You are on page 1of 13

REVIEWS

Dynamic interactions between


transposable elements and their hosts
Henry L. Levin* and John V. Moran‡
Abstract | Transposable elements (TEs) have a unique ability to mobilize to new genomic
locations, and the major advance of second-generation DNA sequencing has provided
insights into the dynamic relationship between TEs and their hosts. It now is clear that TEs
have adopted diverse strategies — such as specific integration sites or patterns of activity
— to thrive in host environments that are replete with mechanisms, such as small RNAs or
epigenetic marks, that combat TE amplification. Emerging evidence suggests that TE
mobilization might sometimes benefit host genomes by enhancing genetic diversity,
although TEs are also implicated in diseases such as cancer. Here, we discuss recent
findings about how, where and when TEs insert in diverse organisms.

Through her pioneering work in maize, Barbara TE‑mediated events to the inter- and intra-individual
McClintock was the first to realize that eukaryotic structural variation in mammalian genomes.
genomes are not static entities but contain transposable TE mobility poses a serious challenge to host fitness.
elements (TEs) that have the ability to move from one Paradoxically, TE insertions that are harmful to the host
chromosomal location to another 1. It now is clear that jeopardize TE survival. Thus, many TEs have evolved
virtually all organisms harbour TEs that have amplified highly specific targeting mechanisms that direct their
in copy number over evolutionary time via DNA or RNA integration to genomic ‘safe havens’, thereby minimizing
intermediates. On occasions, TEs have been sporadically their damage to the host (for example, see REFS 25–29).
co-opted by the host to perform crucial cellular func- Nevertheless, host genomes have evolved potent restric-
tions (for example, see REFS 2–5). However, most TEs are tion mechanisms, such as the methylation of TE DNA
likely to be finely tuned genomic parasites that mobilize sequences and the expression of small RNAs or cytidine
to ensure their own survival6–9.The genomic revolution, deaminases, to restrict TE activity in the germ line and
coupled with new DNA sequencing technologies, now perhaps in somatic cells (for example, see REFS 30–33).
provides an unprecedented wealth of data documenting Interestingly, a growing number of examples suggest
TE content and mobility in a broad array of organisms. that TEs may become activated under certain environ-
In multicellular eukaryotes, TEs must mobilize within mental conditions, such as stress. Stress has been shown
*Section on Eukaryotic
Transposable Elements, gametes or during early development to be transmitted to induce TE transcription, integration or to redirect TE
Laboratory of Gene to future generations. In humans, there are at least 65 integration to alternative target sites34–38. These findings
Regulation and Development, documented cases of diseases resulting from de novo TE are consistent with Barbara McClintock’s hypothesis that
Eunice Kennedy Shriver insertions, and these events account for approximately environmental challenges may induce transposition and
National Institute of Child
Health and Human
1 in 1,000 spontaneous cases of disease5,10. Indeed, new that transposition, in turn, may create genetic diversity
Development, National genomic technologies combined with cell-culture-based to overcome threats to host survival39.
Institutes of Health, Bethesda, experiments have demonstrated that active TEs are more We begin this review with a brief description of
Maryland 20892, USA. prevalent in the human population than was previously the types of TEs and their modes of mobility. We then

Departments of Human
appreciated11–18. A growing body of evidence further describe the latest understanding of TE integration mech-
Genetics and Internal
Medicine and Howard suggests that mammalian TE integration occurs during anisms and how the host defends against these attacks.
Hughes Medical Institute, early development 19–21. In addition, studies of neurogen- Finally, we discuss exciting new research that suggests
University of Michigan esis and some forms of cancer have raised the intriguing that TE mobility may affect the biology of somatic cells.
Medical School, Ann Arbor, possibility that TE activity may have an impact on the From the growing understanding of target-site selec-
Michigan 48109–6518, USA.
e‑mails: henry_levin@nih.gov;
biology of particular somatic cells12,22–24. It is likely that tion to the discovery of new, active TE copies in human
moranj@umich.edu we have observed only the tip of the iceberg and populations, it is clear that the field of transposon biology
doi:10.1038/nrg3030 that we are still underestimating the contribution of continues to yield new insights about genome biology.

NATURE REVIEWS | GENETICS VOLUME 12 | SEPTEMBER 2011 | 615

© 2011 Macmillan Publishers Limited. All rights reserved


REVIEWS

The diversity and abundance of transposons SINE-R–VNTR–Alu elements (SVA elements))47,48 comprise
Mobilization mechanisms. TEs mobilize by remarkably approximately 30% of human genomic DNA sequence59.
diverse replication strategies40 (FIG. 1; TABLE 1). Many Recent research has used a combination of transposon
DNA transposons mobilize by a non-replicative ‘cut display68,69, second-generation DNA sequencing12,15–17 and
and paste’ mechanism, whereby an element-encoded analyses of genomic DNA sequences from the Human
enzyme, the transposase, recognizes sequences at or Structural Variation project 13,70–72, the 1000 Genomes
near TE inverted terminal repeats to cut the TE from its Project 18,73,74 and clinical cohorts14. This research has
existing genomic location and then to paste the excised revealed that L1 presence/absence dimorphisms, as
DNA into a new genomic location41 (FIG. 1a). well as non-allelic recombination between L1 and Alu
Retrotransposons mobilize by the reverse transcrip- elements, account for an appreciable proportion of the
tion of an RNA intermediate; however, different types inter-individual structural variation that is observed
Long terminal repeat of retrotransposons carry out this process by distinct among humans and continue to have a profound effect on
(LTR). A terminal repeated
sequence present at the
mechanisms. Long terminal repeat (LTR) retrotranspo- the human genome (see REF. 5 for a detailed review).
ends of LTR retrotransposons. sons42–44 (FIG. 1b) and non-LTR retrotransposons45 (FIG. 1c)
The LTR contains cis-acting use element-encoded enzymes to mediate their mobility. The diverse patterns of integration sites
sequences that allow In addition, the endonuclease and reverse transcriptase Transposons exhibit a remarkable diversity of integra-
the transcription and
activities of non-LTR retrotransposons also have a cen- tion behaviours. Some TEs preferentially integrate into
polyadenylation of
retrotransposon mRNA. tral role in mobilizing non-autonomous short interspersed gene-dense regions of the genome, whereas others target
LTRs also have crucial roles in elements (SINEs)46–48, certain classes of non-coding regions such as heterochromatin, telomeres or ribosomal
the reverse transcription of RNAs49–52 and messenger RNAs, the latter of which DNA arrays, and some seem to insert throughout the
LTR retrotransposon mRNA. results in the formation of processed pseudogenes53,54. genome. Below, we describe several examples of TE inte-
Short interspersed elements
gration and what is known about how TEs target specific
(SINEs). A family of Transposon activity across species. Examples of DNA sites in genomic DNA.
non-autonomous TEs include Tn5 and Tn7 of Escherichia coli 55,56, P ele-
retrotransposons that require ments of Drosophila melanogaster 57, and Tc1 elements of Integration into gene-rich regions. Many TEs integrate
functional proteins encoded
Caenorhabditis elegans58. Although they thrive in bacteria into gene-rich regions, although they use mechanisms
by long interspersed elements
(LINEs) to mediate their and simpler eukaryotes, DNA TE activity appears to be that prevent the disruption of ORFs. An extreme exam-
retrotransposition. extinct in most mammals, which has fuelled specula- ple is the E. coli Tn7 DNA TE. Tn7 encodes the sequence-
tion that DNA TEs have a limited role in the ongoing specific DNA-binding protein TnsD, which mediates
hAT elements evolution of mammalian genomes59. However, recent Tn7 integration into a specific position in the host chro-
A family of transposons named studies have suggested that DNA TEs, including hAT mosome, termed attnTn7, and thereby avoids damaging
after the hobo, Activator
elements and helitrons, are active in certain bat species60–62. the host genome75,76. A second targeting protein, TnsE,
and Tam3 elements.
Thus, these studies highlight how new DNA sequenc- can alter Tn7 target preference by directing integration
LINE‑1 ing technologies can facilitate fundamental discoveries to plasmids that are transferred between E. coli by conju-
(L1). An abundant family of about the impact of different TE families on genome gation77 or to dsDNA breaks and DNA structures formed
autonomous, long interspersed
evolution and serve as a cautionary note against deriving during DNA replication78. By comparison, the D. mela‑
element (LINE) non-LTR
retrotransposons in general conclusions regarding TE activity from relatively nogaster P element avoids disrupting ORFs by integrat-
mammalian genomes. In few ‘reference’ sequences. ing within 500 bp upstream of transcription start sites of
humans, L1 elements comprise LTR retrotransposons are particularly abundant genes79. However, the mechanism by which P elements
~17% of genomic DNA. in eukaryotes. For example, D. melanogaster contains target these sites requires elucidation.
The vast majority of L1s
are inactive; however, it is
approximately 20 distinct families of LTR retrotranspo- Certain non-LTR retrotransposons encode endonu-
estimated that an average sons that comprise ~1% of the genome63, whereas maize cleases that target specific sites in genomic DNA. For
human genome contains contains ~400 families of LTR retrotransposons that example, the R1 and R2 elements of insects encode
~80–100 active elements. comprise ~75% of the genome64,65. In addition, the mouse sequence-specific endonucleases that cleave at specific
genome contains multiple active LTR-retrotransposon positions within the 28S rDNA locus to initiate target-
Long interspersed elements
(LINEs). A family of families. Indeed, the ongoing retrotransposition of site-primed reverse transcription (TPRT)28,45. However,
autonomous non-long both autonomous LTR retrotransposons and their non- these endonucleases operate by distinct mechanisms.
terminal repeat (non-LTR) autonomous derivatives is estimated to account for R1 encodes an endonuclease that shares sequence simi-
retrotransposons that mobilize approximately 10–12% of sporadic mutations in the larity with an apurinic or apyrimidinic site DNA repair
by retrotransposition.
mouse66. By comparison, there seems to be little LTR endonuclease (APE)80,81, whereas R2 encodes a type IIS
Alu retrotransposon activity in human genomes59; however, restriction endonuclease82. Thus, these elements appar-
An abundant class of short a small number of human endogenous retroviruses are ently have evolved convergent mechanisms to integrate
interspersed elements (SINEs) polymorphic with respect to their presence or absence into ribosomal DNA arrays.
that comprise ~10% of human
at a given genomic location, suggesting that they have LTR retrotransposons also have evolved strategies to
genomic DNA. Alu elements
require the endonuclease and retrotransposed (integrated into new locations) relatively integrate into gene-rich regions while ensuring minimal
reverse transcriptase activities recently in human evolution67. damage to their hosts. For example, the Ty1 and Ty3 ret-
contained within the long Non-LTR retrotransposons are widespread among rotransposons of Saccharomyces cerevisiae specifically
interspersed element 1 (L1) eukaryotes, but have been especially prolific in mam- target gene-free windows that are located immediately
ORF2‑encoded protein to
mediate their mobility. Some
malian genomes. For example, LINE‑1 (long interspersed upstream of RNA polymerase III-transcribed genes,
Alu elements remain active in element 1; abbreviated here to L1) and the non-autonomous such as tRNAs25,27,83,84; Ty3 is directed to integration sites
the human genome. SINEs that they mobilize (for example, Alu and 2–3 bp upstream of such genes by the transcription factor

