EPIGENETIC INHERITANCE

Epigenetic Change Guiding the Selectionof Genetic Variations

The guiding role of development in evolu-tion has been a subject of discussion eversince the pioneering work of Wadding-ton (1957, 1968, 1975) and Schmalhausen(1949). Their basic idea was that selectioncan lead to a change from a stimulus-dependent to a stimulus-independent (orless dependent) phenotypic response. Theprocess leading to the change from a phe-notype whose expression was dependenton an environmental inducer to a consti-tutive expression was called genetic assim-ilation (Waddington 1957; for recent dis-cussion of the idea and its evaluation, seePigliucci et al. 2006; for suggestions em-phasizing its role in the evolution of behav-ior, see Avital and Jablonka 2000; Gottlieb2002).

Ideas about the significance of develop-mental plasticity have recently been strength-ened and extended to provide a generalframework for evolutionary biology (Pigli-ucci 2001; Schlichting and Pigliucci 1998).West-Eberhard (2003) suggested that environ-mentally-induced changes during develop-ment guide the selection of genetic changesthat simulate, stabilize, and ameliorate any det-rimental effects of induced developmentalchanges. She called this developmental guid-ing process, which includes but is not limitedto genetic assimilation, “genetic accommoda-tion.” Induced epiallelic variations that are epi-genetically inherited may enhance the effec-tiveness of assimilation and accommodationprocesses—something that is likely to be par-ticularly important during conditions of stress(Jablonka et al. 1992; Jablonka and Lamb1995, 2005; Pal 1998; Sangster et al. 2004;Badyaev 2005; Siegal and Bergman 2006). Anexample showing the facilitating evolutionaryeffects of epigenetic inheritance was providedby True and Lindquist’s (2000) study, in whichthey compared pairs of yeast strains differingonly in whether or not they carried [PSI�], theprion form of a protein that is involved inmRNA translation. By growing the pairs ofstrains in a variety of conditions, they uncov-ered strain-specific differences between themin colony morphology and growth characteris-

tics. Since the presence of the [PSI�] prionleads to the suppression of nonsense muta-tions, the production of a variety of new pro-tein products in the [PSI�] containing strains(that arose because translation goes beyondthe normal endpoint of functional genes, orbecause stop-codons in the middle of non-functional genes are ignored) was increasedand was beneficial in some conditions. Theepigenetic, selectable variation that is gener-ated in [PSI�] strains might enable a lineage toadapt and “hold” the adaptation until geneticchanges take over; thus, the heritable epige-netic variations in protein architecture pavethe way for genetic adaptation (True et al.2004; Sangster et al. 2004). As a theoreticalmodel has shown, the adaptive effects of such asystem may lead to its evolution even if theresponse is adaptive only once in a millionyears (Masel and Bergman 2003). Selection ofthe epigenetically-based variation generated bythis type of system would be particularly impor-tant in asexual lineages, where the accumula-tion of mutational changes may be slow.

Epigenetic inheritance-driven accommo-dation has probably been important in chro-mosome evolution as well. It may, forexample, have initiated the evolution of di-morphic X and Y chromosomes. Jablonkaand Lamb (1995) suggested that the initialepigenetic silencing of a sex-determining lo-cus could have produced an epigenetic het-eromorphism between chromosomes, whichled to pairing problems in meiosis and con-sequent heterochromatinization and silenc-ing of the homologous region. This wouldhave reduced recombination frequenciesand driven degeneration of the Y chro-mosome; it could also have led to X-chromosome imprinting and dosage com-pensation in mammals (Jablonka 2004a).

Heritable epigenetic variations may alsoplay an important role in the evolution ofchromosomal structures such as centro-meres. Henikoff et al. (2001) have proposedthat the rapid evolution of centromeric se-quences and some centromere-associatedproteins may be driven by an epigenetically-guided arms race (Talbert et al. 2004; Heni-koff and Smith 2007). They suggested thatcentromeres compete to enter the productof female meiosis that will form a gamete

164 Volume 84THE QUARTERLY REVIEW OF BIOLOGY

(rather than a polar body, which is a deadend). Centromeres with DNA sequences thatresult in more efficient spindle fiber attach-ments out-compete others in the race to theprospective egg, thus leading to centromere-associated meiotic drive. However, meioticdrive often has deleterious effects on the or-ganism’s fitness, because deleterious genesthat are linked to the chromosome with thedriving locus—the “strong” centromere, inthis case—reduce individual fitness. The illeffects of centromere-driven meiotic driveare neutralized by the selection of alleles ofcentromere protein genes with a lower bind-ing affinity for the centromeric sequences;hence, centromere-binding proteins evolverapidly and adaptively to counter the “self-ish” centromere sequences. This evolution isdriven by the centromere DNA sequencesand their attached proteins. It is the func-tional epigenotype that must remain stable,whatever the specific identity of the DNAsequence and the proteins at the centro-meric regions.

An important consequence of the genera-tion and evolution of different epigenotypesacross various populations is that, like ge-netic variations, they may initiate reproduc-tive isolation. Differences in chromatin struc-ture that arise by chance or during localadaptation may result in hybrid offspringthat either fail to develop normally orare sterile because the two sets of parentalchromosomes carry incompatible chromatinmarks (Jablonka and Lamb 1995). For exam-ple, incompatibility between parental marksis thought to be the reason why hybrids be-tween two species in the rodent genus Pero-myscus develop abnormally (Vrana et al.2000), and, in plants, crosses between par-ents of different ploidies fail because of thedysfunction of the hybrid endosperm, a tis-sue exhibiting genomic imprinting (Sokolov2006).

The Effects of Epigenetic Variations andEpigenetic Control Mechanisms on the

Generation of Local and SystemicEpigenomic Variation

Although in practice the biases imposedby epigenetic variations, such as methyl-ation marks, on the generation of local

changes in DNA are intertwined with epi-genetic control mechanisms generatingsystemic genomic changes, we need to dis-cuss them separately, for heuristic reasons.It has been known for some time that therates of mutation, transposition, and re-combination are lower in condensed thanin open chromatin (Belyaev and Borodin1982; Jablonka and Lamb 1995), and thatthe movement of transposable elements,which is recognized as a major cause ofgenomic change (Kidwell and Lisch 1997),is markedly influenced by various types ofinternal (genetic) and external (environ-mental) stresses. It is therefore clear thatepigenetic variations bias genetic changes.However, the effect of epigenetic controlmechanisms can go beyond the more orless localized mutational changes inducedby local chromatin variations. Zufall et al.(2005) have suggested that developmen-tally regulated genome rearrangementsbrought about by epigenetic control mech-anisms are an ancient feature of eu-karyotes. If so, it is possible that, duringperiods of stress, the same epigenetic con-trol mechanisms cause global epigenomicmacro-variations that are inherited be-tween generations and that lead to macro-evolutionary changes (Jablonka and Lamb2008; Lamm and Jablonka 2008). Theseepigenetic control mechanisms may un-derlie the systemic changes in the genomethat Goldschmidt (1940) believed drovemacroevolution. Goldschmidt proposedthat macroevolutionary changes are the re-sult of large changes in the genome thatare based either on macromutations (mu-tations in single genes that have very largephenotypic effects) or on systemic muta-tions (changes in the organization of thegenome, such as chromosomal rearrange-ments). Goldschmidt’s ideas used to bederided, but recent data from many bio-logical fronts are changing this attitude(Shapiro 1999; Bateman and DiMichele2002; Fontdevila 2005). Sequence studieshave shown that during plant and animalphylogeny, many developmental geneshave been duplicated and re-used (Gu etal. 2004), and Rodin et al. (2005) havesuggested how epigenetic silencing may

June 2009 165TRANSGENERATIONAL EPIGENETIC INHERITANCE

play a role in this. Epigenetic controlmechanisms probably have a key role inspeciation through polyploidization andhybridization, which are of central impor-tance in plant evolution (Jorgensen 2004;Rapp and Wendel 2005). As we noted ear-lier, recent studies have shown that inmany naturally occurring and experimen-tally constructed polyploids and hybrids,DNA methylation patterns are dramaticallyaltered, and genes in some of the dupli-cated chromosomes are heritably silenced.Following auto- and allo-polyploidization,there is a burst of selectable variation, withall the opportunities for adaptation thatthis provides. This evidence is very much inline with the suggestions of McClintock(1984), who argued that stress leads to areshaping of the genome.

