+ All Categories
Home > Documents > Senescent intimal foam cells are deleterious at all stages of atherosclerosis · of atherosclerosis...

Senescent intimal foam cells are deleterious at all stages of atherosclerosis · of atherosclerosis...

Date post: 21-Jun-2020
Category:
Upload: others
View: 0 times
Download: 0 times
Share this document with a friend
7
(24) and by doing so would position the Xist- coated DNA away from the actively transcribed Xist transcription locus. This would enable other DNA regions on the X chromosome, which are physically linked to these tethered regions, to be brought into closer spatial proximity of the Xist transcription locus. In this way, Xist and its silencing factors can spread to these newly accessible DNA regions on the X chromosome. REFERENCES AND NOTES 1. R. Galupa, E. Heard, Curr. Opin. Genet. Dev. 31, 5766 (2015). 2. E. Splinter et al., Genes Dev. 25, 13711383 (2011). 3. S. S. Rao et al., Cell 159, 16651680 (2014). 4. A. Rego, P. B. Sinclair, W. Tao, I. Kireev, A. S. Belmont, J. Cell Sci. 121, 11191127 (2008). 5. A. Wutz, Nat. Rev. Genet. 12, 542553 (2011). 6. C. M. Clemson, L. L. Hall, M. Byron, J. McNeil, J. B. Lawrence, Proc. Natl. Acad. Sci. U.S.A. 103, 76887693 (2006). 7. J. M. Engreitz et al., Science 341, 1237973 (2013). 8. M. D. Simon et al., Nature 504, 465469 (2013). 9. J. Chaumeil, P. Le Baccon, A. Wutz, E. Heard, Genes Dev. 20, 22232237 (2006). 10. A. Wutz, T. P. Rasmussen, R. Jaenisch, Nat. Genet. 30, 167174 (2002). 11. C. A. McHugh et al., Nature 521, 232236 (2015). 12. C. Chu et al., Cell 161, 404416 (2015). 13. A. Minajigi et al., Science 349, aab2276 (2015). 14. Y. Gruenbaum, A. Margalit, R. D. Goldman, D. K. Shumaker, K. L. Wilson, Nat. Rev. Mol. Cell Biol. 6, 2131 (2005). 15. A. Pombo, N. Dillon, Nat. Rev. Mol. Cell Biol. 16, 245257 (2015). 16. J. Kind, B. van Steensel, Curr. Opin. Cell Biol. 22, 320325 (2010). 17. J. Baron-Benhamou, N. H. Gehring, A. E. Kulozik, M. W. Hentze, Methods Mol. Biol. 257, 135154 (2004). 18. A. Monfort et al., Cell Reports 12, 554561 (2015). 19. A. R. Buxbaum, G. Haimovich, R. H. Singer, Nat. Rev. Mol. Cell Biol. 16, 95109 (2015). 20. L. E. Finlan et al., PLOS Genet. 4, e1000039 (2008). 21. K. L. Reddy, J. M. Zullo, E. Bertolino, H. Singh, Nature 452, 243247 (2008). 22. B. Moindrot et al., Cell Rep. 10.1016/j.celrep.2015.06.053 (2015). 23. W. F. Marshall et al., Curr. Biol. 7, 930939 (1997). 24. J. R. Chubb, S. Boyle, P. Perry, W. A. Bickmore, Curr. Biol. 12, 439445 (2002). ACKNOWLEDGMENTS We thank K. Plath for extensive discussions; A. Collazo for microscopy help; A. Shur, P. Quintero, and V. Grishkevich for technical help; M. Lai for analytical help; J. Engreitz, S. Quinodoz, M. Garber, I. Amit, and J. Rinn for comments on the manuscript; and S. Knemeyer for illustrations. Imaging was performed in the Biological Imaging Facility, and sequencing was performed in the Millard and Muriel Jacobs Genetics and Genomics Laboratory at the California Institute of Technology. C.-K.C. is supported by a NIH National Research Service Award training grant (T32GM07616). This research was funded by the New York Stem Cell Foundation, a NIH Directors Early Independence Award (DP5OD012190), the Edward Mallinckrodt Foundation, Sontag Foundation, Searle Scholars Program, Pew-Steward Scholars program, and funds from the California Institute of Technology. M.G. is a New York Stem Cell FoundationRobertson Investigator. Sequencing data are available online from the National Center for Biotechnology Information Gene Expression Omnibus (www.ncbi.nlm.nih.gov/geo) accession no. GSE80510 (RAP data) and GSE86250 (CLIP data), and additional data and information are available at www.lncRNA.caltech.edu/data.php. SUPPLEMENTARY MATERIALS www.sciencemag.org/content/354/6311/468/suppl/DC1 Materials and Methods Supplementary Text Figs. S1 to S19 References (2549) 5 December 2015; accepted 25 July 2016 Published online 4 August 2016 10.1126/science.aae0047 VASCULAR DISEASE Senescent intimal foam cells are deleterious at all stages of atherosclerosis Bennett G. Childs, 1 Darren J. Baker, 2 Tobias Wijshake, 2,3 Cheryl A. Conover, 4 Judith Campisi, 5,6 Jan M. van Deursen 1,2 * Advanced atherosclerotic lesions contain senescent cells, but the role of these cells in atherogenesis remains unclear. Using transgenic and pharmacological approaches to eliminate senescent cells in atherosclerosis-prone low-density lipoprotein receptordeficient (Ldlr /) mice, we show that these cells are detrimental throughout disease pathogenesis. We find that foamy macrophages with senescence markers accumulate in the subendothelial space at the onset of atherosclerosis, where they drive pathology by increasing expression of key atherogenic and inflammatory cytokines and chemokines. In advanced lesions, senescent cells promote features of plaque instability, including elastic fiber degradation and fibrous cap thinning, by heightening metalloprotease production.Together, these results demonstrate that senescent cells are key drivers of atheroma formation and maturation and suggest that selective clearance of these cells by senolytic agents holds promise for the treatment of atherosclerosis. A therosclerosis initiates when oxidized lipo- protein infiltrates the subendothelial space of arteries, often due to aberrantly elevated levels of apolipoprotein Bcontaining lipo- proteins in the blood ( 1 ). Chemotactic signals arising from activated endothelium and vascu- lar smooth muscle attract circulating monocytes that develop into lipid-loaded foamy macrophages, a subset of which adopt a proinflammatory pheno- type through a mechanism that is not fully un- derstood. The proinflammatory signals lead to additional rounds of monocyte recruitment and accumulation of other inflammatory cells (in- cluding T and B cells, dendritic cells, and mast cells), allowing initial lesions, often termed fatty streaks,to increase in size and develop into plaques (2). Plaque stability, rather than abso- lute size, determines whether atherosclerosis is clinically silent or pathogenic because un- stable plaques can rupture and produce vessel- occluding thrombosis and end-organ damage. Stable plaques have a relatively thick fibrous cap, which largely consists of vascular smooth muscle cells (VSMCs) and extracellular matrix components, partitioning soluble clotting fac- tors in the blood from thrombogenic molecules in the plaque (3). In advanced disease, plaques destabilize when elevated local matrix metallo- protease production degrades the fibrous cap, increasing the risk of lesion rupture and subse- quent thrombosis. Advanced plaques contain cells with markers of senescence, a stress response that entails a per- manent growth arrest coupled to the robust se- cretion of numerous biologically active molecules and is referred to as the senescence-associated secretory phenotype (SASP). The senescence markers include elevated senescence-associated b-galactosidase (SA b-Gal) activity and p16 Ink4a , p53, and p21 expression (4, 5). However, whether and how senescent cells contribute to athero- genesis remains unclear (6, 7). Human plaques contain cells with shortened telomeres, which predispose cells to undergo senescence (8). Con- sistent with a proatherogenic role of senescence is the observation that expression of a loss-of- function telomere-binding protein (Trf2) in VSMCs accelerates plaque growth in the ApoE /mouse model of atherosclerosis, although in vivo evi- dence for increased senescence in plaques was not provided. On the other hand, mice lacking core components of senescence pathways, such as p53, p21, or p19 Arf (7, 911), show accelerated atherosclerosis, implying a protective role for se- nescence. Studies showing that human and mouse polymorphisms that reduce expression of p16 Ink4a and p14 Arf (p19 Arf in mice) correlate with in- creased atheroma risk support this conclusion (7, 12, 13). Thus, whether senescent cells accel- erate or retard atherogenesis is unclear. We used genetic and pharmacological meth- ods of eliminating senescent cells to examine the role of naturally occurring senescent cells at dif- ferent stages of atherogenesis. First, we verified that senescent cells accumulate in low-density lipoprotein receptordeficient (Ldlr /) mice, a model of atherogenesis. We fed 10-week-old Ldlr /mice a high-fat diet (HFD) for 88 days. We then performed SA b-Gal staining, which 472 28 OCTOBER 2016 VOL 354 ISSUE 6311 sciencemag.org SCIENCE 1 Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN 55905, USA. 2 Department of Pediatric and Adolescent Medicine, Mayo Clinic, Rochester, MN 55905, USA. 3 Department of Pediatrics, University of Groningen, University Medical Center Groningen, 9713 AV Groningen, Netherlands. 4 Division of Endocrinology, Metabolism, and Nutrition, Mayo Clinic, Rochester, MN 55905, USA. 5 Buck Institute for Research on Aging, Novato, CA 94945, USA. 6 Life Sciences Division, Lawrence Berkeley National Laboratory, Berkeley, CA 94720, USA. *Corresponding author. Email: [email protected] RESEARCH | REPORTS on June 30, 2020 http://science.sciencemag.org/ Downloaded from
Transcript
Page 1: Senescent intimal foam cells are deleterious at all stages of atherosclerosis · of atherosclerosis Bennett G. Childs,1 Darren J. Baker,2 Tobias Wijshake,2,3 Cheryl A. Conover,4 Judith