616 | SEPTEMBER 2011 | VOLUME 12 www.nature.com/reviews/genetics

© 2011 Macmillan Publishers Limited. All rights reserved


REVIEWS

C &0#VTCPURQUQP D .64TGVTQVTCPURQUQP E 0QP.64TGVTQVTCPURQUQP


n%WVCPFRCUVGo6' 4GRNKECVKXGTGVTQVTCPURQUKVKQP 6CTIGVUKVGRTKOGFTGXGTUGVTCPUETKRVKQP

)CI 4GXGTUGVTCPUETKRVCUG ′764 14( RQN[


#
6TCPURQUCUG
2TQVGCUG +PVGITCUG 14( ′764

6TCPUETKRVKQP
6TCPURQUCUG 6TCPUETKRVKQP
DKPFKPI
40#
40#RQN[OGTCUG++
6TCPURQUCUG 40# 40#RQN[OGTCUG++

6TCPURQUCUG
4GXGTUGVTCPUETKRVKQP

2TKOKPI
CPFTGXGTUG
VTCPUETKRVKQP
'ZEKUKQP
40#
$$
$$
E&0# $$ 777
40# 77
7

.E&0#
14(RTQVGKP
6TCPURQUCUG
5GEQPFUVTCPF
U[PVJGUKUEQORNGVKQP
6CTIGV 6TCPURQTVQH QHKPVGITCVKQP
E&0#VQPWENGWU

+PVGITCUG
+PVGITCVKQP

+PVGITCVKQP

6TCPURQUCUG

Figure 1 | The diverse mechanisms of transposon mobilization. a | DNA transposons. Many DNA transposons are
0CVWTG4GXKGYU^)GPGVKEU
flanked by terminal inverted repeats (TIRs; black arrows), encode a transposase (purple circles), and mobilize by a
‘cut and paste’ mechanism (represented by the scissors). The transposase binds at or near the TIRs, excises the
transposon from its existing genomic location (light grey bar) and pastes it into a new genomic location (dark grey
bar). The cleavages of the two strands at the target site are staggered, resulting in a target-site duplication (TSD)
typically of 4–8 bp (short horizontal black lines flanking the transposable element (TE)) as specified by the
transposase. Retrotransposons (b and c) mobilize by replicative mechanisms that require the reverse transcription
of an RNA intermediate. b | LTR retrotransposons contain two long terminal repeats (LTRs; black arrows) and
encode Gag, protease, reverse transcriptase and integrase activities, all of which are crucial for retrotransposition.
The 5′ LTR contains a promoter that is recognized by the host RNA polymerase II and produces the mRNA of the
TE (the start-site of transcription is indicated by the right-angled arrow). In the first step of the reaction,
Gag proteins (small pink circles) assemble into virus-like particles that contain TE mRNA (light blue), reverse
transcriptase (orange shape) and integrase. The reverse transcriptase copies the TE mRNA into a full-length dsDNA.
SINE-R–VNTR–Alu In the second step, integrase (purple circles) inserts the cDNA (shown by the wide, dark blue arc) into the new
elements target site. Similarly to the transposases of DNA transposons, retrotransposon integrases create staggered cuts at
(SVA elements). Composite, the target sites, resulting in TSDs. c | Non-LTR retrotransposons lack LTRs and encode either one or two ORFs. As for
non-autonomous LTR retrotransposons, the transcription of non-LTR retrotransposons generates a full-length mRNA (wavy, light blue
retrotransposons that also line). However, these elements mobilize by target-site-primed reverse transcription (TPRT). In this mechanism, an
require long interspersed element-encoded endonuclease generates a single-stranded ‘nick’ in the genomic DNA, liberating a 3′-OH that is
element 1 (L1)-encoded
used to prime reverse transcription of the RNA. The proteins that are encoded by autonomous non-LTR
proteins to mediate their
retrotransposons can also mobilize non-autonomous retrotransposon RNAs, as well as other cellular RNAs (see the
mobility. SVA elements are less
abundant than Alu elements,
main text). The TPRT mechanism of a long interspersed element 1 (L1) is depicted in the figure; the new element
and certain families of SVA (dark blue rectangle) is 5′ truncated and is retrotransposition-defective. Some non-LTR retrotransposons lack
elements remain active poly(A) tails at their 3′ ends. The integration of non-LTR retrotransposons can lead to TSDs or small deletions
in the human genome. at the target site in genomic DNA. For example, L1s are generally flanked by 7–20 bp TSDs.

NATURE REVIEWS | GENETICS VOLUME 12 | SEPTEMBER 2011 | 617

© 2011 Macmillan Publishers Limited. All rights reserved


REVIEWS

Table 1 | Classes of transposable elements and their mobility mechanisms


Class of TE Structural features Replication mechanism Variant forms Active examples
DNA transposons • TIRs Transposase-mediated • Some DNA transposons also mobilize via • Tn7 in Escherichia coli
• Transposase excision of donor dsDNA replicative mechanisms • P elements in Drosophila
followed by insertion into • ssDNA transposons lack TIRs: donor ssDNA melanogaster
the target site is inserted into target-site ssDNA, such as for • Tc1 elements in
IS608 of Helicobacter pylori Caenorhabditis elegans
LTR • LTRs Within virus-like particles, • Solo LTRs are commonly found in genomes • Ty1, Ty3 and Ty5 in
retrotransposons • Gag, protease, reverse transcriptase and are a result of LTR–LTR recombination Saccharomyces cerevisiae
reverse copies the mRNA of the TE • Tf1 and Tf2 in
transcriptase and into a full-length cDNA; Schizosaccharomyces
integrase integrase inserts the pombe
cDNA into target sites • Tnt1 in tobacco
Non-LTR • One or two ORFs An element-encoded • Non-autonomous, non-LTR retrotransposons • L1 in human, mouse, and
retrotransposons • 5′ truncations endonuclease mediates (for example, Alu and SVA elements, as other mammals
and inversion/ TPRT. The endonuclease well as other eukaryotic SINEs) rely on the • I factor in D.
deletion (for nicks the DNA at the endonuclease and reverse transcriptase of melanogaster
mammalian target site and uses the 3′ an autonomous non-LTR retrotransposon to • Zorro3 in
L1 elements) nicked end for the primer mediate retrotransposition Candida albicans
• Some end in poly(A) as it reverse transcribes TE • The L1 retrotransposition machinery can • R1 and R2 in insects
tails (for example, mRNA also mobilize mRNAs (to generate processed
L1s); others do not pseudogenes) and certain non-coding RNAs
(for example, R2) (for example, the U6 snRNA)
L1, long interspersed element 1; LTR, long terminal repeat; SINE, short interspersed element; snRNA, small nuclear RNA; SVA, SINE-R–VNTR–Alu; TE, transposable
element; TIR, terminal inverted repeat; TPRT, target-site-primed reverse transcription.

complexes TFIIIB and TFIIIC85,86 (FIG. 2a). However, in fructose-1,6-bisphosphatase (fbp1) promoter is induced
the case of the snR6 non-coding RNA gene, which does when the activating transcription factor Atf1 binds to
not depend on TFIIIC for its expression, the TFIIIB an 8 bp upstream activating sequence (UAS1)98. Tf1
factors Brf1 and TATA-box binding protein (TBP) are integration generally occurs 30 bp or 40 bp downstream
sufficient to direct Ty3 integration87,88. of UAS1; however, mutating six nucleotides of UAS1 or
Ty1 integrates into a ~700 bp window upstream deleting the atf1 gene disrupts Tf1 integration specificity,
of tRNA genes with a periodicity of 80 bp27,89 (FIG. 2b). causing integration to occur throughout the plasmid26.
Although the factors that direct Ty1 to tRNA genes remain Although cells lacking Atf1 show little reduction in the
unknown, the unusual periodicity of integration depends overall Tf1 retrotransposition frequency 99, the above
on the amino-terminal domain of Bdp1, which is another data, as well as the finding that Atf1 forms a complex
TFIIIB factor 90. The ability to integrate upstream of RNA with Tf1 integrase, indicate that specific transcription
polymerase III-transcribed genes can also regulate host factors such as Atf1 can play a crucial part in directing
and TE gene expression. For example, Ty1 and Ty3 inser- Tf1 integration to a specific target site. Notably, experi-
tions can stimulate the transcription of downstream RNA ments conducted with a synthetic promoter revealed
polymerase III-transcribed genes and transcription of that RNA polymerase II transcription is not sufficient
the RNA polymerase III target genes can reciprocate by to target Tf1 integration99.
repressing Ty1 transcription91,92. Clearly, determining how The recent development of second-generation
Ty1 targets integration sites and exploring how integration sequencing technology has allowed the in vivo examina-
alters gene regulation remain areas for future study. tion of Tf1 integration sites en masse. Characterization
The ability to target RNA polymerase III-transcribed of 73,125 Tf1 integration events from four independ-
genes is not unique to LTR retrotransposons. For exam- ent experiments revealed a highly reproducible pattern:
ple, the Dictyostelium discoideum non-LTR retrotranspo- approximately 95% of integration events are clustered
son DRE (also known as TRE5A) preferentially inserts upstream of ORFs96. Interestingly, the most frequently
Target-site-primed reverse ~48 bp upstream of tRNA genes, whereas the retrotrans- targeted promoters are associated with genes that are
transcription
(TPRT). The mechanism of
poson Tdd3 (also known as TRE3A) inserts downstream induced by environmental stresses. The targeting of
mobility that is generally used of tRNA genes29,93. Indeed, experimental evidence sug- genes that respond to stress, coupled with the ability
by long interspersed elements gests that the TRE5A ORF1‑encoded protein directly of Tf1 to induce the expression of adjacent genes26, sug-
(LINEs) and short interspersed interacts with subunits of TFIIIB to direct its integration gests that Tf1 integration has the potential to improve
elements (SINEs). An
to tRNA genes94. the survival of specific cells that are exposed to environ-
endonuclease, encoded by
the LINE, nicks genomic The Schizosaccharomyces pombe retrotransposon mental stress. Likewise, the transcription of Tf2, another
DNA to expose a 3′-OH at Tf1 preferentially integrates into the promoters of RNA LTR retrotransposon in S. pombe, is induced by oxidative
the target site that can be used polymerase II-transcribed genes and provides another and osmotic stress or by growth in low oxygen concen-
as a primer to initiate the example of how TEs target gene-rich regions 95–97 trations34,100. Clearly, understanding the consequences of
reverse transcription of the
(FIG. 2c). Tf1 integration has been studied by examining stress-induced retrotransposition will yield insights about
retrotransposon RNA by a
LINE-encoded reverse its integration into promoters contained within extra- how TE mobility can lead to genetic diversity, which may
transcriptase. chromosomal replicating plasmids26. For example, the affect the ability of an organism to cope with stress.

618 | SEPTEMBER 2011 | VOLUME 12 www.nature.com/reviews/genetics

© 2011 Macmillan Publishers Limited. All rights reserved


REVIEWS

Finally, certain retroviruses, which are descended fused to GFP colocalize with heterochromatin108, which
from LTR retrotransposons101, also exhibit preferential suggests that the chromodomain has a principal role in
integration in gene-rich regions. For example, HIV‑1 directing integration. Indeed, fusion of one such chro-
preferentially integrates into RNA polymerase II- modomain to the C terminus of Tf1 integrase directs
transcribed genes, whereas murine leukaemia virus integration of this TE to heterochromatin108.
shows a strong integration preference near transcrip- The Ty5 LTR retrotransposon also targets gene-
tional start sites 102–104. Structural and biochemical poor regions in S. cerevisiae. Approximately 90% of Ty5
data demonstrate that HIV‑1 integrase interacts with integration events occur within the silent mating type
the cellular lens epithelium-derived growth factor loci or near silent heterochromatin at telomeres109–111
(LEDGF; also known as p75) host factor, and there is (FIG. 2d). Genetic and biochemical experiments indi-
evidence that this interaction has an important role in cate that a nine-amino-acid targeting domain in the
proviral DNA integration105,106. Ty5 integrase C terminus directly binds to a structural
component of heterochromatin, Sir4, to target integra-
Integration into heterochromatin. Some TEs target tion35,112,113. Moreover, fusing Sir4 to the DNA bind-
heterochromatic sequences that contain relatively few ing domain of the LexA repressor protein causes Ty5
genes. For example, chromoviruses, which are related to integration to be redirected to LexA binding sites114.
Ty3–Gypsy LTR retrotransposons, reside in the hetero- Thus, the Ty5 integrase targeting domain, by inter-
chromatin of eukaryotes from fungi to vertebrates107. acting with Sir4, directs integration to heterochroma-
The integrase enzymes of these viruses contain a tin. Interestingly, genetic and biochemical evidence
chromodomain near the carboxyl terminus that is indicate that the Ty5 targeting domain evolved its
related to the heterochromatin protein HP1, which interaction with Sir4 by mimicking residues in a host
binds to histone H3 methylated at lysine 9 (REF. 107). factor, Esc1p, that binds to the same amino acids of
Furthermore, chromovirus integrase chromodomains Sir4 (REF. 115).

