chromatin-modifying genetic interventions suppress age … › content › pnas › 113 › 40 ›...

6
Chromatin-modifying genetic interventions suppress age-associated transposable element activation and extend life span in Drosophila Jason G. Wood a , Brian C. Jones a , Nan Jiang a , Chengyi Chang a , Suzanne Hosier a , Priyan Wickremesinghe a , Meyrolin Garcia a , Davis A. Hartnett a , Lucas Burhenn a , Nicola Neretti a , and Stephen L. Helfand a,1 a Department of Molecular Biology, Cell Biology, and Biochemistry, Brown University, Providence, RI 02912 Edited by Nancy M. Bonini, University of Pennsylvania, Philadelphia, PA, and approved August 5, 2016 (received for review March 28, 2016) Transposable elements (TEs) are mobile genetic elements, highly enriched in heterochromatin, that constitute a large percentage of the DNA content of eukaryotic genomes. Aging in Drosophila mel- anogaster is characterized by loss of repressive heterochromatin structure and loss of silencing of reporter genes in constitutive heterochromatin regions. Using next-generation sequencing, we found that transcripts of many genes native to heterochromatic regions and TEs increased with age in fly heads and fat bodies. A dietary restriction regimen, known to extend life span, repressed the age-related increased expression of genes located in heterochro- matin, as well as TEs. We also observed a corresponding age-associ- ated increase in TE transposition in fly fat body cells that was delayed by dietary restriction. Furthermore, we found that manipulating genes known to affect heterochromatin structure, including overex- pression of Sir2, Su(var)39, and Dicer-2, as well as decreased expres- sion of Adar, mitigated age-related increases in expression of TEs. Increasing expression of either Su(var)39 or Dicer-2 also led to an increase in life span. Mutation of Dicer-2 led to an increase in DNA double-strand breaks. Treatment with the reverse transcriptase inhib- itor 3TC resulted in decreased TE transposition as well as increased life span in TE-sensitized Dicer-2 mutants. Together, these data sup- port the retrotransposon theory of aging, which hypothesizes that epigenetically silenced TEs become deleteriously activated as cel- lular defense and surveillance mechanisms break down with age. Furthermore, interventions that maintain repressive heterochroma- tin and preserve TE silencing may prove key to preventing damage caused by TE activation and extending healthy life span. aging | heterochromatin | transposable elements | dietary restriction | silencing T here is mounting evidence that preservation of the epigenetic structure of chromatin is important to maintaining healthy aging, and loss of this chromatin structure may be a hallmark of the aging process (reviewed in ref. 1). In aging yeast, cells lose characteristic silencing in heterochromatic regions, resulting in deleterious age-related consequences such as sterility, and replica- tive life span is regulated by the histone H4K16 acetylation mark (2, 3). In Caenorhabditis elegans, genetic manipulation of chromatin factors affecting histone methylation and acetylation directly affect life span (4). In Drosophila, the integrity of constitutive repressive heterochromatin declines with age at both the genomic and cellular level (5), with one consequence of these changes being an age-re- lated loss of gene silencing in repressive constitutive heterochro- matin regions in multiple tissues of the fly (6). In mice, changes in epigenetic chromatin marks are seen with age, and age-related phenotypes of behavior, memory, and learning are observed on genetic manipulation of certain histone marks (1). A breakdown in heterochromatin organization has also been observed during repli- cative senescence of human cells in culture, with expression and copy number of transposable elements (TEs) increasing (7, 8). TEs are mobile genetic elements that make up a large per- centage of eukaryotic genomes and that are particularly enriched in regions of heterochromatin (reviewed in ref. 9). Because TEs function by either excising or copying themselves from the ge- nome and integrating into new genomic locations, there is a high potential for deleterious effects on cellular function. Over- expressing TE RNAs is sufficient to cause cytotoxicity, and inhibiting expression of these TEs causes reversal of both se- nescence and cytotoxic phenotypes (10, 11). TE activation has been observed with age in mice (8, 12), as well as C. elegans (13), yeast (14), Drosophila (15, 16), and senescent human tissue culture cells (7). To combat the genomic threat of activation of TEs and loss of heterochromatic gene silencing, somatic cells have evolved mecha- nisms to suppress TE transcription, primarily through establish- ment and maintenance of repressive heterochromatin at TE sites (17). In Drosophila, the small interfering RNA (siRNA) pathway, through the proteins Dicer-2 and Argonaute 2 (AGO2), recruits the histone H3K9 methyltransferase Su(var)39 to catalyze formation of repressive heterochromatin at TE sites (1820). Mutation of Dicer- 2, the double-stranded RNase responsible for generating siRNAs, results in TE reactivation and heterochromatin disorganization (19, 20). The RNA-editing enzyme ADAR (adenosine deaminase act- ing on RNA) is thought to modify the effectiveness of siRNA- directed heterochromatinization, and therefore the efficacy of TE silencing, by editing the dsRNA substrates used for siRNA biogenesis, thereby preventing proper processing by Dicer-2 (21). Sir2/Sirt1, the founding member of the sirtuin family of proteins, is a protein deacetylase important in maintaining gene Significance Most eukaryotic genomes contain abundant transposable ele- ments (TEs), mobile DNA elements that can replicate and move within the genome. Because of the deleterious nature of active TEs, cells have mechanisms to suppress and prevent TE activation, including formation of repressive heterochromatin. In this report, we show that many TEs become activated with age in Dro- sophila, and this activation is prevented by dietary restriction, an intervention known to extend life span. We also show TE acti- vation is blocked by genetic manipulations that stabilize hetero- chromatin and increase life span. This study provides evidence that a breakdown in TE silencing and repression may be a con- tributing factor to aging, and preventing TE activation may be a significant method of ameliorating the diseases of aging. Author contributions: J.G.W., B.C.J., and S.L.H. designed research; J.G.W., B.C.J., N.J., C.C., S.H., P.W., M.G., D.A.H., L.B., and S.L.H. performed research; J.G.W., B.C.J., and N.N. analyzed data; and J.G.W. and S.L.H. wrote the paper. The authors declare no conflict of interest. This article is a PNAS Direct Submission. See Commentary on page 11069. 1 To whom correspondence should be addressed. Email: [email protected]. This article contains supporting information online at www.pnas.org/lookup/suppl/doi:10. 1073/pnas.1604621113/-/DCSupplemental. www.pnas.org/cgi/doi/10.1073/pnas.1604621113 PNAS | October 4, 2016 | vol. 113 | no. 40 | 1127711282 GENETICS SEE COMMENTARY Downloaded by guest on August 3, 2020