(24) and by doing so would position the Xist-coated DNA away from the actively transcribedXist transcription locus. This would enable otherDNA regions on the X chromosome, which arephysically linked to these tethered regions, tobe brought into closer spatial proximity of theXist transcription locus. In this way, Xist and itssilencing factors can spread to these newlyaccessible DNA regions on the X chromosome.

REFERENCES AND NOTES

1. R. Galupa, E. Heard, Curr. Opin. Genet. Dev. 31, 57–66 (2015).2. E. Splinter et al., Genes Dev. 25, 1371–1383 (2011).3. S. S. Rao et al., Cell 159, 1665–1680 (2014).4. A. Rego, P. B. Sinclair, W. Tao, I. Kireev, A. S. Belmont, J. Cell

Sci. 121, 1119–1127 (2008).5. A. Wutz, Nat. Rev. Genet. 12, 542–553 (2011).6. C. M. Clemson, L. L. Hall, M. Byron, J. McNeil, J. B. Lawrence,

Proc. Natl. Acad. Sci. U.S.A. 103, 7688–7693 (2006).7. J. M. Engreitz et al., Science 341, 1237973 (2013).8. M. D. Simon et al., Nature 504, 465–469 (2013).9. J. Chaumeil, P. Le Baccon, A. Wutz, E. Heard, Genes Dev. 20,

2223–2237 (2006).10. A. Wutz, T. P. Rasmussen, R. Jaenisch, Nat. Genet. 30, 167–174

(2002).11. C. A. McHugh et al., Nature 521, 232–236 (2015).12. C. Chu et al., Cell 161, 404–416 (2015).13. A. Minajigi et al., Science 349, aab2276 (2015).14. Y. Gruenbaum, A. Margalit, R. D. Goldman, D. K. Shumaker,

K. L. Wilson, Nat. Rev. Mol. Cell Biol. 6, 21–31 (2005).15. A. Pombo, N. Dillon, Nat. Rev. Mol. Cell Biol. 16, 245–257

(2015).16. J. Kind, B. van Steensel, Curr. Opin. Cell Biol. 22, 320–325 (2010).17. J. Baron-Benhamou, N. H. Gehring, A. E. Kulozik, M. W. Hentze,

Methods Mol. Biol. 257, 135–154 (2004).18. A. Monfort et al., Cell Reports 12, 554–561 (2015).19. A. R. Buxbaum, G. Haimovich, R. H. Singer, Nat. Rev. Mol. Cell

Biol. 16, 95–109 (2015).20. L. E. Finlan et al., PLOS Genet. 4, e1000039 (2008).21. K. L. Reddy, J. M. Zullo, E. Bertolino, H. Singh, Nature 452,

243–247 (2008).22. B. Moindrot et al., Cell Rep. 10.1016/j.celrep.2015.06.053

(2015).23. W. F. Marshall et al., Curr. Biol. 7, 930–939 (1997).24. J. R. Chubb, S. Boyle, P. Perry, W. A. Bickmore, Curr. Biol. 12,

439–445 (2002).

ACKNOWLEDGMENTS

We thank K. Plath for extensive discussions; A. Collazo formicroscopy help; A. Shur, P. Quintero, and V. Grishkevich fortechnical help; M. Lai for analytical help; J. Engreitz, S. Quinodoz,M. Garber, I. Amit, and J. Rinn for comments on the manuscript;and S. Knemeyer for illustrations. Imaging was performedin the Biological Imaging Facility, and sequencing was performed inthe Millard and Muriel Jacobs Genetics and Genomics Laboratoryat the California Institute of Technology. C.-K.C. is supported by aNIH National Research Service Award training grant(T32GM07616). This research was funded by the New York StemCell Foundation, a NIH Director’s Early Independence Award(DP5OD012190), the Edward Mallinckrodt Foundation,Sontag Foundation, Searle Scholars Program, Pew-StewardScholars program, and funds from the California Institute ofTechnology. M.G. is a New York Stem Cell Foundation–RobertsonInvestigator. Sequencing data are available online from theNational Center for Biotechnology Information Gene ExpressionOmnibus (www.ncbi.nlm.nih.gov/geo) accession no. GSE80510(RAP data) and GSE86250 (CLIP data), and additional data andinformation are available at www.lncRNA.caltech.edu/data.php.