C6[VCTIGVKPI Figure 2 | Mechanisms that position integration.


6(+++$ a | The Ty3 targeting mechanism. Integration of Ty3
+PVGITCUG 6(+++% occurs one or two base pairs upstream of tRNA genes.
V40#IGPG This pattern requires Brf1 and TATA-box binding protein
(TBP), which are components of the transcription factor
complex TFIIIB that recruits integrase (grey oval) to
the target site. b | The Ty1 targeting mechanism. The
transcription factor Bdp1 is a component of TFIIIB and is
D6[VCTIGVKPI required to recruit the chromatin remodelling complex
+PVGITCUG Isw2 (light blue semicircle). Orange cylinders indicate
nucleosomes and black lines represent DNA. Integration
6(+++$ of Ty1 occurs with an 80 bp periodicity in a 700 bp
+UY 6(+++% window upstream of tRNA genes. The periodicity
V40#IGPG requires both Bdp1 and Isw2. c | The mechanism of Tf1
DRYKPFQY
targeting. Tf1 integrates into promoters that are
QHKPVGITCVKQP transcribed by RNA polymerase II. Transcription factors,
such as Atf1, bind to the promoter and recruit integrase
to the insertion sites. d | The mechanism of Ty5
integration. In the absence of a stress condition, the
E6HVCTIGVKPI
+PVGITCUG targeting domain of integrase is phosphorylated (P).
#VH This directs the integration to heterochromatin by
binding Sir4 (green ovals), a structural component
2TQOQVGT )GPG of the heterochromatin. When cells are exposed to
6TCPUETKRVKQPHCEVQTDKPFKPIUKVG environmental stress the integrase is dephosphorylated
and integration occurs in gene-rich regions.

F6[VCTIGVKPI
0QUVTGUU 5VTGUU
+PVGITCUG
6CTIGVKPI
FQOCKP 2

2
5KT 5KT 5KT 5KT 5KT 5KT

*GVGTQEJTQOCVKP )GPGTKEJTGIKQP

0CVWTG4GXKGYU^)GPGVKEU
NATURE REVIEWS | GENETICS VOLUME 12 | SEPTEMBER 2011 | 619

© 2011 Macmillan Publishers Limited. All rights reserved


REVIEWS

Although the ability of Ty5 integrase to target het- Dispersed patterns of integration. In contrast to some
erochromatin suggests that the TE dictates target-site TEs that use elegant mechanisms for targeted integration
integration, there are also indications that TE–host into specific regions of the genome, other TEs appear to
interactions can alter Ty5 target-site preference. Mass lack target-site specificity. For example, L1s and the non-
spectroscopy revealed that phosphorylation of the inte- autonomous elements that they mobilize are interspersed
grase targeting domain at Ser1095 is crucial for binding throughout the genome59. Indeed, ~30% of engineered
Sir4 (REF. 35) and mutating Ser1095 redirected integra- human L1 retrotransposition events in cultured cells, and
tions to expressed regions of the genome. Although the a similar proportion of recently discovered full-length,
host-encoded kinase has not been identified, studies dimorphic human-specific L1s, are near or within the
using a phospho-specific antibody indicate that stresses, introns of genes13,50,123,124. Because protein-coding genes
such as nitrogen deprivation, can downregulate Ser1095 constitute ~40% of the human genome125,126, these find-
phosphorylation35. Thus, stress conditions may alter ings suggest a lack of robust mechanisms that are used by
the phosphorylation state of Ty5 integrase, thereby L1s or the host to prevent L1 retrotransposition into genes.
redirecting the Ty5 integration specificity. This elegant The interspersed nature of L1 and Alu sequences
example provides a plausible mechanism for how stress probably reflects the fact that the L1 endonuclease has
can alter transposon mobilization in a manner that relatively weak target-site specificity, preferentially
might provide an advantage for the host. It remains to be cleaving the sequence 5′-TTTT/A‑3′ (and variants of
determined whether retargeting Ty5 to gene-rich regions that sequence) to initiate TPRT80,121,127,128. Interestingly,
benefits Ty5 by allowing newly retrotransposed copies although ‘young’ Alu and L1 insertions have similar
to reside in permissive expression contexts, or benefits interspersed integration patterns, cytogenetic studies and
the host by generating genetic diversity, thus offering the examination of the human genome reference sequence
potential for the host to adapt to stress. have revealed that evolutionarily ‘older’ L1s and Alus have
distinct genomic distributions59,129. Older L1s preferen-
Integration into telomeres. Some TEs exclusively integrate tially reside in gene-poor, AT‑rich sequences, whereas
at or near telomeric chromosome ends. For example, the older Alus preferentially reside in gene-rich, GC‑rich
HeTA, TART and TAHRE non-LTR retrotransposons regions of the genome59.
comprise the ends of D. melanogaster chromosomes The distinct distributions of older L1s and Alus are
and probably substitute for the function of telomerase likely to result from post-integration selective processes
in maintaining chromosome end integrity 2,116,117. The that have operated on the genome for millions of years59.
SART1 and TRAS1 non-LTR retrotransposons may have However, how these skewed distributions arose remains
a similar role in Bombyx mori 118. The proteins encoded a mystery. Some researchers have suggested that Alus
by TART, TAHRE, SART1 and TRAS1 have an APE-like may possibly have an advantageous, albeit undefined,
domain118,119, and it is likely that the SART1 and TRAS1 role in gene-rich regions of the genome59. Others have
endonuclease proteins direct their integration into telom- suggested that L1 retrotransposition events into genic
eric repeats118; however, the functional role of the putative regions may exert a greater fitness cost to the host than
endonuclease domains encoded by TART and TAHRE for Alu insertions130. If so, negative selection would lead
remains unknown. to the removal of detrimental L1 alleles from the popula-
Excitingly, recent studies have revealed that certain tion. Consistent with this hypothesis, data suggest that
retrotransposons can target telomeric sequences for inte- evolutionarily recent human full-length L1 insertions
gration. For example, by an alternative endonuclease- are detrimental to the host 131,132, whereas in vitro studies
independent retrotransposition mechanism, human L1 have revealed that L1s contain cis-acting sequences that
retrotransposons containing missense mutations in the can reduce gene expression133,134. Clearly, further studies
L1 endonuclease active site can integrate at endogenous are needed to explain how the distributions of L1 and
DNA lesions and dysfunctional telomeres in Chinese Alu elements have diverged over evolutionary time.
hamster ovary cell lines that are deficient for p53 func- Despite their interspersed distribution, a small body
tion and for factors that are important in the non- of evidence suggests that there may be preferred, albeit
homologous end-joining pathway of DNA repair 120,121. rare, L1 integration sites. For example, independent
Similarly, members of the Penelope clade of retrotrans- L1‑mediated retrotransposon insertions (that is, an
posons, which encode a reverse transcriptase that lacks SVA element and an Alu element) at the same nucleotide
an obvious endonuclease domain, reside at telomeres in position in the Bruton agammaglobulinaemia tyrosine
organisms from four eukaryotic kingdoms122. The RNAs kinase (BTK) gene have resulted in two sporadic cases of
encoded by these terminal Penelope elements also con- X‑linked agammaglobulinaemia135. Similarly, independ-
tain sequences that are complementary to telomeric DNA ent L1 and Alu insertions associated with colorectal and
sequences, suggesting that base pairing between the TE desmoid tumours, respectively, have occurred at the same
RNA and telomeric ssDNA is crucial for integration. nucleotide position in the adenomatous polyposis coli
Interestingly, both of the above cases can be considered (APC) gene22,135,136. Additionally, two independent Alu
Desmoid tumours as a type of RNA-mediated DNA repair that seems curi- insertions at the same nucleotide position in the F9 gene
Soft tissue tumours that can ously similar to the mechanism used by telomerase120,122. have caused haemophilia B135,137. Thus, it would not be
arise in the abdomen as well
as in other parts of the body.
Future studies could elucidate whether host factors are surprising to find that chromatin structure and acces-
They are typically benign crucial for the localization of these retrotransposons sibility have an impact on L1‑mediated retrotransposon
and grow slowly. to DNA lesions and/or chromosomal termini. target preference138.

620 | SEPTEMBER 2011 | VOLUME 12 www.nature.com/reviews/genetics

© 2011 Macmillan Publishers Limited. All rights reserved


REVIEWS

How the host defends against transposons REFS 30,147–152); here we briefly summarize common
Although many TEs have evolved mechanisms to limit themes that have emerged from the above studies.
genome damage, TE integration still poses a potential Endo-siRNAs have the potential to inhibit TE mobil-
threat to the host. Thus, it is not surprising that host ity through the post-transcriptional disruption of trans-
organisms have evolved many diverse mechanisms poson mRNA. For example, ‘trigger’ dsRNAs can be
RNA-directed DNA to combat TE activity. However, the host must be derived from: the complementary inverted terminal
methylation able to discriminate TE sequences from host genes to repeats in DNA transposons, structured mRNA tran-
(RdDM). A pathway in which accomplish this feat. Below we discuss the mechanistic scripts or overlapping regions contained within conver-
24 nucleotide small RNAs
strategies used by the host to restrict TE mobilization. gent transcription units30,147,148,153. The resultant dsRNAs
interact with a de novo
methyltransferase to mediate can then be processed into ~21–24 nt endo-siRNAs by
the methylation and DNA methylation. Cytosine methylation (to 5‑methyl- members of the Dicer family of proteins30,154 (FIG. 3a).
transcriptional silencing of cytosine) is an important DNA modification in eukary- These endo-siRNAs are loaded onto an Argonaute protein,
homologous genomic loci otes with genomes >500 Mb, which include vertebrates, and the ‘passenger’ RNA strand (typically the sense
in plants.
flowering plants and some fungi. Most cytosine meth- strand of a TE) is degraded. The remaining complex
Small interfering RNAs ylation in plants and mammals, and almost all cytosine of an ssRNA and Argonaute is called the RNA-induced
(siRNAs). Small (~21–24 methylation in the fungus Neurospora crassa, occurs silencing complex (RISC); the RNA directs RISC to com-
nucleotide) RNAs that are within repetitive elements and is correlated with the plementary sequences in target mRNAs, leading to their
generated from dsRNA
transcriptional repression of retrotransposons in somatic post-transcriptional degradation. Importantly, the RNAi
‘triggers’ by Dicer-dependent
and Dicer-independent and germline cells139,140. machinery has the capability to inhibit any TE that gen-
mechanisms. They bind to Experiments in mammals and plants have demon- erates a dsRNA trigger that can serve as a substrate for
Argonaute proteins and guide strated that the global demethylation of genomic DNA the RNAi machinery.
the resultant complex to strongly reactivates TE transcription141–144. For exam- By a different mechanism, piRNAs can be generated
complementary mRNAs
to mediate silencing.
ple, deletion of the DNA cytosine‑5‑methyltransferase from genomic loci that encode long precursor RNAs
3‑like (Dnmt3l) gene in mice leads to the loss of de novo containing the remnants of different families of TEs151.
PIWI-interacting RNAs cytosine methylation of both LTR and non-LTR retro- In general, processing of these precursor RNAs leads to
(piRNAs). A family of small transposons, reactivation of TE expression in sper- the production of a mature ~24–35 nt piRNAs (FIG. 3b).
(~24–35 nucleotide) RNAs
matocytes and spermatogonia and meiotic catastrophe A subfamily of Argonaute proteins, known as the PIWI
that are processed from
piRNA precursor mRNAs. The in male germ cells145. Determining whether TE mobi- clade of proteins, predominantly binds to mature antisense
mature piRNAs interact with lization is directly responsible for the meiotic defects piRNAs and directs them to complementary sequences
specialized Argonaute proteins requires further study. Moreover, recent data demon- in TE mRNA. An endonuclease activity that is associated
(from the PIWI clade), to strate that the inactivation of cytosine methylation in with the PIWI protein cleaves the TE mRNA to release a
mediate RNA silencing.
Arabidopsis thaliana causes a burst of retrotransposon sense-strand piRNA, which can interact with other PIWI
Dicer and DNA TE activity, resulting in substantial increases clade proteins. Binding of this complex to the original
A family of RNase III proteins in TE copy number 144. Thus, epigenetic mechanisms act piRNA precursor RNA then re-iterates this amplification
that possess an endonuclease to control the expression, and perhaps the mobility, of cycle through a ‘ping-pong’ mechanism151,155. In addition
activity that can process
various TEs. to this type of mechanism that restricts TE mobility in the
dsRNA ‘triggers’ into small
interfering RNAs (siRNAs) or Multiple lines of evidence indicate that DNA meth- germ line, recent studies suggest that specialized piRNA
microRNAs (miRNAs). ylation inhibits TE transcription. Patterns of DNA pathways that do not operate via a ping-pong mechanism
methylation are established during gametogenesis might restrict somatic TE activity 32,155–157.
Argonaute proteins and are mediated by DNMT3A and the non-catalytic Examples of TEs that are controlled by small-RNA-
Proteins that bind to small
RNAs and are the defining
paralogue DNMT3L in mammals, but how TEs are based mechanisms include Tc1 transposons in C. elegans
component of the recognized as methylation substrates requires further and P elements in D. melanogaster 153,158. Also, 21 nt siR-
RNA-induced silencing study 146. By comparison, during plant development, NAs, which are derived from the Athila family of LTR
complex (RISC); they have an small, 24 nucleotide (nt) RNAs target paralogous DNA retrotransposons in the vegetative nucleus of pollen
ssRNA binding domain (PAZ)
sequences that share high levels of homology (such as grains in A. thaliana, are delivered to the sperm cells to
and a ribonuclease domain
(PIWI). The small RNAs guide TEs) for cytosine methylation. The mechanism of RNA- inhibit the expression of transposons that, in principle,
Argonaute proteins to target directed DNA methylation (RdDM) is not fully understood could mobilize in the germ line159.
mRNAs in order to mediate but it appears to require the canonical RNA interference Small-RNA-based mechanisms may also be crucial
post-transcriptional (RNAi) machinery (see below and FIG. 3), the DNA for silencing mammalian L1 elements. For example,
degradation and/or
translational silencing.
methyltransferase DRM2 and two plant-specific RNA an antisense promoter located within the human L1 5′
polymerases, Pol IV and Pol V146. UTR allows the production of an antisense RNA that, in
PIWI clade of proteins principle, could base pair with sense-strand L1 mRNA
A specialized class of Small RNAs inhibit TEs. Small-RNA-based mechanisms to establish a dsRNA substrate for Dicer160. Furthermore,
Argonaute proteins that
— including those involving endogenous small interfering mouse mutants lacking the murine PIWI family proteins
interact with PIWI-interacting
RNAs (piRNAs) to mediate RNAs (endo-siRNAs) and PIWI-interacting RNAs (piRNAs)) MILI or MIWI2 exhibit a loss of methylation of L1 and
transposable element — also act to defend eukaryotic cells against TEs. The intracisternal A particle (IAP) LTR retrotransposon
silencing. Members include: mechanisms by which these small RNAs are gener- DNA; this loss correlates with their transcriptional acti-
PIWI, Aubergine and Argonaute ated and how they inhibit TEs remain an active area of vation in male germ cells161. Similarly, mice lacking a
3 in D. melanogaster; MIWI1,
MIWI2 and MILI in mice, and
investigation in various model organisms. Mechanistic MILI-interacting protein, Tudor-containing protein 1,
HIWI1, HIWI2, HIWI3 and HILI details regarding these processes can be found in exhibit a similar loss of methylation of L1 DNA and a
in humans. many outstanding reviews on this topic (for example, reactivation of L1 expression162. Finally, mouse mutants