Upload: others

Post on 07-Jul-2020

0 views

Category:

Documents


0 download

TRANSCRIPT

Page 1: Chromatin-modifying genetic interventions suppress age … › content › pnas › 113 › 40 › 11277.full.pdf · Chromatin-modifying genetic interventions suppress age-associated

Chromatin-modifying genetic interventions suppressage-associated transposable element activationand extend life span in DrosophilaJason G. Wooda, Brian C. Jonesa, Nan Jianga, Chengyi Changa, Suzanne Hosiera, Priyan Wickremesinghea,Meyrolin Garciaa, Davis A. Hartnetta, Lucas Burhenna, Nicola Nerettia, and Stephen L. Helfanda,1

aDepartment of Molecular Biology, Cell Biology, and Biochemistry, Brown University, Providence, RI 02912

Edited by Nancy M. Bonini, University of Pennsylvania, Philadelphia, PA, and approved August 5, 2016 (received for review March 28, 2016)

Transposable elements (TEs) are mobile genetic elements, highlyenriched in heterochromatin, that constitute a large percentage ofthe DNA content of eukaryotic genomes. Aging in Drosophila mel-anogaster is characterized by loss of repressive heterochromatinstructure and loss of silencing of reporter genes in constitutiveheterochromatin regions. Using next-generation sequencing, wefound that transcripts of many genes native to heterochromaticregions and TEs increased with age in fly heads and fat bodies. Adietary restriction regimen, known to extend life span, repressedthe age-related increased expression of genes located in heterochro-matin, as well as TEs. We also observed a corresponding age-associ-ated increase in TE transposition in fly fat body cells that was delayedby dietary restriction. Furthermore, we found that manipulatinggenes known to affect heterochromatin structure, including overex-pression of Sir2, Su(var)3–9, and Dicer-2, as well as decreased expres-sion of Adar, mitigated age-related increases in expression of TEs.Increasing expression of either Su(var)3–9 or Dicer-2 also led to anincrease in life span. Mutation of Dicer-2 led to an increase in DNAdouble-strand breaks. Treatment with the reverse transcriptase inhib-itor 3TC resulted in decreased TE transposition as well as increasedlife span in TE-sensitized Dicer-2mutants. Together, these data sup-port the retrotransposon theory of aging, which hypothesizes thatepigenetically silenced TEs become deleteriously activated as cel-lular defense and surveillance mechanisms break down with age.Furthermore, interventions that maintain repressive heterochroma-tin and preserve TE silencing may prove key to preventing damagecaused by TE activation and extending healthy life span.

aging | heterochromatin | transposable elements | dietary restriction |silencing

There is mounting evidence that preservation of the epigeneticstructure of chromatin is important to maintaining healthy

aging, and loss of this chromatin structure may be a hallmark ofthe aging process (reviewed in ref. 1). In aging yeast, cells losecharacteristic silencing in heterochromatic regions, resulting indeleterious age-related consequences such as sterility, and replica-tive life span is regulated by the histone H4K16 acetylation mark (2,3). In Caenorhabditis elegans, genetic manipulation of chromatinfactors affecting histone methylation and acetylation directly affectlife span (4). In Drosophila, the integrity of constitutive repressiveheterochromatin declines with age at both the genomic and cellularlevel (5), with one consequence of these changes being an age-re-lated loss of gene silencing in repressive constitutive heterochro-matin regions in multiple tissues of the fly (6). In mice, changes inepigenetic chromatin marks are seen with age, and age-relatedphenotypes of behavior, memory, and learning are observed ongenetic manipulation of certain histone marks (1). A breakdown inheterochromatin organization has also been observed during repli-cative senescence of human cells in culture, with expression andcopy number of transposable elements (TEs) increasing (7, 8).TEs are mobile genetic elements that make up a large per-

centage of eukaryotic genomes and that are particularly enriched

in regions of heterochromatin (reviewed in ref. 9). Because TEsfunction by either excising or copying themselves from the ge-nome and integrating into new genomic locations, there is ahigh potential for deleterious effects on cellular function. Over-expressing TE RNAs is sufficient to cause cytotoxicity, andinhibiting expression of these TEs causes reversal of both se-nescence and cytotoxic phenotypes (10, 11). TE activation hasbeen observed with age in mice (8, 12), as well as C. elegans (13),yeast (14), Drosophila (15, 16), and senescent human tissue culturecells (7).To combat the genomic threat of activation of TEs and loss of

heterochromatic gene silencing, somatic cells have evolved mecha-nisms to suppress TE transcription, primarily through establish-ment and maintenance of repressive heterochromatin at TE sites(17). In Drosophila, the small interfering RNA (siRNA) pathway,through the proteins Dicer-2 and Argonaute 2 (AGO2), recruits thehistone H3K9 methyltransferase Su(var)3–9 to catalyze formationof repressive heterochromatin at TE sites (18–20). Mutation of Dicer-2, the double-stranded RNase responsible for generating siRNAs,results in TE reactivation and heterochromatin disorganization (19,20). The RNA-editing enzyme ADAR (adenosine deaminase act-ing on RNA) is thought to modify the effectiveness of siRNA-directed heterochromatinization, and therefore the efficacy ofTE silencing, by editing the dsRNA substrates used for siRNAbiogenesis, thereby preventing proper processing by Dicer-2(21). Sir2/Sirt1, the founding member of the sirtuin family ofproteins, is a protein deacetylase important in maintaining gene

Significance

Most eukaryotic genomes contain abundant transposable ele-ments (TEs), mobile DNA elements that can replicate and movewithin the genome. Because of the deleterious nature of activeTEs, cells havemechanisms to suppress and prevent TE activation,including formation of repressive heterochromatin. In this report,we show that many TEs become activated with age in Dro-sophila, and this activation is prevented by dietary restriction, anintervention known to extend life span. We also show TE acti-vation is blocked by genetic manipulations that stabilize hetero-chromatin and increase life span. This study provides evidencethat a breakdown in TE silencing and repression may be a con-tributing factor to aging, and preventing TE activation may be asignificant method of ameliorating the diseases of aging.

Author contributions: J.G.W., B.C.J., and S.L.H. designed research; J.G.W., B.C.J., N.J., C.C.,S.H., P.W., M.G., D.A.H., L.B., and S.L.H. performed research; J.G.W., B.C.J., and N.N. analyzeddata; and J.G.W. and S.L.H. wrote the paper.