SUPPLEMENTARY MATERIALS

www.sciencemag.org/content/354/6311/468/suppl/DC1Materials and MethodsSupplementary TextFigs. S1 to S19References (25–49)

5 December 2015; accepted 25 July 2016Published online 4 August 201610.1126/science.aae0047

VASCULAR DISEASE

Senescent intimal foam cellsare deleterious at all stagesof atherosclerosisBennett G. Childs,1 Darren J. Baker,2 Tobias Wijshake,2,3 Cheryl A. Conover,4

Judith Campisi,5,6 Jan M. van Deursen1,2*

Advanced atherosclerotic lesions contain senescent cells, but the role of these cellsin atherogenesis remains unclear. Using transgenic and pharmacological approachesto eliminate senescent cells in atherosclerosis-prone low-density lipoproteinreceptor–deficient (Ldlr–/–) mice, we show that these cells are detrimental throughoutdisease pathogenesis. We find that foamy macrophages with senescence markersaccumulate in the subendothelial space at the onset of atherosclerosis, where they drivepathology by increasing expression of key atherogenic and inflammatory cytokinesand chemokines. In advanced lesions, senescent cells promote features of plaqueinstability, including elastic fiber degradation and fibrous cap thinning, by heighteningmetalloprotease production. Together, these results demonstrate that senescent cells arekey drivers of atheroma formation and maturation and suggest that selective clearanceof these cells by senolytic agents holds promise for the treatment of atherosclerosis.

Atherosclerosis initiates when oxidized lipo-protein infiltrates the subendothelial spaceof arteries, often due to aberrantly elevatedlevels of apolipoprotein B–containing lipo-proteins in the blood (1). Chemotactic signals

arising from activated endothelium and vascu-lar smooth muscle attract circulating monocytesthat develop into lipid-loaded foamy macrophages,a subset of which adopt a proinflammatory pheno-type through a mechanism that is not fully un-derstood. The proinflammatory signals lead toadditional rounds of monocyte recruitment andaccumulation of other inflammatory cells (in-cluding T and B cells, dendritic cells, and mastcells), allowing initial lesions, often termed “fattystreaks,” to increase in size and develop intoplaques (2). Plaque stability, rather than abso-lute size, determines whether atherosclerosisis clinically silent or pathogenic because un-stable plaques can rupture and produce vessel-occluding thrombosis and end-organ damage.Stable plaques have a relatively thick fibrouscap, which largely consists of vascular smoothmuscle cells (VSMCs) and extracellular matrixcomponents, partitioning soluble clotting fac-tors in the blood from thrombogenic moleculesin the plaque (3). In advanced disease, plaquesdestabilize when elevated local matrix metallo-protease production degrades the fibrous cap,

increasing the risk of lesion rupture and subse-quent thrombosis.Advanced plaques contain cells with markers

of senescence, a stress response that entails a per-manent growth arrest coupled to the robust se-cretion of numerous biologically active moleculesand is referred to as the senescence-associatedsecretory phenotype (SASP). The senescencemarkers include elevated senescence-associatedb-galactosidase (SA b-Gal) activity and p16Ink4a,p53, and p21 expression (4, 5). However, whetherand how senescent cells contribute to athero-genesis remains unclear (6, 7). Human plaquescontain cells with shortened telomeres, whichpredispose cells to undergo senescence (8). Con-sistent with a proatherogenic role of senescenceis the observation that expression of a loss-of-function telomere-binding protein (Trf2) in VSMCsaccelerates plaque growth in the ApoE–/– mousemodel of atherosclerosis, although in vivo evi-dence for increased senescence in plaques wasnot provided. On the other hand, mice lackingcore components of senescence pathways, suchas p53, p21, or p19Arf (7, 9–11), show acceleratedatherosclerosis, implying a protective role for se-nescence. Studies showing that human and mousepolymorphisms that reduce expression of p16Ink4a

and p14Arf (p19Arf in mice) correlate with in-creased atheroma risk support this conclusion(7, 12, 13). Thus, whether senescent cells accel-erate or retard atherogenesis is unclear.We used genetic and pharmacological meth-

ods of eliminating senescent cells to examine therole of naturally occurring senescent cells at dif-ferent stages of atherogenesis. First, we verifiedthat senescent cells accumulate in low-densitylipoprotein receptor–deficient (Ldlr–/–) mice,a model of atherogenesis. We fed 10-week-oldLdlr–/– mice a high-fat diet (HFD) for 88 days.We then performed SA b-Gal staining, which

472 28 OCTOBER 2016 • VOL 354 ISSUE 6311 sciencemag.org SCIENCE

1Department of Biochemistry and Molecular Biology, MayoClinic, Rochester, MN 55905, USA. 2Department of Pediatricand Adolescent Medicine, Mayo Clinic, Rochester, MN 55905,USA. 3Department of Pediatrics, University of Groningen,University Medical Center Groningen, 9713 AV Groningen,Netherlands. 4Division of Endocrinology, Metabolism, andNutrition, Mayo Clinic, Rochester, MN 55905, USA. 5BuckInstitute for Research on Aging, Novato, CA 94945, USA.6Life Sciences Division, Lawrence Berkeley NationalLaboratory, Berkeley, CA 94720, USA.*Corresponding author. Email: [email protected]

RESEARCH | REPORTSon June 30, 2020

http://science.sciencemag.org/

Dow

nloaded from

Page 2: Senescent intimal foam cells are deleterious at all stages of atherosclerosis · of atherosclerosis Bennett G. Childs,1 Darren J. Baker,2 Tobias Wijshake,2,3 Cheryl A. Conover,4 Judith

revealed that senescent cells are present inatherosclerotic lesions but not in the normaladjacent vasculature or aortas of Ldlr–/– micefed a low-fat diet (LFD) (fig. S1A). In addition,plaque-rich aortic arches had elevated transcriptlevels of p16Ink4a, p19Arf, and various canonicalSASP components, including the matrix metallo-proteases Mmp3 and Mmp13 and the inflam-matory cytokines Il1a and Tnfa (fig. S1B). Toeliminate senescent cells from plaques, we usedp16-3MR (3MR) mice (14), a transgenic modelthat expresses the herpes simplex virus thymidinekinase under the control of the p16Ink4a genepromoter and kills p16Ink4a+ senescent cells uponadministration of ganciclovir (GCV). Plaques ofLdlr–/–;3MR mice fed a HFD for 88 days andthen treated short term with GCV had low levelsof SA b-Gal activity compared with Ldlr–/– mice

