7
DOI: 10.1126/science.1157535 , 1496 (2008); 320 Science et al. Yasemin Sancak, Acid Signaling to mTORC1 The Rag GTPases Bind Raptor and Mediate Amino www.sciencemag.org (this information is current as of December 9, 2008 ): The following resources related to this article are available online at http://www.sciencemag.org/cgi/content/full/320/5882/1496 version of this article at: including high-resolution figures, can be found in the online Updated information and services, http://www.sciencemag.org/cgi/content/full/1157535/DC1 can be found at: Supporting Online Material found at: can be related to this article A list of selected additional articles on the Science Web sites http://www.sciencemag.org/cgi/content/full/320/5882/1496#related-content http://www.sciencemag.org/cgi/content/full/320/5882/1496#otherarticles , 16 of which can be accessed for free: cites 28 articles This article 2 article(s) on the ISI Web of Science. cited by This article has been http://www.sciencemag.org/cgi/content/full/320/5882/1496#otherarticles 2 articles hosted by HighWire Press; see: cited by This article has been http://www.sciencemag.org/cgi/collection/ecology Ecology : subject collections This article appears in the following http://www.sciencemag.org/about/permissions.dtl in whole or in part can be found at: this article permission to reproduce of this article or about obtaining reprints Information about obtaining registered trademark of AAAS. is a Science 2008 by the American Association for the Advancement of Science; all rights reserved. The title Copyright American Association for the Advancement of Science, 1200 New York Avenue NW, Washington, DC 20005. (print ISSN 0036-8075; online ISSN 1095-9203) is published weekly, except the last week in December, by the Science on December 9, 2008 www.sciencemag.org Downloaded from

DOI: 10.1126/science.1157535 , 1496 (2008); 320 Science et al

  • Upload
    lykiet

  • View
    214

  • Download
    0

Embed Size (px)

Citation preview

Page 1: DOI: 10.1126/science.1157535 , 1496 (2008); 320 Science et al

DOI: 10.1126/science.1157535 , 1496 (2008); 320Science

et al.Yasemin Sancak,Acid Signaling to mTORC1The Rag GTPases Bind Raptor and Mediate Amino

www.sciencemag.org (this information is current as of December 9, 2008 ):The following resources related to this article are available online at

http://www.sciencemag.org/cgi/content/full/320/5882/1496version of this article at:

including high-resolution figures, can be found in the onlineUpdated information and services,

http://www.sciencemag.org/cgi/content/full/1157535/DC1 can be found at: Supporting Online Material

found at: can berelated to this articleA list of selected additional articles on the Science Web sites

http://www.sciencemag.org/cgi/content/full/320/5882/1496#related-content

http://www.sciencemag.org/cgi/content/full/320/5882/1496#otherarticles, 16 of which can be accessed for free: cites 28 articlesThis article

2 article(s) on the ISI Web of Science. cited byThis article has been

http://www.sciencemag.org/cgi/content/full/320/5882/1496#otherarticles 2 articles hosted by HighWire Press; see: cited byThis article has been

http://www.sciencemag.org/cgi/collection/ecologyEcology

: subject collectionsThis article appears in the following

http://www.sciencemag.org/about/permissions.dtl in whole or in part can be found at: this article

permission to reproduce of this article or about obtaining reprintsInformation about obtaining

registered trademark of AAAS. is aScience2008 by the American Association for the Advancement of Science; all rights reserved. The title

CopyrightAmerican Association for the Advancement of Science, 1200 New York Avenue NW, Washington, DC 20005. (print ISSN 0036-8075; online ISSN 1095-9203) is published weekly, except the last week in December, by theScience

on

Dec

embe

r 9,

200

8 w

ww

.sci

ence

mag

.org

Dow

nloa

ded

from

Page 2: DOI: 10.1126/science.1157535 , 1496 (2008); 320 Science et al

in others, such as adult liver, it does not sub-stantially affect protein secretory function butrather controls select transcriptional programssuch as lipogenesis. Preservation of the normalhepatic lipid profile suggests that compoundsthat inhibit XBP1 activation in the liver mayreduce serum lipids without causing hepaticsteatosis in patients with dyslipidemias.

Given XBP1’s known function as a keymediator of the UPR, it was surprising that itsfunction in regulating lipogenesis was unre-lated to the ER stress response. Indeed, apoB-100 folding and secretion, as well as the overallhepatocyte protein secretory function, were min-imally compromised by loss of XBP1, likelybecause XBP1 independent basal chaperonegene expression is sufficient to accommodatemoderate secretory loads. Interestingly, IRE1a,the upstream activator of XBP1, was constitu-tively active in the Xbp1∆ liver, suggestingthe presence of a negative feedback loop thatprecisely maintains XBP1s protein levels evenin the absence of ER stress. The nature of thissignal, and its relationship to the ER stressresponse and to the activation of XBP1 in the

liver by carbohydrate feeding, require furtherinvestigation.

References and Notes1. H. N. Ginsberg, Y. L. Zhang, A. Hernandez-Ono, Obesity

14 (suppl. 1), 41S (2006).2. F. Foufelle, P. Ferre, Biochem. J. 366, 377 (2002).3. D. Ron, P. Walter, Nat. Rev. Mol. Cell Biol. 8, 519

(2007).4. A. L. Shaffer et al., Immunity 21, 81 (2004).5. A. H. Lee, N. N. Iwakoshi, L. H. Glimcher, Mol. Cell. Biol.