The authors declare no conflict of interest.

This article is a PNAS Direct Submission.

See Commentary on page 11069.1To whom correspondence should be addressed. Email: [email protected].

This article contains supporting information online at www.pnas.org/lookup/suppl/doi:10.1073/pnas.1604621113/-/DCSupplemental.

www.pnas.org/cgi/doi/10.1073/pnas.1604621113 PNAS | October 4, 2016 | vol. 113 | no. 40 | 11277–11282

GEN

ETICS

SEECO

MMEN

TARY

Dow

nloa

ded

by g

uest

on

Aug

ust 3

, 202

0

Page 2: Chromatin-modifying genetic interventions suppress age … › content › pnas › 113 › 40 › 11277.full.pdf · Chromatin-modifying genetic interventions suppress age-associated

silencing in heterochromatin regions of yeast and flies throughdeacetylation of specific histone lysines required for establishmentand spreading of heterochromatin (3, 6, 22). Increasing Sir2 ex-pression has been shown to repress the normal age-related loss ofgene silencing in heterochromatin regions in both yeast and flies(3, 6), as well as the loss of gene silencing seen in mammalianembryonic stem cells undergoing genotoxic stress (23). In additionto their roles in heterochromatin formation and maintenance, thesepathways have also been linked to aging. Decreasing activity of thesiRNA pathway through Ago2 and Dicer-2 mutants leads to de-repression of TE activity and shortened life span (15, 24). Sir2 has acentral role in aging, with increased gene dosage shown to extendlife span in yeast (2) and in nine of 10 studies in nematodes and flies(25–28). Hypomorphs of Adar, a negative regulator of the siRNApathway, also exhibit an extension of life span (21). These pathwaysshare an ability to affect heterochromatin formation and life span.Here we report a characterization of the heterochromatic tran-

scriptome in adult Drosophila tissues and show activation of TEsand increased transposition with age. We provide evidence that the

aging process is characterized by a failure to properly control andrepress expression of heterochromatin-based genes and TEs in thesoma. We show that genetic interventions positively modifying het-erochromatin structure or negatively modulating TE activity extendmetazoan life span. Finally, we show that attenuating TE ac-tivity pharmacologically decreases transposition and extends life spanin Dicer-2 mutants. This study offers the first comprehensive char-acterization of the effect of known life span-extending interventionson heterochromatic regions and explores new interventions forcontrolling age-associated TE reactivation.

ResultsExpression of Genes Located Within Heterochromatin Increases withAge and Is Attenuated Under Dietary Restriction. Because hetero-chromatic regions change in structure with age, specifically infemale heads and fat bodies (5), we reasoned that genes locatedwithin these regions may also show changes in silencing or geneexpression with age. We used RNA-seq to assay the expression of the∼250 genes natively located within the Drosophila heterochromatin

−4 0 4Value

heads

−3 0 3Value

fat body

TABOR_I

Invader4_LTR

Gypsy10_LTR

TART_B1

I_DM

BEL_LTR

G6_DM

HETA

DIVER_LTR

DIVER_I

BATUMI_I

BURDOCK_I

DOC

QUASIMODO2−LTR

BEL_I

TRANSPAC_LTR

BLOOD_I

Gypsy12_LTR

DMTOM1_LTR

BLOOD_LTR

BS

NOMAD_I

MDG1_LTR

MDG1_I

DOC6_DM

PLACW_DM

Gypsy3_LTR

ACCORD_LTR

BATUMI_LTR

R2_DM

ACCORD_I

Gypsy3_I

Gypsy1−LTR_DM

Gypsy6_LTR

Gypsy1−I_DM

Copia2_I

Copia_LTR

Copia_I

DOC5_DMTRANSIB3HETAGypsy8_IQUASIMODO2−LTRACCORD2_LTRHMSBEAGLE_IGypsy1−LTR_DMTransib5BATUMI_LTRTRANSPAC_IG6_DMDOCFROGGER_IP−1_DYDOC6_DMTransib−N1_DMGypsy_DSDM176_ITOM_ITABOR_IDM1731_LTRNINJA_LTRMDG1_LTRIDEFIX_IBEL_LTRInvader5_LTRMariner2_DMPOGODM1731_IFW2_DMChouto_IGypsy3_IFW3_DMGypsy8_LTRHELENA_RTCopia1−LTR_DMG7_DMInvader3_LTROSVALDO_IGypsy_LTRDM176_LTRACCORD2_IBURDOCK_LTRGypsy12_LTRI_DMACCORD_LTRZAM_LTRDMTOM1_LTRGypsy1−I_DMGypsy7_LTRACCORD_IBARI_DMBica_LTRTLD2Gypsy6_LTRMAX_LTRCopia2_ICopia_ICopia_LTRPLACW_DM

HC DR

HC DR

heads

HC DR

log 2 f

old

chan

ge(o

ld/y

oung

)

HC DR

fat body

CG17163

CaMKI

myoglianin

CG10465

CG17486

eIF4G

CG3262

CG2177

CG32017

CG17082

CG11360

Ephrin

fog

CG12567

CG32016

CG40263

CG40191

CG9935

CG32350

CG17494

CG31997

CG2316

Rad23

Tif−IA

CG32230

CG33978

gus

CG40006

alpha−Cat

plexA

Yeti

Gprk1

Kif3C

CG3107

CG17167

Rfabg

CG6675

CG17018

−2 0 2Value

−4 0 4Value

Ephrin

tio

pan

Nipped−B

CG40263

p120ctn

CG40178

Sox102F

CG11360

Actbeta

CaMKI

RhoGAP102A

CG12547

CG40498

Asator

Atf6

CG41320

FucTC

Rfabg

CG17082

Cht3

CG1674

cals

Alg−2

Gfat1

alpha−Cat

zfh2

Caps

CG41378

Spf45

Pur−alpha

CG31997

sv

CG42402

Syt7

CG17167

CG40053

A B C D

Fig. 1. DR attenuates age-related loss of silencingin heterochromatin genes and TEs. (A and B) Heatmap shows log2 fold change (old/young) values forheterochromatin genes in both (A) heads and (B) fatbodies. (Left) Log2 fold change under HC conditions.(Right) Log2 fold change under DR. Heat map showsgenes with log2 fold changes greater than 0.263(1.2× fold change) under HC conditions that are lo-cated within annotated heterochromatin regions(29). Ninety-five percent of shown genes in headsand 89% in fat bodies are suppressed by DR. (C andD) Heat map shows log2 fold change (old/young) ofTE expression for both HC (Left) and DR (Right) dietin both (C) heads and (D) fat bodies. Heat mapshows TEs with log2 fold changes greater than 0.322(1.25× fold change) under HC conditions. Eighty-seven percent of shown TEs were suppressed by DRin both heads and fat bodies.