Although we do not yet know how epi-genetic control systems are involved in thegeneration of such systemic mutations,processes based on pairing, such as themechanisms seen in ciliates (in which pair-ing with scanRNAs determines which se-quences are degraded) and during meioticmis-pairing (in which unpaired regions aredeleted or heterochromatinized) may berecruited under conditions of genomicand ecological stress. Molinier et al. (2006,and personal communication) showed thatexposing Arabidopsis to UV-C radiation inone generation caused a heritable increasein the recombination rate of the wholepopulation of irradiated plants for at leastfour generations. This might be an exam-ple of an induced, pairing-based systemic,epigenetic change. Jablonka and Lamb(1995, 2008) suggested that there may havebeen selection for specific heritable epige-netic responses based on pairing, whichare determined by the type of stress (e.g.,direct radiation-induced damage to DNA,nutritional stress, heat stress), its severity,and its probability of reoccurrence.

Evolutionary Constraints and AffordancesImposed by Epigenetic Inheritance

Cellular EISs were a precondition for theevolution of complex multicellular organ-isms with specialized cell lineages, becausecells in such lineages have to maintain and

transmit their determined states, even whenthe conditions that initiated them are longpast. Since the cells that give rise to the nextgeneration of organisms need to have anuncommitted state, and efficient EISs couldjeopardize this, EISs must have imposed astrong constraint on the evolution of ontog-eny. There are several features of develop-ment that may be outcomes of selection toprevent cells with inappropriate epigeneticlegacies from founding the next generation.For example, the difficulty of reversing someepigenetic states, the early segregation andquiescent state of the germline of many ani-mal groups, and the massive changes inchromatin structure that occur during mei-osis and gamete production, may all be theresult of selection against transmitting theepigenetic “memories” associated with thedevelopmental changes and chance epimu-tations that would prevent a zygote fromstarting its development in a totipotentepigenetic state (Jablonka and Lamb1995, 2005). Recently, Pepper et al. (2007)have suggested that serial differentiation—the sequence of differentiation that startswith self-renewing somatic stem cells andproceeds through several non-self-renewing,transient, amplifying cell stages before end-ing with terminally differentiated cells—isalso a strategy that evolved to avoid thesomatic selection of selfish genetic and epi-genetic variations.

Jablonka and Lamb (2006b) argued thatthe constraints and affordances of epige-netic control systems and epigenetic inher-itance played a crucial role in all eight ofthe major evolutionary transitions identi-fied by Maynard Smith and Szathmary(1995). For example, the transition fromindependent genes to long chromosomeswas probably dependent on epigenetic in-heritance based upon chromatin marking,which maintains patterns of gene activityfollowing DNA replication. Epigenetic con-trol mechanisms may also have been im-portant in the transition from prokaryotesto eukaryotes—a transition that was associ-ated with processes of endosymbiogenesis.It is likely that massive and heritable inac-tivation of large parts of the symbiont-to-begenome, as well as the employment of

166 Volume 84THE QUARTERLY REVIEW OF BIOLOGY

mechanisms of structural inheritance thatenabled the integrity of the basic structureof the symbiont-to-be membranes to per-sist, were involved in this transition.

The Evolution of EISs: Their Origin andSelection

In light of the growing volume of workand the theoretical considerations thatsuggest that nongenetic mechanisms of in-formation transfer play key roles in evolu-tion, the evolutionary origin of nongeneticinheritance systems is of fundamental in-terest. There have been some theoreticaland comparative studies that have ad-dressed the evolution of EISs, but not allaspects have been explored. The evolution-ary origins of DNA methylation have beenconsidered, and several different hypothe-ses for the advantages it conferred havebeen suggested (Bestor 1990; Bird 1995;Regev et al. 1998; Colot and Rossignol1999; Mandrioli 2004). There are also sev-eral comparative studies of histone evolu-tion (Sandman et al. 1998; Felsenfeld andGroudine 2003), and the evolution ofRNAi systems for defence against genomicparasites and as regulators in an ancientRNA world has been suggested (Ceruttiand Casas-Mollano 2006).

Specific developmental processes thatinvolve epigenetic inheritance, such asgenomic imprinting and X-chromosomeinactivation, have also been subjects of evo-lutionary study (e.g., Jablonka and Lamb1995; Lyon 1998; Haig 2002; Jablonka2004a; Wolf and Hager 2006). However,other developmental processes that de-pend on EISs (paramutation and stress-induced epigenomic alterations, for exam-ple) have not yet received much attentionfrom evolutionary biologists, and the adap-tive significance—if any—of epigeneticmechanisms leading to systemic changesduring periods of genomic and ecologicalstresses is at present an open question.

The stability of epigenetic transmissionis likely to be an evolved trait that dependson the relative cost of error and the costof development (Rando and Verstrepen2007). Epigenetic recall may be selectivelysuperior to full epigenetic inheritance in

environments that change every few gener-ations because the cost of response-errorthat occurs when memory is perfect is re-duced, as is the cost of development-from-scratch, which occurs when reset is com-plete and full induction is required. Thetransmission of epigenetic engrams thatlead to an inducer-requiring yet facilitatedresponse may therefore often be an opti-mal compromise between the danger of atyrannically good memory, on the onehand, and the expensive response-delaythat comes with ”forgetting” too thor-oughly, on the other. Direct evidence forepigenetic recall is needed, however, andtheoretical exploration through modelingmight point to biological systems with strat-egies that would qualify them as good tar-gets for empirical research.

Epigenetic inheritance should be fa-vored in fluctuating environmental condi-tions that last for more than one genera-tion (but not for very long) and may beparticularly important in the type of envi-ronments experienced by many microor-ganisms (Lachmann and Jablonka 1996;Balaban et al. 2004; Lewis 2007; Rando andVerstrepen 2007). In such fluctuating en-vironments, efficient epigenetic inheri-tance is likely to evolve (i) if the parentalenvironment carries reliable informationabout the offspring’s environment (Jab-lonka et al. 1995), (ii) when the responseto induction is lengthy and incurs a veryhigh cost (Lachmann and Jablonka 1996),and (iii) when recall is not an option orincurs too high a cost.

theoretical and practicalimplications

Incorporating epigenetic inheritance intoevolutionary theory extends the scope of evo-lutionary thinking and leads to notions ofheredity and evolution that incorporate de-velopment. Dobzhansky’s definition of evo-lution as “a change in the genetic composi-tion of populations” (1937, p.11) appears tobe too narrow because it does not incorpo-rate all sources of heritable variations. Bothevolution and heredity need to be redefined.Jablonka and Lamb (2007a,b,c) suggestedthat evolution should be redefined as the

June 2009 167TRANSGENERATIONAL EPIGENETIC INHERITANCE

set of processes that lead to changes in the natureand frequency of heritable types in a population,and heredity as the developmental reconstruc-tion processes that link ancestors and descen-dants and lead to similarity between them.These deliberately broad redefinitions al-low evolutionary possibilities denied by the“Modern Synthesis” version of evolutionarytheory, which states that variations areblind, are genetic (nucleic acid-based), andthat saltational events do not significantly con-tribute to evolutionary change (Mayr 1982).The epigenetic perspective challenges all theseassumptions, and it seems that a new extendedtheory, informed by developmental studiesand epigenetic inheritance, and incorporatingDarwinian, Lamarckian, and saltational frame-works, is going to replace the Modern Synthe-sis version of evolution (Jablonka and Lamb2005, 2007c). We believe, therefore, that theimpact of epigenetics and epigenetic inheri-tance on evolutionary theory and the philoso-phy of biology will be profound.