23, 7448 (2003).6. D. Acosta-Alvear et al., Mol. Cell 27, 53 (2007).7. A. M. Reimold et al., Nature 412, 300 (2001).8. A. H. Lee, G. C. Chu, N. N. Iwakoshi, L. H. Glimcher,

EMBO J. 24, 4368 (2005).9. A. M. Reimold et al., Genes Dev. 14, 152 (2000).10. R. Sriburi, S. Jackowski, K. Mori, J. W. Brewer, J. Cell Biol.

167, 35 (2004).11. N. O. Davidson, G. S. Shelness, Annu. Rev. Nutr. 20, 169

(2000).12. M. M. Hussain, J. Iqbal, K. Anwar, P. Rava, K. Dai,

Front. Biosci. 8, s500 (2003).13. M. C. Schotz, A. Scanu, I. H. Page, Am. J. Physiol. 188,

399 (1957).14. S. J. Stone et al., J. Biol. Chem. 279, 11767 (2004).15. X. X. Yu et al., Hepatology 42, 362 (2005).16. J. M. Ntambi et al., Proc. Natl. Acad. Sci. U.S.A. 99,

11482 (2002).17. P. Cohen et al., Science 297, 240 (2002).

18. L. Abu-Elheiga, W. Oh, P. Kordari, S. J. Wakil, Proc. Natl.Acad. Sci. U.S.A. 100, 10207 (2003).

19. J. D. Horton, J. L. Goldstein, M. S. Brown, J. Clin. Invest.109, 1125 (2002).

20. H. C. Towle, E. N. Kaytor, H. M. Shih, Annu. Rev. Nutr.17, 405 (1997).

21. M. Miyazaki et al., J. Biol. Chem. 279, 25164 (2004).22. H. Basciano, L. Federico, K. Adeli, Nutr. Metab. 2, 5

(2005).23. Supported by NIH grants AI32412 and P01 AI56296 (L.H.G.),

NIH grants DK48873 and DK56626 (D.E.C.), and the EllisonMedical Foundation (L.H.G.). We thank K. Rajewsky forprovidingMx1-cremice, J. Goldstein and M. Brown for SREBPantibodies, R. Milne for apoB antibody, K. Mori for ATF6aantibody, E. Fisher for advice on pulse-chase experiments,M. Wu and J. Wei for help with FPLC analyses, D. Hu forhistologic analyses, K. Heidtman for excellent technicalassistance, and M. Wein and W. Garrett for critical readingof the manuscript. L.H.G. has equity in Bristol-Myers Squibband has filed a patent regarding methods for regulatinghepatic lipogenesis with XBP1.

Supporting Online Materialwww.sciencemag.org/cgi/content/full/320/5882/1492/DC1Materials and MethodsFigs. S1 to S5Tables S1 to S4References

19 March 2008; accepted 17 April 200810.1126/science.1158042

The Rag GTPases Bind Raptor andMediate Amino Acid Signalingto mTORC1Yasemin Sancak,1,2 Timothy R. Peterson,1,2 Yoav D. Shaul,1,2 Robert A. Lindquist,1,2Carson C. Thoreen,1,2 Liron Bar-Peled,1 David M. Sabatini1,2,3*

The multiprotein mTORC1 protein kinase complex is the central component of a pathway thatpromotes growth in response to insulin, energy levels, and amino acids and is deregulated incommon cancers. We find that the Rag proteins—a family of four related small guanosinetriphosphatases (GTPases)—interact with mTORC1 in an amino acid–sensitive manner and arenecessary for the activation of the mTORC1 pathway by amino acids. A Rag mutant that isconstitutively bound to guanosine triphosphate interacted strongly with mTORC1, and itsexpression within cells made the mTORC1 pathway resistant to amino acid deprivation. Conversely,expression of a guanosine diphosphate–bound Rag mutant prevented stimulation of mTORC1by amino acids. The Rag proteins do not directly stimulate the kinase activity of mTORC1, but,like amino acids, promote the intracellular localization of mTOR to a compartment that alsocontains its activator Rheb.

The mTOR complex 1 (mTORC1) branchof the mammalian target of rapamycin(mTOR) pathway is a major driver of

cell growth in mammals and is deregulated inmany common tumors (1). It is also the targetof the drug rapamycin, which has generatedconsiderable interest as an anticancer therapy.

Diverse signals regulate the mTORC1 path-way, including insulin, hypoxia, mitochondrialfunction, and glucose and amino acid availa-bility. Many of these are integrated upstream ofmTORC1 by the tuberous sclerosis complex(TSC1-TSC2) tumor suppressor, which acts asan important negative regulator of mTORC1through its role as a guanosine triphosphatase(GTPase)–activating protein (GAP) for Rheb, asmall guanosine triphosphate (GTP)–bindingprotein that potently activates the protein ki-nase activity of mTORC1 (2). Loss of eitherTSC protein causes hyperactivation of mTORC1signaling, even in the absence of many of theupstream signals that are normally required to

maintain pathway activity. A notable excep-tion is the amino acid supply, as the mTORC1pathway remains sensitive to amino acid star-vation in cells lacking either TSC1 or TSC2(3–5).