NATURE REVIEWS | GENETICS VOLUME 12 | SEPTEMBER 2011 | 621

© 2011 Macmillan Publishers Limited. All rights reserved


REVIEWS

CUK40#RCVJYC[ DRK40#RCVJYC[
40# 40#
40#RQN[OGTCUG

&0#VTCPURQUQP RK40#ENWUVGT

2TQEGUUKPI 2TQEGUUKPICPFDKPFKPIVQ2+9+ENCFGRTQVGKP

#7$
′
FU40#nVTKIIGTo
′ ′ ′ /CVWTGRK40#
&KEGTOGFKCVGFIGPGTCVKQP
QHUOCNNFQWDNGUVTCPF
UK40#U

#7$
&KEGT
+PEQTRQTCVKQPQHCUKPING ′ ′
UVTCPFUK40#
CPVKUGPUG ′ ′ 6TCPURQUQPO40#
VQVJG6'O40# KPVQ4+5%
#TIQPCWVG
%NGCXCIGCPFRTQEGUUKPI
′ ′ 4+5%

#7$ #)1

$KPFKPIQH6'O40# ′ ′ #ORNKȮECVKQPE[ENG ′ ′

′ ′ 0GYUGEQPFCT[RK40# 5GPUGUVTCPF
VTCPURQUQPRK40#
#)1

′ ′
%NGCXCIGQHO40# ′ ′ RK40#RTGEWTUQTO40#
O40#FGITCFCVKQP
D[EGNNWNCTGP\[OGU
%NGCXCIG

Figure 3 | The degradation of transposon mRNA by RNAi. a | The small interfering RNA (siRNA) pathway.
0CVWTG4GXKGYU^)GPGVKEU
dsRNA ‘triggers’ (represented by the hairpin), which are derived from the inverted terminal repeats of a DNA
transposon in the case illustrated, are processed and then cleaved into 21–24 nucleotide (nt) siRNAs by the Dicer
family of proteins (light green shape). A single-strand siRNA (short red line), complementary to the transposon mRNA,
is selectively incorporated into the Argonaute (dark green shape)-containing RNA-induced silencing complex (RISC).
The siRNA directs RISC to complementary sequences in the transposon mRNA (long red line), leading to its
post-transcriptional degradation. The figure is drawn based on concepts presented in REFS 147,148,152. b | The
PIWI-interacting RNA (piRNA) pathway. A primary piRNA transcript (wavy, blue line) generated from a piRNA cluster
(blue rectangles) that contains sequences derived from transposable elements (TEs; dark blue rectangle) is processed
into mature 24–35 nt piRNAs (small blue line). Binding of the mature piRNA by the PIWI or Aubergine (AUB) proteins
allows it to be directed to complementary sequences in TE mRNA (red line). Endonucleolytic cleavage of the mRNA
(represented by the scissors), 10 nt from the 5′ end of the small RNA, and 3′ cleavage/processing liberates a secondary
sense-strand transposon piRNA (small red line), which associates with the Argonaute 3 (AGO3) protein. The binding of
this complex to complementary sequences in the original precursor piRNA, followed by endonucleolytic cleavage,
regenerates an antisense piRNA that can be directed to TE mRNA. This iterative cycle (known as a ‘ping-pong’ cycle)
can lead to the destruction of transposon mRNA in the germ line. The piRNA model was redrawn based on concepts
and models presented in REFS 30,151,157. The example illustrated is for Drosophila melanogaster; however, a similar
pathway probably operates in mammalian cells (see the main text for details).

piRNA cluster lacking the non-canonical Maelstrom protein, a compo- above examples provide compelling evidence that small-
A genomic DNA locus that nent of the nuage complex (a germline-specific perinu- RNA-based pathways probably control the expression
encodes PIWI-interacting RNA clear structure) that may be important for small RNA of certain TEs in the mammalian germ line.
(piRNA) precursor RNAs. Many biogenesis, exhibit derepression of L1 transcription, Finally, it is noteworthy that other antisense-RNA-
piRNA clusters contain sense
and antisense sequences that
an increase in the levels of L1 ribonucleoprotein par- based mechanisms may be involved in TE silencing. For
are derived from mobile ticle intermediates in spermatids and a chromosomal example, antisense transcripts from S. cerevisiae Ty1
genetic elements. synapsis defect during male meiosis163. Together, the elements reduce Ty1 integrase and reverse transcriptase

622 | SEPTEMBER 2011 | VOLUME 12 www.nature.com/reviews/genetics

© 2011 Macmillan Publishers Limited. All rights reserved


REVIEWS

recent work on the reverse transcriptase encoded by the


6TCPURQUKVKQPKPRTGEWTUQTU R2 non-LTR retrotransposon of B. mori demonstrated
QHICOGVGUQTRGTJCRU that this enzyme is more processive than the reverse
ICOGVGUVJGOUGNXGU
)GTONKPGGXGPVU transcriptases encoded by retroviruses168. Alternatively,
L1 5′ truncation might result if host factors cause the dis-
sociation of the L1 reverse transcriptase from the nascent
cDNA and/or degrade the L1 mRNA during integration.
6TCPURQUKVKQPKP
GCTN[FGXGNQROGPV
In these scenarios, to generate a full-length insertion the
L1 reverse transcriptase would need to complete inte-
gration before the TPRT intermediate is recognized as
5QOCVKEGXGPVU DNA damage by the host 50,169. Indeed, proteins that are
involved in the non-homologous end-joining pathway of
DNA repair seem to restrict the retrotransposition
of a zebrafish LINE‑2 element in DT40 chicken cells170,
6TCPURQUKVKQPKP
NCVGTFGXGNQROGPV whereas members of DNA excision repair pathway
(such as the ERCC1–XPF complex) might restrict L1
6WOQWTU 0GWTQPCNRTGEWTUQTEGNNU retrotransposition in cultured human cells171.
Finally, in addition to recognizing the L1 integration
Figure 4 | Timing of transposition. Germline transposable element (TE) integration
0CVWTG4GXKGYU^)GPGVKEU intermediate as a form of DNA damage, recent data sug-
events can result from TE mobility in cells that give rise to gametes or from TE mobility
post-fertilization during early development. Embryonic TE mobility in cells that do not
gest that retrotransposition indicator cassettes, delivered
contribute to the germ line or mobility at later developmental stages can, in principle, by engineered L1s in human embryonic carcinoma cell
lead to somatic TE integration events. The overlapping brackets signify that some TE lines, can be epigenetically silenced during or imme-
insertions in early development can contribute to the germ line, whereas others may diately after their integration into genomic DNA172.
not. Endogenous long interspersed element 1 (L1) retrotransposition events can occur Because L1 is an ancient ‘stowaway’ in mammalian
in certain tumours, and experiments using engineered human L1s suggest that L1 genomes, it is likely that the host has evolved multiple
retrotransposition may also occur during mammalian neurogenesis. Examples of the mechanisms to combat L1 mobility at discrete steps in
developmental timing of TE integration events are described in the main text. the retrotransposition pathway and that some of these
mechanisms operate in a context-dependent manner.
Continued studies should reveal new and more diverse
protein levels by a post-translational mechanism; this host mechanisms to restrict TE mobility.
leads to the inhibition of Ty1 mobility and thus controls
Ty1 copy number 164. Because S. cerevisiae lacks RNAi Developmental triggers of transposition
machinery, these results suggest that genomes have Despite mechanisms to combat TE mobility, TEs con-
evolved other RNA-dependent strategies to 'tame' TEs. tinue to thrive in many host genomes. Thus, TEs must
have evolved ways to either overwhelm or counteract
Aicardi–Goutieres
Cytosine deaminases and DNA repair factors restrict these host defences. TEs must mobilize in germ cells or
syndrome
A rare, autosomal recessive TEs. Proteins that are involved in nucleic-acid metabo- during early development to ensure their survival (FIG. 4).
genetic disorder that leads to lism and/or DNA repair can also restrict TE mobility. However, some TEs can mobilize in somatic cells, provid-
brain dysfunction as well as For example, members of the apolipoprotein B mRNA- ing a potential mechanism to generate intra-individual
other symptoms. The early editing enzyme 3 (APOBEC3) family of cytidine deami- genetic variation.
onset form of the disease can
be caused by mutations in the
nases can restrict the retrotransposition of various retro-
TREX1 gene and is usually fatal. viruses and LTR and non-LTR retrotransposons33. For Transposition in the germ line or during early devel-
retroviruses and LTR retrotransposons, APOBEC3 pro- opment. D. melanogaster P elements provide one of the
Hybrid dysgenesis teins generally deaminate cytidines during the first strand best-studied cases of cell-type-specific transposition173.
In Drosophila melanogaster,
cDNA synthesis, which leads to either cDNA degradation P element transposition occurs when females lacking P
a sterility-inducing syndrome
that is induced by the or the integration of a mutated provirus. The mechanisms elements mate with males carrying P elements; P ele-
mobilization of P elements by which particular APOBEC3 proteins restrict non-LTR ment mobilization can cause hybrid dysgenesis in the
in crosses between females retrotransposons still require elucidation. Similarly, over- offspring. In the reciprocal cross, eggs from females
lacking P elements and expression of the 3′-repair exonuclease 1 (Trex1) gene, containing P elements produce a repressor protein and
males carrying P elements.
mutations in which cause Aicardi–Goutieres syndrome, piRNAs that inhibit P elements transposition57,158. The
Virus-like particle can inhibit L1 and IAP retrotransposition in cultured repressor is an alternatively spliced, truncated form of
(VLP). A cytoplasmic particle cell assays165,166 but the mechanism of TREX1‑mediated the transposase. Importantly, the repressor not only
that comprises long terminal TE repression requires elucidation. controls which crosses produce germline integration
repeat (LTR) retrotransposon
Other mechanisms are also likely to restrict the but also inhibits transposition in the soma.
mRNA, the LTR retrotranspo-
son-encoded proteins and mobility of non-LTR retrotransposons. For example, The D. melanogaster Gypsy element is another exam-
host factors that are required the overwhelming majority of L1 elements in mamma- ple of a TE that exhibits tissue-specific control174,175.
for reverse transcription lian genomes are 5′ truncated and are thus essentially Gypsy transcription is induced in somatic follicle cells
of LTR retrotransposon mRNA. ‘dead on arrival’ because they cannot synthesize proteins that surround the oocyte. The TE mRNA assembles
LTR retrotransposon mRNA is
reverse transcribed into a
that are crucial for retrotransposition167. It has been pro- into virus-like particles that are thought to traffic to the
double-stranded cDNA posed that 5′ truncation may be due to the low proces- oocyte to carry out transposition. It remains unclear
within VLPs. sivity of the L1‑encoded reverse transcriptase. However, whether the transfer of Gypsy virus-like particles to the

