+ All Categories
Home > Documents > Transcriptional regulation of the stress response by mTOR

Transcriptional regulation of the stress response by mTOR

Date post: 12-Nov-2023
Category:
Upload: upf
View: 0 times
Download: 0 times
Share this document with a friend
12
(332), re2. [DOI: 10.1126/scisignal.2005326] 7 Science Signaling López-Rodríguez (1 July 2014) Jose Aramburu, M. Carmen Ortells, Sonia Tejedor, Maria Buxadé and Cristina Transcriptional regulation of the stress response by mTOR ` This information is current as of 2 July 2014. The following resources related to this article are available online at http://stke.sciencemag.org. Article Tools http://stke.sciencemag.org/cgi/content/full/sigtrans;7/332/re2 Visit the online version of this article to access the personalization and article tools: References http://stke.sciencemag.org/cgi/content/full/sigtrans;7/332/re2#otherarticles This article cites 187 articles, 82 of which can be accessed for free: Glossary http://stke.sciencemag.org/glossary/ Look up definitions for abbreviations and terms found in this article: Permissions http://www.sciencemag.org/about/permissions.dtl Obtain information about reproducing this article: the American Association for the Advancement of Science; all rights reserved. by Association for the Advancement of Science, 1200 New York Avenue, NW, Washington, DC 20005. Copyright 2008 (ISSN 1937-9145) is published weekly, except the last week in December, by the American Science Signaling on July 2, 2014 stke.sciencemag.org Downloaded from on July 2, 2014 stke.sciencemag.org Downloaded from
Transcript

(332), re2. [DOI: 10.1126/scisignal.2005326] 7Science SignalingLópez-Rodríguez (1 July 2014) Jose Aramburu, M. Carmen Ortells, Sonia Tejedor, Maria Buxadé and Cristina

Transcriptional regulation of the stress response by mTOR`

This information is current as of 2 July 2014. The following resources related to this article are available online at http://stke.sciencemag.org.

Article Tools http://stke.sciencemag.org/cgi/content/full/sigtrans;7/332/re2

Visit the online version of this article to access the personalization and article tools:

References http://stke.sciencemag.org/cgi/content/full/sigtrans;7/332/re2#otherarticles

This article cites 187 articles, 82 of which can be accessed for free:

Glossary http://stke.sciencemag.org/glossary/

Look up definitions for abbreviations and terms found in this article:

Permissions http://www.sciencemag.org/about/permissions.dtl

Obtain information about reproducing this article:

the American Association for the Advancement of Science; all rights reserved. byAssociation for the Advancement of Science, 1200 New York Avenue, NW, Washington, DC 20005. Copyright 2008

(ISSN 1937-9145) is published weekly, except the last week in December, by the AmericanScience Signaling

on July 2, 2014 stke.sciencem

ag.orgD

ownloaded from

on July 2, 2014

stke.sciencemag.org

Dow

nloaded from

R E V I E W

T R A N S C R I P T I O N

Transcriptional regulation of the stress responseby mTORJose Aramburu,1* M. Carmen Ortells,2 Sonia Tejedor,1 Maria Buxadé,1 Cristina López-Rodríguez1*

D

The kinase mammalian target of rapamycin (mTOR) is a central regulator of cell growth and prolifer-ation that integrates inputs from growth factor receptors, nutrient availability, intracellular ATP (aden-osine 5′-triphosphate), and a variety of stressors. Since early works in the mid-1990s uncovered the role ofmTOR in stimulating protein translation, this kinase has emerged as a rather multifaceted regulator ofnumerous processes. Whereas mTOR is generally activated by growth- and proliferation-stimulatingsignals, its activity can be reduced and even suppressed when cells are exposed to a variety of stressconditions. However, cells can also adapt to stress while maintaining their growth capacity and mTORfunction. Despite knowledge accumulated on how stress represses mTOR, less is known about mTOR in-fluencing stress responses. In this review, we discuss the capability of mTOR, in particular mTOR com-plex 1 (mTORC1), to activate stress-responsive transcription factors, and we outline open questions forfuture investigation.