The mechanisms through which amino acidssignal to mTORC1 remain mysterious. It is areasonable expectation that proteins that sig-nal the availability of amino acids to mTORC1are also likely to interact with it, but, so far,no good candidates have been identified. Be-cause most mTORC1 purifications rely onantibodies to isolate mTORC1, we wonderedif in previous work antibody heavy chainsobscured, during SDS–polyacrylamide elec-trophoresis (SDS-PAGE) analysis of purifiedmaterial, mTORC1-interacting proteins of 45to 55 kD. Indeed, using a purification strategythat avoids this complication (6), we identifiedthe 44-kD RagC protein as copurifying withoverexpressed raptor, the defining componentof mTORC1 (7–10).

RagC is a Ras-related small GTP-bindingprotein and one of four Rag proteins in mam-mals (RagA, RagB, RagC, and RagD). RagAand RagB are very similar to each other andare orthologs of budding yeast Gtr1p, whereasRagC and RagD are similar and are orthologsof yeast Gtr2p (11–13). In yeast and in humancells, the Rag and Gtr proteins function as het-erodimers consisting of one Gtr1p-like (RagAor RagB) and one Gtr2p-like (RagC or RagD)component (14, 15). The finding that RagCcopurifies with raptor was intriguing to us be-cause, in yeast, Gtr1p and Gtr2p regulate theintracellular sorting of the Gap1p amino acidpermease (16) and microautophagy (17), pro-cesses modulated by amino acid levels and

1Whitehead Institute for Biomedical Research and Depart-ment of Biology, Massachusetts Institute of Technology (MIT),Nine Cambridge Center, Cambridge, MA 02142, USA. 2MITCenter for Cancer Research, 77 Massachusetts Avenue,Cambridge, MA 02139, USA. 3Broad Institute, Seven Cam-bridge Center, Cambridge, MA 02142, USA.

*To whom correspondence should be addressed. E-mail:[email protected]

13 JUNE 2008 VOL 320 SCIENCE www.sciencemag.org1496

REPORTS

on

Dec

embe

r 9,

200

8 w

ww

.sci

ence

mag

.org

Dow

nloa

ded

from

Page 3: DOI: 10.1126/science.1157535 , 1496 (2008); 320 Science et al

the TOR pathway (18–20). The Gtr proteinshave been proposed to act downstream or inparallel to TORC1 in yeast because their over-expression induces microautophagy even inthe presence of rapamycin, which normally sup-presses it (17).

To verify our identification of RagC as anmTORC1-interacting protein, we expressedraptor with different pairs of Rag proteins inhuman embryonic kidney (HEK)-293T cells.Consistent with the Rags functioning as het-erodimers, raptor copurified with RagA-C orRagB-C, but not with RagA-B or the Rap2Acontrol protein (Fig. 1A). Because the nucle-otide loading state of most GTP-binding pro-teins regulates their functions, we generatedRagB, RagC, and RagD mutants predicted(14, 16, 17) to be restricted to the GTP- or gua-nosine diphosphate (GDP)–bound conforma-tions (for simplicity, we call these mutantsRagBGTP, RagBGDP, etc.) (6). When expressedwith mTORC1 components, Rag heterodimerscontaining RagBGTP immunoprecipitated with

more raptor and mTOR than did complexescontaining wild-type RagB or RagBGDP (Fig.1B). The GDP-bound form of RagC increasedthe amount of copurifying mTORC1, so thatRagBGTP-CGDP recovered the highest amountof endogenous mTORC1 of any heterodimertested (Fig. 1C). Giving an indication of thestrength of the mTORC1-RagBGTP-CGDP asso-ciation, in this same assay, we could not detectcoimmunoprecipitation of mTORC1 with Rheb1(Fig. 1C), an established interactor and acti-vator of mTORC1 (1). When expressed alone,raptor, but not mTOR, associated with RagBGTP-DGDP, which suggests that raptor is the keymediator of the Rag-mTORC1 interaction (Fig.1D). Consistent with this, rictor, an mTOR-interacting protein that is only part of mTORC2(1), did not copurify with any Rag heterodimer(Fig. 1C and fig. S1). Last, highly purified rap-tor interacted in vitro with RagB-D and, to alarger extent, with RagBGTP-DGDP, which in-dicates that the Rag-raptor interaction is mostlikely direct (Fig. 1E).

We tested whether various Rag heterodimersaffected the regulation of the mTORC1 path-way within human cells. In HEK-293T cells,expression of the RagBGTP-DGDP heterodimer,which interacted strongly with mTORC1, notonly activated the pathway, but also made itinsensitive to deprivation for leucine or totalamino acids, as judged by the phosphorylationstate of the mTORC1 substrate T389 of S6K1(Fig. 2, A and B). The wild-type RagB-Cheterodimer had milder effects than RagBGTP-CGDP, making the mTORC1 pathway insensi-tive to leucine deprivation, but not to the strongerinhibition caused by total amino acid starvation(Fig. 2, A and B). Expression of RagBGDP-DGTP, a heterodimer that did not interact withmTORC1 (Fig. 1, C and D), had dominant-negative effects, as it eliminated S6K1 phospho-rylation in the presence, as well as absence, ofleucine or amino acids (Fig. 2, A and B). Ex-pression of RagBGDP alone also suppressedS6K1 phosphorylation (fig. S2). These resultssuggest that the activity of themTORC1 pathway