(fig. S1C), indicating efficient clearance of senes-cent cells. Examination of the plaques by trans-mission electron microscopy (TEM) revealedthat three morphologically distinct cell types—elongated, vacuolated cells located in the endo-thelial layer; spindly foam cells with histologicalproperties of VSMCs; and large foamy cells re-sembling lipid-loaded macrophages—producedX-galactosidase (X-Gal) crystals (Fig. 1A). We referto these cells as endothelial-like, foamy VSMC–like, and foamy macrophage–like cells, respec-tively, because cells within plaques change shapeand lineage markers, precluding accurate assess-ment of cell origin (15, 16). All three senescent celltypes were efficiently eliminated by treatmentwith GCV (Fig. 1A).To assess the impact senescent cells have

on plaque development, we placed 10-week-old

Ldlr–/–;3MR mice on a HFD for 88 days andsimultaneously treated them with GCV or ve-hicle during this period (Fig. 1B) to intermittent-ly remove p16Ink4a+ cells. To control for potentialeffects of GCV, independent of 3MR expression,we also examined GCV-treated Ldlr–/– mice ona HFD. En face staining of descending aortaswith Sudan IV revealed that plaque burdenwas ~60% lower in GCV-treated Ldlr–/–;3MRmice than in vehicle-treated Ldlr–/–;3MR or GCV-treated Ldlr–/– mice, owing to decreases in bothplaque number and size (Fig. 1C). Similarly, GCV-treated Ldlr–/–;3MRmice showed reduced plaqueburden and destruction of aortic elastic fibersbeneath the neointima in the brachiocephalicartery (fig. S2, A to C), a site that rapidly de-velops advanced atherosclerotic plaques (17).Compared with vehicle-treated Ldlr–/–;3MRmice,

SCIENCE sciencemag.org 28 OCTOBER 2016 • VOL 354 ISSUE 6311 473

X-Gal + cell types in atherosclerotic plaques in Ldlr –/–

Ldlr–/–;;3MR, HFD +GCV (n=4)

X-g

al+

cells

in p

laq

ues

(%

)

Ldlr–/–, HFD +GCV (n=3) Endothelium-like cell Foamy VSMC-like cell Foamy macrophage-like cell

0

20

40

60

80

100

120

140

160

180

0

1

2

3

4

5

6

0

5

10

15

20

25

30

Sudan IV

Ldlr–/–;3MR

HFD + GCV

HFD +Veh

Ldlr –/–

HFD +GCV

***

Ldlr–/–, HFD +GCV (n =9) Ldlr–/–;3MR, HFD +Veh (n = 6)

Pla

qu

es p

er d

esce

nd

ing

ao

rta

Ave

rag

e p

laq

ue

size

x10

3 (µ

m2 )

*** *

***

Ldlr–/–;3MR, HFD +GCV (n = 10)

21 70 158

LFD HFD + Veh

LFD HFD

+GCV

Age (days)

* *

Abd

omin

al a

orta

T

hora

cic

aort

a

Su

dan

IV-p

osi

tive

are

a (%

)

0

5

10

15

20

25

"! #! $!

** ***

**

VSMC -like

Mac -like

EC -like

LFD HFD

+GCV

Ldlr –/–:

Ldlr–/–;3MR:

Fig. 1. p16Ink4a+ senescent cells drive formation of atherosclerotic plaques.(A) (Left) X-Gal electron microscopy images showing three types of senescentcells in plaques of Ldlr–/– mice on a HFD for 88 days. Cell outlines are tracedby dashed yellow lines. Endothelial-like cells are elongated and adjacent tothe lumen. VSMC-like cells are elongated spindle-shaped cells or irregularlyshaped ramified cells. Macrophage-like cells are highly vacuolated circular cells.(Right) Senescent cell quantification in plaques. Mac, macrophage; EC, endo-

thelial cell. (B) (Left) Experimental design for testing the effect of senescent cellclearance on atherogenesis. Veh, Vehicle. (Right) Sudan IV–stained descendingaortas (not including the aortic arch). (C) Quantification of total descending aortaplaque burden, number, and lesion size. Scale bars: 2 mm (A); 500 nm [(A), insets].Data represent mean ± SEM (error bars) [biological n is indicated on graphsand refers to individual plaques in (A) (one per mouse) and aortas in (C)]. *P <0.05; **P < 0.01; ***P < 0.001 (unpaired two-tailed t tests with Welch’s correction).

RESEARCH | REPORTSon June 30, 2020

http://science.sciencemag.org/

Dow

nloaded from

Page 3: Senescent intimal foam cells are deleterious at all stages of atherosclerosis · of atherosclerosis Bennett G. Childs,1 Darren J. Baker,2 Tobias Wijshake,2,3 Cheryl A. Conover,4 Judith

GCV-treated Ldlr–/–;3MR mice expressed loweramounts of p16Ink4a mRNA and other senescence-marker mRNAs in aortic arches, confirming thatp16Ink4a+ senescent cells were efficiently clearedby GCV (fig. S3A). Expression of 3MR, as measuredby reverse transcription quantitative polymerasechain reaction (RT-qPCR) analysis of monomericred fluorescent protein transcripts, increased inHFD-fed mice but remained at baseline levelswith GCV treatment. Complementary en faceSA b-Gal staining of aortas confirmed thatp16Ink4a+ senescent cells were effectively cleared(fig. S3B). GCV-treatment of Ldlr–/– mice didnot alter SA b-Gal staining or other senescencemarkers (fig. S3, B and C). GCV-treated Ldlr–/–

and Ldlr–/–;3MR mice did not differ in bodyweight, fat mass, and fat deposit weight (fig. S4,A to D). Circulating monocytes, lymphocytes,platelets, and neutrophils, all of which are involvedin atherogenesis, were unaffected (fig. S4, E to H).Levels of atherogenic lipids in the blood of GCV-

treated Ldlr–/– and Ldlr–/–;3MR mice and vehicle-treated Ldlr–/–;3MR mice were highly elevatedcompared with levels in LFD-fed controls, withno differences between the distinct HFD-fedcohorts (fig. S4I). Thus, the atheroprotective ef-fect in GCV-treated Ldlr–/–;3MR mice is due tothe killing of p16Ink4a+ senescent cells ratherthan changes in feeding habits, blood lipids, orcirculating immunocytes. Reductions in plaqueburden, number, and size observed with p16-3MRwere reproducible with two independent trans-genic systems [INK-ATTAC (fig. S5) (18, 19) andINK-Nitroreductase (NTR) (fig. S6)] designedto kill p16Ink4a+ senescent cells through distinctmechanisms, as well as with the senolytic drugABT263, which inhibits the anti-apoptotic pro-teins Bcl-2 and Bcl-xL and selectively kills senes-cent cells (fig. S7) (20).To investigate how senescent cells drive plaque

initiation and growth, we focused on atherogen-esis onset at lesion-prone sites of the vascula-

ture (21). After just 9 days on an atherogenic diet,Ldlr–/– mice had overtly detectable fatty-streaklesions solely in the inner curvature of the aorticarch (Fig. 2A and fig. S8A). Surprisingly, theseearly lesions were highly positive for SA b-Galactivity (Fig. 2A). By contrast, Ldlr–/– mice con-taining 3MR and treated daily with GCV duringthe 9-day HFD-feeding period had low levels ofSA b-Gal activity and much smaller fatty streaks(Fig. 2A). Histological examination by TEM ofthe SA b-Gal–stained samples revealed that fattystreaks of HFD-fed Ldlr−/− mice consisted of fociof foam cell macrophages arranged in mono- ormultilayers (Fig. 2, B and C). The lesions hadintact elastic fibers and no fibrous cap. X-Galcrystals were detectable exclusively in foam cellmacrophages, irrespective of lesion size (Fig. 2,B and D). Foam cell macrophages in foci of 9-daylesions of Ldlr–/–;3MR mice receiving daily, high-dose GCV were rarely arranged in multilayers andhad a much lower incidence of crystals (Fig. 2, C