11278 | www.pnas.org/cgi/doi/10.1073/pnas.1604621113 Wood et al.

Dow

nloa

ded

by g

uest

on

Aug

ust 3

, 202

0

Page 3: Chromatin-modifying genetic interventions suppress age … › content › pnas › 113 › 40 › 11277.full.pdf · Chromatin-modifying genetic interventions suppress age-associated

compartments (29) from young (10 d) and old (40 d) female fly headsand fat bodies. The vast majority of cells in the head are neurons andglial cells. The fly fat body is the functional homolog of mammalianadipose tissue and liver, and a central organ in the fly immune sys-tem. We found a number of genes located in heterochromatic re-gions increased expression with age in both wild-type heads and fatbodies (Fig. 1 A and B). Gene ontology analysis reveals no sig-nificantly enriched pathways among these genes, suggesting theage-related increase in expression of these genes is likely to be aconsequence of loss of heterochromatic silencing, rather than aspecific compensatory or correlative response tied to aging.Dietary restriction (DR) is an intervention known to extend

life span and delay numerous age-related phenotypes. We pre-viously observed that DR can oppose age-related loss of genesilencing of reporter genes in constitutive heterochromatin (6).On this basis, we examined whether the age-dependent increasein expression of native genes in the constitutive heterochromatinregions were also suppressed under a DR regimen. The vast ma-jority (95% in heads, 89% in fat bodies) of heterochromatin genesthat increased expression with age on high-calorie food increasedless or decreased in expression with age on DR food [high calorie(HC) vs. DR] in both heads (Fig. 1A) and fat bodies (Fig. 1B).Although we observed a similar fraction of euchromatin genes thatincreased expression with age, this pattern of suppression by DRwas not maintained in the most highly up-regulated euchromatingenes (Fig. S1 A and B), suggesting DR has a selective effect onheterochromatin genes. Similar to previous studies using reportergenes in both yeast (2) and flies (6), these data suggest that geneexpression in heterochromatic regions becomes dysregulated withage, and that a known life span-extending intervention, DR, canprevent this.

TE Expression Increases with Age and Is Attenuated Under DR.One ofthe most notable attributes of heterochromatin is the abundantenrichment of TEs within these regions. The Drosophila genomeis rich in retroviral-like long terminal repeat (LTR) and longinterspersed nuclear elements (LINEs) (30), and activation ofboth LINEs and LTRs has been observed in the fly nervous systemand fat body during aging (15, 16). Because heterochromatin in-tegrity declines with age in multiple model systems (1), we hy-pothesized that a loss of heterochromatin in regions highly enrichedwith TEs would show a correlated increase in TE expression withage. To test this hypothesis, we examined the expression profile ofTEs with age and diet in female fly heads and fat bodies, usingwhole transcriptome RNA-seq. Similar to our observations fornative heterochromatin genes, we also found many TEs that in-creased their expression with age, in both heads and fat bodies(Fig. 1 C and D). We observed a high degree of overlap in the TEsthat increased expression most with age between these two tissues,most of which were retrotransposons (Fig. S2A). In both tissuetypes, as observed with the heterochromatin genes, DR attenu-ated the age-related increase in expression observed in HC food(Fig. 1 C and D, right “DR” columns). TEs that had the highestincrease in expression with age on HC food tended to increaseless or even decrease in expression with age under DR conditions(87% lower in DR relative to HC in both heads and fat bodies;Dataset S1). These data suggest that, similar to reporter genes inheterochromatin (2, 6) and native heterochromatin genes (Fig. 1 Aand B), TE silencing is lost with age, and DR opposes this.

TE Transposition Increases with Age and Is Delayed by DR. An age-dependent increase in TE expression and transposition in theDrosophila brain associated with decline in neural function hasbeen reported (15). Similar increases in TE expression levels andcopy numbers have been observed in aging mouse tissue (8). Toassay for age-related transposition in our flies, we used the gypsy-TRAP reporter system (15). This system is designed such thatgreen fluorescent protein (GFP) becomes expressed when an

endogenous gypsy retrotransposon jumps into an engineered sitecontaining known hotspot sequences from the ovo locus (31). Wemonitored flies as they aged and visually sorted them into eitherlow, medium, or high categories based on the number of GFP-positive cells observed in the fat body (Fig. 2A). Longitudinal flycohorts were assayed for transposition every 7 d on either HCor DR foods (Fig. 2B). We observed a clear age-associated in-crease in transposition, and DR delayed this increase relative to HCcontrols (Fig. 2 B and C). Similar results were seen in two additionalgenetic backgrounds (Fig. S3 A–D). We examined the life spans ofthese three different genetic background gypsy-TRAP lines andobserved that life span correlated with the timing of increasedtransposition. Lines exhibiting earlier transposition showed shorterlife span, and those with later transposition longer life span(Fig. S3E). Similarly, the delay in age-related transpositionresulting from DR correlated with the ability of DR to extend lifespan in each line. To ensure the increased GFP expression weobserved with age was caused by transposition rather than age-related disruption in the GAL80/GAL4/GFP reporter system, weexamined a gypsy-TRAP line with a mutant ovo locus not sus-ceptible to transposition and observed no significant increase inGFP expression with age (Fig. S3F). Rather than a constant linearincrease in GFP expression with time, the gypsy-TRAP assayshowed a clear age-associated increase in TE mobilization, consis-tent with the TE theory of aging, which posits an age-related loss ofnormal cellular surveillance mechanisms and consequent increase indeleterious TE activity. The relationship between the timing of in-creased gypsy transposition and life span further supports TE mo-bilization as an age-associated phenomenon.