As we noted earlier, it is now recognizedthat epigenetic inheritance is relevant forecology, and new methods and approachesto the research questions to which it pointsshould be developed (Bossdorf et al.2008). The relevance of epigenetic varia-tions to biodiversity in our rapidly chang-ing world is also of obvious interest andclearly has to be explored.

A discussion of the implications of epige-netic studies for medicine is beyond thescope of this review, but since epigenetic de-fects can be transmitted between genera-tions of cells and individuals, we direct thereader’s attention to some recent reviews.Baylin and Jones (2007) review the epigenet-ics of cancer, and Zoghbi and Beaudet(2007) review diseases caused by defects inchromatin marking and imprinting. The epi-genetic aspects of metabolic diseases andtheir transgenerational effects are also beingintensely studied (see Bateson et al. 2004;Gluckman and Hanson 2005; Gluckman et

al. 2007; Petronis 2004, 2006). The epidemi-ological aspects of epigenetic inheritancewere reviewed by Jablonka (2004b), and theimportance of epigenetics for aging researchhas been discussed by Vanyushin (1973),Holliday (1984), Lamb (1994), and Issa(2000). The recently reported ability ofpathogenic microorganisms to evolve herita-ble epigenetic resistance to medication (e.g.,antibiotics) may be of major medical impor-tance (Adam et al 2008), and the relevanceof epigenetic inheritance for therapeuticcloning and nuclear transplantation in ani-mals, including humans, is self-evident (seeJaenisch and Gurdon 2007).

Heredity is a fundamental property of liv-ing organisms. It is therefore not surprisingthat, in the beginning of the last century, therediscovery of Mendel’s laws and the chro-mosomal mechanisms underlying them ledto profound changes in all branches of biol-ogy. Today, at the dawn of the 21st century,another aspect of heredity—epigenetic in-heritance and the epigenetic control mech-anisms underlying it—is being unravelled.Like the early 20th-century discoveries, it,too, is driving a great expansion and trans-formation in our understanding of biology.

acknowledgments

We would like to thank the many colleagues whogenerously provided us with greatly needed informa-tion and feedback: Nathalie Q. Balaban, Daniel Beny-shek, Renee Borges, Graham C. Burdge, GiacomoCavalli, Vincent Colot, Gyorgy Csaba, Scott Gilbert,Janine Guespin-Michel, Rudolf Hagemann, LuisaHirschbein, David Martin, Marjorie Matzke, FredMeins, Jean Molinier, John Parker, Minoo Rassoulza-degan, Wolf Reik, Yih-Horng Shiao, Michael Skinner,Nadine Vastenhouw, Robert A. Waterland, andGuangtian Zou. We also thank Maya Raz for her con-tribution to the graphics. We are especially grateful toMarion Lamb for reading (and re-reading) the wholemanuscript, and for her invaluable critical commentsand constructive suggestions. Finally, we would like tothank Massimo Pigliucci and two anonymous review-ers for their useful and critical comments.

REFERENCES

Adam M., Murali B., Glenn N. O., Potter S. S. 2008.Epigenetic inheritance based evolution of antibi-

otic resistance in bacteria. BMC Evolutionary Biol-ogy 8:52.

168 Volume 84THE QUARTERLY REVIEW OF BIOLOGY

Agrawal A. A., Laforsch C., Tollrian R. 1999. Trans-generational induction of defences in animals andplants. Nature 401(6748):60–63.

Alleman M., Doctor J. 2000. Genomic imprinting inplants: observations and evolutionary implica-tions. Plant Molecular Biology 43(2–3):147–161.

Allen J. A., editor. 2004. Issue on plant polyploidy.Biological Journal of the Linnean Society 82(4):409–700.

Allen N. D., Norris M. L., Surani M. A. 1990. Epige-netic control of transgene expression and imprint-ing by genotype-specific modifiers. Cell 61(5):853–861.

Allis C. D., Jenuwein T., Reinberg D., Caparros M.-L.,editors. 2007. Epigenetics. Cold Spring Harbor(NY): Cold Spring Harbor Laboratory Press.

Allshire R. C., Selker E. U. 2007. Fungal models forepigenetic research: Schizosaccharomyces pombe andNeurospora crassa. Pages 101–125 in Epigenetics, ed-ited by C. D. Allis et al. Cold Spring Harbor (NY):Cold Spring Harbor Laboratory Press.

Anway M. D., Cupp A. S., Uzumcu M., Skinner M. K.2005. Epigenetic transgenerational actions of en-docrine disruptors and male fertility. Science308(5727):1466–1469.

Anway M. D., Leathers C., Skinner M. K. 2006a. En-docrine disruptor vinclozolin induced epigenetictransgenerational adult-onset disease. Endocrinol-ogy 147(12):5515–5523.

Anway M. D., Memon M. A., Uzumcu M., SkinnerM. K. 2006b. Transgenerational effect of the en-docrine disruptor vinclozolin on male spermato-genesis. Journal of Andrology 27(6):868–879.

Avital E., Jablonka E. 2000. Animal Traditions: Behav-ioural Inheritance in Evolution. Cambridge (UK):Cambridge University Press.

Badyaev A. V. 2005. Stress-induced variation in evolu-tion: from behavioural plasticity to genetic assim-ilation. Proceedings of the Royal Society of London,Series B: Biological Sciences 272(1566):877–886.

Balaban N. Q., Merrin Q. J., Chait R., Kowalik L.,Leibler S. 2004. Bacterial persistence as a pheno-typic switch. Science 305(5690):1622–1625.

Bantignies F., Cavalli G. 2006. Cellular memory anddynamic regulation of polycomb group proteins.Current Opinion in Cell Biology 18(3):275–283.

Barlow D. P., Bartolomei M. S. 2007. Genomic im-printing in mammals. Pages 357–373 in Epigenetics,edited by C. D. Allis et al. Cold Spring Harbor(NY): Cold Spring Harbor Laboratory Press.

Bateman R. M., DiMichele W. A. 2002. Generatingand filtering major phenotypic novelties: neo-Goldschmidtian saltation revisited. Pages 109–159in Developmental Genetics and Plant Evolution, editedby Q. C. B. Cronk et al. London (UK): Taylor &Francis.

Bateson P., Barker D., Clutton-Brock T., Deb D.,

D’Udine B., Foley R. A., Gluckman P., et al. 2004.Developmental plasticity and human health. Na-ture 430(6998):419–421.

Baulcombe D. C. 2007. Amplified silencing. Science315(5809):199–200.

Baxa U., Cassese T., Kajava A. V., Steven A. C. 2006.Structure, function, and amyloididogenesis of fun-gal prions: filament polymorphisms and prionsvariants. Advances in Protein Chemistry 73:125–180

Baylin S. B., Jones P. A. 2007. Epigenetic determi-nants of cancer. Pages 457–476 in Epigenetics, ed-ited by C. D. Allis et al. Cold Spring Harbor (NY):Cold Spring Harbor Laboratory Press.

Bean C. J., Schaner C. E., Kelly W. G. 2004. Meioticpairing and imprinted X chromatin assembly inCaenorhabditis elegans. Nature Genetics 36(1):100–105.

Beisson J., Sonneborn T. M. 1965. Cytoplasmic inher-itance of the organization of the cell cortex inParamecium aurelia. Proceedings of the National Acad-emy of Sciences USA 53(2):275–282.

Belyaev D. K., Borodin P. M. 1982. The influence ofstress on variation and its role in evolution. Biolo-gisches Zentralblatt 100:705–714.

Belyaev D. K., Ruvinsky A. O., Borodin P. M. 1981a.Inheritance of alternative states of the fused genein mice. Journal of Heredity 72:107–112.

Belyaev D. K., Ruvinsky A. O., Trut L. N. 1981b. In-herited activation-inactivation of the star gene infoxes: its bearing on the problem of domestica-tion. Journal of Heredity 72(4):267–274.