ow

on July 2, 2014

stke.sciencemag.org

nloaded from

mTOR Complexes in Mammalian Cells

The serine/threonine kinase target of rapamycin (TOR) belongs to thefamily of phosphatidylinositol 3-kinase (PI3K)–related kinases (PIKKs)and is a main activator of biosynthetic processes needed for cell growthin all eukaryotic organisms (1). TOR is the most frequently used acronymfor this kinase, but earlier works also used the name FRAP, for FK506-binding protein 12–rapamycin–associated protein. The acronym mTORrefers to “mammalian TOR,” although recently “mechanistic target ofrapamycin” is commonly used. The kinase mTOR functions in larger mul-tiprotein complexes: mTOR complex 1 (mTORC1) and mTOR complex 2(mTORC2). mTORC1 is defined by regulatory-associated protein of mTOR(Raptor) (2, 3), whereas mTORC2 contains the proteins rapamycin-insensitive companion of mTOR (Rictor) and stress-activated mitogen-activated protein kinase (MAPK)–interacting protein 1 (Sin1) (4, 5). Theseand additional mTOR-interacting proteins regulate its activity and speci-ficity toward various substrates (6). mTORC1 is activated by growth fac-tors, nutrients, and energy and promotes cell growth by enhancing thetranslation rate of diverse proteins by activating the ribosomal S6 subunitkinases (S6K) 1 and 2 (1, 7, 8) and inactivating the translation repressorprotein eukaryotic translation initiation factor 4E (eIF4E)–binding protein1 (4E-BP1) (9). mTORC1 also enhances protein synthesis indirectly byincreasing the activity of RNA polymerases I and III, which transcribegenes encoding ribosomal and transfer RNAs (10).

In addition, mTOR influences diverse transcription factors and the ex-pression of gene products involved in the control of metabolism, ribosomalbiogenesis, growth, and proliferation (11–16). mTORC2 was originallyshown to be essential for the function of the actin cytoskeleton (4, 17),but is now also known to regulate cell growth, differentiation, proliferation,and lipid homeostasis (18, 19). At least part of the growth-promoting ac-tivity of mTORC2 is mediated by activating and stabilizing the kinase Akt[also known as protein kinase B (PKB)], which in turn enhances the ac-tivity of mTORC1 (20, 21). A defining feature of mTOR is its inhibitionby rapamycin (22, 23), a compound originally isolated from the bacterium

1Department of Experimental and Health Sciences, Universitat Pompeu Fabra,Barcelona 08003, Spain. 2Centre for Genomic Regulation and UniversitatPompeu Fabra, Barcelona 08003, Spain.*Corresponding author. E-mail: [email protected] (J.A.); [email protected] (C.L.-R.)

Streptomyces hygroscopicus that binds to the intracellular chaperoneFK506-binding protein 1A, 12 kD (FKBP12). The rapamycin-FKBP12complex in turn binds with high affinity to the FKBP12-rapamycin bind-ing (FRB) domain in TOR. This domain is accessible to the rapamycin-FKBP12 complex in mTORC1, but not in mTORC2. Structural studiesshow that mTORC1 complexes are dimeric, with two molecules of mTORand Raptor per complex (24). This work, together with earlier biochem-ical analysis, shows that rapamycin-FKBP12 alters the conformation ofthe mTOR-Raptor complex, weakening their interaction and destabilizingthe mTORC1 dimer (2, 24). Notably, activation of mTORC1 by nutrientscauses a conformational change in the mTOR-Raptor complex that makesit more sensitive to the destabilizing effect of rapamycin (2). Rapamycin-FKBP12 rapidly suppresses the activity of mTORC1 toward many, al-though not all, of its substrates (25–27). However, when cells are incubatedwith rapamycin for long periods (hours to days), rapamycin-FKBP12 canbind to newly synthesized mTOR before it can assemble into new mTORC1and mTORC2 complexes, precluding the regeneration of their pools andinhibiting both mTORC1- and mTORC2-dependent functions (28).

Stress-Sensitive Checkpoints in mTOR Activity

The activity of mTOR is sensitive to complex signaling networks, and thenumber of proteins that at some point can influence mTOR function isprobably in the hundreds (29). From this perspective, it is conceivable thatdiverse sources of stress may affect mTOR complexes by acting on differentcomponents in these networks, and indeed, there are several well-characterizedregulators and signaling circuits that inhibit the activity of mTOR understress (30–33) (Fig. 1, A and B).

A substantial part of mTORC1 inhibitory inputs are channeled throughthe tuberous sclerosis (TSC) proteins TSC1 and TSC2 (34, 35). mTORC1activity is primarily regulated by its associated protein Ras homolog en-riched in brain (Rheb), a small guanosine triphosphatase (GTPase) that,in its guanosine triphosphate (GTP)–loaded state, activates mTORC1. Rhebenables the activation of mTORC1 in response to growth factors and,together with the Ragulator complex, to amino acids (36), a process thatoccurs at the lysosome surface (37–39). TSC2 acts as a GTPase-activatingprotein (GAP) and promotes GTP hydrolysis to convert Rheb-GTP to Rheb-GDP (guanosine diphosphate), thereby inactivating mTORC1 (40). TSC2exists in a heterodimeric complex together with TSC1, which has no GAP