NATURE REVIEWS | GENETICS VOLUME 12 | SEPTEMBER 2011 | 623

© 2011 Macmillan Publishers Limited. All rights reserved


REVIEWS

oocyte occurs via an enveloped particle (similarly to suggesting a role for the insertion in cancer develop-
retroviruses) or by a form of endocytosis. However, the ment 22. Together with the transgenic L1 experiments
Flamenco locus encodes piRNAs that silence Gypsy ele- (discussed above), these findings establish that somatic
ments in follicle cells, thereby preventing the spread of TE mobility can lead to phenotypic changes in the host.
these TEs to the surrounding germ cells155. Intriguingly, several lines of evidence suggest that
Relatively little is known about the developmental somatic L1 retrotransposition may also occur in the
timing of L1 retrotransposition in mammals. The sheer mammalian nervous system (reviewed in REF. 5). First,
numbers of L1 and Alu retrotransposons that populate an engineered human L1 can retrotranspose in neuro-
mammalian genomes provide immediate evidence that genic zones of the brain in transgenic mice24 when its
they mobilize in the germ line. Various studies, using expression is driven by a promoter contained within
endogenous and engineered L1s, provide strong experi- its native 5′ UTR184. Second, engineered human L1s can
mental evidence to support this assertion (also reviewed retrotranspose in cultured rat neuronal progenitor cells
in REF. 5). For example, full-length mouse L1 RNA and (NPCs), in human embryonic stem cell-derived NPCs
the mouse L1 ORF1‑encoded protein are co-expressed and at low levels in human fetal-derived NPCs23,24. Third,
in leptotene and zygotene spermatocytes during meiotic sensitive multiplex quantitative PCR experiments have
prophase176. In addition, the mouse ORF1‑encoded pro- suggested a modest increase in L1 copy number in
tein is expressed in the cytoplasm during specific stages post-mortem brain tissue when compared to heart and
of development in oocytes177. Similarly, human oocytes liver tissue that was derived from the same individual23.
express L1 RNA and support the retrotransposition of Finally, the retrotransposition of an engineered human
an engineered human L1 element 178. Finally, transgenic L1 is elevated in a mouse model of Rett syndrome (a
mouse experiments demonstrated that an engineered neurodevelopmental disorder), and induced pluripotent
human L1 retrotransposon, which was driven from the stem cells derived from Rett syndrome patients exhibit
promoter of the mouse RNA polymerase II large subunit an increase in L1 DNA copy number when compared
gene, could retrotranspose in male germ cells179. to normal controls, suggesting a potential increase in
Unexpectedly, a growing body of experimental evi- endogenous L1 retrotransposition185.
dence suggests that L1 retrotransposition might occur The above studies strongly suggest that certain neu-
frequently during early development (FIG. 4) (also ronal cells may be permissive for L1 retrotransposition.
reviewed in REF. 5). For example, human embryonic stem However, additional research is needed to truly under-
cells can express L1 RNA and ORF1‑encoded protein, stand the effect of L1 retrotransposition in the brain. For
and can accommodate the retrotransposition of engi- example, recent advances in DNA sequencing technol-
neered L1s, albeit at lower levels than in other types of ogy should provide a means to directly test whether the
transformed human cells19,180. In addition, studies of a L1 DNA copy number changes that were detected in
male patient with X‑linked choroideraemia revealed that quantitative PCR experiments represent actual de novo
his mother had mosaicism for the mutagenic L1 inser- endogenous retrotransposition events or instead result
tion in both germline and somatic tissues20. Thus, the from other forms of genomic instability that have been
initial retrotransposition event must have occurred dur- reported in neurons186,187. Similarly, it remains unclear
ing early embryogenesis in the mother. Finally, recent whether endogenous L1 retrotransposition events rep-
transgenic experiments conducted in rats and mice led resent a type of ‘genomic noise’ or whether they have any
to the conclusion that most L1 retrotransposition occurs functional impact on neuronal development. Finally, it
during early embryogenesis and that most of the resultant remains a mystery why neuronal cells may accommo-
events are not heritable21. Intriguingly, these data suggest date L1 retrotransposition at apparently higher levels
that L1 ribonucleoprotein particles can be deposited into than other somatic cells. Nonetheless, these studies have
zygotes by either the sperm or the egg to undergo retro- unveiled a new area of investigation that is likely to be
transposition during early development, thereby provid- the subject of future work.
ing a possible mechanism to generate somatic mosaicism
and intra-individual genetic variation (see below). Deregulated L1 retrotransposition in cancer cells. A
growing body of evidence suggests that L1 retrotrans-
Somatic transposition. Classical experiments in maize position may become deregulated in certain cancers. For
revealed that DNA TE activity in somatic tissues could example, early studies revealed that hypomethylation of
lead to variegated corn colour phenotypes1,181. Since the L1 promoter is correlated with increased L1 expres-
then, somatic TE events have been reported in other sion and/or the production of the L1 ORF1‑encoded
organisms. For example, it is well established that Tc1 protein in certain tumours188–190. Moreover, engineered
transposition in the Bergerac strain of C. elegans pref- human L1s readily retrotranspose in a variety of trans-
erentially occurs in somatic cells182. Similarly, a recent formed human and mouse cell lines but generally
study has revealed that somatic transposition of the show lower levels of retrotransposition activity in nor-
Hatvine1‑rrm DNA TE — into the promoter region of mal human cells such as fibroblasts (for example, see
the VvTFL1A gene of the grapevine cultivar Carnigan REFS 11,191,192). Consistent with this, recent findings
— affects the grapevine branching pattern and the size using second-generation DNA sequencing revealed
X-linked choroideraemia
of fruit clusters183. Also, a mutagenic L1 insertion was a total of nine de novo L1 retrotransposition events in
A recessive degenerative identified in the APC gene in tumour tissue, but not in 6 out of 20 examined non-small-cell lung tumours12.
retinal disease. the surrounding tissue, of a patient with colon cancer, Intriguingly, the tumours that contained the new L1

624 | SEPTEMBER 2011 | VOLUME 12 www.nature.com/reviews/genetics

© 2011 Macmillan Publishers Limited. All rights reserved


REVIEWS

insertions also exhibited a specific genome-wide hypo- and how the host responds to this ever-present threat,
methylation signature, which is consistent with the we might unveil the dynamic forces that shape our
notion that altering the epigenome can create a permis- genomes. Indeed, we are now able to critically evaluate
sive environment for L1 expression and/or retrotrans- the McClintock doctrine, and future experiments could
position and perhaps for the retrotransposition of other provide valuable insight into whether the increases in
classes of non-LTR retrotransposons. Clearly, further TE transcription that are caused by environmental stress
innovations in DNA sequencing of heterogeneous cell lead to higher levels of TE integration and whether these
populations will be crucial to reveal patterns of TE activ- insertions have an impact on host phenotypes and/or
ity in diverse tumours. Then the challenge will be to survival.
determine whether all these TE insertions are ‘passenger’ It remains a curiosity why sequences without any
mutations that are a consequence of the altered cellular apparent purpose continue to thrive in genomes. What is
milieu of cancer cells or whether some act as ‘drivers’ to clear is that an understanding of TE biology is necessary
promote tumorigenesis. to understand genome biology. It is intriguing to specu-
late that some phenotypic differences among organisms
Closing remarks or between individuals are due to the effects of TEs.
It is undeniable that TEs have played an important part These speculations require rigorous experimental tests.
in structuring genomes and generating genetic diversity. However, the coming years should be an exciting time
By understanding how, when and where TEs integrate, for TE biology.