BA

0

25

50

75

100

5 12 19 26 33 42 49 High Med Low

0 25 50 75

100

5 12 19 26 33 42 49

0

25

50

75

100

0 10 20 30 40 50

Per

cent

sta

inin

gP

erce

nt m

ediu

m o

r hig

h

Age (days)

Age (days)

C

Low

Med

ium

Hig

h

Staininglevel

High calorie

Dietary restriction

DRHigh Calorie

Fig. 2. DR delays age-related increase in TE transposition. (A) Gypsy-TRAP flies(genotype: w1118; If/+; r4-GAL4, UAS-GFP/tub-ovo-GAL80) were categorized aslow (<20 GFP-positive cells), medium (20–100), or high (>100) based on numberof GFP-positive cells observed in fat body (representative images shown). Thedemarcation between low and medium categories is clear; most flies have ei-ther fewer than 10 cells staining, or more than 50. (Scale bar, 200 μm.) (B)Percentage of observed flies falling into each category at indicated points forHC (Top) and DR (Bottom) diets (n = ∼120 per cohort). (C) Transposition (de-fined here as medium- or high-category GFP staining) increases with age and isdelayed by DR. Percentage of flies falling into either medium or high categoriesis displayed with time for both HC and DR diets. Log-rank P < 10−10.

Wood et al. PNAS | October 4, 2016 | vol. 113 | no. 40 | 11279

GEN

ETICS

SEECO

MMEN

TARY

Dow

nloa

ded

by g

uest

on

Aug

ust 3

, 202

0

Page 4: Chromatin-modifying genetic interventions suppress age … › content › pnas › 113 › 40 › 11277.full.pdf · Chromatin-modifying genetic interventions suppress age-associated

Age-Related Increases in TE Expression Are Attenuated by GeneticInterventions That Affect Heterochromatin Structure. Our data dem-onstrate a correlation between changes in heterochromatin struc-ture with age and expression and transposition of TEs during normalaging. This led us to examine whether genetically manipulatingheterochromatin structure and maintenance, either directly orindirectly, could also affect the age-related changes in expres-sion of TEs. To test our hypothesis, we genetically increased theactivity of two important systems that maintain repressive het-erochromatin in somatic cells: the siRNA pathway and Sir2. Usingtransgenic fly lines, we increased gene dosage of Sir2, Dicer-2, andSu(var)3–9 and also used an Adar hypomorph, interventions weexpect to stabilize heterochromatin. Each of these genes offers aunique and complementary approach to simultaneously manipu-lating heterochromatic regions and enhancing TE silencing.We assayed age-dependent loss of silencing of TEs in each of

these various transgenic [Sir2, Su(var)3–9, and Dicer-2] or hypo-morphic (Adar) fly lines compared with their respective controls.The TEs that showed the greatest increase in expression with agein the control lines were overwhelmingly attenuated or decreasedwith age by these genetic interventions [Fig. 3 A–D; Sir2, 72%;Adar, 84%, Su(var)3–9, 92%; Dicer-2, 68%]. Interestingly, we again

observed significant overlap in the TEs that increased with ageamong the different genetic backgrounds (Fig. S2B). Together,these data suggest that increased expression of Sir2, Dicer-2, or Su(var)3–9, as well as decreased gene dosage of Adar, all prevent theage-related loss of silencing of TEs observed in wild-type and con-trol lines, suggesting a common link between heterochromatin fac-tors and proper maintenance of TE silencing.TE activation is known to disrupt gene expression and destabilize

the genome. Increased DNA damage has been observed in aging fatbody nuclei, as assayed by γH2Av immunostaining (16). Our findingof a repression of TE expression with increased Dicer-2 expressionled us to investigate whether genomic integrity was similarly com-promised in animals with disrupted RNAi/heterochromatin path-ways and increased TE activity. Similar to depletion of AGO2 in fatbody cells (16), we observed an increase in double-strand breaks inDicer-2 mutants compared with controls in fat body nuclei (Fig.S4A), suggesting deleterious genomic consequences when properTE silencing is disrupted.

Genetic Interventions That Affect Heterochromatin Structure Also AffectLife Span. In addition to their effects of delaying age-related het-erochromatin gene and TE silencing loss, both increased Sir2

Sir2

−3 0 3Value

EtO

HR

U

Adar

wt

hyp

Su(var)3-9

wt

SV

2

−4 0 4Value

EtO

H

RU

Dicer-2A B C D

MAX_LTRBS2Stalker2_LTRCopia1−LTR_DMTIRANT_IGypsy3_IGypsy9_IGypsy5_IG6_DMUlysses_ITIRANT_LTRDMRT1CDOC5_DMBaggins1BATUMI_IDMRT1AACCORD2_LTRGypsy6_LTRGypsy_IZAM_LTRGypsy1−I_DMG_DMDMRT1BGypsy_LTRGypsy5_LTRMAX_IBARI_DMBATUMI_LTRChimpo_LTRBica_LTRCIRCER1_DMCopia2_ICopia_LTRR2_DMCopia_I

DIVER2_LTRGypsy4_LTRDIVER_IGypsy10_LTRTIRANT_LTRGypsy3_IACCORD_LTRDMLTR5Copia1−I_DMGypsy4_IInvader1_LTRGypsy5_LTRACCORD2_LTRGypsy1−I_DMTAHREGypsy6A_LTRChimpo_LTRGypsy6_LTRGypsy1−LTR_DMTRANSPAC_LTRBEL_LTRGypsy5_IMDG3_ICopia2_IGypsy12_LTRUlysses_IMAX_IBica_LTRR1_DMMAX_LTRBS3_DMHETACopia_LTRChouto_IBATUMI_IACCORD_ICopia_IBATUMI_LTR

BEL_LTRStalker2_LTRROO_ITRANSPAC_LTRGypsy7_LTRGypsy5_INOMAD_IZAM_LTRMDG1_IMDG3_LTRTABOR_LTRI_DMDOCACCORD_LTRBATUMI_IGypsy3_IR2_DMROO_LTRFW_DMDM1731_LTRG2_DMTABOR_IG7_DMBURDOCK_LTRTARTBSDIVER2_LTRBATUMI_LTRGypsy1−I_DMMDG1_LTRPLACW_DMCopia2_IGypsy1−LTR_DMGypsy6_LTRCopia_ICopia_LTR

Chouto_LTRTART_B1M4DMFW_DMACCORD2_LTRG6_DMDM412B_LTRFB4_DMDIVER2_LTRSTALKER4_ITOM_ININJA_IQUASIMODO_IGypsy4_IDMRT1BIDEFIX_LTRIVK_DMBLOOD_IBaggins1DM412Gypsy2_LTRDMCR1ANOMAD_LTRTARTTV1IGypsy8_LTRBARI_DMCopia2_IGypsy12A_LTRGypsy6_LTRGypsy8_IROVER−I_DMBica_LTRGypsy5_IMDG1_IDM176_IGypsy2−LTR_DMROO_ITRANSIB2TRANSIB3BATUMI_IBSMAX_LTRMAX_IDM297_II_DMDOC5_DMTABOR_IGypsy11_IStalker2_LTRACCORD2_IGypsy9_IZAM_IBATUMI_LTRGypsy12_LTRCopia_ICopia_LTR

log2 fold change (old/young)

log2 fold change (old/young)