474 28 OCTOBER 2016 • VOL 354 ISSUE 6311 sciencemag.org SCIENCE

Fig. 2. Intimal senescent foamymacrophages form duringearly atherogenesis and fosterproduction of proatherogenicfactors. (A) (Left) Schematic ofthe inner curvature of the aorticarch. LV, left ventricle. (Middle)Examples of SA b-Gal–stained9-day fatty streaks with andwithout senescent cell clearanceand quantification. (Right)Measurements of streak size.Treatment involved the adminis-tration of 25 mg/kg of GCV oncedaily. BCA, brachiocephalicartery. (B) TEM images of Ldlr–/–

mice after 9-day HFD feeding,showing fatty-streak foci withX-Gal–positive foam cellmacrophages (artificial coloringarticulates cell boundaries inthe multilayer). (C) Quantificationof multilayer foci in day-9 fattystreaks with and without senes-cent cells. (D) Quantification offoam cell macrophages with X-Galcrystal–containing vesicles with-out and with clearance. (E) (Left)Representative SA b-Gal–stained12-day fatty streaks without andwith GCV treatment (25 mg/kgof GCV three times daily) for thelast 3 days. (Right) Quantificationof lesion burden. (F) RT-qPCRanalysis of senescence markerexpression in fatty streaks collectedfrom Ldlr–/– and Ldlr–/–;3MRmice on a 12-day HFD and treatedwith GCV for the last 3 days.Scale bars: 1 mm (A) and (E);2 mm (B); and 500 nm [(B), insets].Bar graphs represent mean ± SEM(error bars) [biological n is indicated directly on all graphs and refers to individual aortic arches in (A) and (C) to (E) or dissected aortic arch innercurvatures in (F)]. *P < 0.05; **P < 0.01; ***P < 0.001 (unpaired two-tailed t tests with Welch’s correction).

0

0.2

0.4

0.6

0.8

1

1.2

1.4

&'()% **+(,%

0

0.2

0.4

0.6

0.8

1

1.2

0

10

20

30

40

50

60

70

0

0.5

1

1.5

2

2.5

Str

eak

size

(%

of

arch

su

rfac

e)

Ldlr–/– +GCV

Ldlr–/–;3MR +GCV

***

n =

6n

= 7

Inner curvature

BCA Ldlr–/–;3MR + GCV Ldlr–/– + GCV

SA- -GAL stained aortic arches after 9-day HFD

LV

Inner curvature

Ldlr–/–;3MR + GCV

Ldlr–/– + GCV

0

0.4

0.8

1.2

1.6

2

n =

3n

= 3

*

X-G

al+

foam

y m

acro

ph

ages

(%

)

**

Mu

ltila

yer

foci

per

sec

tio

n

n =

6 n =

7

*

Multilayer fatty-streak lesion

Fo

ld c

han

ge

in in

ner

cu

rvat

ure

p16 Vcam1 Mcp1 Tnfα Il1α Mmp12 Mmp13

** * *

*** * ***

n =

6n

= 6

n =

6n

= 7

Fatty streak

Monolayer fatty-streak lesion

Str

eak

size

(%

of

arch

su

rfac

e)

β

Ink4a

RESEARCH | REPORTSon June 30, 2020

http://science.sciencemag.org/

Dow

nloaded from

Page 4: Senescent intimal foam cells are deleterious at all stages of atherosclerosis · of atherosclerosis Bennett G. Childs,1 Darren J. Baker,2 Tobias Wijshake,2,3 Cheryl A. Conover,4 Judith

and D). Elevated SA b-Gal activity in fatty streakscorrelated with increased levels of p16Ink4a andvarious other senescence markers, includingMmp3, Mmp13, Il1a, and Tnfa (fig. S8B). Nine-day treatment of HFD-fed Ldlr–/– mice withABT263 confirmed that senolysis reduces ath-erogenesis onset (fig. S8C).To determine how senescent foamy macro-

phages contribute to early atherogenesis, weestablished 9-day fatty streaks in Ldlr–/– andLdlr–/–;3MR mice and then administered high-dose GCV for 3 days while continuing to feed themice a HFD. Short-term clearance of senescentcells markedly reduced streak size and SA b-Galpositivity (Fig. 2E). TEM images showed thatcleared foci were drastically remodeled, withacellular debris retained in the subendotheliumand few foamy macrophages containing X-Galcrystals (fig. S8, D and E). RT-qPCR revealed astark reduction in p16Ink4a and SASP components(including Mmp3, Mmp13, Il1a, and Tnfa), aswell as two key molecular drivers of monocyterecruitment (the chemokine Mcp1 and the leuko-cyte receptor Vcam1), whose expression is partlydriven by Tnfa (Fig. 2F). These data suggestthat subendothelial senescent foamy macro-phages arising in early lesions enhance Tnfa-mediated Vcam1 expression, as well as theMcp1gradient, to perpetuate monocyte recruitmentfrom the blood.

Next, we examined the role of senescent cellsin the maturation of benign plaques to complexadvanced lesions. Although mouse models foratherosclerosis do not develop clinical symp-toms associated with plaque ruptures, plaquematuration in these mice can be studied usingsurrogate markers of plaque instability, includ-ing fibrous cap thinning (22, 23), decreased col-lagen deposition, elastic fiber degradation, andplaque calcification (24). To assess the effect ofsenescent cell clearance on the maturation ofexisting plaques, we employed late-disease senes-cent cell clearance protocols in which we placedLdlr/– and Ldlr –/–;3MRmice on a HFD for 88 daysto create plaques, followed by 100 days of GCVtreatment. During GCV treatment, mice werefed a HFD or LFD to promote continued plaqueadvancement or stasis, respectively (Fig. 3A andfig. S9). Ldlr–/–;3MR mice maintained on theHFD and receiving GCV showed attenuated dis-ease progression, as evidenced by a lower plaquenumber and size compared with GCV-treatedLdlr–/– or vehicle-treated Ldlr–/–;3MR controls (Fig.3A). Whereas plaques of GCV-treated Ldlr–/–;3MRmice on a LFD had markedly reduced Sudan IVstaining compared with plaques of control mice,the lesion-covered aortic area did not change(fig. S10A), even though 3MR-mediated senescentcell killing was confirmed by SA b-Gal staining(fig. S10B) and RT-qPCR for senescence markers