1. McClintock, B. The origin and behavior of mutable loci 21. Kano, H. et al. L1 retrotransposition occurs mainly in 37. Hirochika, H. Activation of tobacco retrotransposons
in maize. Proc. Natl Acad. Sci. USA 36, 344–355 embryogenesis and creates somatic mosaicism. during tissue culture. EMBO J. 12, 2521–2528 (1993).
(1950). Genes Dev. 23, 1303–1312 (2009). 38. Courtial, B. et al. Tnt1 transposition events are
2. Levis, R. W., Ganesan, R., Houtchens, K., Tolar, L. A. & References 19–21 provide evidence that L1s can induced by in vitro transformation of Arabidopsis
Sheen, F. M. Transposons in place of telomeric repeats retrotranspose during early embryonic development. thaliana, and transposed copies integrate into genes.
at a Drosophila telomere. Cell 75, 1083–1093 (1993). 22. Miki, Y. et al. Disruption of the APC gene by a Mol. Genet. Genomics 265, 32–42 (2001).
3. Agrawal, A., Eastman, Q. M. & Schatz, D. G. retrotransposal insertion of L1 sequence in a colon 39. McClintock, B. The significance of responses of the
Transposition mediated by RAG1 and RAG2 and its cancer. Cancer Res. 52, 643–645 (1992). genome to challenge. Science 226, 792–801 (1984).
implications for the evolution of the immune system. 23. Coufal, N. G. et al. L1 retrotransposition in human 40. Craig, N. L., Craigie, R., Gellert, M. & Lambowitz, A. M.
Nature 394, 744–751 (1998). neural progenitor cells. Nature 460, 1127–1131 (eds) Mobile DNA II (ASM Press, Washington DC, 2002).
4. Feschotte, C. Transposable elements and the evolution (2009). 41. Kleckner, N. Regulation of transposition in bacteria.
of regulatory networks. Nature Rev. Genet. 9, 24. Muotri, A. R. et al. Somatic mosaicism in neuronal Annu. Rev. Cell Biol. 6, 297–327 (1990).
397–405 (2008). precursor cells mediated by L1 retrotransposition. 42. Garfinkel, D. J., Boeke, J. D. & Fink, G. R. Ty element
5. Beck, C. R., Garcia-Perez, J. L., Badge, R. M. & Nature 435, 903–910 (2005). transposition: reverse transcriptase and virus-like
Moran, J. V. LINE‑1 elements in structural variation 25. Sandmeyer, S. Integration by design. Proc. Natl Acad. particles. Cell 42, 507–517 (1985).
and disease. Annu. Rev. Genomics Hum. Genet. Sci. USA 100, 5586–5588 (2003). 43. Boeke, J. D., Garfinkel, D. J., Styles, C. A. & Fink, G. R.
18 Jul 2011 (doi:10.1146/annurev-genom‑ 26. Leem, Y. E. et al. Retrotransposon Tf1 is targeted to Ty elements transpose through an RNA intermediate.
082509‑141802). pol II promoters by transcription activators. Mol. Cell Cell 40, 491–500 (1985).
6. Orgel, L. E., Crick, F. H. & Sapienza, C. Selfish DNA. 30, 98–107 (2008). 44. Eichinger, D. J. & Boeke, J. D. The DNA intermediate
Nature 288, 645–646 (1980). This paper demonstrates a mechanism by which in yeast Ty1 element transposition copurifies with
7. Doolittle, W. F. & Sapienza, C. Selfish genes, the the S. pombe Tf1 retrotransposon can target RNA virus-like particles: cell-free Ty1 transposition. Cell 54,
phenotype paradigm and genome evolution. Nature polymerase II-transcribed genes. 955–966 (1988).
284, 601–603 (1980). 27. Devine, S. E. & Boeke, J. D. Integration of the yeast 45. Luan, D. D., Korman, M. H., Jakubczak, J. L. &
8. Bestor, T. H. Sex brings transposons and genomes into retrotransposon Ty1 is targeted to regions upstream Eickbush, T. H. Reverse transcription of R2Bm RNA is
conflict. Genetica 107, 289–295 (1999). of genes transcribed by RNA polymerase III. primed by a nick at the chromosomal target site: a
9. Hickey, D. A. Selfish DNA: a sexually-transmitted Genes Dev. 10, 620–633 (1996). mechanism for non-LTR retrotransposition. Cell 72,
nuclear parasite. Genetics 101, 519–531 (1982). 28. Eickbush, T. H. in Mobile DNA II (eds Craig, N. L., 595–605 (1993).
10. Goodier, J. L. & Kazazian, H. H. Jr. Retrotransposons Craigie, R., Gellert, M. & Lambowitz, A. M.) 813–835 46. Kajikawa, M. & Okada, N. LINEs mobilize SINEs in the
revisited: the restraint and rehabilitation of parasites. (ASM Press, Washington DC, 2002). eel through a shared 3′ sequence. Cell 111, 433–444
Cell 135, 23–35 (2008). 29. Winckler, T., Dingermann, T. & Glockner, G. (2002).
11. Moran, J. V. et al. High frequency retrotransposition Dictyostelium mobile elements: strategies to amplify 47. Dewannieux, M., Esnault, C. & Heidmann, T.
in cultured mammalian cells. Cell 87, 917–927 in a compact genome. Cell. Mol. Life Sci. 59, LINE-mediated retrotransposition of marked Alu
(1996). 2097–2111 (2002). sequences. Nature Genet. 35, 41–48 (2003).
12. Iskow, R. C. et al. Natural mutagenesis of human 30. Malone, C. D. & Hannon, G. J. Small RNAs as 48. Hancks, D. C., Goodier, J. L., Mandal, P. K.,
genomes by endogenous retrotransposons. Cell 141, guardians of the genome. Cell 136, 656–668 (2009). Cheung, L. E. & Kazazian, H. H. Jr. Retrotransposition
1253–1261 (2010). 31. Bestor, T. H. & Bourc’his, D. Transposon silencing and of marked SVA elements by human L1s in cultured
13. Beck, C. R. et al. LINE‑1 retrotransposition activity in imprint establishment in mammalian germ cells. cells. Hum. Mol. Genet. 2 Jun 2011 (doi:10.1093/
human genomes. Cell 141, 1159–1170 (2010). Cold Spring Harb. Symp. Quant. Biol. 69, 381–387 hmg/ddr245).
14. Huang, C. R. et al. Mobile interspersed repeats are (2004). 49. Garcia-Perez, J. L., Doucet, A. J., Bucheton, A.,
major structural variants in the human genome. 32. Golden, D. E., Gerbasi, V. R. & Sontheimer, E. J. Moran, J. V. & Gilbert, N. Distinct mechanisms for
Cell 141, 1171–1182 (2010). An inside job for siRNAs. Mol. Cell 31, 309–312 trans-mediated mobilization of cellular RNAs by
15. Witherspoon, D. J. et al. Mobile element scanning (2008). the LINE‑1 reverse transcriptase. Genome Res. 17,
(ME-Scan) by targeted high-throughput sequencing. 33. Chiu, Y. L. & Greene, W. C. The APOBEC3 cytidine 602–611 (2007).
BMC Genomics 11, 410 (2010). deaminases: an innate defensive network opposing 50. Gilbert, N., Lutz, S., Morrish, T. A. & Moran, J. V.
16. Hormozdiari, F. et al. Alu repeat discovery and exogenous retroviruses and endogenous retroelements. Multiple fates of L1 retrotransposition intermediates
characterization within human genomes. Genome Res. Annu. Rev. Immunol. 26, 317–353 (2008). in cultured human cells. Mol. Cell. Biol. 25,
21, 840–849 (2011). 34. Sehgal, A., Lee, C. Y. & Espenshade, P. J. SREBP controls 7780–7795 (2005).
17. Ewing, A. D. & Kazazian, H. H. Jr. High-throughput oxygen-dependent mobilization of retrotransposons in 51. Buzdin, A. et al. A new family of chimeric retrotranscripts
sequencing reveals extensive variation in human- fission yeast. PLoS Genet. 3, e131 (2007). formed by a full copy of U6 small nuclear RNA fused to
specific L1 content in individual human genomes. 35. Dai, J., Xie, W., Brady, T. L., Gao, J. & Voytas, D. F. the 3′ terminus of L1. Genomics 80, 402–406 (2002).
Genome Res. 20, 1262–1270 (2010). Phosphorylation regulates integration of the yeast Ty5 52. Weber, M. J. Mammalian small nucleolar RNAs are
18. Ewing, A. D. & Kazazian, H. H. Jr. Whole-genome retrotransposon into heterochromatin. Mol. Cell 27, mobile genetic elements. PLoS Genet. 2, e205 (2006).
resequencing allows detection of many rare LINE‑1 289–299 (2007). 53. Wei, W. et al. Human L1 retrotransposition: cis
insertion alleles in humans. Genome Res. 21, This paper demonstrates that TEs can respond to preference versus trans complementation. Mol. Cell.
985–990 (2011). stress by changing their target sites. When cells Biol. 21, 1429–1439 (2001).
19. Garcia-Perez, J. L. et al. LINE‑1 retrotransposition in lack access to nitrogen, Ty5 no longer integrates 54. Esnault, C., Maestre, J. & Heidmann, T. Human LINE
human embryonic stem cells. Hum. Mol. Genet. 16, into heterochromatin but instead targets coding retrotransposons generate processed pseudogenes.
1569–1577 (2007). sequences in the genome. Nature Genet. 24, 363–367 (2000).
20. van den Hurk, J. A. et al. L1 retrotransposition can 36. Grandbastien, M. A. et al. Stress activation and genomic 55. Reznikoff, W. S. Tn5 transposition: a molecular tool
occur early in human embryonic development. impact of Tnt1 retrotransposons in Solanaceae. for studying protein structure-function. Biochem. Soc.
Hum. Mol. Genet. 16, 1587–1592 (2007). Cytogenet. Genome Res. 110, 229–241 (2005). Trans. 34, 320–323 (2006).