Fig. 3. Manipulation of genes in heterochromatinpathways affects TE expression. Heat maps of log2

fold change (old/young) of TEs for the following gen-otypes and their respective controls: (A) elav-Gen-eSwitch > UAS-EP2300 (Sir2 overexpression); (B) Adarhypomorph vs. wild-type; (C) Su(var)3–9 overexpression[SV2 and SV3 lines overexpress Su(var)3–9 from abasally expressed UAS promoter 160–180% abovecontrol; Fig. S4B]; and (D) tubulin-GeneSwitch >UAS-Dicer-2 (Dicer-2 overexpression). (Left) Controlcondition. (Right) Transgenic, hypomorph, or RU486-induced condition. The percentage of shown TEs thatare suppressed by the indicated genotype are(A) 72%, (B) 84%, (C) 92%, and (D) 68%. All TEs withlog2 fold change (old/young) greater than 0.322 (1.25×fold change) in the control condition are shown.

11280 | www.pnas.org/cgi/doi/10.1073/pnas.1604621113 Wood et al.

Dow

nloa

ded

by g

uest

on

Aug

ust 3

, 202

0

Page 5: Chromatin-modifying genetic interventions suppress age … › content › pnas › 113 › 40 › 11277.full.pdf · Chromatin-modifying genetic interventions suppress age-associated

expression and decreased Adar expression have been shown to ex-tend life span in flies (21, 25), whereas Dicer-2 and Ago2 mutantsare short-lived (15, 24). Given our findings presented earlier, weexamined whether increased expression of Su(var)3–9 or Dicer-2also have a positive effect on life span. We found that two differentSu(var)3–9 transgenic lines, SV2 and SV3, which constitutively in-crease Su(var)3–9 expression to 160–180% of the wild-type level inthe absence of a driver (Fig. S4B), extend life span significantlycompared with their genetically matched control (Fig. 4A). Simi-larly, increasing Dicer-2 expression in adult life in either all tissues(Fig. 4B) or selectively in adult neurons (Fig. 4C), using the condi-tional inducible GeneSwitch system, was able to extend life span,with flies fed the gene-activating drug RU486 outliving a geneticallyidentical control cohort fed only the diluent control EtOH. Togetherwith previously published data (21, 25), these observed life spanextensions suggest a coordinated response integrating maintenanceof heterochromatin structure, TE suppression, and longevity.

Pharmacological Suppression of Transposable Element Activity DelaysAge-Related Transposition and Extends Life Span. To test whetherthe age-related effects we observed were a result of increased TEactivity, we sought to directly modulate transposition pharmaco-logically. We used the reverse transcriptase inhibitor lamivudine(3TC) to directly block TE transposition. Using the gypsy-TRAPreporter system in the fat body, we observed that flies treated with3TC had a significant delay in age-related transposition comparedwith controls (Fig. 5A). This effect was observed whether the drugwas administered constantly throughout life (Fig. 5A) or startedduring midadulthood at 20 d of age (Fig. S4C).As our evidence suggests that maintenance of TE silencing is

important for ensuring longevity, we tested whether attenuating TEactivity directly could extend life span. In the Dicer-2L811fsX mutant

background, which exhibits high levels of TE activity becauseof defective RNAi silencing (32), we found that administra-tion of 3TC was able to extend life span significantly comparedwith controls (Fig. 5B). Together, these data suggest that het-erochromatin-altering pathways that extend life span may do soat least in part by suppressing TE activity to ensure healthylongevity, and confirm that suppression of TE activation islikely to be an attractive target mechanism for pharmacologicalintervention.

DiscussionAging is characterized by a breakdown in homeostasis through-out multiple cellular and organismal systems. Genomic and epi-genomic stability is vital for somatic cells to maintain proper geneexpression and silencing. A number of studies have shown changesin chromatin structure during organismal aging in various modelsystems, especially in heterochromatin (1, 2). Although hetero-chromatin regions contain some genes, a likely reason for thecentral importance of maintaining repressive heterochromatin isthat heterochromatin is replete with TEs, and unchecked TE ac-tivation has particularly damaging ramifications for genomic sta-bility. Indeed, the expression and mobilization of TEs is disruptiveenough that cells have evolved RNAi pathways responsible forsilencing and preventing TE activation. These cellular defensesagainst TE activation include siRNA-directed induction andmaintenance of repressive heterochromatin to prevent TE ex-pression, and degradation of target transcripts. siRNA pathwaymutants exhibit both an activation of TEs and a shortening of lifespan (15, 24). Here we provide evidence that many heterochro-matin genes and TEs do in fact increase expression with age in twodifferent tissues. Increased transposition has been previouslyreported in the aging fly brain (15). Similarly, we also observe anincrease in somatic TE transposition with age in the fat body.DNA double-strand breaks increase with age in the fat body (16),and we show this DNA damage also accumulates at a younger agein Dicer-2 mutant flies with high levels of TEs. The age-relatedincrease in TE transcript levels as well as TE mobilization weresuppressed by DR. Together, these data suggest that an increase inTE expression and transposition is an important hallmark of so-matic aging, and preventing this increased TE activity is likely topreserve genomic stability and cellular homeostasis.We demonstrate in a whole organism model that aging is as-

sociated with a loss of silencing of both TEs and genes nativelylocated in heterochromatin regions, with a resultant increase inTE mobilization and an associated increase in DNA damage. It

BA

0

20

40

60

80

100

0 20 40 60 80 100

Per

cent

Aliv

e

Age (days)

EtOH RU486

0

20

40

60

80

100

0 20 40 60 80

Per

cent

Aliv

e

Age (days)

WT SV2 SV3

0

20

40

60

80

100

Per

cent

Aliv

e

0 20 40 60 80 100 Age (days)

C

EtOH RU486

tub-GS > UAS-Dcr-2

elav-GS > UAS-Dcr-2

Fig. 4. Increasing expression of Su(var)3–9 or Dicer-2 increases life span.(A) SV2 and SV3 lines [overexpressing Su(var)3–9] are long-lived comparedwith controls. Survivorship of adult female flies is shown (n = 250). WT meanlife span = 40.6 d, median = 44 d; SV2 mean = 58.5 d, median = 60 d, 36%median extension, log-rank P < 10−10; SV3 mean = 58.3 d, median = 60 d,36% median extension, P < 10−10. (B) Whole-adult animal Dicer-2 over-expressing flies are long-lived compared with controls. Survivorship of tu-bulin-GeneSwitch > UAS-Dicer-2 adult females (n = 250) is shown, eitherwith EtOH (uninduced) or 200 μM RU486 (induced expression). EtOH mean =52.3 d, median = 54 d; RU486 mean = 63.5 d, median = 66 d, 22% medianextension, P < 10−10. (C) Flies expressing Dicer-2 in adult neurons are long-livedcompared with controls. Survivorship of elav-GeneSwitch > UAS-Dicer-2 adultfemales is shown, for EtOH (uninduced) or 500 μM RU486 (induced) (n = 250).EtOH mean = 53.8 d, median = 56 d; RU486 mean = 61.7 d, median = 66 d,18% median extension, P < 10−10. All life span experiments were repeatedwith similar results; representative experiments shown.