www.SCIENCESIGNALING.org 1 July 2014 Vol 7 Issue 332 re2 1

R E V I E W

on July 2, 2014 stke.sciencem

ag.orgD

ownloaded from

activity but is needed to stabilize TSC2 (41, 42). The GAP activity of theTSC1/TSC2 complex is assisted by a third component of TSC, Tre2-Bub2-cdc16 (TBC) 1 domain family, member 7 (TBC1D7) (43). The GAPactivity of TSC2 is adjusted through inhibitory and activating phospho-rylations. TSC2 phosphorylation by the growth factor–activated kinaseAkt/PKB disrupts its association with TSC1 and causes the dissociationof the TSC complex from the lysosome, enabling the regeneration of Rheb-GTP and the assembly of mTORC1 with the Ragulator complex and Rheb(39, 44). TSC can also be inactivated through phosphorylation of TSC2 bythe p90 ribosomal protein S6 kinase a-1 (RSK1) (45). On the other hand,TSC is activated by phosphorylations mediated by glycogen synthase kinase3b (GSK3b) (46) and adenosine 5′monophosphate (AMP)–activated kinase(AMPK) in response to energy stress and reactive oxygen species (ROS)(47, 48). AMPK is a major sensor of energy stress and is activated inresponse to the increase in the intracellular ratio of AMP and adenosine5′-diphosphate (ADP) to adenosine 5′-triphosphate (ATP) (49–51). AnothermTORC1-suppressive mechanism under energy stress is the inhibitory

www.SCIENCESIGNALING.or

phosphorylation of Rheb by the complexformed by p38b with the p38-regulated/activated protein kinase [PRAK, also knownas the MAPK-activated protein kinase 5(MK5)] (52). TSC2 activity can also be en-hanced by the DNA damage and hypoxia-inducible protein regulated in developmentand DNA damage responses (REDD1), alsoknown as DNA damage–inducible transcript4 (DDIT4) (30, 53), which facilitates the dis-sociation of TSC2 from the scaffold protein14-3-3 (54). TSC2 is also activated by theREDD1 homolog REDD2 (30, 53) and bysestrins 1 and 2 induced by the transcrip-tion factor p53 in response to DNA damage(55). The activity of mTORC1 can also bedecreased under energy stress conditionsthrough the inhibitory phosphorylation ofRaptor at Ser722/792 by AMPK (56) and theAMPK-related kinase MAP/microtubuleaffinity-regulating kinase 4 (MARK4) (57).

TSC2-activating and inhibitory circuitsare also regulated by changes in the abun-dance of their components. For instance, inresponse to DNA damage, the tumor sup-pressor and transcriptional regulator p53can enhance the expression of genes encod-ing mTORC1 repressors, including TSC2,AMPK, REDD1, sestrin 2, and the PI3K an-tagonist phosphatase and tensin homolog(PTEN) (58). REDD1 is also induced in ap53-independent manner in response to en-ergy stress (59) and by the hypoxia-induciblefactor (HIF) during hypoxia (60, 61), inwhich mitochondrial ATP synthesis is com-promised while production of mitochondrialROS is enhanced.

Activation of mTORC2 depends on PI3Ksignaling (26, 62) and would be expectedto be sensitive to perturbations of this path-way under stress. Notably, mTORC2 is par-tially repressed, although not fully inactivated,upon phosphorylation of its essential compo-

nent Rictor by the mTORC1-activated S6K1 and by negative feedbackthrough mTORC1 on insulin receptor signaling (63–65). Another intriguingaspect of mTORC2 is its positive regulation by TSC2, an effect that is in-dependent of the GAP activity of TSC1/2 and inhibition of mTORC1 (66).The finding that mTORC1 can attenuate mTORC2 activity does not mean,though, that mTORC2 is not functioning in cells with active mTORC1. Rather,both complexes are active in growing and proliferating cells (26, 67) andshould be viewed as continuously communicating and balancing each other(Fig. 1B).

Regulation of Stress Response Transcription Factorsby mTOR

Adaptive mechanisms to survive and maintain functionality under stressoften involve changes in gene expression patterns. Although a large num-ber of transcription regulators are directly or indirectly sensitive to pertur-bations of the intracellular milieu, which in itself could be used by cells to

A

B C

mTOREnergy deficit

Glucose deprivation

Amino acid deficiency

Hypoxia

ROS

DNA damage

Heat shock

Osmotic stress

ER stress

Growth factor signaling

Nutrients (amino acids, glucose)

Energy sufficiencyNutrient uptake

Translation

Ribosome biogenesis

Biosynthesis

Proliferation

mTORC1

HIF-1Adaptation to

hypoxia

Nrf2Resistance to

ROS

HSF1Resistance to

heat stress

NFAT5Adaptation toosmotic stress

p53Genomeintegrity

FOXO Homeostasis

mTORC1

Rheb

Ragulator

MARK4PRAK

mTORC2

TSC2 AMPK

TSC2TSC1

Energy deficit

Growth factordeficiency

Amino aciddeficiency

Hypoxia

ROS

DNA damage

Fig. 1. mTOR sensitivity to stressors, and stress-responsive transcription factors stimulated by mTOR.