NATURE REVIEWS | GENETICS VOLUME 12 | SEPTEMBER 2011 | 625

© 2011 Macmillan Publishers Limited. All rights reserved


REVIEWS

56. Peters, J. E. & Craig, N. L. Tn7: smarter than we 82. Yang, J., Malik, H. S. & Eickbush, T. H. Identification of 102. Mitchell, R. S. et al. Retroviral DNA integration:
thought. Nature Rev. Mol. Cell Biol. 2, 806–814 the endonuclease domain encoded by R2 and other ASLV, HIV, and MLV show distinct target site
(2001). site-specific, non-long terminal repeat preferences. PLoS Biol. 2, e234 (2004).
57. Rio, D. C. in Mobile DNA II (eds Craig, N. L., Craigie, R., retrotransposable elements. Proc. Natl Acad. Sci. USA 103. Ciuffi, A. Mechanisms governing lentivirus integration
Gellert, M. & Lambowitz, A. M.) 484–518 (ASM Press, 96, 7847–7852 (1999). site selection. Curr. Gene Ther. 8, 419–429 (2008).
Washington DC, 2002). 83. Bushman, F. D. Targeting survival: integration site 104. Ciuffi, A. & Bushman, F. D. Retroviral DNA integration:
58. Plasterk, R. H. The Tc1/mariner transposon family. selection by retroviruses and LTR-retrotransposons. HIV and the role of LEDGF/p75. Trends Genet. 22,
Curr. Top. Microbiol. Immunol. 204, 125–143 (1996). Cell 115, 135–138 (2003). 388–395 (2006).
59. Lander, E. S. et al. Initial sequencing and analysis of 84. Lesage, P. & Todeschini, A. L. Happy together: 105. Engelman, A. & Cherepanov, P. The lentiviral integrase
the human genome. Nature 409, 860–921 (2001). the life and times of Ty retrotransposons and their binding protein LEDGF/p75 and HIV‑1 replication.
60. Ray, D. A., Pagan, H. J., Thompson, M. L. & hosts. Cytogenet. Genome Res. 110, 70–90 (2005). PLoS Pathog. 4, e1000046 (2008).
Stevens, R. D. Bats with hATs: evidence for recent DNA 85. Kirchner, J., Connolly, C. M. & Sandmeyer, S. B. 106. Poeschla, E. M. Integrase, LEDGF/p75 and HIV
transposon activity in genus Myotis. Mol. Biol. Evol. Requirement of RNA polymerase III transcription replication. Cell. Mol. Life Sci. 65, 1403–1424 (2008).
24, 632–639 (2007). factors for in vitro position-specific integration of a 107. Malik, H. S. & Eickbush, T. H. Modular evolution of the
61. Pritham, E. J. & Feschotte, C. Massive amplification of retroviruslike element. Science 267, 1488–1491 integrase domain in the Ty3/Gypsy class of LTR
rolling-circle transposons in the lineage of the bat (1995). retrotransposons. J. Virol. 73, 5186–5190 (1999).
Myotis lucifugus. Proc. Natl Acad. Sci. USA 104, 86. Chalker, D. L. & Sandmeyer, S. B. Ty3 integrates 108. Gao, X., Hou, Y., Ebina, H., Levin, H. L. & Voytas, D. F.
1895–1900 (2007). within the region of RNA polymerase III Chromodomains direct integration of retrotransposons
62. Ray, D. A. et al. Multiple waves of recent DNA transcription initiation. Genes Dev. 6, 117–128 to heterochromatin. Genome Res. 18, 359–369
transposon activity in the bat, Myotis lucifugus. (1992). (2008).
Genome Res. 18, 717–728 (2008). 87. Yieh, L., Hatzis, H., Kassavetis, G. & Sandmeyer, S. B. 109. Zou, S. & Voytas, D. F. Silent chromatin determines
References 60–62 reveal that certain DNA Mutational analysis of the transcription factor IIIB-DNA target preference of the Saccharomyces
transposons are active in the Myotis genus of bats. target of Ty3 retroelement integration. J. Biol. Chem. retrotransposon Ty5. Proc. Natl Acad. Sci. USA 94,
63. Bowen, N. J. & McDonald, J. F. Drosophila 277, 25920–25928 (2002). 7412–7416 (1997).
euchromatic LTR retrotransposons are much younger 88. Yieh, L., Kassavetis, G., Geiduschek, E. P. & 110. Zou, S., Wright, D. A. & Voytas, D. F.
than the host species in which they reside. Genome Sandmeyer, S. B. The Brf and TATA-binding protein The Saccharomyces Ty5 retrotransposon family is
Res. 11, 1527–1540 (2001). subunits of the RNA polymerase III transcription associated with origins of DNA replication at the
64. Schnable, P. S. et al. The B73 maize genome: factor IIIB mediate position-specific integration of telomeres and the silent mating locus HMR. Proc. Natl
complexity, diversity, and dynamics. Science 326, the gypsy-like element, Ty3. J. Biol. Chem. 275, Acad. Sci. USA 92, 920–924 (1995).
1112–1115 (2009). 29800–29807 (2000). 111. Zou, S., Ke, N., Kim, J. M. & Voytas, D. F.
65. SanMiguel, P. et al. Nested retrotransposons in the 89. Bachman, N., Eby, Y. & Boeke, J. D. Local definition of The Saccharomyces retrotransposon Ty5 integrates
intergenic regions of the maize genome. Science 274, Ty1 target preference by long terminal repeats and preferentially into regions of silent chromatin at the
765–768 (1996). clustered tRNA genes. Genome Res. 14, 1232–1247 telomeres and mating loci. Genes Dev. 10, 634–645
66. Maksakova, I. A. et al. Retroviral elements and their (2004). (1996).
hosts: insertional mutagenesis in the mouse germ line. 90. Bachman, N., Gelbart, M. E., Tsukiyama, T. & 112. Gai, X. & Voytas, D. F. A single amino acid change in
PLoS Genet. 2, e2 (2006). Boeke, J. D. TFIIIB subunit Bdp1p is required for the yeast retrotransposon Ty5 abolishes targeting to
67. Moyes, D., Griffiths, D. J. & Venables, P. J. Insertional periodic integration of the Ty1 retrotransposon and silent chromatin. Mol. Cell 1, 1051–1055 (1998).
polymorphisms: a new lease of life for endogenous targeting of Isw2p to S. cerevisiae tDNAs. Genes Dev. 113. Xie, W. et al. Targeting of the yeast Ty5
retroviruses in human disease. Trends Genet. 23, 19, 955–964 (2005). retrotransposon to silent chromatin is mediated by
326–333 (2007). 91. Bolton, E. C. & Boeke, J. D. Transcriptional interactions between integrase and Sir4p. Mol. Cell.
68. Badge, R. M., Alisch, R. S. & Moran, J. V. ATLAS: a interactions between yeast tRNA genes, flanking Biol. 21, 6606–6614 (2001).
system to selectively identify human-specific L1 genes and Ty elements: a genomic point of view. 114. Zhu, Y., Dai, J., Fuerst, P. G. & Voytas, D. F.
insertions. Am. J. Hum. Genet. 72, 823–838 (2003). Genome Res. 13, 254–263 (2003). Controlling integration specificity of a yeast
69. Sheen, F. M. et al. Reading between the LINEs: 92. Kinsey, P. T. & Sandmeyer, S. B. Adjacent pol II and pol retrotransposon. Proc. Natl Acad. Sci. USA 100,
human genomic variation induced by LINE‑1 III promoters: transcription of the yeast 5891–5895 (2003).
retrotransposition. Genome Res. 10, 1496–1508 retrotransposon Ty3 and a target tRNA gene. Nucleic 115. Brady, T. L., Fuerst, P. G., Dick, R. A., Schmidt, C. &
(2000). Acids Res. 19, 1317–1324 (1991). Voytas, D. F. Retrotransposon target site selection by
70. Kidd, J. M. et al. Mapping and sequencing of 93. Hofmann, J. et al. Transfer RNA genes from imitation of a cellular protein. Mol. Cell. Biol. 28,
structural variation from eight human genomes. Dictyostelium discoideum are frequently associated 1230–1239 (2008).
Nature 453, 56–64 (2008). with repetitive elements and contain consensus boxes 116. George, J. A., Traverse, K. L., DeBaryshe, P. G.,
71. Kidd, J. M. et al. A human genome structural variation in their 5′ and 3′-flanking regions. J. Mol. Biol. 222, Kelley, K. J. & Pardue, M. L. Evolution of diverse
sequencing resource reveals insights into mutational 537–552 (1991). mechanisms for protecting chromosome ends by
mechanisms. Cell 143, 837–847 (2010). 94. Chung, T., Siol, O., Dingermann, T. & Winckler, T. Drosophila TART telomere retrotransposons.
72. Korbel, J. O. et al. Paired-end mapping reveals Protein interactions involved in tRNA gene-specific Proc. Natl Acad. Sci. USA 107, 21052–21057 (2010).
extensive structural variation in the human genome. integration of Dictyostelium discoideum non-long 117. Biessmann, H. et al. HeT‑A, a transposable element
Science 318, 420–426 (2007). terminal repeat retrotransposon TRE5‑A. Mol. Cell. specifically involved in “healing” broken chromosome
73. Durbin, R. M. et al. A map of human genome variation Biol. 27, 8492–8501 (2007). ends in Drosophila melanogaster. Mol. Cell. Biol. 12,
from population-scale sequencing. Nature 467, 95. Behrens, R., Hayles, J. & Nurse, P. Fission yeast 3910–3918 (1992).
1061–1073 (2010). retrotransposon Tf1 integration is targeted to 5′ ends 118. Fujiwara, H., Osanai, M., Matsumoto, T. & Kojima, K. K.
74. Mills, R. E. et al. Mapping copy number variation by of open reading frames. Nucleic Acids Res. 28, Telomere-specific non-LTR retrotransposons and
population-scale genome sequencing. Nature 470, 4709–4716 (2000). telomere maintenance in the silkworm, Bombyx mori.
59–65 (2011). 96. Guo, Y. & Levin, H. L. High-throughput sequencing Chromosome Res. 13, 455–467 (2005).
References 12–18 and 70–74 used of retrotransposon integration provides a 119. Gladyshev, E. A. & Arkhipova, I. R. A subtelomeric
second-generation DNA sequencing and/or modern saturated profile of target activity in non-LTR retrotransposon Hebe in the bdelloid rotifer
genomic approaches to demonstrate that TEs Schizosaccharomyces pombe. Genome Res. 20, Adineta vaga is subject to inactivation by deletions
continue to have an ongoing impact on human 239–248 (2010). but not 5′ truncations. Mob. DNA 1, 12 (2010).
genome structural variation. This study describes the sequencing of a saturated 120. Morrish, T. A. et al. Endonuclease-independent LINE‑1
75. Kuduvalli, P. N., Rao, J. E. & Craig, N. L. Target DNA set of insertion sites that are actively targeted retrotransposition at mammalian telomeres. Nature
structure plays a critical role in Tn7 transposition. by a TE. 446, 208–212 (2007).
EMBO J. 20, 924–932 (2001). 97. Singleton, T. L. & Levin, H. L. A long terminal repeat 121. Morrish, T. A. et al. DNA repair mediated by
76. Craig, N. L. in Mobile DNA II (eds Craig, N. L., retrotransposon of fission yeast has strong endonuclease-independent LINE‑1 retrotransposition.
Craigie, R., Gellert, M. & Lambowitz, A. M.) 423–456 preferences for specific sites of insertion. Eukaryotic Nature Genet. 31, 159–165 (2002).
(ASM Press, Washington DC, 2002). Cell 1, 44–55 (2002). 122. Gladyshev, E. A. & Arkhipova, I. R. Telomere-
77. Wolkow, C. A., DeBoy, R. T. & Craig, N. L. Conjugating 98. Neely, L. A. & Hoffman, C. S. Protein kinase A and associated endonuclease-deficient Penelope-like
plasmids are preferred targets for Tn7. Genes Dev. 10, mitogen-activated protein kinase pathways retroelements in diverse eukaryotes. Proc. Natl Acad.
2145–2157 (1996). antagonistically regulate fission yeast fbp1 Sci. USA 104, 9352–9357 (2007).
78. Peters, J. E. & Craig, N. L. Tn7 transposes proximal to transcription by employing different modes of action References 120 and 122 reveal pathways by which
DNA double-strand breaks and into regions where at two upstream activation sites. Mol. Cell. Biol. 20, retrotransposons can use telomeric sequences as
chromosomal DNA replication terminates. Mol. Cell 6, 6426–6434 (2000). integration substrates.
573–582 (2000). 99. Majumdar, A., Chatterjee, A. G., Ripmaster, T. L. & 123. Gilbert, N., Lutz-Prigge, S. & Moran, J. V.
79. Bellen, H. J. et al. The Drosophila gene disruption Levin, H. L. The determinants that specify the Genomic deletions created upon LINE‑1
project: progress using transposons with distinctive integration pattern of retrotransposon Tf1 in the fbp1 retrotransposition. Cell 110, 315–325 (2002).
site-specificities. Genetics 188,731–743 (2011). promoter of Schizosaccharomyces pombe. J. Virol. 124. Symer, D. E. et al. Human L1 retrotransposition is
80. Feng, Q., Moran, J., Kazazian, H. & Boeke, J. D. 85, 519–529 (2010). associated with genetic instability in vivo. Cell 110,
Human L1 retrotransposon encodes a conserved 100. Chen, D. et al. Global transcriptional responses of 327–338 (2002).
endonuclease required for retrotransposition. Cell 87, fission yeast to environmental stress. Mol. Biol. Cell 125. Harrow, J. et al. GENCODE: producing a reference
905–916 (1996). 14, 214–229 (2003). annotation for ENCODE. Genome Biol. 7 (Suppl. 1), 4
81. Feng, Q., Schumann, G. & Boeke, J. D. 101. Malik, H. S., Henikoff, S. & Eickbush, T. H. Poised for (2006).
Retrotransposon R1Bm endonuclease cleaves the contagion: evolutionary origins of the infectious 126. Coffey, A. J. et al. The GENCODE exome: sequencing
target sequence. Proc. Natl Acad. Sci. USA 95, abilities of invertebrate retroviruses. Genome Res. 10, the complete human exome. Eur. J. Hum. Genet. 19,
2083–2088 (1998). 1307–1318 (2000). 827–831 (2011).