Belyaev D. K., Ruvinsky A. O., Agulnik A. I., AgulnikS. I. 1983. Effect of hydrocortisone on the fre-quency and inheritance of the fused gene in mice.Genetika 64(4):275–281.

Benkemoun L., Saupe S. J. 2006. Prion proteins asgenetic material in fungi. Fungal Genetics and Biol-ogy 43(12):789–803.

Bennett S. T., Wilson A. J., Esposito L., Bouzekri N.,Undlien D. E., Cucca F., Nistico L., et al. 1997.Insulin VNTR allele-specific effect in type 1 diabe-tes depends on identity of untransmitted paternalallele. Nature Genetics 17(3):350–352.

Berger S. L. 2007. The complex language of chromatinregulation during transcription. Nature 447(7143):407–412.

Bestor T. H. 1990. DNA methylation: evolution ofa bacterial immune function into a regulatorof gene expression and genome structure inhigher eukaryotes. Philosophical Transactions of theRoyal Society of London, Series B: Biological Sciences326(1235):179–187.

Bird A. P. 1995. Gene number, noise reduction andbiological complexity. Trends in Genetics 11(3):94–100.

Blasco M. A. 2007. The epigenetic regulation of mam-

June 2009 169TRANSGENERATIONAL EPIGENETIC INHERITANCE

malian telomeres. Nature Reviews Genetics 8(4):299–309.

Blewitt M. E., Vickaryous N. K., Paldi A., Koseki H.,Whitelaw E. 2006. Dynamic reprogramming ofDNA methylation at an epigenetically sensitive al-lele in mice. PLoS Genetics 2(4):399–405.

Bossdorf O., Richards C. L., Pigliucci M. 2008. Epige-netics for ecologists. Ecology Letters 11(2):106–115.

Brink R. A. 1973. Paramutation. Annual Review of Ge-netics 7:129–152.

Buiting K., Gro� S., Lich C., Gillessen-Kaesbach G.,El-Maarri O., Horsthemke B. 2003. Epimutationsin Prader-Willi and Angelman syndromes: a mo-lecular study of 136 patients with an imprintingdefect. American Journal of Human Genetics 72(3):571–577.

Burdge G. C., Slater-Jefferies J., Torrens C., PhillipsE. S., Hanson M. A., Lillycrop K. A. 2007. Dietaryprotein restriction of pregnant rats in the F0 gen-eration induces altered methylation of hepaticgene promoters in the adult male offspring in theF1 and F2 generations. British Journal of Nutrition97(3):435–439.

Buss L. W. 1987. The Evolution of Individuality. Prince-ton (NJ): Princeton University Press.

Campbell P., editor. 2007. Follow the curve: the forcesthat shape cell membranes. Nature 447(7143 In-sight):353–500.

Casadesus J., Low D. 2006. Epigenetic gene regulationin the bacterial world. Microbiology and MolecularBiology Reviews 70(3):830–856.

Cavalier-Smith T. 2004. The membranome and mem-brane heredity in development and evolution.Pages 335–351 in Organelles, Genomes and EukaryotePhylogeny, edited by R. P. Hirt and D. S. Horner.Boca Raton (FL): CRC Press.

Cerutti H., Casas-Mollano J. A. 2006. On the originand functions of RNA-mediated silencing: fromprotists to man. Current Genetics 50(2):81–99.

Chandler V. L. 2007. Paramutation: from maize tomice. Cell 128(4):641–645.

Chandler V. L., Eggleston W. B., Dorweiler J. E. 2000.Paramutation in maize. Plant Molecular Biology43(2–3):121–145.

Chang H.-S., Anway M. D., Rekow S. S., Skinner M. K.2006. Transgenerational epigenetic imprinting ofthe male germline by endocrine disruptor expo-sure during gonadal sex determination. Endocri-nology 147(12):5524–5541.

Chien P., Weissman J. S., DePace A. H. 2004. Emerg-ing principles of conformation-based prion inher-itance. Annual Review of Biochemistry 73:617–656.

Chong S., Vickaryous N., Ashe A., Zamudio N.,Youngson N., Hemley S., Stopka T., et al. 2007.Modifiers of epigenetic reprogramming show pa-ternal effects in the mouse. Nature Genetics 39(5):614–622.

Cikos S., Vesela J., Il’kova G., Rehak P., Czikkova S.,Koppel J. 2005. Expression of beta adrenergic re-ceptors in mouse oocytes and preimplantationembryos. Molecular Reproduction and Development71(2):145–153.

Colot V., Rossignol J. L. 1999. Eukaryotic DNA meth-ylation as an evolutionary device. BioEssays 21(5):402–411.

Cormier S., Vandormael-Pournin S., Babinet C.,Cohen-Tannoudji M. 2004. Developmental ex-pression of the Notch signaling pathway genesduring mouse preimplantation development. GeneExpression Patterns 4(6):713–717.

Costa Y., Speed R. M., Gautier P., Semple C. A., Ma-ratou K., Turner J. M. A., Cooke H. J. 2006. MouseMAELSTROM: the link between meiotic silencingof unsynapsed chromatin and microRNA path-way? Human Molecular Genetics 15(15):2324–2334.

Crews D., Gore A. C., Hsu T. S., Dangleben N. L.,Spinetta M., Schallert T., Anway M. D., SkinnerM. K. 2007. Transgenerational epigenetic imprintson mate preference. Proceedings of the NationalAcademy of Sciences USA 104(14):5942–5946.

Cropley J. E., Suter C. M., Beckman K. B., MartinD. I. K. 2006. Germ-line epigenetic modificationof the murine Avy allele by nutritional supplemen-tation. Proceedings of the National Academy of SciencesUSA 103(46):17308–17312.

Csaba G., Karabelyos C., Inczefi-Gonda A., PallingerE. 2005. Three-generation investigation on seroto-nin content in rat immune cells long after�-endorphin exposure in late pregnancy. Hormoneand Metabolic Research 37:172–177.

Cullen B. R. 2004. Transcription and processingof human microRNA precursors. Molecular Cell16(6):861–865.

Cullis C. A. 2005. Mechanisms and control of rapidgenomic changes in flax. Annals of Botany 95(1):201–206.

Delage Y., Goldsmith M. 1912. The Theories of Evolution.Trans. Andre Tridon. New York: B. W. Huebsch.

Dobzhansky T. 1937. Genetics and the Origin of Species.New York: Columbia University Press.

Dubnau D., Losick R. 2006. Bistability in bacteria.Molecular Microbiology 61(3):564–572.

Dubrova Y. E. 2003. Radiation-induced transgenera-tional instability. Oncogene 22(45):7087–7093.

Ekwall K. 2007. ‘Arc’ escorts siRNAs in heterochro-matin assembly. Nature Structural and Molecular Bi-ology 14(3):178–179.

Felsenfeld G., Groudine M. 2003. Controlling thedouble helix. Nature 421(6921):448–453.

Flanagan J. M., Popendikyte V., Pozdniakovaite N.,Sobolev M., Assadzadeh A., Schumacher A., Zan-geneh M., et al. 2006. Intra- and interindividualepigenetic variation in human germ cells. Ameri-can Journal of Human Genetics 79(1):67–84.

170 Volume 84THE QUARTERLY REVIEW OF BIOLOGY

Flintoft L., editor. 2007. Focus on: epigenetics. NatureReviews Genetics 8(4):245–314.

Fontdevila A. 2005. Hybrid genome evolution bytransposition. Cytogenetic and Genome Research110(1):49–55.

Fothergill P. G. 1952. Historical Aspects of Organic Evo-lution. London (UK): Hollis and Carter.

Frankel J. 1989. Pattern Formation Ciliate Studies andModels. New York: Oxford University Press.