(A) mTOR integrates signaling from growth factor, nutrient, and energy sensors to stimulate growth-promoting processes and can be inhibited by diverse types of stress conditions. (B) A schematic viewof mTORC1 inhibition by the TSC1/2 complex in response to diverse stressors, by other negative reg-ulators (such as AMPK, PRAK, and MARK4) under energy stress, or through deactivation of the Ragu-lator complex under amino acid deprivation. (C) Stress-responsive transcription factors that can bestimulated by mTORC1.

g 1 July 2014 Vol 7 Issue 332 re2 2

R E V I E W

on July 2, 2014 stke.sciencem

ag.orgD

ownloaded from

react to various stressors, certain transcription factors have critical rolesin stress responses. For example, HIF-1a, ROS-activated nuclear factor(erythroid-derived 2)–like 2 (NFE2L2 or Nrf2), hypertonicity-activatednuclear factor of activated T cells 5 (NFAT5), heat shock response factor(HSF1), DNA damage–activated p53, and the homeostasis regulatorforkhead box O (FOXO) proteins orchestrate stress-specific adaptation re-sponses and are sensitive to mTOR activity (Fig. 1C).

mTORC1 and HIF-1aThe hypoxia response factor HIF is a heterodimer formed by a constitutivesubunit, HIF-1b, and an inducible subunit, HIF-1a. HIF-1a is continuous-ly synthesized, but under normoxic conditions, it is rapidly degraded (68).HIF-1b can also dimerize with the HIF-1a homolog HIF-2a, which is alsoregulated through continuous synthesis and degradation. Although bothHIF-1a and HIF-2a can activate hypoxia-protective responses, they donot have entirely overlapping functions (69, 70). Under normoxic condi-tions, HIF-1a is inhibited by two oxygen-dependent, ROS-sensitive typesof enzymes: prolyl hydroxylases (PHDs) and factor inhibiting HIF-1 (FIH).Both proteins use molecular oxygen and a-ketoglutarate to hydroxylatekey residues in HIF-1a. PHD-mediated hydroxylation of Pro402 andPro564 in HIF-1a targets it for ubiquitylation by the von Hippel–Lindau(VHL) E3 ubiquitin ligase complex and subsequent proteasomal degrada-tion. FIH hydroxylates Asn803, a modification that inhibits HIF-1 transcrip-tional activity, although it does not cause its degradation (71, 72). Hypoxiacauses the activation of HIF-1–dependent responses by suppressing PHDand FIH. Decreasing oxygen concentration to 0.5 to 2% causes a rapidaccumulation of HIF-1a and expression of its target genes (73, 74). Amajor effect of activating HIF-1a is the switch in the mode of energy produc-tion in the mitochondria from respiration-dependent to glycolysis-dependentATP synthesis (75). This aspect is quite interesting because it means thatHIF-1a is an effective regulator of energy and metabolite resources. In-deed, it is now clear that some cell types, such as activated lymphocytesand tumor cells, also use HIF-1a to boost glucose uptake and glycolysisduring normoxia as well to respond to increased biosynthetic and ener-getic demands (76, 77).

mTORC1 can enhance HIF-1a activity in both normoxia and hypoxia.During normoxia, and despite the activity of PHDs, mTORC1 facilitatesthe accumulation of HIF-1a in normal and tumor cells by increasing itstranslation rate (76–80). mTORC1 can also enhance HIF-1a protein abun-dance during hypoxia and chemically induced hypoxia-like conditions(81, 82) and increase its transcriptional activity under hypoxia by facilitat-ing the interaction of HIF-1a with the transcriptional coactivator p300(83). mTORC1 activity can be inhibited during hypoxia; one mechanismby which this occurs is through HIF-1a–induced expression of the geneencoding mTORC1 inhibitor REDD1 (54, 60, 84), which suggests recip-rocal regulation and a balance between mTOR activity and the intensity ofthe hypoxia response.

mTORC1 and Nrf2The transcription factor Nrf2 responds to increases in ROS concentrationsand enhances the expression of genes encoding diverse proteins involvedin maintaining cellular redox balance, such as glutathione-synthesizing en-zymes, thioredoxin reductases, or peroxiredoxins (85). The abundanceof Nrf2 is controlled by a repressor, Kelch-like ECH-associated protein1 (Keap1), which facilitates the ubiquitylation and subsequent proteasome-mediated degradation of Nrf2 (86–88). Increased intracellular ROS inducesthe oxidation of various cysteines in Keap1, triggering a conformation-al change that causes it to dissociate from Nrf2, which then escapes theubiquitylation-degradation cycle and induces antioxidant response genes(85). Activation of Nrf2 in the absence of overt oxidative stress can also

have important effects, as seen in tumors bearing mutations in eitherKEAP1 or NFE2L2 that disrupt the repression of Nrf2 by Keap1 (85, 89).Enhanced Nrf2 activity in tumors increases survival not only to endogenousROS but also to chemotherapeutic drugs and confers stronger antiapoptoticdefenses and cell proliferation capability (90).