A

celllysate

IP: HA

raptor

mTOR

rictor

+ - - - ---HA-Rheb1: + - - - -

HA-RagB: - GTP GTP GDP GDP-HA-RagC: GTP GDP GTP GDP--

raptor

mTOR

rictor

HA-Rap2A

HA-Rheb1

HA-RagB

HA-RagC

HA-Rap2A:

D

HA-GST-RagB: GTP GDP GTP GTP

FLAG-RagD: GDP GTP GDP GDP

FLAG-RagD

myc-raptor

myc-mTOR

+ +myc-mTOR: + -myc-raptor: + + +-

FLAG-RagD

myc-raptor

myc-mTOR

celllysate

IP: FLAG

HA-GST-RagB

HA-GST-RagB

myc-raptor: + + + +HA-Rap2A: + - - -

HA-Rag: - A+B A+C B+C

myc-raptor

myc-raptor

IP: HA HA-RagB

HA-RagC

HA-RagA

HA-RagB

HA-RagC

HA-RagA

celllysate

HA-Rap2A

HA-Rap2A

B CHA-GST-RagB: GTP GDP-

FLAG--RagD: -

myc-mTOR + myc-raptor: + ++

FLAG--Rap2A: --++ +

+

FLAG--RagD

FLAG-RagD

FLAG-Rap2A

celllysate

HA-GST-RagB

IP: FLAG HA-GST-RagB

FLAG--Rap2A

myc-raptor

myc-mTOR

myc-raptor

myc-mTOR

WT

+-

Rhe

b1

Rag

BG

TP-D

GD

PR

agB

-DR

ap2A

1051

% input

FLAG-raptor

HA-Rap2AHA-Rheb1

HA-RagB

HA-RagD

in vitrobindingassay

E

Fig. 1. Interaction of Rag heterodimerswith recombinant and endogenous mTORC1in a manner that depends on the nucleotidebinding state of RagB. In (A) through (D)HEK-293T cells were transfected with theindicated cDNAs in expression vectors, celllysates were prepared, and lysates and he-magglutinin (HA)– or FLAG-tagged immuno-precipitates were analyzed by immunoblottingfor the amounts of the specified recombi-nant or endogenous proteins. (E) In vitrobinding of purified FLAG-raptor with wild-type RagB-D or RagBGTP-DGDP.

www.sciencemag.org SCIENCE VOL 320 13 JUNE 2008 1497

REPORTS

on

Dec

embe

r 9,

200

8 w

ww

.sci

ence

mag

.org

Dow

nloa

ded

from

Page 4: DOI: 10.1126/science.1157535 , 1496 (2008); 320 Science et al

under normal growth conditions depends onendogenous Rag function.

To verify the actions of the Rags in a morephysiological setting than that achieved by tran-sient cDNA transfection, we generated HEK-293T cell lines stably expressing Rheb1, RagB,or RagBGTP (attempts to generate lines stably ex-pressing RagBGDP failed). Under normal growthconditions, these cells were larger than controlcells and had higher levels of mTORC1 path-way activity (Fig. 3A). Unlike transient Rheb1overexpression (Fig. 2, A and B), stable ex-pression did not make the mTORC1 pathwayinsensitive to leucine or amino acid starvation(Fig. 3, B and C), consistent with evidence thattransiently overexpressed Rheb may have non-physiological consequences on amino acid sig-naling to mTORC1 (4, 5). Stable expressionof a Rheb1GTP mutant was also unable to makethe mTORC1 pathway resistant to amino aciddeprivation (fig. S3). In contrast, stable expres-sion of RagBGTP eliminated the sensitivity of themTORC1 pathway to leucine or total amino acidwithdrawal, whereas that of wild-type RagBovercame sensitivity to leucine but not to amino

acid starvation (Fig. 3, B and C). Thus, transientor stable expression of the appropriate Rag mu-tants is sufficient to put the mTORC1 pathwayinto states that mimic the presence or absence ofamino acids.

To determine if the Rag mutants affect sig-naling to mTORC1 from inputs besides aminoacids, we tested whether in RagBGTP-expressingcells themTORC1 pathwaywas resistant to otherperturbations known to inhibit it. This was notthe case, as oxidative stress, mitochondrial in-hibition, or energy deprivation still reduced S6K1phosphorylation in these cells (fig. S4). Moreover,in HEK-293E cells, expression of RagBGTP-DGDP

did not maintainmTORC1 pathway activity in theabsence of insulin (Fig. 2C). Expression of thedominant-negative RagBGDP-DGTP heterodimerdid, however, block insulin-stimulated phospho-rylation of S6K1 (Fig. 2C), as did amino acidstarvation (Fig. 2D). Thus, although RagBGTP

expression mimics amino acid sufficiency, itcannot substitute for other inputs that mTORC1normally monitors.