0

25

50

75

100

0 20 40 60

Per

cent

Aliv

e

Age (days)

Control 3TC

0

10

20

30

40

50

60

Control 3TC

Per

cent

GF

P p

ositi

ve a

t 30

days

A B

Dcr-2L811fsX

Fig. 5. Administration of lamivudine (3TC) suppresses retrotranposition andextends Dicer-2 mutant life span. (A) Percentage of gypsy-TRAP flies withmore than 30 fat body cells positive for GFP expression at 30 d of age forboth untreated (control) and 10 μM 3TC treated fly cohorts. Fisher’s exacttest P < 0.0001. (B) 10 μM 3TC treated flies are long-lived compared withuntreated controls in the Dicer-2L811fsX genetic background. Survivorship ofadult female flies is shown (n = 250). Control mean = 43.0 d, median = 45 d;3TC mean = 46.4 d, median = 48 d, 6.6% median extension, log-rank P <0.0001. Life span was repeated with similar results.

Wood et al. PNAS | October 4, 2016 | vol. 113 | no. 40 | 11281

GEN

ETICS

SEECO

MMEN

TARY

Dow

nloa

ded

by g

uest

on

Aug

ust 3

, 202

0

Page 6: Chromatin-modifying genetic interventions suppress age … › content › pnas › 113 › 40 › 11277.full.pdf · Chromatin-modifying genetic interventions suppress age-associated

is highly likely that the pathways and processes of heterochro-matin maintenance, RNAi, and gene and TE silencing representpotential homeostatic failure points that lead to the phenotypesof aging. Here we have also presented genetic evidence thatthese pathways are linked to aging, as manipulating the expres-sion of several genes involved in maintaining heterochromatinstructure, TE silencing, and RNAi efficacy directly suppressesage-related TE expression as well as extends life span. Further-more, we show that DR, an environmental intervention thatextends life span robustly and delays most age-related pheno-types, also opposes the loss of silencing we observe during nor-mal aging and delays age-dependent transposition, furtherlending support to the idea that these processes are associatedwith and may be relevant to aging. We also demonstrate thatinhibition of retrotransposon activity is strongly correlated withextension of life span. Our data support the retrotransposon theoryof aging model, where proper maintenance of silent heterochro-matin and prevention of TE activation are key to guaranteeing ge-nomic stability and homeostasis during aging and ensuring healthylongevity. Furthermore, our demonstration that dietary (DR), ge-netic, and pharmacological interventions that reduce the age-relatedincreases in TE activity can also extend life span suggests new andnovel pathways for the development of interventions designed toextend healthy life span.

Materials and MethodsGypsy-TRAP Assay. Flies from the gypsy-TRAP line were aged for 5 d andinitially examined for the number of GFP-positive cells, then examined every7 d until age 49 d, using a fluorescent dissecting microscope. Flies wereseparated into three categories based on visual inspection, according to howmany fat body cells were GFP positive: low (<20 cells), medium (20–100 cells),and high (>100 cells). The same cohort of flies was examined longitudinallythroughout each of the experiments.

RNA-seq. Total RNA from the indicated genotypes was collected from∼100 fly heads or ∼50 dissected fat bodies, using the miRvana RNA prep

kit (Ambion). Then, 100 ng total RNA was used as input for strandedRNA-seq library construction, using the Ovation Universal RNA-seq kit(Nugen) with Drosophila rRNA depletion module. Libraries were sequencedon an Illumina HiSEq. 2500 in 1 × 50 bp mode. All samples were preparedusing at least three individually isolated biological replicates for eachcondition.

Bioinformatics. Standard RNA-seq analysis was performed using Tophat 2.0for sequence alignment, followed by gene-based counting using the easy-RNASeq R package from Bioconductor. Finally, trimmed mean of M-values(TMM) normalization and fold change calculation was performed usingedgeR, with a generalized linear model (GLM) correcting for batch effects,as described in the edgeR vignette. To account for repetitive sequencingreads and perform TE quantification, RepEnrich (33) was used to generate acount table of reads for each TE, and edgeR was used to normalize, calculatefold changes, and perform differential expression testing. The Repbase an-notation was used as input for RepEnrich.

Heat maps were generated by ranking the TEs or genes in order of log2

fold change of expression with age (old/young) for the reference (control/HC) condition and then plotting log2 fold change with age for all conditions.To show consistent trends in our data and include genes/TEs that behavedconsistently but did not meet the threshold of statistical significance, wedisplayed genes or TEs where log2 fold change was greater than 0.322 (1.25×fold; TEs) or 0.263 (1.2x fold; heterochromatin genes). Plots were generatedusing the heatmap.2 function in the gplots2 R package.

See SI Materials andMethods for additional details on fly stocks andmethods.

ACKNOWLEDGMENTS. We thank Will Lightfoot and Gillian Horwitz fortechnical assistance and Christoph Schorl and the Brown University GenomicsCore Facility for sequencing. We also thank Joshua Dubnau, Robert Reenan,Kristen Johansen, and the Bloomington Drosophila Stock center for flystocks and Robert Reenan and John Sedivy for helpful discussions. This workwas supported in part by National Institute on Aging (NIA) T32 TrainingGrant AG41688 and Fellowship Award AG47736 (to B.C.J.); NIA GrantsAG16667 and AG24353, a Glenn/American Federation for Aging Research(AFAR) Breakthroughs in Gerontology award, NIH Program Project GrantAG51449, and Grant P30AI042853 from the Providence/Boston Center forAIDS Research (to S.L.H.); Ellison/AFAR Postdoctoral Fellowship awards (toJ.G.W. and N.J.); a Nathan Shock Center Pilot Grant (to J.G.W.); and NIHGrants AG28753 and AG50582 (to N.N.).