Genereux D. P., Miner B. E., Bergstrom C. T., LairdC. D. 2005. A population-epigenetic model to in-fer site-specific methylation rates from double-stranded DNA methylation patterns. Proceedings ofthe National Academy of Sciences USA 102(16):5802–5807.

Ginsburg S., Jablonka E. 2008. Epigenetic learning innon-neural organisms. Journal of Biosciences 33(4).

Gluckman P., Hanson M. 2005. The Fetal Matrix: Evo-lution, Development and Disease. Cambridge (UK):Cambridge University Press.

Gluckman P. D., Hanson M. A., Beedle A. S. 2007.Non-genomic transgenerational inheritance ofdisease risk. BioEssays 29(2):145–154.

Goldschmidt R. B. 1940. The Material Basis of Evolution.New Haven (CT): Yale University Press.

Gottlieb G. 2002. Developmental-behavioral initiationof evolutionary change. Psychological Review 109(2):211–218.

Grandjean V., Hauck Y., Beloin C., Le Hegarat F.,Hirschbein L. 1998. Chromosomal inactivation ofBacillus subtilis exfusants: a prokaryotic model ofepigenetic regulation. Biological Chemistry 379(4–5):553–557.

Grant-Downton R. T., Dickinson H. G. 2005. Epige-netics and its implications for plant biology. 1.The epigenetic network in plants. Annals of Botany96(7):1143–1164.

Grant-Downton R. T., Dickinson H. G. 2006. Epige-netics and its implications for plant biology. 2.The ‘epigenetic epiphany’: epigenetics, evolutionand beyond. Annals of Botany 97(1):11–27.

Grewal S. I. S., Jia S. T. 2007. Heterochromatin revis-ited. Nature Reviews Genetics 8(1):35–46.

Grimes G., Aufderheide K. 1991. Cellular aspects ofpattern formation: the problem of assembly.Monographs in Developmental Biology 22:1–94.

Groth A., Rocha W., Verreault A., Almouzni G. 2007.Chromatin challenges during DNA replicationand repair. Cell 128(4):721–733.

Gu Z., Rifkin S. A., White K. P., and Li W.-H. 2004.Duplicate genes increase gene expression diversitywithin and between species. Nature Genetics 36(6):577–579.

Hadchouel M., Farza H., Simon D., Tiollais P., Pour-cel C. 1987. Maternal inhibition of hepatitis Bsurface antigen gene expression in transgenic

mice correlates with de novo methylation. Nature329(6138):454–456.

Haig D. 2002. Genomic Imprinting and Kinship. NewBrunswick (NJ): Rutgers University Press.

Haig D. 2004. The (dual) origin of epigenetics. ColdSpring Harbor Symposia on Quantitative Biology 69:67–70.

Hajkova P., Erhardt S., Lane N., Haaf T., El-Maarri O.,Reik W., Walter J., Surani M. A. 2002. Epigeneticreprogramming in mouse primordial germ cells.Mechanisms of Development 117(1–2):15–23.

Henderson I. R., Jacobsen S. E. 2007. Epigenetic in-heritance in plants. Nature 447(7143):418–424.

Henikoff S., Ahmad K., Malik H. S. 2001. The centro-mere paradox: stable inheritance with rapidlyevolving DNA. Science 293(5532):1098–1102.

Henikoff S., Smith M. M. 2007. Histone variants andepigenetics. Pages 249–264 in Epigenetics, editedby C. D. Allis et al. Cold Spring Harbor (NY): ColdSpring Harbor Laboratory Press.

Himber C., Dunoyer P., Moissiard G., RitzenthalerC., Voinnet O. 2003. Transitivity-dependent and-independent cell-to-cell movement of RNA si-lencing. EMBO Journal 22(17):4523– 4533.

Hitchins M. P., Wong J. J. L., Suthers G., Suter C. M.,Martin D. I. K., Hawkins N. J., Ward R. L. 2007.Brief report: inheritance of a cancer-associatedMLH1 germ-line epimutation. New England Journalof Medicine 356(7):697–705.

Holliday R. 1984. The significance of DNA methyl-ation in cellular aging. Pages 269–283 in MolecularBiology of Aging, edited by A. D. Woodhead et al.New York: Plenum Press.

Holliday R. 1994. Epigenetics—an overview. Develop-mental Genetics 15:453–457.

Holliday R. 2002. Epigenetics come of age in thetwenty-first century. Journal of Genetics 81(1):1–4.

Holliday R. 2006. Epigenetics: a historical overview.Epigenetics 1(2):76–80.

Huettel B., Kanno T., Daxinger L., Bucher E., van derWinden J., Matzke A. J., Matzke M. 2007. RNA-directed DNA methylation mediated by DRD1 andPol IVb: a versatile pathway for transcriptional genesilencing in plants. Biochimica et Biophysica Acta-GeneStructure and Expression 1769(5–6):358–374.

Issa J. P. 2000. CpG-island methylation in aging andcancer. Current Topics in Microbiology and Immunol-ogy 249:101–118.

Jablonka E. 2004a. The evolution of the peculiaritiesof mammalian sex chromosomes: an epigeneticview. BioEssays 26(12):1327–1332.

Jablonka E. 2004b. Epigenetic epidemiology. Interna-tional Journal of Epidemiology 33(5):929–935.

Jablonka E., Lachmann M., Lamb M. J. 1992. Evi-dence, mechanisms and models for the inheri-tance of acquired characters. Journal of TheoreticalBiology 158(2):245–268.

June 2009 171TRANSGENERATIONAL EPIGENETIC INHERITANCE

Jablonka E., Lamb M. J. 1989. The inheritance ofacquired epigenetic variations. Journal of Theoreti-cal Biology 139(1):69–83.

Jablonka E., Lamb M. J. 1995. Epigenetic Inheritance andEvolution: The Lamarckian Dimension. Oxford (UK):Oxford University Press.

Jablonka E., Lamb M. J. 2002. The changing conceptof epigenetics. Annals of the New York Academy ofSciences 981:82–96.

Jablonka E., Lamb M. J. 2005. Evolution in Four Dimen-sions: Genetic, Epigenetic, Behavioral, and SymbolicVariation in the History of Life. Cambridge (MA):MIT Press.

Jablonka E., Lamb M. J. 2006a. Evolutionary epigenet-ics. Pages 252–264 in Evolutionary Genetics: Conceptsand Case Studies, edited by C. W. Fox and J. B. Wolf.New York: Oxford University Press.

Jablonka E., Lamb M. J. 2006b. The evolution ofinformation in the major transitions. Journal ofTheoretical Biology 239(2):236–246.

Jablonka E., Lamb M. J. 2007a. Precis of Evolution inFour Dimensions. Behavioral and Brain Sciences 30:353–365.

Jablonka E., Lamb M. J. 2007b. Bridging the gap: thedevelopmental aspects of evolution. Behavioral andBrain Sciences 30:378–392.

Jablonka E., Lamb M. J. 2007c. The expanded evolu-tionary synthesis—a response to Godfrey-Smith,Haig, and West-Eberhard. Biology and Philosophy22(3):453–472.

Jablonka E., Lamb M. J. 2008. The epigenome inevolution: beyond the modern synthesis. VOGisHerald 12:242–254.

Jablonka E., Oborny B., Molnar I., Kisdi E., HofbauerJ., Czaran T. 1995. The adaptive advantage of phe-notypic memory in changing environments. Philo-sophical Transactions of the Royal Society of London,Series B: Biological Sciences 350(1332):133–141.

Jaenisch R., Gurdon J. 2007. Nuclear transplantationand the reprogramming of the genome. Pages415–434 in Epigenetics, edited by C. D. Allis et al.Cold Spring Harbor (NY): Cold Spring HarborLaboratory Press.

Jirtle R. L., Skinner M. K. 2007. Environmental epi-genomics and disease susceptibility. Nature ReviewsGenetics 8(4):253–262.

Jollos V. Experimentelle Protistenstudien 1. 1921. Un-tersuchungen uber Variabilitat und Vererbungbei Infusorien. Archiv fur Protistenkunde 43:1–222.