This evidence for a primary role of the Ragproteins in amino acid signaling to mTORC1

raised the question of where, within the pathwaythat links amino acids to mTORC1, the Ragproteins might function. The existence of theRag-mTORC1 interaction (Fig. 1), the effects onamino acid signaling of the Rag mutants (Figs. 2and 3), and the sensitivity to rapamycin of theS6K1 phosphorylation induced by RagBGTP (fig.S4), strongly suggested that the Rag proteinsfunction downstream of amino acids and up-stream of mTORC1. To verify this, we tookadvantage of the established finding that cyclo-heximide reactivates mTORC1 signaling in cellsstarved for amino acids by blocking proteinsynthesis and thus boosting the levels of theintracellular amino acids sensed by mTORC1(21–23). Thus, if the Rag proteins act upstream ofamino acids, cycloheximide should overcome theinhibitory effects of the RagBGDP-CGTP hetero-dimer on mTORC1 signaling, but if they aredownstream, cycloheximide should not reactivatethe pathway. The results were clear: cyclo-heximide treatment of cells reversed the inhi-bition of mTORC1 signaling caused by leucinedeprivation, but not that caused by expressionof RagBGDP-CGTP (fig. S5). Given the place-

A

C

B

+-insulin: +- +- +-Rap2A

RagBGDP

+RagCGTP

RagBGTP

+RagCGDPRheb1

FLAG-S6K1 &transfected

cDNAs

HA-GST-Rap2A

HA-GST-RagB

HA-GST-RagC

HA-GST-Rheb1

S6K1

P -T389-S6K1

cell lysate

D

cell lysate

insulin (nM):

+-amino acids: + +- -0 10 150 0 10 150

S6K1

P -T389-S6K1

cell lysate

S6K1

P -T389-S6K1

HA-GST-Rap2A

HA-GST-RagB

HA-GST-RagC

+-leucine: +-+- +-Rap2A

RagBGDP

+RagCGTP

RagBGTP

+RagCGDP Rheb1

transfectedcDNAs

FLAG-S6K1 &

+-

RagB+

RagC

HA-GST-Rheb1

IP: FLAG

IP: FLAG

+- +- +- +-Rap2A

RagBGTP

+RagCGDP Rheb1

FLAG-S6K1 &

S6K1

P -T389-S6K1

HA-GST-Rap2AHA-GST-Rheb1

HA-GST-RagC

HA-GST-RagBcell lysate

transfectedcDNAs

IP: FLAG

+-

RagBGDP

+RagCGTP

RagB+

RagC

amino acids:

Fig. 2. Effects of overexpressed RagBGTP-containing heterodimers on themTORC1 pathway and its response to leucine, amino acids, or insulin. Effectsof expressing the indicated proteins on the phosphorylation state ofcoexpressed S6K1 in response to deprivation and stimulation with (A)leucine, (B) total amino acids, or (C) insulin. Cell lysates were prepared fromHEK-293T cells deprived for 50 min of serum and of (A) leucine or (B) aminoacids and, then, where indicated, stimulated with leucine or amino acids for10 min. HEK-293E cells (C) were deprived of serum for 50 min and, where

indicated, stimulated with 150 nM insulin for 10 min. Lysates and FLAG-immunoprecipitates were analyzed for the levels of the specified proteins andthe phosphorylation state of S6K1. (D) Effects of amino acid deprivation on insulin-mediated activation of mTORC1. HEK-293E cells were starved forserum and amino acids or just serum for 50 min, and where specified, stimulated with 10 or 150 nM insulin. Cell lysates were analyzed for the leveland phosphorylation state of S6K1.

13 JUNE 2008 VOL 320 SCIENCE www.sciencemag.org1498

REPORTS

on

Dec

embe

r 9,

200

8 w

ww

.sci

ence

mag

.org

Dow

nloa

ded

from

Page 5: DOI: 10.1126/science.1157535 , 1496 (2008); 320 Science et al

ment of the Rag proteins downstream of aminoacids and upstream of mTORC1, we deter-mined whether amino acids regulate the Rag-mTORC1 interaction within cells. Initial testsusing transiently coexpressed Rag proteins andmTORC1 components did not reveal any reg-ulation of the interaction. Because we reasonedthat pronounced overexpression might over-come the normal regulatory mechanisms thatoperate within the cell, we developed an assay(6), based on a reversible chemical cross-linker,that allows us to detect the interaction of stablyexpressed FLAG-tagged Rag proteins withendogenous mTORC1. With this approach,we readily found that amino acids, but notinsulin, promote the Rag-mTORC1 interactionwhen we used either FLAG-tagged RagB or

RagD to isolate mTORC1 from cells (Fig. 3Dand fig. S6A). As the GTP-loading state of theRag proteins also regulates the Rag-mTORC1interaction (Fig. 1), we determined whether ami-no acids modulate the amount of GTP bound toRagB. Indeed, amino acid stimulation of cellsincreased the GTP loading of RagB (Fig. 3E).Consistent with this, amino acids did not furtheraugment the already high level of interactionbetween mTORC1 and the RagBGTP mutant(Fig. 3D).