(Fig. 3B). Regardless of diet, senescent cell clear-ance reduced expression of inflammatory cytokines(Fig. 3B) and monocyte recruitment factors (fig.S10C). GCV treatment decreased expressionof matrix metalloproteases linked to plaque de-stabilization, includingMmp3, Mmp12, andMmp13(25, 26) (Fig. 3B), which suggests that senescentcell elimination stabilizes the fibrous cap.To investigate this and other features of plaque

maturation, we conducted histopathology onplaques collected from the above cohorts. WhenLdlr–/– mice originally fed a HFD for 88 dayswere left on the diet for an additional 100 days,their descending aorta plaques showed reducedcap thickness, diminished collagen content (byMasson’s trichrome staining), and more disrupt-ed aortic elastic fibers (by Voerhoff von Giesonstaining) in comparison with those of LFD-fedmice (Fig. 4, A and B, and fig. S11). In contrast,all of these markers of plaque instability weredecreased with clearance of p16Ink4a+ cells, re-gardless of diet, during the 100-day GCV treat-ment period. Similarly, clearance of p16Ink4a+ cellsincreased cap thickness and collagen contentin brachiocephalic arteries from mice revertedto LFD (fig. S12). We extended these studies byswitching Ldlr–/–;3MR and Ldlr–/– mice after88 days of HFD feeding to a LFD with GCV injec-tions for 35 days (fig. S13A). In this experiment,senescent cell elimination preserved fibrous cap

SCIENCE sciencemag.org 28 OCTOBER 2016 • VOL 354 ISSUE 6311 475

Fig. 3. Removal of p16Ink4a+

cells in established plaquesperturbs the proatherogenicmicroenvironment. (A) (Left)Experimental design. (Middle)Sudan IV–stained descendingaortas (not including the arch).(Right) Quantification of SudanIV+ areas and plaque number.(B) RT-qPCR for senescencemarkers in aortic arches fromindicated cohorts. Aortic archesfrom Ldlr–/–;3MR females fed aLFD until 258 days of age andtreated with Veh for the last100 days were used to assessbaseline expression levels.Treatments in (A) and (B)involved daily injections of5 mg/kg of GCV (or Veh) for5 days, followed by 14 daysoff; this cycle was repeated for100 days. Bar graphs representmean ± SEM (error bars)[biological n is indicated directlyon all graphs and refers to indi-vidual aortas in (A) or aorticarches in (B)]. *P < 0.05; **P <0.01; ***P < 0.001 [analysis ofvariance (ANOVA) with Sidak’spost-hoc correction for family-wise error in (A) and unpairedtwo-tailed t test with Welch’scorrection in (B)].

0

2

4

6

8

10

12

14

16

!" #" $" %" &" '" (" )" *" !+"

0

5

10

15

20

25

HFD +Veh

0

10

20

30

40

50

Ldlr–/–, HFD +GCV (n = 6)

Ldlr –/–; 3MR, HFD +Veh (n = 6)

Su

dan

IV+

area

(%

of

aort

a)

** **

Ldlr –/–; 3MR, HFD +GCV (n = 6)

Pla

qu

e n

um

ber

* **

21

LFD

70 158

HFD

Age (days)

HFD +GCV

258

Sudan IV HFD +GCV

HFD + GCV

HFD + Veh

Ldlr–/–;3MR Ldlr –/–

Abd

omin

al

Tho

raci

c

LFD HFD

Ldlr –/–;3MR, LFD throughout +Veh (last 100 days) (n = 3 mice) Ldlr –/–, LFD +GCV (n = 5 mice) Ldlr –/–;3MR, LFD +GCV (n = 3 mice) Ldlr –/–, HFD +GCV (n = 5 mice) Ldlr –/–;3MR, HFD +GCV (n = 4 mice)

*** ** * *** ** ** *

*** *** *** **

Fo

ld-c

han

ge

in a

ort

ic a

rch

p16 Ink4a p19 Arf Il1α

**

Mmp3 Mmp2 Mmp13 Mmp12 Mmp9 Mmp1

** ** *

* *

** **

Tnfα

*** *

**

**

Ldlr–/–;3MR:

Ldlr–/–: HFD

+GCV

RESEARCH | REPORTSon June 30, 2020

http://science.sciencemag.org/

Dow

nloaded from

Page 5: Senescent intimal foam cells are deleterious at all stages of atherosclerosis · of atherosclerosis Bennett G. Childs,1 Darren J. Baker,2 Tobias Wijshake,2,3 Cheryl A. Conover,4 Judith

thickness (fig. S13, B and C). Furthermore, le-sional foamy macrophage–like cell content wasreduced, whereas VSMC-like cell content increased(fig. S13, D and E), resulting in plaques with ahigher VSMC-like/macrophage-like cell ratio, amarker for greater stability (fig. S13F) (27). Clear-ance markedly reduced monocyte attachmentto plaque-adjacent endothelium (fig. S13G), thussupporting our conclusion from early fatty-streakexperiments that enhanced monocyte chemo-taxis may partially explain the proatherogenic

nature of senescent cells. These results stronglysuggest that eliminating p16Ink4a+ cells promotesplaque stability.To further investigate the mechanism by which

senescent cells drive atherogenesis, we tested thepossibility that senescent cells in plaques expressproatherogenic factors. We dissected lesion-bearing tissue from HFD-fed Ldlr–/–;ATTACmice,prepared single-cell suspensions, and exploitedp16Ink4a-dependent expression of green fluores-cent protein (GFP) by ATTAC to collect GFP+ se-

nescent and GFP– nonsenescent cell populationsfor analysis by RT-qPCR (Fig. 4C). Senescent cellsexpressed a broad spectrum of proatherogenicfactors, including Il1a, Tnfa, Mmp3, Mmp12,Mmp13, Mcp1, and Vcam1 (Fig. 4C). A subset ofthese factors (including Il1a,Mmp12,Mmp13, andMcp1) was expressed at markedly elevated levelscompared with expression in nonsenescent cells.Using both transgenic and pharmacological

approaches to clear p16Ink4a+ cells without inter-fering with the senescence program, we showed

476 28 OCTOBER 2016 • VOL 354 ISSUE 6311 sciencemag.org SCIENCE

0

5

10

15

20

25

30

0

5

10

15

20

25

30

!" #" $"

Mas

son’

s Tr

ichr

ome

H–E

Ldlr –/– Ldlr –/–;3MR

88 d HFD followed by 100 d LFD +GCV

Ldlr–/–, +GCV Ldlr –/–;3MR, +GCV

Fib

rou

s ca

p t

hic

knes

s (µ

m)