Nrf2 is sensitive to mTOR activity, although different studies show dif-ferent effects of mTOR on Nrf2. Rapamycin represses Nrf2-dependentantioxidant responses in renal carcinoma cells (91), suggesting a positiveregulation by mTORC1. Activation of Nrf2 by its release from Keap1 andinduction of cytoprotective target genes are also enhanced by mTORC1during selective autophagy (92). However, a study in HepG2 hepatocarci-noma cells showed that although rapamycin reduced basal abundance ofNrf2, it did not inhibit its accumulation in response to (R)-a-lipoic acid, adithiol redox-active compound that induces Nrf2-dependent gene expres-sion (93), which suggests that oxidative stress can increase Nrf2 indepen-dently from mTORC1. On the other hand, prolonged treatment of humanfibroblasts with rapamycin reduced Keap1 abundance and enhanced Nrf2accumulation (94). Therefore, mTORC1 can have both positive and neg-ative effects on Nrf2 in different cell types and experimental settings. Aswill be discussed further in the following sections, mTORC1 can also en-hance ROS production in epithelial stem cells and hematopoietic stemcells (HSCs) (95, 96), which suggests that the potential protective functionof mTORC1 may be offset by its ROS-promoting effects in some cell typesand conditions.

mTORC1 and HSF1HSF1 is a heat stress–activated transcription factor that is ubiquitouslyexpressed in mammalian cells and exists as a monomeric inactive formin unstressed cells (97). Heat, as well as heat-independent stressors, such asH2O2, low pH, and hypoosmotic and hyperosmotic stress, activates HSF1by inducing its trimerization, which enables it to bind DNA (97). HSF1induces the expression of genes encoding various heat shock proteins(HSPs), including HSP70, to chaperone other proteins at risk of aggrega-tion and misfolding (98). In turn, accumulating HSP70 induces negativefeedback on HSF1 by binding HSF1 and repressing its transcriptional ac-tivity (99, 100). HSF1 can also play substantial roles in addition to its stress-responsive function, as shown by the recent identification of numerous genesregulated by HSF1 that support oncogenic processes in tumor cells (101).

Short-term heat shock has been shown to activate the kinase S6K down-stream of mTORC1 (102, 103) and enhance the transcriptional activity ofHSF1 and its induction of HSPs in a human cancer cell line by phosphoryl-ating it in Ser326 (103). The later study showed that heat shock–enhancedS6K1 phosphorylation and activation of HSF1 were rapamycin-sensitive, sug-gesting the involvement of mTORC1. Moreover, a recent screening fortumor-suppressing compounds showed that translation inhibitors, includ-ing rapamycin and other inhibitors of the PI3K-mTOR pathway, preventedHSF1 from binding DNA in various cancer cell types (104), which pro-vides another mechanism by which mTOR activity may enhance HSF1function. However, a study in Saccharomyces cerevisiae showed thatsustained activation of yeast HSF1 inhibited TOR signaling (105). Al-though both studies were done in very different organisms, they suggestthat mTORC1-activated HSF1 could negatively feedback on mTORC1to modulate its activity.

mTORC1 and NFAT5NFAT5 belongs to the family of proteins with a Rel-like DNA binding do-main, which also comprises calcineurin-activated NFAT1, NFAT2, NFAT3,and NFAT4 and nuclear factor kB (NF-kB) (106, 107). One of the mostextensively characterized functions of NFAT5 is its ability to protect cellsfrom osmotic stress caused by excessive extracellular tonicity, which it

www.SCIENCESIGNALING.org 1 July 2014 Vol 7 Issue 332 re2 3

R E V I E W

on July 2, 2014 stke.sciencem

ag.orgD

ownloaded from

does by inducing various chaperones, osmolite transporters, and en-zymes that enable cells to survive and adapt to prolonged osmotic stress(108, 109). Osmotic stress can inhibit the activity of mTORC1 and mTORC2complexes, although the degree of inhibition differs with stress intensityand duration. Exposure of diverse cell lines to intense, short-term osmoticstress (more than 600 mosmol/kg for up to 1 hour) causes a substantialdecrease in the phosphorylation of major mTORC1 and mTORC2 targets,such as S6K1 or Akt (110–112). Intriguingly, one work reported that a30-min pulse of hyperosmotic stress (800 mosmol/kg) enhanced the ac-tivity of S6K1 in fibroblasts, although whether this was due to increasedmTORC1 activity was not tested (113). Although the inhibitory effect ofosmostress on mTOR has been known for years (30, 31), the mechanismsinvolved have not been fully elucidated. The ability of AMPK to respondto diverse stressors, including osmotic stress (51), would make it a possi-ble candidate to inhibit mTORC1 under osmostress. However, recent stu-dies do not support a main role for AMPK in regulating mTORC1 in cellsexposed to hypertonicity (67, 114). Earlier studies showed that inhibitionof S6K1 and Akt in cells exposed to high, short-term osmostress was me-diated by a calyculin A–sensitive phosphatase (110, 111). In this regard, itwas recently shown that short-term osmotic stress activated the intrinsickinase activity of mTORC1 through the phosphorylation of Raptor byc-Jun N-terminal kinase (JNK), but still inhibited the phosphorylation ofS6K1 and 4E-BP1 in cells (114). Calyculin A abrogated the inhibition ofS6K1 and 4E-BP1 and enabled their robust phosphorylation by mTORC1under osmotic stress (114). These findings indicate that high-intensity os-motic stress can interfere with signaling pathways downstream of mTORC1without inhibiting its kinase activity. On the other hand, mammalian cellsexposed to milder hypertonic conditions (500 mosmol/kg) exhibited amoderate inhibition of mTOR signaling (67), but maintained the functionof mTOR complexes to sustain protein synthesis, cell growth, and cell pro-liferation under osmostress (67, 115).