1. Wood JG, Helfand SL (2013) Chromatin structure and transposable elements in or-ganismal aging. Front Genet 4(December):274.

2. Feser J, Tyler J (2011) Chromatin structure as a mediator of aging. FEBS Lett 585(13):2041–2048.

3. Dang W, et al. (2009) Histone H4 lysine 16 acetylation regulates cellular lifespan.Nature 459(7248):802–807.

4. Benayoun BA, Pollina EA, Brunet A (2015) Epigenetic regulation of ageing: Linkingenvironmental inputs to genomic stability. Nat Rev Mol Cell Biol 16(10):593–610.

5. Wood JG, et al. (2010) Chromatin remodeling in the aging genome of Drosophila.Aging Cell 9(6):971–978.

6. Jiang N, et al. (2013) Dietary and genetic effects on age-related loss of gene silencingreveal epigenetic plasticity of chromatin repression during aging. Aging (Albany, NY)5(11):813–824.

7. De Cecco M, et al. (2013) Genomes of replicatively senescent cells undergo globalepigenetic changes leading to gene silencing and activation of transposable ele-ments. Aging Cell 12(2):247–256.

8. De Cecco M, et al. (2013) Transposable elements become active and mobile in thegenomes of aging mammalian somatic tissues. Aging (Albany, NY) 5(12):867–883.

9. Slotkin RK, Martienssen R (2007) Transposable elements and the epigenetic regula-tion of the genome. Nat Rev Genet 8(4):272–285.

10. Wang J, et al. (2011) Inhibition of activated pericentromeric SINE/Alu repeat transcription insenescent human adult stem cells reinstates self-renewal. Cell Cycle 10(17):3016–3030.

11. Ambati J, Fowler BJ (2012) Mechanisms of age-related macular degeneration. Neuron75(1):26–39.

12. Van Meter M, et al. (2014) SIRT6 represses LINE1 retrotransposons by ribosylatingKAP1 but this repression fails with stress and age. Nat Commun 5:5011.

13. Dennis S, Sheth U, Feldman JL, English KA, Priess JR (2012) C. elegans germ cells showtemperature and age-dependent expression of Cer1, a Gypsy/Ty3-related retro-transposon. PLoS Pathog 8(3):e1002591.

14. Maxwell PH, Burhans WC, Curcio MJ (2011) Retrotransposition is associated withgenome instability during chronological aging. Proc Natl Acad Sci USA 108(51):20376–20381.

15. Li W, et al. (2013) Activation of transposable elements during aging and neuronaldecline in Drosophila. Nat Neurosci 16(5):529–531.

16. Chen H, Zheng X, Xiao D, Zheng Y (2016) Age-associated de-repression of retro-transposons in the Drosophila fat body, its potential cause and consequence. AgingCell 15(3):542–552.

17. Holoch D, Moazed D (2015) RNA-mediated epigenetic regulation of gene expression.Nat Rev Genet 16(2):71–84.

18. Schotta G, et al. (2002) Central role of Drosophila SU(VAR)3-9 in histone H3-K9methylation and heterochromatic gene silencing. EMBO J 21(5):1121–1131.

19. Ghildiyal M, Zamore PD (2009) Small silencing RNAs: An expanding universe. Nat RevGenet 10(2):94–108.

20. Castel SE, Martienssen RA (2013) RNA interference in the nucleus: Roles for smallRNAs in transcription, epigenetics and beyond. Nat Rev Genet 14(2):100–112.

21. Savva YA, et al. (2013) RNA editing regulates transposon-mediated heterochromaticgene silencing. Nat Commun 4:2745.

22. Rosenberg MI, Parkhurst SM (2002) Drosophila Sir2 is required for heterochromaticsilencing and by euchromatic Hairy/E(Spl) bHLH repressors in segmentation and sexdetermination. Cell 109(4):447–458.

23. Oberdoerffer P, et al. (2008) SIRT1 redistribution on chromatin promotes genomicstability but alters gene expression during aging. Cell 135(5):907–918.

24. Lim DH, et al. (2011) The endogenous siRNA pathway in Drosophila impacts stressresistance and lifespan by regulating metabolic homeostasis. FEBS Lett 585(19):3079–3085.

25. Whitaker R, et al. (2013) Increased expression of Drosophila Sir2 extends life span in adose-dependent manner. Aging (Albany, NY) 5(9):682–691.

26. Viswanathan M, Guarente L (2011) Regulation of Caenorhabditis elegans lifespan bysir-2.1 transgenes. Nature 477(7365):E1–E2.

27. Burnett C, et al. (2011) Absence of effects of Sir2 overexpression on lifespan inC. elegans and Drosophila. Nature 477(7365):482–485.

28. Hoffmann J, Romey R, Fink C, Yong L, Roeder T (2013) Overexpression of Sir2 in theadult fat body is sufficient to extend lifespan of male and female Drosophila. Aging(Albany, NY) 5(4):315–327.

29. Riddle NC, et al. (2011) Plasticity in patterns of histone modifications and chromo-somal proteins in Drosophila heterochromatin. Genome Res 21(2):147–163.

30. Charlesworth B, Langley CH (1989) The population genetics of Drosophila transpos-able elements. Annu Rev Genet 23:251–287.

31. Dej KJ, Gerasimova T, Corces VG, Boeke JD (1998) A hotspot for the Drosophila gypsyretroelement in the ovo locus. Nucleic Acids Res 26(17):4019–4025.

32. Ghildiyal M, et al. (2008) Endogenous siRNAs derived from transposons and mRNAs inDrosophila somatic cells. Science 320(5879):1077–1081.

33. Criscione SW, Zhang Y, Thompson W, Sedivy JM, Neretti N (2014) Transcriptional land-scape of repetitive elements in normal and cancer human cells. BMC Genomics 15(1):583.

34. Boeke J, et al. (2010) Phosphorylation of SU(VAR)3-9 by the chromosomal kinase JIL-1.PLoS One 5(4):e10042.

35. Yang JS, et al. (2011) OASIS: Online application for the survival analysis of lifespanassays performed in aging research. PLoS One 6(8):e23525.

11282 | www.pnas.org/cgi/doi/10.1073/pnas.1604621113 Wood et al.

Dow

nloa

ded

by g

uest

on

Aug

ust 3

, 202

0