88 d HFD

Ldlr –/–;3MR

Ldlr –/–;3MR

**

88 d HFD

88 d HFD

+ 100 d LFD

188 d HFD

n= 6

n= 6

n=

7

n= 1

0 n=

10

* **

88 d HFD followed by 100 d HFD +GCV V

on G

ieso

n

ns

p16 p19 EGFP Il1 Mmp3 Mmp13 Mmp12 Tnf Mcp1

mR

NA

exp

ress

ion

: G

FP

+vs

GF

P–

LV

FACS of digested atherogenic aortic

arches of Ldlr–/–;INK-ATTAC mice

Vcam1

GFP– cells

Analysis of proatherogenic

factors

GFP+ cells

Ldlr –/– Ldlr –/–;3MR

*

**

*

*

*

* *

GFP– cells (n = 4 mice) GFP+ cells (n = 4 mice)

ArfInk4a

Fig. 4. Senescent cells promote plaque instability by elevating metallo-protease production. (A) Representative sections from descending aortaplaques of mice with the indicated genotypes, treatments, diets, and histologicalstainings. Red dashed lines trace the fibrous cap; red arrowheads indicate rup-tured aortic elastic fibers. H–E, hematoxylin and eosin. (B) Quantification of fibrouscap thickness in plaques from (A). (C) (Top) Experimental overview. (Bottom)

RT-qPCR analysis of senescence markers in GFP+ and GFP– cells. EGFP, enhancedgreen fluorescent protein. Bar graphs represent mean ± SEM (error bars)[biological n is indicated directly on all graphs and refers to individual descendingaorta mouse plaques in (B) and flow-sorted cell fractions isolated from individualmice in (C)]. *P < 0.05; **P < 0.01; ns, not significant [ANOVA with Sidak’s post-hoc correction for familywise error in (B) and ratio paired t test in (C)].

RESEARCH | REPORTSon June 30, 2020

http://science.sciencemag.org/

Dow

nloaded from

Page 6: Senescent intimal foam cells are deleterious at all stages of atherosclerosis · of atherosclerosis Bennett G. Childs,1 Darren J. Baker,2 Tobias Wijshake,2,3 Cheryl A. Conover,4 Judith

that senescent cells are uniformly deleteriousthroughout atherogenesis. Very early fatty streakscontain abundant senescent foam cell macro-phages, which create an environment conduciveto further lesion growth by up-regulating inflam-matory cytokines and monocyte chemotacticfactors. Removal of p16Ink4a+ foamy macrophagesfrom fatty streaks led to marked lesion regres-sion. In contrast, advanced plaques contain threemorphologically distinct senescent cell types thatnot only drive lesion maturation through inflam-mation and monocyte chemotaxis but also pro-mote extracellular matrix degradation. Clearingsenescent cells from advanced lesions inhibitsboth plaque growth and maladaptive plaque re-modeling processes associated with plaque rup-ture, including fibrous cap thinning and elasticfiber degeneration. Furthermore, senescent cellsin lesions show heightened expression of keySASP factors and effectors of inflammation,monocyte chemotaxis, and proteolysis, includingIl1a, Mcp1, Mmp12, and Mmp13. These data sug-gest that senescent cells can directly influencecore proatherogenic processes through spe-cific secreted factors. By comparison, the ex-pression of other factors such as Mmp3, Tnfa,and Vcam1 is reduced with senolysis, but thesefactors are not enriched in p16Ink4a+ cells, whichimplies that senescent cells also can influence theproatherogenic milieu indirectly. Collectively, ourresults show that senescent cells drive athero-sclerosis at all stages through paracrine activityand raise the possibility that druglike moleculesthat remove senescent cells from patients with-out toxic side effects could contribute to ther-apeutic management of the disease.

REFERENCES AND NOTES

1. I. Tabas, G. García-Cardeña, G. K. Owens, J. Cell Biol. 209,13–22 (2015).

2. C. Weber, H. Noels, Nat. Med. 17, 1410–1422 (2011).3. K. Sakakura et al., Heart Lung Circ. 22, 399–411 (2013).4. I. Gorenne, M. Kavurma, S. Scott, M. Bennett, Cardiovasc. Res.

72, 9–17 (2006).5. T. Minamino et al., Circulation 105, 1541–1544 (2002).6. J. C. Wang, M. Bennett, Circ. Res. 111, 245–259 (2012).7. D. Muñoz-Espín, M. Serrano, Nat. Rev. Mol. Cell Biol. 15,

482–496 (2014).8. J. Wang et al., Circulation 132, 1909–1919 (2015).9. A. K. Khanna, J. Biomed. Sci. 16, 66 (2009).10. J. Mercer, N. Figg, V. Stoneman, D. Braganza, M. R. Bennett,

Circ. Res. 96, 667–674 (2005).11. H. González-Navarro et al., J. Am. Coll. Cardiol. 55, 2258–2268

(2010).12. K. Wouters et al., PLOS ONE 7, e32440 (2012).13. C. L. Kuo et al., Arterioscler. Thromb. Vasc. Biol. 31, 2483–2492

(2011).14. M. Demaria et al., Dev. Cell 31, 722–733 (2014).15. S. Feil et al., Circ. Res. 115, 662–667 (2014).16. L. S. Shankman et al., Nat. Med. 21, 628–637 (2015).17. H. Williams, J. L. Johnson, K. G. Carson, C. L. Jackson,

Arterioscler. Thromb. Vasc. Biol. 22, 788–792 (2002).18. D. J. Baker et al., Nature 479, 232–236 (2011).19. D. J. Baker et al., Nature 530, 184–189 (2016).20. J. Chang et al., Nat. Med. 22, 78–83 (2016).21. Y. Nakashima, E. W. Raines, A. S. Plump, J. L. Breslow,

R. Ross, Arterioscler. Thromb. Vasc. Biol. 18, 842–851(1998).

22. J. L. Johnson, C. L. Jackson, Atherosclerosis 154, 399–406(2001).

23. M. C. Clarke et al., Nat. Med. 12, 1075–1080 (2006).24. N. Maldonado et al., Am. J. Physiol. Heart Circ. Physiol. 303,

H619–H628 (2012).

25. C. Silvestre-Roig et al., Circ. Res. 114, 214–226 (2014).26. S. M. Ghaderian, R. Akbarzadeh Najar, A. S. Tabatabaei Panah,

Coron. Artery Dis. 21, 330–335 (2010).27. A. V. Finn, M. Nakano, J. Narula, F. D. Kolodgie, R. Virmani,

Arterioscler. Thromb. Vasc. Biol. 30, 1282–1292 (2010).

ACKNOWLEDGMENTS

We thank R.-M. Laberge and M. Demaria for sharing data on thesenolytic properties of ABT263, as well as N. David, Y. Poon,M. Hofker, B. van de Sluis, and the van Deursen lab for helpfuldiscussions. This work was supported by a grant from thePaul F. Glenn Foundation (to J.M.v.D. and D.J.B.) and NIH grantsR01CA96985 and CA168709 (to J.M.v.D.). J.M.v.D. and J.C. arecofounders of Unity Biotechnology, a company developing

senolytic medicines including small molecules that selectivelyeliminate senescent cells. J.M.v.D., D.J.B., B.G.C., and J.C. areco-inventors on patent applications licensed to or filed by UnityBiotechnology. The p16-3MR mice are available from J.C. undera material transfer agreement. INK-ATTAC and INK-NTR mice areavailable from J.M.v.D. under a material transfer agreement.