To determine whether the Rag proteins arenecessary for amino acids to activate themTORC1pathway, we used combinations of lentivirallydelivered short hairpin RNAs (shRNAs) to sup-press RagA and RagB or RagC and RagD atthe same time. Loss of RagA and RagB also led

to the loss of RagC and RagD and vice versa,which suggests that, within cells, the Ragproteins are unstable when not in hetero-dimers (Fig. 3F). In cells with a reduction inthe expression of all the Rag proteins, leucine-stimulated phosphorylation of S6K1 was strong-ly reduced (Fig. 3G). The role of the Rag proteinsappears to be conserved in Drosophila cells asdouble-stranded RNA–mediated suppressionof the Drosophila orthologs of RagB or RagCeliminated amino acid–induced phosphoryl-ation of dS6K (Fig. 3H). Consistent with ami-no acids being necessary for activation ofmTORC1 by insulin, a reduction in Rag ex-pression also suppressed insulin-stimulatedphosphorylation of S6K1 (fig. S6B). Thus, theRag proteins appear to be both necessary and

G

P -T389-S6K1

S6K1

Rag

C_1

+R

agD

_1

Rag

C_2

+R

agD

_2

luci

f.

+-leucine: +- +-

lentiviralshRNA:

F

RagB

RagA

RagD

RagC

raptor

mTOR

luci

f.

luci

f.lentiviralshRNA: R

agC

_1 +

Rag

D_1

Rag

C_2

+ R

agD

_2

Rag

A_1

+ R

agB

_1

Rag

A_2

+ R

agB

_2

A B

S6K1

P -T389-S6K1

mTOR

raptor

+-leucine: +- +-+-

lowexposure

highexposure

Rap2A Rheb1RagBcells expressing: RagBGTP

FLAG-Rap2AFLAG-Rheb1

FLAG-RagB

C

P -T389-S6K1

Rap2A

Rheb1

RagB

RagB

GTP

cell diameter (µm)10 12 14 16 18 20 22

Rap2A Rheb1

RagB

RagBGTPRap2A

10 12 14 16 18 20 22

D E

H

GFP

dRag

B_1

dS6K

dsRNA: dRag

B_2

dRag

C_1

dRag

C_2

+- +- +-+-amino acids: +- -T398-dS6KP

celllysate

FLAG-Rag

raptor

mTOR

IP:FLAG

raptor

mTOR

FLAG-Rag

+ +-amino acids: +- +-cells expressing: RagB RagBGTPRagD-

P -T389-S6K1

A+- +- +-+-amino acids:

Rap2A Rheb1RagBcells expressing: RagBGTP

lowexposure

highexposure

S6K1

mTOR

raptor

FLAG-Rap2AFLAG-Rheb1

FLAG-RagB

amino acids: +-

%GTP: 63±844±4

GDP

GTPIP:

RagB

origin

Fig. 3. Insensitivity of the mTORC1 pathway to amino acid deprivation in cells stablyexpressing RagBGTP. (A) Cell size distributions (graphs) and S6K1 phosphorylation(immunoblot) of cells stably expressing RagB, Rheb1, RagGTP, or Rap2A. Mean cell diameters(mm) ± SD are Rap2A, 16.05 ± 0.07; Rheb1, 16.79 ± 0.06; RagB, 16.40 ± 0.08; andRagBGTP, 16.68 ± 0.06 (n = 4 and P < 0.0008 for all comparisons to Rap2A-expressingcells). HEK-293T cells transduced with lentiviruses encoding the specified proteins weredeprived for 50 min for serum and (B) leucine or (C) total amino acids, and, whereindicated, restimulated with leucine or amino acids for 10 min. Cell lysates were analyzed for the levels of the specified proteins and thephosphorylation state of S6K1. (D) Amino acid–stimulated interaction of the Rag proteins with mTORC1. HEK-293T cells stably expressing FLAG-tagged RagB, RagD, or RagBGTP were starved for amino acids and serum for 50 min and, where indicated, restimulated with amino acids for 10 min.Cells were then processed with a chemical cross-linking assay, and cell lysates and FLAG immunoprecipitates were analyzed for the amounts of theindicated proteins. (E) Effects of amino acid stimulation on GTP loading of RagB. Values are means ± SD for n = 3 (P < 0.02 for increase in GTPloading caused by amino acid stimulation). (F) Abundance of RagA, RagB, RagC, and RagD in HeLa cells expressing the indicated shRNAs. (G) S6K1phosphorylation in HeLa cells expressing shRNAs targeting RagC and RagD. Cells were deprived of serum and leucine for 50 min, and, whereindicated, were restimulated with leucine for 10 min. (H) Effects of double-stranded RNA (dsRNA)–mediated knockdowns of Drosophila orthologs ofRagB or RagC on amino acid–induced phosphorylation of dS6K.

www.sciencemag.org SCIENCE VOL 320 13 JUNE 2008 1499

REPORTS

on

Dec

embe

r 9,

200

8 w

ww

.sci

ence

mag

.org

Dow

nloa

ded

from

Page 6: DOI: 10.1126/science.1157535 , 1496 (2008); 320 Science et al

sufficient for mediating amino acid signaling tomTORC1.