An earlier study in yeast had already shown that TOR promoted sur-vival under salt stress (116). In mammalian cells, mTORC1 stimulated theexpression of several osmoprotective gene products under moderate osmo-stress and enhanced histone acetylation in their promoters and the re-cruitment of NFAT5 and RNA polymerase II (67). mTORC1 was found toinfluence the expression of a subset of NFAT5-regulated genes as well asseveral NFAT5-independent ones, suggesting that mTOR influences othermechanisms or factors in the osmotic stress response (67). One such NFAT5-dependent osmotic stress–responsive gene encodes REDD1, the abun-dance of which is increased at both the mRNA and protein levels by mTOR.REDD1 can inhibit mTORC1 activity in various stress contexts (54, 61),but intriguingly, its induction by osmotic stress does not appear to inhibitmTORC1 (67). Hypertonic stress can enhance mitochondrial ROS pro-duction (117, 118), which can inhibit mTORC1 (119). Because REDD1attenuates the generation of mitochondrial ROS (120), it is possible thatthe mTORC1 inhibitory function of REDD1 under osmotic stress couldbe compensated by a protective antioxidant effect.

mTORC1 and p53The tumor suppressor transcription factor p53 is well known for its respon-siveness to DNA damage and its ability to prevent cellular transformationby inducing gene products that control the cell cycle, DNA repair, senes-cence, and apoptosis (121–123). Among other targets, p53 induces the ex-pression of various genes encoding proteins capable of inhibiting mTORactivity directly or indirectly [including PTEN, AMPK, TSC2, REDD1,sestrins, and TIGAR (TP53-induced glycolysis and apoptosis regulator)(58)], thus slowing down cell growth and attenuating biosynthetic pro-cesses that could favor the expansion of altered cells. Through AMPK-mediated phosphorylation of p53 at Ser15, p53 can also repress growth

and proliferation under energy stress and in the absence of DNA damage(124). Besides these stress-activated functions, p53 can influence mTORactivity by modulating glucose utilization and energy production byinhibiting various steps in glycolysis and enhancing mitochondrial res-piration (125) and by providing antioxidant defenses through the in-duction of proteins that are involved in ROS neutralization, such assestrins (126, 127).

As found for other stress-responsive transcription factors, p53 is alsosensitive to mTORC1 activity. mTORC1 can increase p53 activity by en-hancing its translation rate (128, 129) or by activating S6K1 that then se-questers and neutralizes the E3 ubiquitin protein ligase mouse doubleminute 2 (MDM2), a p53 repressor (130). It is interesting that increasedactivation of mTOR in TSC- or REDD1-deficient cells increases their sen-sitivity to stressors that activate p53; thus, in a sense, mTORC1 can beviewed as an amplifier of the proapoptotic function of p53 under stress.A similar stress sensitization mechanism in TSC1 mutant cells functionsthrough the enhanced mTORC1-dependent translation of alternative read-ing frame (ARF), a cell cycle repressor and p53 activator (131). In addition,mTORC1 can enhance p53-dependent induction of replicative senescencein cells subjected to moderate DNA damage and also in nonstressed cells(132, 133). The stimulatory effect of mTORC1 on p53 could be one reasonwhy TSC-deficient cells that maintain p53 functionality mostly give rise tobenign tumors (128, 134).

mTOR complexes and FOXOTranscription factors of the FOXO family are important regulators ofcellular homeostasis that play protective roles under diverse types of stressbut can also enhance cell death mechanisms (135). In different contexts,FOXO transcription factors can induce the expression of genes encodingproteins that arrest the cell cycle, activate apoptosis, enhance antioxidantresponses, promote autophagy, or modulate receptor signaling, includingsignaling in mTOR-mediated pathways (135).

Stress conditions that stimulate FOXO activity are generally inhibitoryfor mTOR complexes. Several kinases that activate FOXO, such as JNKin response to ROS (136), AMPK under energy stress (137, 138), and MK5(or PRAK) in response to DNA damage (139), can either inhibit mTORC1directly or inhibit upstream signaling to mTORC1 and mTORC2. FOXO canalso oppose mTORC1 by inducing its negative regulator sestrin 3 (140). Con-versely, by activating Akt (141, 142) or serum- and glucocorticoid-regulatedkinase 1 (SGK1) (143, 144), mTORC2 represses FOXO activity. In glio-blastoma tumors, mTORC2 also opposes FOXO by inhibiting its deacet-ylation by Sirt1 (145). Intriguingly, FOXO promotes the expression of thegene encoding PI3K and enhances PI3K signaling in certain tumor cells(146) as well as the expression of the gene encoding the mTORC2 com-ponent Rictor (140), which suggests that FOXO can induce its own neg-ative feedback through the expression of several repressors.