SUPPLEMENTARY MATERIALS

www.sciencemag.org/content/354/6311/472/suppl/DC1Materials and MethodsFigs. S1 to S13References (28, 29)

11 March 2016;accepted 29 September 201610.1126/science.aaf6659

APE GENETICS

Chimpanzee genomic diversityreveals ancient admixturewith bonobosMarc de Manuel,1* Martin Kuhlwilm,1* Peter Frandsen,2,3* Vitor C. Sousa,4,5

Tariq Desai,6 Javier Prado-Martinez,1,7 Jessica Hernandez-Rodriguez,1

Isabelle Dupanloup,4,5 Oscar Lao,8,9 Pille Hallast,7,10 Joshua M. Schmidt,11

José María Heredia-Genestar,1 Andrea Benazzo,12 Guido Barbujani,12

Benjamin M. Peter,13 Lukas F. K. Kuderna,1 Ferran Casals,1 Samuel Angedakin,14

Mimi Arandjelovic,14 Christophe Boesch,14 Hjalmar Kühl,14 Linda Vigilant,14

Kevin Langergraber,15 John Novembre,13 Marta Gut,8 Ivo Gut,8 Arcadi Navarro,1,8,16

Frands Carlsen,3 Aida M. Andrés,11 Hans. R. Siegismund,2 Aylwyn Scally,6

Laurent Excoffier,4,5 Chris Tyler-Smith,9 Sergi Castellano,11 Yali Xue,9

Christina Hvilsom,3† Tomas Marques-Bonet1,8,16†

Our closest living relatives, chimpanzees and bonobos, have a complex demographichistory. We analyzed the high-coverage whole genomes of 75 wild-born chimpanzeesand bonobos from 10 countries in Africa. We found that chimpanzee populationsubstructure makes genetic information a good predictor of geographic origin at countryand regional scales. Multiple lines of evidence suggest that gene flow occurred frombonobos into the ancestors of central and eastern chimpanzees between 200,000 and550,000 years ago, probably with subsequent spread into Nigeria-Cameroon chimpanzees.Together with another, possibly more recent contact (after 200,000 years ago), bonoboscontributed less than 1% to the central chimpanzee genomes. Admixturethus appears to have been widespread during hominid evolution.

Compared with our knowledge of the ori-gins and population history of humans,much less is known about the extant spe-cies closest to humans, chimpanzees (Pantroglodytes) and bonobos (Pan paniscus).

Unraveling the demographic histories of our closestliving relatives provides an opportunity for com-parisons with our own history and thus for study-ing processes that might have played a recurringrole in hominid evolution. Because of a paucity offossil records (1), our understanding of the demo-graphic history of the Pan genus has primarilyrelied on population genetic data frommitochon-drial genomes (2, 3), nuclear fragments (4, 5), andmicrosatellites (6, 7). More recently, the analysisof whole-genome sequences from chimpanzeesand bonobos has hinted at a complex evolution-ary history for the four taxonomically recognizedchimpanzee subspecies (8). However, although

chimpanzees and bonobos hybridize in captivity(9), the extent of interbreeding among chimpan-zee subspecies and between chimpanzees andbonobos in the wild remains unclear.We analyzed 75 complete genomes from the

Pan genus, of which 40 were sequenced for thisproject to a mean sequence coverage of 25-fold.Our samples span 10 African countries, from thewesternmost to the easternmost regions of thechimpanzee range (Fig. 1A). We discovered 32%more variable sites than previously identified (8, 10),highlighting the value of our sampling scheme.Different lines of evidence suggest larger histor-ical effective population sizes in central chim-panzees, including haplotype diversity in eachsubspecies (fig. S5), Y chromosome diversity (fig.S3), fixation index (FST)–based phylogenies (fig.S16), and genome-wide linkage disequilibrium(fig S6). An analysis of the long-term demographic

SCIENCE sciencemag.org 28 OCTOBER 2016 • VOL 354 ISSUE 6311 477

resubmitted 6 September 2016Accepted

RESEARCH | REPORTSon June 30, 2020

http://science.sciencemag.org/

Dow

nloaded from

Page 7: Senescent intimal foam cells are deleterious at all stages of atherosclerosis · of atherosclerosis Bennett G. Childs,1 Darren J. Baker,2 Tobias Wijshake,2,3 Cheryl A. Conover,4 Judith

Senescent intimal foam cells are deleterious at all stages of atherosclerosisBennett G. Childs, Darren J. Baker, Tobias Wijshake, Cheryl A. Conover, Judith Campisi and Jan M. van Deursen

DOI: 10.1126/science.aaf6659 (6311), 472-477.354Science 

, this issue p. 472Scienceblood clots. Experimental removal of the senescent cells had beneficial effects at both stages of the disease.promoted the expression of matrix metalloproteases implicated in the rupture of atherosclerotic plaque, which can lead to stages of atherosclerosis, senescent foam cells promoted the expression of inflammatory cytokines. At later stages, theythe role of senescent lipid-loaded macrophages (so-called ''foam cells'') in the pathogenesis of atherosclerosis. At early

examinedet al.Cells enter a state of senescence in response to certain stresses. Studying mouse models, Childs Wreaking havoc while (growth-)arrested

ARTICLE TOOLS http://science.sciencemag.org/content/354/6311/472

MATERIALSSUPPLEMENTARY http://science.sciencemag.org/content/suppl/2016/10/27/354.6311.472.DC1

CONTENTRELATED

http://stke.sciencemag.org/content/sigtrans/8/393/ra91.fullhttp://stke.sciencemag.org/content/sigtrans/8/393/pc23.fullhttp://stke.sciencemag.org/content/sigtrans/9/409/ra2.fullhttp://stm.sciencemag.org/content/scitransmed/8/323/323ra13.fullhttp://stm.sciencemag.org/content/scitransmed/7/275/275ra20.fullhttp://stke.sciencemag.org/content/sigtrans/6/293/ra83.fullhttp://stke.sciencemag.org/content/sigtrans/9/419/ra29.fullhttp://stm.sciencemag.org/content/scitransmed/6/239/239sr1.fullhttp://stm.sciencemag.org/content/scitransmed/8/333/333ra50.full

REFERENCES

http://science.sciencemag.org/content/354/6311/472#BIBLThis article cites 29 articles, 12 of which you can access for free

PERMISSIONS http://www.sciencemag.org/help/reprints-and-permissions

Terms of ServiceUse of this article is subject to the

is a registered trademark of AAAS.ScienceScience, 1200 New York Avenue NW, Washington, DC 20005. The title (print ISSN 0036-8075; online ISSN 1095-9203) is published by the American Association for the Advancement ofScience

Copyright © 2016, American Association for the Advancement of Science

on June 30, 2020

http://science.sciencemag.org/

Dow

nloaded from


Recommended