Unlike Rheb (24, 25), the Rag heterodimersdid not directly stimulate the kinase activityof mTORC1 in vitro (fig. S7), so we consid-ered the possibility that the Rag proteins reg-ulate the intracellular localization of mTOR.mTOR is found on the endomembrane systemof the cell, including the endoplasmic reticu-lum, Golgi apparatus, and endosomes (26, 27).The intracellular localization of endogenousmTOR, as revealed with an antibody that wevalidated recognizes mTOR in immuno-fluorescence assays (fig. S8), was strikinglydifferent in cells deprived of amino acids thanin cells starved and briefly restimulated withamino acids (Fig. 4A and fig. S11) or growingin fresh complete media (fig. S9). In starvedcells, mTOR was in tiny puncta throughout thecytoplasm, whereas in cells stimulated withamino acids for as little as 3 min, mTOR lo-calized to the perinuclear region of the cell, tolarge vesicular structures, or to both (Fig. 4A).Rapamycin did not block the change in mTOR

localization induced by amino acids (Fig. 4A),which indicated that it is not a consequence ofmTORC1 activity but rather may be one ofthe mechanisms that underlies mTORC1 acti-vation. The amino acid–induced change in mTORlocalization required expression of the Rag pro-teins and of raptor (Fig. 4, B and C), and aminoacids also regulated the localization of raptor(fig. S10).

In cells overexpressing RagB, Rheb1, orRheb1GTP, mTOR behaved as in control cells,its localization changing upon amino acid stimu-lation from small puncta to the perinuclear regionand vesicular structures (Fig. 4D). In contrast, incells overexpressing the RagBGTP mutant thateliminates the amino acid sensitivity of themTORC1 pathway, mTOR was already presenton the perinuclear and vesicular structures inthe absence of amino acids, and became evenmore localized to them upon the addition ofamino acids (Fig. 4D). Thus, there is a correla-tion, under amino acid–starvation conditions,between the activity of the mTORC1 pathwayand the subcellular localization of mTOR,

which implies a role for Rag-mediated mTORtranslocation in the activation of mTORC1 inresponse to amino acids.

We failed to find an established marker ofthe endomembrane system that colocalized withmTOR in amino acid–starved cells. However, incells stimulated with amino acids, mTOR in theperinuclear region and on the large vesicularstructures overlapped with Rab7 (Fig. 5A), whichindicated that a substantial fraction of mTORtranslocated to the late endosomal and lysosomalcompartments in amino acid–replete cells. Incells expressing RagBGTP, mTOR was present onthe Rab7-positive structures even in the absenceof amino acids (Fig. 5B).

The perinuclear region and vesicular struc-tures on which mTOR appears after amino acidstimulation are similar to the Rab7-positivestructures where green fluorescent protein (GFP)–tagged Rheb localizes in human cells (28, 29).Unlike mTOR, however, amino acids did notappreciably affect the localization of Rheb, asGFP-Rheb1 colocalized withDiscosoma red flu-orescent protein (DsRed)–labeled Rab7 (DsRed-

Fig. 4. Rag-dependentregulation by aminoacids of the intracel-lular localizationofmTOR.(A) HEK-293T cells werestarved for serum andamino acids for 50 minor starved and then re-stimulated with aminoacids for the indicatedtimes in the presence orabsence of rapamycin.Cells were then processedin an immunofluores-cence assay to detectmTOR (green), costainedwith 4′,6′-diamidino-2-phenylindole (DAPI) forDNA content (blue), andimaged. Of these cells,80 to 90% exhibited themTOR localization patternshown. (B) and (C) mTORlocalization in HEK-293Tcells expressing the in-dicated shRNAs and de-prived and restimulatedwith amino acids as in(A). Immunoblot of rap-tor expression levels. (D)mTOR localization inHEK-

293T cells stably expressing RagB, Rheb1, RagBGTP, or Rheb1GTP and deprivedand restimulated with amino acids as in (A).

13 JUNE 2008 VOL 320 SCIENCE www.sciencemag.org1500

REPORTS

on

Dec

embe

r 9,

200

8 w

ww

.sci

ence

mag

.org

Dow

nloa

ded

from

Page 7: DOI: 10.1126/science.1157535 , 1496 (2008); 320 Science et al

Rab7) in the presence or absence of aminoacids (Fig. 5C). Unfortunately, it is currentlynot possible to compare, in the same cells, thelocalization of endogenous mTOR with that ofRheb, because the signal for GFP-Rheb orendogenous Rheb is lost after fixed cells arepermeabilized to allow access to intracellularantigens (28, 29). Nevertheless, given that bothmTOR and Rheb are present in Rab7-positivestructures after amino acid stimulation, we pro-pose that amino acids might control the activityof the mTORC1 pathway by regulating, throughthe Rag proteins, the movement of mTORC1 tothe same intracellular compartment that con-tains its activator Rheb (see model in Fig. 5D).This would explain why activators of Rheb,like insulin, do not stimulate the mTORC1 path-way when cells are deprived of amino acids andwhy Rheb is necessary for amino acid–dependentmTORC1 activation (4) (fig. S12). When Rhebis highly overexpressed, some might becomemislocalized and inappropriately encounterand activate mTORC1, which could explainwhy Rheb overexpression, but not loss of TSC1or TSC2, makes the mTORC1 pathway insen-sitive to amino acids (4, 5).

In conclusion, the Rag GTPases bind rap-tor, are necessary and sufficient to mediateamino acid signaling to mTORC1, and medi-ate the amino acid–induced relocalization ofmTOR within the endomembrane system of

the cell. Given the prevalence of cancer-linkedmutations in the pathways that control mTORC1(1), it is possible that Rag function is also de-regulated in human tumors.