Jorgensen R. A. 2004. Restructuring the genome inresponse to adaptive challenge: McClintock’s boldconjecture revisited. Cold Spring Harbor Symposia onQuantitative Biology 69:349–354.

Karpen G. H., Hawley R. S. 2007. Epigenetic regula-tion of chromosome inheritance. Pages 265–289in Epigenetics, edited by C. D. Allis et al. Cold

Spring Harbor (NY): Cold Spring Harbor Labora-tory Press.

Kidwell M. G., Lisch D. 1997. Transposable elementsas sources of variation in animals and plants. Pro-ceedings of the National Academy of Sciences USA94(15):7704–7711.

Kim V. N. 2006. Small RNAs just got bigger: Piwi-interacting RNAs (piRNAs) in mammalian testes.Genes and Development 20(15):1993–1997.

Kolle S., Stojkovic M., Prelle K., Waters M., Wolf E.,Sinowatz F. 2001. Growth hormone (GH)/GH re-ceptor expression and GH-mediated effects dur-ing early bovine embryogenesis. Biology of Repro-duction 64:1826–1834.

Kussell E., Leibler S. 2005. Phenotypic diversity, pop-ulation growth, and information in fluctuating en-vironments. Science 309(5743):2075–2078.

Lachmann M., Jablonka E. 1996. The inheritance ofphenotypes: an adaptation to fluctuating environ-ments. Journal of Theoretical Biology 181:1–9.

Lamb M. J. 1994. Epigenetic inheritance and aging.Reviews in Clinical Gerontology 4(2):97–105.

Lamm E., Jablonka E. 2008. The nurture of nature:hereditary plasticity in evolution. Philosophical Psy-chology 21(3):305–319.

Laurent M., Charvin G., Guespin-Michel J. 2005. Bi-stability and hysteresis in epigenetic regulation ofthe lactose operon. Since Delbruck, a long seriesof ignored models. Cellular and Molecular Biology51(7):583–594.

Levites E. V. 2000. Epigenetic variability as a source ofbiodiversity and a factor of evolution. Pages 73–75in Biodiversity and Dynamics of Ecosystems in NorthEurasia, Volume 1, Part 3. Novosibirsk (Russia):Institute of Cytology and Genetics SB RAS.

Levy A. A., Feldman M. 2004. Genetic and epigeneticreprogramming of the wheat genome upon al-lopolyploidization. Biological Journal of the LinneanSociety 82(4):607–613.

Li E., Bird A. 2007. DNA methylation in mammals.Pages 341–356 in Epigenetics, edited by C. D. Alliset al. Cold Spring Harbor (NY): Cold Spring Har-bor Laboratory Press.

Lindegren C. C. 1949. The Yeast Cell, its Genetics andCytology. Saint Louis (MO): Educational Publishers.

Lindqvist C., Janczak A. M., Natt D., Baranowska I.,Lindqvist N., Wichman A., Lundeberg J., Lind-berg J., Torjesen P. A., Jensen P. 2007. Transmis-sion of stress-induced learning impairment andassociated brain gene expression from parents tooffspring in chickens. PLoS ONE 2(4):e364.

Little J. B. 2003. Genomic instability and bystandereffects: a historical perspective. Oncogene 22(45):6978–6987.

Logan C. A. 2007. Overheated rats, race, and thedouble gland: Paul Kammerer, endocrinology and

172 Volume 84THE QUARTERLY REVIEW OF BIOLOGY

the problem of somatic induction. Journal of theHistory of Biology 40(4):683–725.

Long L., Lin X., Zhai J., Kou H., Yang W., Liu B. 2006.Heritable alteration in DNA methylation patternoccurred specifically at mobile elements in riceplants following hydrostatic pressurization. Bio-chemical and Biophysical Research Communications340(2):369–376.

Lyon M. F. 1998. X-chromosome inactivation: a re-peat hypothesis. Cytogenetics and Cell Genetics 80:133–137.

Malagnac F., Silar P. 2003. Non-Mendelian determi-nants of morphology in fungi. Current Opinion inMicrobiology 6(6):641–645.

Malagnac F., Silar P. 2006. Regulation, cell differen-tiation and protein-based inheritance. Cell Cycle5(22):2584–2587.

Mandrioli M. 2004. Epigenetic tinkering and evolu-tion: is there any continuity in the role of cytosinemethylation from invertebrates to vertebrates? Cel-lular and Molecular Life Sciences 61(19–20):2425–2427.

Marcus E., editor. 2007. Issue on epigenetics. Cell128(4):627–802.

Martin C., Zhang Y. 2007. Mechanisms of epigeneticinheritance. Current Opinion in Cell Biology 19(3):266–272.

Masel J., Bergman A. 2003. The evolution of the evolv-ability properties of the yeast prion [PSI�]. Evolu-tion 57(7):1498–1512.

Matzke M. A., Birchler J. A. 2005. RNAi-mediatedpathways in the nucleus. Nature Reviews Genetics6(1):24–35.

Matzke M. A., Scheid O. M. 2007. Epigenetic regula-tion in plants. Pages 167–189 in Epigenetics, editedby C. D. Allis et al. Cold Spring Harbor (NY): ColdSpring Harbor Laboratory Press.

Maynard Smith J. 1990. Models of a dual inheritancesystem. Journal of Theoretical Biology 143(1):41–53.

Maynard Smith J., Szathmary E. 1995. The Major Tran-sitions in Evolution. Oxford (UK) and New York:W. H. Freeman Spektrum.

Mayr E. 1982. The Growth of Biological Thought: Diver-sity, Evolution, and Inheritance. Cambridge (MA):Belknap Press.

McClintock B. 1984. The significance of responses ofthe genome to challenge. Science 226(4676):792–801.

Meister G., Tuschl T. 2004. Mechanisms of gene silenc-ing by double-stranded RNA. Nature 431(7006):343–349.

Meyer E., Chalker D. L. 2007. Epigenetics of ciliates.Pages 127–150 in Epigenetics, edited by C. D. Alliset al. Cold Spring Harbor (NY): Cold Spring Har-bor Laboratory Press.

Misteli T. 2007. Beyond the sequence: cellular orga-nization of genome function. Cell 128:787–800.

Molinier J., Ries G., Zipfel C., Hohn B. 2006. Trans-generation memory of stress in plants. Nature 442:1046–1049.

Morris C. A., Moazed D. 2007. Centromere assemblyand propagation. Cell 128(4):647–650.

Mortusewicz O., Schermelleh L., Walter J., CardosoM. C., Leonhardt H. 2005. Recruitment of DNAmethyltransferase I to DNA repair sites. Proceedingsof the National Academy of Sciences USA 102(25):8905–8909.

Nanney D. L. 1958. Epigenetic control systems. Pro-ceedings of the National Academy of Sciences USA44(7):712–717.

Nanney D. L. 1968. Cortical patterns in cellular mor-phogenesis: differences in cortical patterns in cil-iates may be hereditary, but independent of genicdifferences. Science 160(3827):496–502.

Nanney D. L. 1985. Heredity without genes: ciliateexplorations of clonal heredity. Trends in Genetics1:295–298.

Naz R. K., Sellamuthu R. 2006. Receptors in sperma-tozoa: are they real? Journal of Andrology 27(5):627–636.

Newbold R. R., Padilla-Banks E., Jefferson W. N. 2006.Adverse effects of the model environmental estro-gen diethylstilbestrol are transmitted to subse-quent generations. Endocrinology 147:S11–S17.

Novick A., Weiner M. 1957. Enzyme induction as anall-or-none phenomenon. Proceedings of the Na-tional Academy of Sciences USA 43(7):553–566.

Nowacki M., Vijayan V., Zhou Y., Schotanus K., DoakT. G., Landweber L. F. 2008. RNA-mediated epige-netic programming of a genome-rearrangementpathway. Nature 451(7175):153–159.