626 | SEPTEMBER 2011 | VOLUME 12 www.nature.com/reviews/genetics

© 2011 Macmillan Publishers Limited. All rights reserved


REVIEWS

127. Jurka, J. Sequence patterns indicate an enzymatic 153. Sijen, T. & Plasterk, R. H. Transposon silencing in the 176. Branciforte, D. & Martin, S. L. Developmental and cell
involvement in integration of mammalian retroposons. Caenorhabditis elegans germ line by natural RNAi. type specificity of LINE‑1 expression in mouse testis:
Proc. Natl Acad. Sci. USA 94, 1872–1877 (1997). Nature 426, 310–314 (2003). implications for transposition. Mol. Cell. Biol. 14,
128. Cost, G. J. & Boeke, J. D. Targeting of human 154. Tabara, H. et al. The rde‑1 gene, RNA interference, 2584–2592 (1994).
retrotransposon integration is directed by the specificity and transposon silencing in C. elegans. Cell 99, 177. Trelogan, S. A. & Martin, S. L. Tightly regulated,
of the L1 endonuclease for regions of unusual DNA 123–132 (1999). developmentally specific expression of the first
structure. Biochemistry 37, 18081–18093 (1998). 155. Malone, C. D. et al. Specialized piRNA pathways act in open reading frame from LINE‑1 during mouse
129. Korenberg, J. R. & Rykowski, M. C. Human genome germline and somatic tissues of the Drosophila ovary. embryogenesis. Proc. Natl Acad. Sci. USA 92,
organization: Alu, LINES, and the molecular structure Cell 137, 522–535 (2009). 1520–1524 (1995).
of metaphase chromosome bands. Cell 53, 391–400 156. Olivieri, D., Sykora, M. M., Sachidanandam, R., 178. Georgiou, I. et al. Retrotransposon RNA expression
(1988). Mechtler, K. & Brennecke, J. An in vivo RNAi assay and evidence for retrotransposition events in human
130. Brookfield, J. F. Selection on Alu sequences? identifies major genetic and cellular requirements for oocytes. Hum. Mol. Genet. 18, 1221–1228 (2009).
Curr. Biol. 11, R900–R901 (2001). primary piRNA biogenesis in Drosophila. EMBO J. 29, 179. Ostertag, E. M. et al. A mouse model of human L1
131. Boissinot, S., Entezam, A. & Furano, A. V. Selection 3301–3317 (2010). retrotransposition. Nature Genet. 32, 655–660
against deleterious LINE‑1‑containing loci in the 157. Zamore, P. D. Somatic piRNA biogenesis. EMBO J. 29, (2002).
human lineage. Mol. Biol. Evol. 18, 926–935 (2001). 3219–3221 (2010). 180. Macia, A. et al. Epigenetic control of retrotransposon
132. Boissinot, S., Davis, J., Entezam, A., Petrov, D. & 158. Brennecke, J. et al. An epigenetic role for maternally expression in human embryonic stem cells. Mol. Cell.
Furano, A. V. Fitness cost of LINE‑1 (L1) activity in inherited piRNAs in transposon silencing. Science Biol. 31, 300–316 (2011).
humans. Proc. Natl Acad. Sci. USA 103, 9590–9594 322, 1387–1392 (2008). 181. Federoff, N. in Mobile DNA II (eds. Craig, N. L.,
(2006). 159. Slotkin, R. K. et al. Epigenetic reprogramming and Craigie, R., Gellert, M. & Lambowitz, A. M.)
133. Han, J. S., Szak, S. T. & Boeke, J. D. Transcriptional small RNA silencing of transposable elements in 997–1007 (ASM Press, Washington DC, 2002).
disruption by the L1 retrotransposon and implications pollen. Cell 136, 461–472 (2009). 182. Emmons, S. W. & Yesner, L. High-frequency excision
for mammalian transcriptomes. Nature 429, References 155, 156, 158 and 159 highlight some of transposable element Tc1 in the nematode
268–274 (2004). pathways by which small RNAs can inhibit TE Caenorhabditis elegans is limited to somatic cells.
134. Perepelitsa-Belancio, V. & Deininger, P. mobility in either the germ line or soma. Cell 36, 599–605 (1984).
RNA truncation by premature polyadenylation 160. Yang, N. & Kazazian, H. H. Jr. L1 retrotransposition 183. Fernandez, L., Torregrosa, L., Segura, V., Bouquet, A.
attenuates human mobile element activity. Nature is suppressed by endogenously encoded small & Martinez-Zapater, J. M. Transposon-induced gene
Genet. 35, 363–366 (2003). interfering RNAs in human cultured cells. Nature activation as a mechanism generating cluster shape
135. Conley, M. E., Partain, J. D., Norland, S. M., Struct. Mol. Biol. 13, 763–771 (2006). somatic variation in grapevine. Plant J. 61, 545–557
Shurtleff, S. A. & Kazazian, H. H. Jr. Two independent 161. Kuramochi-Miyagawa, S. et al. DNA methylation of (2010).
retrotransposon insertions at the same site within the retrotransposon genes is regulated by Piwi family 184. Swergold, G. D. Identification, characterization, and
coding region of BTK. Hum. Mutat. 25, 324–325 members MILI and MIWI2 in murine fetal testes. cell specificity of a human LINE‑1 promoter. Mol. Cell.
(2005). Genes Dev. 22, 908–917 (2008). Biol. 10, 6718–6729 (1990).
136. Halling, K. C. et al. Hereditary desmoid disease in a 162. Reuter, M. et al. Loss of the Mili-interacting Tudor 185. Muotri, A. R. et al. L1 retrotransposition in neurons is
family with a germline Alu I repeat mutation of the domain-containing protein‑1 activates transposons modulated by MeCP2. Nature 468, 443–446 (2010).
APC gene. Hum. Hered. 49, 97–102 (1999). and alters the Mili-associated small RNA profile. References 23, 24 and 185 suggest that
137. Vidaud, D. et al. Haemophilia B due to a de novo Nature Struct. Mol. Biol. 16, 639–646 (2009). engineered human L1s, and perhaps endogenous
insertion of a human-specific Alu subfamily member 163. Soper, S. F. et al. Mouse maelstrom, a component of L1s, can retrotranspose in somatic cells of the
within the coding region of the factor IX gene. nuage, is essential for spermatogenesis and transposon mammalian nervous system.
Eur. J. Hum. Genet. 1, 30–36 (1993). repression in meiosis. Dev. Cell 15, 285–297 (2008). 186. Rehen, S. K. et al. Chromosomal variation in neurons
138. Cost, G. J., Golding, A., Schlissel, M. S. & Boeke, J. D. References 161–163 highlight how the small RNA of the developing and adult mammalian nervous
Target DNA chromatinization modulates nicking by L1 machinery influences the expression, and perhaps system. Proc. Natl Acad. Sci. USA 98, 13361–13366
endonuclease. Nucleic Acids Res. 29, 573–577 (2001). mobility, of certain TEs in mammalian cells. (2001).
139. Selker, E. U. et al. The methylated component of the 164. Matsuda, E. & Garfinkel, D. J. Posttranslational 187. Westra, J. W. et al. Neuronal DNA content variation
Neurospora crassa genome. Nature 422, 893–897 interference of Ty1 retrotransposition by antisense (DCV) with regional and individual differences in the
(2003). RNAs. Proc. Natl Acad. Sci. USA 106, 15657–15662 human brain. J. Comp. Neurol. 518, 3981–4000
140. Yoder, J. A., Walsh, C. P. & Bestor, T. H. (2009). (2010).
Cytosine methylation and the ecology of intragenomic This paper reveals a novel RNA-based mechanism 188. Belancio, V. P., Roy-Engel, A. M. & Deininger, P. L.
parasites. Trends Genet. 13, 335–340 (1997). that inhibits Ty1 retrotransposition in S. cerevisiae. All y’all need to know ‘bout retroelements in cancer.
141. Goll, M. G. & Bestor, T. H. Eukaryotic cytosine 165. Crow, Y. J. et al. Mutations in the gene encoding the Semin. Cancer Biol. 20, 200–210 (2010).
methyltransferases. Annu. Rev. Biochem. 74, 3′‑5′ DNA exonuclease TREX1 cause Aicardi-Goutieres 189. Alves, G., Tatro, A. & Fanning, T. Differential
481–514 (2005). syndrome at the AGS1 locus. Nature Genet. 38, methylation of human LINE‑1 retrotransposons in
142. Schaefer, C. B., Ooi, S. K., Bestor, T. H. & Bourc’his, D. 917–920 (2006). malignant cells. Gene 176, 39–44 (1996).
Epigenetic decisions in mammalian germ cells. Science 166. Stetson, D. B., Ko, J. S., Heidmann, T. & Medzhitov, R. 190. Asch, H. L. et al. Comparative expression of the
316, 398–399 (2007). TREX1 prevents cell-intrinsic initiation of LINE‑1 p40 protein in human breast carcinomas
143. Maksakova, I. A., Mager, D. L. & Reiss, D. autoimmunity. Cell 134, 587–598 (2008). and normal breast tissues. Oncol. Res. 8, 239–247
Keeping active endogenous retroviral-like elements in 167. Grimaldi, G. & Singer, M. F. Members of the KpnI (1996).
check: the epigenetic perspective. Cell. Mol. Life Sci. family of long interspersed repeated sequences join 191. Kubo, S. et al. L1 retrotransposition in nondividing
65, 3329–3347 (2008). and interrupt alpha-satellite in the monkey genome. and primary human somatic cells. Proc. Natl Acad.
144. Tsukahara, S. et al. Bursts of retrotransposition Nucleic Acids Res. 11, 321–338 (1983). Sci. USA 103, 8036–8041 (2006).
reproduced in Arabidopsis. Nature 461, 423–426 168. Eickbush, T. H. & Jamburuthugoda, V. K. The diversity 192. Shi, X., Seluanov, A. & Gorbunova, V. Cell divisions are
(2009). of retrotransposons and the properties of their reverse required for L1 retrotransposition. Mol. Cell. Biol. 27,
This paper used modern genomic approaches to transcriptases. Virus Res. 134, 221–234 (2008). 1264–1270 (2007).
reveal that certain classes of LTR retrotransposon 169. Babushok, D. V. & Kazazian, H. H. Jr. Progress in
are mobilized in a DDM1 (decrease in DNA understanding the biology of the human mutagen Acknowledgements
methylation 1) mutant background. It provides LINE‑1. Hum. Mutat. 28, 527–539 (2007). We thank J. Kim, J. Garcia-Perez and members of the Moran
an example of how epigenetic changes can lead 170. Suzuki, J. et al. Genetic evidence that the non- laboratory for critical reading of the manuscript. H.L.L. was
to TE mobility. homologous end-joining repair pathway is involved in supported in part by the Intramural Research Program of the
145. Bourc’his, D. & Bestor, T. H. Meiotic catastrophe and LINE retrotransposition. PLoS Genet. 5, e1000461 US National Institutes of Health (NIH) from the Eunice
retrotransposon reactivation in male germ cells (2009). Kennedy Shriver National Institute of Child Health and
lacking Dnmt3L. Nature 431, 96–99 (2004). 171. Gasior, S. L., Roy-Engel, A. M. & Deininger, P. L. Human Development. He received additional support from
146. Law, J. A. & Jacobsen, S. E. Establishing, maintaining ERCC1/XPF limits L1 retrotransposition. DNA Repair the Intramural AIDS Targeted Antiviral Program. J.V.M. was
and modifying DNA methylation patterns in plants (Amst.) 7, 983–989 (2008). supported in part by grants from the NIH (GM060518 and
and animals. Nature Rev. Genet. 11, 204–220 (2010). 172. Garcia-Perez, J. L. et al. Epigenetic silencing of GM082970) and is an Investigator of the Howard Hughes
147. van Rij, R. P. & Berezikov, E. Small RNAs and the engineered L1 retrotransposition events in human Medical Institute.
control of transposons and viruses in Drosophila. embryonic carcinoma cells. Nature 466, 769–773
Trends Microbiol. 17, 163–171 (2009). (2010). Competing interests statement
148. Ghildiyal, M. & Zamore, P. D. Small silencing RNAs: This paper describes a host mechanism that is able The authors declare competing financial interests: see Web
an expanding universe. Nature Rev. Genet. 10, to epigenetically silence reporter genes delivered version for details.
94–108 (2009). into genomic DNA by L1 retrotransposition.
149. Slotkin, R. K. & Martienssen, R. Transposable 173. Rio, D. C. Regulation of Drosophila P element
elements and the epigenetic regulation of the genome. transposition. Trends Genet. 7, 282–287 (1991). FURTHER INFORMATION
Nature Rev. Genet. 8, 272–285 (2007). 174. Pelisson, A. et al. Gypsy transposition correlates Henry L. Levin’s homepage: https://science.nichd.nih.gov/
150. Czech, B. & Hannon, G. J. Small RNA sorting: matchmaking with the production of a retroviral envelope-like confluence/display/pcrm/Henry+Levin
for Argonautes. Nature Rev. Genet. 12, 19–31 (2011). protein under the tissue-specific control of the John V. Moran’s homepage at the Howard Hughes Medical
151. Aravin, A. A., Hannon, G. J. & Brennecke, J. The Piwi- Drosophila flamenco gene. EMBO J. 13, 4401–4411 Institute: http://www.hhmi.org/research/investigators/
piRNA pathway provides an adaptive defense in the (1994). moran_bio.html
transposon arms race. Science 318, 761–764 (2007). 175. Prud’homme, N., Gans, M., Masson, M., Terzian, C. & John V. Moran’s homepage at the University of Michigan:
152. Fischer, S. E. Small RNA-mediated gene silencing Bucheton, A. Flamenco, a gene controlling the gypsy http://www.hg.med.umich.edu/faculty/john-v-moran-phd
pathways in C. elegans. Int. J. Biochem. Cell Biol. 42, retrovirus of Drosophila melanogaster. Genetics 139, ALL LINKS ARE ACTIVE IN THE ONLINE PDF
1306–1315 (2010). 697–711 (1995).

NATURE REVIEWS | GENETICS VOLUME 12 | SEPTEMBER 2011 | 627

© 2011 Macmillan Publishers Limited. All rights reserved

You might also like