References and Notes1. D. A. Guertin, D. M. Sabatini, Cancer Cell 12, 9 (2007).2. D. D. Sarbassov, S. M. Ali, D. M. Sabatini, Curr. Opin. Cell

Biol. 17, 596 (2005).3. M. P. Byfield, J. T. Murray, J. M. Backer, J. Biol. Chem.

280, 33076 (2005).4. T. Nobukuni et al., Proc. Natl. Acad. Sci. U.S.A. 102,

14238 (2005).5. E. M. Smith, S. G. Finn, A. R. Tee, G. J. Browne,

C. G. Proud, J. Biol. Chem. 280, 18717 (2005).6. Materials and methods are available as supporting

material on Science Online.7. D.-H. Kim et al., Cell 110, 163 (2002).8. R. Loewith et al., Mol. Cell 10, 457 (2002).9. K. P. Wedaman et al., Mol. Biol. Cell 14, 1204

(2003).10. K. Hara et al., Cell 110, 177 (2002).11. M. Bun-Ya, S. Harashima, Y. Oshima, Mol. Cell. Biol. 12,

2958 (1992).12. A. Schurmann, A. Brauers, S. Massmann, W. Becker,

H. G. Joost, J. Biol. Chem. 270, 28982 (1995).13. E. Hirose, N. Nakashima, T. Sekiguchi, T. Nishimoto,

J. Cell Sci. 111, 11 (1998).14. N. Nakashima, E. Noguchi, T. Nishimoto, Genetics 152,

853 (1999).15. T. Sekiguchi, E. Hirose, N. Nakashima, M. Ii, T. Nishimoto,

J. Biol. Chem. 276, 7246 (2001).16. M. Gao, C. A. Kaiser, Nat. Cell Biol. 8, 657 (2006).17. F. Dubouloz, O. Deloche, V. Wanke, E. Cameroni,

C. De Virgilio, Mol. Cell 19, 15 (2005).18. J. B. Kunz, H. Schwarz, A. Mayer, J. Biol. Chem. 279,

9987 (2004).

19. K. J. Roberg, N. Rowley, C. A. Kaiser, J. Cell Biol. 137,1469 (1997).

20. E. J. Chen, C. A. Kaiser, J. Cell Biol. 161, 333 (2003).21. A. Beugnet, A. R. Tee, P. M. Taylor, C. G. Proud, Biochem.

J. 372, 555 (2003).22. G. R. Christie, E. Hajduch, H. S. Hundal, C. G. Proud,

P. M. Taylor, J. Biol. Chem. 277, 9952 (2002).23. D. J. Price, R. A. Nemenoff, J. Avruch, J. Biol. Chem. 264,

13825 (1989).24. Y. Sancak et al., Mol. Cell 25, 903 (2007).25. X. Long, Y. Lin, S. Ortiz-Vega, K. Yonezawa, J. Avruch,

Curr. Biol. 15, 702 (2005).26. R. M. Drenan, X. Liu, P. G. Bertram, X. F. Zheng, J. Biol.

Chem. 279, 772 (2004).27. M. Mavrakis, J. Lippincott-Schwartz, C. A. Stratakis,

I. Bossis, Autophagy 3, 151 (2007).28. K. Saito, Y. Araki, K. Kontani, H. Nishina, T. Katada,

J. Biochem. 137, 423 (2005).29. C. Buerger, B. DeVries, V. Stambolic, Biochem. Biophys.

Res. Commun. 344, 869 (2006).30. Supported by grants from the NIH (R01 CA103866 and

AI47389), Department of Defense (W81XWH-07-0448),and W.M. Keck Foundation to D.M.S. as well as an EMBOfellowship to Y.D.S. We thank T. Kang for the preparationof FLAG-raptor and members of the Sabatini lab forhelpful suggestions.

Supporting Online Materialwww.sciencemag.org/cgi/content/full/1157535/DC1Materials and MethodsFigs. S1 to S12References

10 March 2008; accepted 13 May 2008Published online 22 May 2008;10.1126/science.1157535Include this information when citing this paper.

Fig. 5. Amino acids promote the localization of mTOR to a Rab7-positivecompartment that also contains Rheb. (A) mTOR and Rab7 localization in cellsdeprived or stimulated with amino acids. HEK-293T cells transiently trans-fected with a cDNA for DsRed-Rab7 were starved for serum and amino acidsfor 50 min and, where indicated, stimulated with amino acids for 10min. Cellswere then processed to detect mTOR (green), Rab7 (red), and DNA content(blue), and imaged. Two examples are shown of mTOR localization in the

presence of amino acids. (B) HEK-293T cells stably expressing RagBGTP andtransiently transfected with a cDNA for DsRed-Rab7 were treated andprocessed as in (A). (C) Rheb1 and Rab7 localization in cells deprived orstimulated with amino acids. HEK-293T cells transiently transfected with 1 to2 ng of cDNAs for GFP-Rheb1 and DsRed-Rab7 were treated as in (A), processedto detect Rheb1 (green), Rab7 (red), and DNA content (blue), and imaged. (D)Model for role of Rag GTPases in signaling amino acid availability to mTORC1.

www.sciencemag.org SCIENCE VOL 320 13 JUNE 2008 1501

REPORTS

on

Dec

embe

r 9,

200

8 w

ww

.sci

ence

mag

.org

Dow

nloa

ded

from