Pak J., Fire A. 2007. Distinct populations of primaryand secondary effectors during RNAi in C. elegans.Science 315(5809):241–244.

Pal C. 1998. Plasticity, memory and the adaptive land-scape of the genotype. Proceedings of the Royal Societyof London, Series B: Biological Sciences 265(1403):1319–1323.

Palauqui J. C., Elmayan T., Pollien J. M., Vaucheret H.1997. Systemic acquired silencing: transgene-specific post-transcriptional silencing is transmit-ted by grafting from silenced stocks to non-silenced scions. EMBO Journal 16(15):4738–4745.

Pantaleon M., Whiteside E. J., Harvey M. B., BarnardR. T., Waters M. J., Kaye P. L. 1997. Functionalgrowth hormone (GH) receptors and GH are ex-pressed by preimplantation mouse embryos: a rolefor GN in early embryogenesis? Proceedings of theNational Academy of Sciences USA 94(10):5125–5130.

Pavelka J., Koudelova J. 2001. Inheritance of atemperature-modified phenotype of the short an-tennae (sa) mutation in a moth, Ephestia kuehniella(Lepidoptera: Pyralidae). Journal of Heredity 92(3):234–242.

June 2009 173TRANSGENERATIONAL EPIGENETIC INHERITANCE

Pepper J. W., Sprouffske K., Maley C. C. 2007. Animalcell differentiation patterns suppress somatic evo-lution. PloS Computational Biology 3(12):e250.

Petronis A. 2004. The origin of schizophrenia: geneticthesis, epigenetic antithesis, and resolving synthe-sis. Biological Psychiatry 55(10):965–970.

Petronis A. 2006 Epigenetics and twins: three variationson the theme. Trends in Genetics 22(7):347–350.

Pigliucci M. 2001. Phenotypic Plasticity: Beyond Natureand Nurture. Baltimore (MD): John Hopkins Uni-versity Press.

Pigliucci M., Murren C. J., Schlichting C. D. 2006.Phenotypic plasticity and evolution by genetic as-similation. Journal of Experimental Biology 209(12):2362–2367.

Pikaard C. S. 2000. The epigenetics of nucleolar dom-inance. Trends in Genetics 16(11):495–500.

Pikaard C. S. 2003. Nucleolar dominance. Pages 399–402 in Nature Encyclopedia of the Human Genome,Volume 4, edited by D. N. Cooper. London (UK):Nature Publishing Group.

Polo S. E., Roche D., Almouzni G. 2006. New histoneincorporation marks sites of UV repair in humancells. Cell 127(3):481–493.

Popova N. K. 2006. From genes to aggressive behav-ior: the role of serotonergic system. BioEssays28(5):495–503.

Prusiner S. B. 1998. Prions. Proceedings of the NationalAcademy of Sciences USA 95(23):13363–13383.

Rando O. J., Verstrepen K. J. 2007. Timescales ofgenetic and epigenetic inheritance. Cell 128(4):655–668.

Rapp R. A., Wendel J. F. 2005. Epigenetics and plantevolution. New Phytologist 168(1):81–91.

Rassoulzadegan M., Grandjean V., Gounon P., Vin-cent S., Gillot I., Cuzin F. 2006. RNA-mediatednon-mendelian inheritance of an epigeneticchange in the mouse. Nature 441(7092):469–474.

Regev A., Lamb M. J., Jablonka E. 1998. The role ofDNA methylation in invertebrates: developmentalregulation or genome defense? Molecular Biologyand Evolution 15(7):880–891.

Richards E. J. 2006. Inherited epigenetic variation—revisiting soft inheritance. Nature Reviews Genetics7(5):395–401.

Richerson P. J., Boyd R. 2005. Not by Genes Alone: HowCulture Transformed Human Evolution. Chicago(IL): University of Chicago Press.

Ringrose L., Paro R. 2004. Epigenetic regulation ofcellular memory by the polycomb and trithoraxgroup proteins. Annual Review of Genetics 38:413–443.

Ringrose L., Paro R. 2007. Polycomb/trithorax re-sponse elements and epigenetic memory of cellidentity. Development 134(2):223–232.

Roberts B. T., Wickner R. B. 2003. Heritable activity:

a prion that propagates by covalent autoactivation.Genes and Development 17(17):2083–2087.

Rodin S. N., Parkhomchuk D. V., Riggs A. D. 2005.Epigenetic changes and repositioning determinethe evolutionary fate of duplicated genes. Biochem-istry (Moscow) 70(5):559–567.

Ruden D. M., Xiao L., Garfinkel M. D., Lu X. 2005.Hsp90 and environmental impacts on epigeneticstates: a model for the trans-generational effects ofdiethylstibesterol on uterine development andcancer. Human Molecular Genetics 14(Review Issue1):R149–R155.

Ruvinsky A. O., Lobkov Y. I., Belyaev D. K. 1983a.Spontaneous and induced activation of genes af-fecting the phenotypic expression of glucose6-phosphate dehydrogenase in Daphnia pulex. 1.Intraclonal variations in the electrophoretic mo-bility of G6PD. Molecular & General Genetics 189(3):485–489.

Ruvinsky A. O., Lobkov Y. I., Belyaev D. K. 1983b.Spontaneous and induced activation of genes af-fecting the phenotypic expression of glucose6-phosphate dehydrogenase in Daphnia pulex 2.Glucose-induced changes in the electrophoreticmobility of G6PD. Molecular & General Genetics 189:490–494.

Salmon A., Ainouche M. L., Wendel J. F. 2005. Ge-netic and epigenetic consequences of recent hy-bridization and polyploidy in Spartina (Poaceae).Molecular Ecology 14(4):1163–1175.

Sandman K., Pereira S. L., Reeve J. N. 1998. Diversityof prokaryotic chromosomal proteins and the or-igin of the nucleosome. Cellular and Molecular LifeSciences 54(12):1350–1364.

Sangster T. A., Lindquist S., Queitsch C. 2004. Undercover: causes, effects and implications of Hsp90-mediated genetic capacitance. BioEssays 26(4):348–362.

Sano H., Kamada I., Youssefian S., Katsumi M.,Wabiko H. 1990. A single treatment of rice seed-lings with 5-azacytidine induces heritable dwarf-ism and undermethylation of genomic DNA. Mo-lecular and General Genetics 220(3):441–447.

Schacter D. L. 2001. Forgotten Ideas, Neglected Pioneers:Richard Semon and the Story of Memory. Philadelphia(PA): Psychology Press.

Schermelleh L., Haemmer A., Spada F., Rosing N.,Meilinger D., Rothbauer U., Cardoso M., Leon-hardt H. 2007. Dynamics of Dnmt1 interactionwith the replication machinery and its role inpostreplicative maintenance of DNA methylation.Nucleic Acids Research 35(13):4301–4312.

Schlichting C. D., Pigliucci M. 1998 Phenotypic Evolu-tion: A Reaction Norm Perspective. Sunderland (MA):Sinauer.

Schmalhausen I. I. 1949. Factors of Evolution: The The-

174 Volume 84THE QUARTERLY REVIEW OF BIOLOGY

ory of Stabilizing Selection. Philadelphia (PA): Black-iston.

Schuettengruber B., Chourrout D., Vervoort M., Le-blanc B., Cavalli G. 2007. Genome regulation bypolycomb and trithorax proteins. Cell 128(4):735–745.

Shapiro J. A. 1999. Genome system architecture andnatural genetic engineering in evolution. Annals ofthe New York Academy of Sciences 870:23–35.

Shiao Y. H., Crawford E. B., Anderson L. M., Patel P.,Ko K. 2005. Allele-specific germ cell epimutationin the spacer promoter of the 45S ribosomal RNAgene after Cr(III) exposure. Toxicology and AppliedPharmacology 205(3):290–296.

Shiu P. K. T., Raju N. B., Zickler D., Metzenberg R. L.2001. Meiotic silencing by unpaired DNA. Cell107(7):905–916.

Shorter J., Lindquist S. 2005. Prions as adaptive con-duits of memory and inheritance. Nature ReviewsGenetics 6(6):435–450.

Siegal M. L., Bergman A. 2006. Canalization. Pages235–251 in Evolutionary Genetics, edited by C. W.Fox and J. B. Wolf. New York: Oxford UniversityPress.

Smits W. K., Kuipers O. P., Veening J. W. 2006. Phe-notypic variation in bacteria: the role of feedbackregulation. Nature Reviews Microbiology 4(4):259–271.

Sokolov V. A. 2006. Imprinting in plants. Russian Jour-nal of Genetics 42(9):1043–1052.

Steele E. J., Lindley R. A., Blanden R. V. 1998.Lamarck’s Signature: How Retrogenes are ChangingDarwin’s Natural Selection Paradigm. Reading (MA):Perseus Books.

Talbert P. B., Bryson T., Henikoff S. 2004. Adaptiveevolution of centromere proteins in plants andanimals. Journal of Biology 3:18.

True H. L., Lindquist S. L. 2000. A yeast prion pro-vides a mechanism for genetic variation and phe-notypic diversity. Nature 407(6803):477–483.

True H. L., Berlin I., Lindquist S. L. 2004. Epigeneticregulation of translation reveals hidden geneticvariation to produce complex traits. Nature431(7005):184–187.

Trut L. N., Plyusnina I. Z., Oskina I. N. 2004. Anexperiment on fox domestication and debatableissues of evolution of the dog. Russian Journal ofGenetics 40(6):644–655.

Tuite M. F., Cox B. S. 2006. The [PSI�] prion of yeast:a problem of inheritance. Methods 39(1):9–22.

Turner J. M. A., Mahadevaiah S. K., Fernandez-Capetillo O., Nussenzweig A., Xu X. L., DengC.-X., Burgoyne P. S. 2005. Silencing of unsyn-apsed meiotic chromosomes in the mouse. NatureGenetics 37(1):41–47.

Uptain S. M., Lindquist S. 2002. Prions as protein-

based genetic elements. Annual Review of Microbi-ology 56:703–741.

Van der Heijden G. W., Derijck A., Ramos L., GieleM., Van der Vlag J., de Boer P. 2006. Transmissionof modified nucleosomes from the mouse malegermline to the zygote and subsequent remodel-ing of paternal chromatin. Developmental Biology298(2):458–469.

Vanyushin B. F. 2006a. DNA methylation and epige-netics. Russian Journal of Genetics 42(9):985–997.

Vanyushin B. F. 2006b. DNA methylation in plants.Current Topics in Microbiology and Immunology 301:67–122.

Vanyushin B. F., Nemirovsky L. E., Klimenko V. V.,Vasiliev V. K., Belozersky A. N. 1973. The5-methylcytosine in DNA of rats. Tissue and agespecificity and the changes induced by hydrocor-tisone and other agents. Gerontologia 19(3):138–152.

Vastenhouw N. L., Brunschwig K., Okihara K. L., Mul-ler F., Tijsterman M., Plasterk R. H. A. 2006. Long-term gene silencing by RNAi. Nature 442(7105):882.

Vesela J., Rehak P., Mihalik J., Czikkova S., Pokorny J.,Koppel J. 2003. Expression of serotonin receptorsin mouse oocytes and preimplantation embryos.Physiological Research 52(2):223–228.

Vrana P. B., Fossella J. A., Matteson P., del Rio T.,O’Neill M. J., Tilghman S. M. 2000. Genetic andepigenetic incompatibilities underlie hybrid dys-genesis in Peromyscus. Nature Genetics 25(1):120–124.

Waddington C. H. 1957. The Strategy of the Genes: ADiscussion of Some Aspects of Theoretical Biology. Lon-don (UK): Allen & Unwin.

Waddington C. H. 1968. The basic ideas of biology.Pages 1–32 in Towards a Theoretical Biology: Prolego-mena, edited by C. H. Waddington. Edinburgh(UK): Edinburgh University Press.

Waddington C. H. 1975. The Evolution of an Evolution-ist. Edinburgh (UK): Edinburgh University Press.

Waterland R. A., Travisano M., Tahiliani K. G. 2007.Diet-induced hypermethylation at agouti viable yel-low is not inherited transgenerationally throughthe female. FASEB Journal 21(12):3380–3385.

Waterland R. A., Travisano M., Tahiliani K. G.,Rached M. T., Mirza S. 2008. Methyl donor sup-plementation prevents transgenerational amplifi-cation of obesity. International Journal of Obesity 32:1373–1379.

Waters E. R., Schaal B. A. 1996. Heat shock induces aloss of rRNA-encoding DNA repeats in Brassicanigra. Proceedings of the National Academy of SciencesUSA 93(4):1449–1452.

Weaver I. C. G., Cervoni N., Champagne F. A.,D’Alessio A. C., Sharma S., Seckl J. R., Dymov S.,Szyf M., Meaney M. J. 2004. Epigenetic program-

June 2009 175TRANSGENERATIONAL EPIGENETIC INHERITANCE

ming by maternal behavior. Nature Neuroscience7(8):847–854.

West-Eberhard M. J. 2003. Developmental Plasticity andEvolution. New York: Oxford University Press.

Wickner R. B. 1994. [URE3] as an altered URE2protein: evidence for a prion analog in Saccharo-myces cerevisiae. Science 264(5158):566–569.

Wickner R. B., Edskes H. K., Ross E. D., Pierce M. M.,Baxa U., Brachmann A., Shewmaker F. 2004.Prion genetics: new rules for a new kind of gene.Annual Review of Genetics 38:681–707.

Wolf J. B., Hager R. 2006. A maternal–offspring co-adaptation theory for the evolution of genomicimprinting. PLoS Biology 4(12):2238–2243.

Wu C.-T., Morris J. R. 2001. Genes, genetics, andepigenetics: a correspondence. Science 293(5532):1103–1105.

Wu T. C. J., Wang L., Wan Y.-J. Y. 1992. Expression ofestrogen receptor gene in mouse oocyte and dur-ing embryogenesis. Molecular Reproduction and De-velopment 33(4):407–412.

Yi H., Riddle N. C., Stokes T. L., Woo H.-R., RichardsE. J. 2004. Induced and natural epigenetic varia-tion. Cold Spring Harbor Symposia on QuantitativeBiology 69:155–160.

Zacharioudakis I., Gligoris T., Tzamarias D. 2007. Ayeast catabolic enzyme controls transcriptionalmemory. Current Biology 17(23):2041–2046.

Zaratiegui M., Irvine D. V., Martienssen R. A. 2007. Noncoding RNAs and gene silencing. Cell 128(4):763–776.

Zhivotovsky L. A. 2002. A model of the early evolutionof soma-to-germline feedback. Journal of TheoreticalBiology 216(1):51–57.

Zoghbi H. Y., Beaudet A. L. 2007. Epigenetics andhuman disease. Pages 435–456 in Epigenetics, ed-ited by C. D. Allis et al. Cold Spring Harbor (NY):Cold Spring Harbor Laboratory Press.

Zordan R. E., Galgoczy D. J., Johnson A. D. 2006. Epi-genetic properties of white-opaque switching in Can-dida albicans are based on a self-sustaining transcrip-tional feedback loop. Proceedings of the NationalAcademy of Sciences USA 103(34):12807–12812.

Zuckerkandl E., Cavalli G. 2007. Combinatorial epi-genetics, “junk DNA,” and the evolution of com-plex organisms. Gene 390(1–2):232–242.

Zufall R. A., Robinson T., Katz L. A. 2005. Evolutionof developmentally regulated genome rearrange-ments in eukaryotes. Journal of Experimental Zoology,Part B: Molecular and Developmental Evolution304B(5):448–455.

176 Volume 84THE QUARTERLY REVIEW OF BIOLOGY