However, although these observations reveal some degree of antago-nism between FOXO and both mTOR complexes, there are also specificcontexts in which FOXO can benefit from the ability of mTORC1 to at-tenuate mTORC2. In T lymphocytes, excess mTORC1 activation throughdeletion of TSC1 leads to reduced mTORC2 function along with a sub-stantial reduction in mTORC2 and Akt-dependent repressive phosphorylationof FOXO1 and FOXO3 in response to T cell receptor (TCR) activation (147).TSC1-deficient T lymphocytes also exhibit a stronger induction of apoptosisupon TCR activation and greater expression of several FOXO-regulatedgenes encoding proteins that promote apoptosis and cell cycle arrest, suchas Bcl-2–like protein 11 (BCL2L11) and cyclin-dependent kinase inhibi-tor 1A (CDKN1A, also known as p21Cip1) (147). Additionally, rapamycintreatment in human colon cancer cell lines inactivates FOXO concomitant-ly with increased Akt activity (148).

www.SCIENCESIGNALING.org 1 July 2014 Vol 7 Issue 332 re2 4

R E V I E W

Negative effects of mTOR during stress responses

Although mTORC1 can be beneficial for the stress response, augmentedmTORC1 activity can be detrimental. For instance, TSC2-deficient cellscannot shut down mTORC1 when their DNA is damaged and die fasterthan wild-type cells because mTORC1 enhances the accumulation of p53(128). Likewise, TSC2-deficient cells poorly survive glucose depriva-tion because they cannot cope with the energy drain caused by mTORC1-

on July 2, 2014 stke.sciencem

ag.orgD

ownloaded from

driven protein synthesis (149). Studies inflies show that enhancing TORC1 activityby overexpressing Rheb increases theirsensitivity to oxidative stress (150), and otherstudies in flies and mice reveal that excessTORC1 activity worsens neuronal deteri-oration caused by huntingtin aggregates,whereas inhibition of TORC1 activates anautophagic response that clears protein ag-gregates and improves neuron survival(151, 152). These findings indicate that ex-cessive mTORC1 activity could overloadmechanisms of protein quality control, asconfirmed in a recent article showing thatTSC2-deficient fibroblasts exhibit de-creased mRNA translation fidelity (153).

Increased activation of mTORC1 by thesuppression of TSC1 or overexpression ofRheb also impairs hematopoiesis and self-renewal of HSCs (96, 154). This repressiveeffect correlates with increased mitochon-drial biogenesis and increased amounts ofROS, and the neutralization of ROS re-stores HSC function in Tsc1-deficient mice(96). mTORC1 also increases ROS in nor-mal oral keratinocytes and epithelial stemcells by decreasing the abundance of mito-chondrial manganese superoxide dismutase(MnSOD) (95). These effects seem at oddswith the ability of mTORC1 to activate anti-oxidant defenses through Nrf2 describedin other cells (92), suggesting that the bal-ance between the ROS-protective and ROS-promoting activities of mTORC1 may differsubstantially in different cell types and mi-croenvironments.

If reducing mTORC1 activity can helpcells to resist certain stresses, is it then par-adoxical that mTORC1 can also stimulateprotective stress responses? Both possibil-ities can be reconciled by proposing that par-tial suppression of mTORC1 may protectstressed cells by reducing the rate of bio-synthesis and the cellular energy demandwhile enabling sufficient mTORC1 activi-ty to support prosurvival stress responses(Fig. 2, A and B). As described above, ex-amples where the sustained activation ofmTORC1 reduces resistance to stress includeits roles in increasing mitochondrial ROSproduction; enhancing p53’s antiproliferative,proapoptotic, and senescence-promoting

functions; exacerbating ATP depletion in nutrient-restricted cells; or disrupt-ing proteostasis through excessive protein translation (95, 128, 149, 153). Acommon theme in these effects may be the persistence of energy-drainingmTORC1-driven biosynthetic processes and the overloading of mechanismsfor eliminating endogenous noxious and waste products (Fig. 2C). On theother hand, the stimulation of stress resistance responses by mTORC1 isobserved in cells where mTORC1 is not constitutively augmented, underconditions that can either maintain or transiently enhance its activity [such

A

mTOR thresholdrequired forcell growth

mTORactivity

Stress response

Intense stressModerate stress

Time

mTORactivity

Stress response (i.e., NFAT5)

mTORactivity

Stress response

Moderate stress

RapamycinTorin1

Normally regulated mTOR

Stress Stress

Protective responsesare insufficient

to cope withfurther stress due

to overactive mTOR

ElevatedmTOR activity

ROS production

Biogenesis rate

ATP expenditure

p53

mTORC1

B Constitutively active mTORC

Partially active mTORcould enhance

protective responses:

HIF-1 HSF1

Nrf2 NFAT5

ReducedmTOR activity

ROS production

Biogenesis rate

ATP expenditure

mTORC1

Protection from stress Increased sensitivity to stress

Fig. 2. Potential scenario compatible with mTOR being partially inhibited under stress but still capable of

enhancing adaptive stress responses. (A) Moderate stress intensity can cause a partial suppression ofmTOR activity and initiate stress responses with induction of stress-adaptation genes. Even if mTORactivity fell below the threshold required to sustain rapid proliferation or growth rate, cells could stillhave sufficient mTOR to enhance protective responses and facilitate a faster adaptation to stress. Phar-macological mTOR inhibitors such as rapamycin or Torin1 can prevent mTOR from enhancing stress re-sponses, as shown for HIF-1a, HSF1, Nrf2, or NFAT5. High-intensity stress can suppress mTOR so that itcannot further stimulate some stress responses. In this model, lack of an enhanced stress responsemediated by mTOR may not be necessarily detrimental for cells, and they could survive stress—despiteinducing a less robust response—by decreasing high-energy expending and stress-sensitizing bio-synthetic processes. (B) In cells with normally regulated mTORC1, partial suppression of mTORC1 bystress could attenuate processes that can negatively affect stress resistance. Remaining mTORC1 activitywould contribute to enhance transcriptionally regulated stress protection programs. (C) By contrast, in cellsunable to suppress mTOR activity, such as TSCmutants or cells overexpressing Rheb, the combination ofstress with the inability to attenuate energy expenditure and the overloading of repair or disposal of dam-aged components would worsen sensitivity to stress. Even if some stress adaptation responses could bestimulated by mTORC1, they would not be sufficient to counteract stress. In addition, overactive mTORC1can enhance the activation of p53 in response to various stressors, driving cells to senescence or death.

www.SCIENCESIGNALING.org 1 July 2014 Vol 7 Issue 332 re2 5

R E V I E W

stke.sciencemD

ownloaded from

as hypoxia, xenophagy, and short-term heat shock (79, 82, 83, 92, 103)]or reduce it [such as hyperosmotic stress (67)]. These observations maysuggest that sustained, increased activity of mTORC1 sensitizes cells toseveral types of stress, whereas moderate, regulated activity of mTORC1would favor stress adaptation responses (Fig. 2, B and C). Here, it mustbe noted that stress conditions under which mTORC1 can stimulate tran-scriptional responses [such as hypoxia (82, 83), hyperosmotic stress (67),or DNA damage (130)] can also inhibit mTORC1 (60, 67, 112, 134). Thissuggests that the extent of mTORC1 activity under increasing stress inten-sity will be important for determining its capacity to enhance protectiveresponses. These examples suggest that careful analysis of mTOR func-tion under moderate stress conditions could uncover other mTOR-sensitivetranscriptionally regulated processes. For instance, severe nutrient depriva-tion or intense endoplasmic reticulum (ER) stress can suppress mTOR, buta milder intensity of these stressors could allow enough mTOR activity tosupport relevant cell functions. In this regard, mTORC1 antagonizes the in-duction of the ER stress–responsive factors activating transcription fac-tor 4 (ATF4), ATF6, and CCAAT/enhancer binding protein homologousprotein (CHOP) (155), and emerging evidence suggests that a bidirectionalcrosstalk exists between mTOR complexes and the unfolded protein re-sponse (156).

Toward Future Directions: A Place for mTOR Complexes in theMap of Stress Responses?

As discussed above, mTORC1 can stimulate a number of stress-activatedtranscriptional responses, and mTORC2 can also enhance certain stress re-sponses, as shown in yeast and mammalian tumors. In S. cerevisiae, TORC2enhances cell survival to low amounts of DNA damage by stimulating theactivity of the SGK1-related kinases Ypk1 and Ypk2 (157), and suppressionof TORC2 in Schizosaccharomyces pombe impairs survival under hydrogen

on July 2, 2014 ag.org

peroxide stress, salt stress, and DNA dam-age (158–160). Furthermore, in a positivefeedforward loop with Akt and the atypicalRIO kinases RIOK1 and RIOK2, mTORC2increases survival of mouse glioblastomacells to the DNA-damaging drugs doxoru-bicin and temozolomide (161).

Intensity and duration could be importantvariables in the effect of stress on mTOR ac-tivity and its capacity to enhance protectiveresponses. A given quantity of stress is likelyto have a different impact on cellular func-tions and elicit different responses depend-ing on a cell’s state of biosynthesis andgrowth; for example, cells undergoing activegrowth and intense metabolic activity maybenefit from the ability of mTOR to enhancestress survival responses. In this regard, it isinteresting that different types of tumorsoften exhibit augmented mTOR signaling(162) together with enhanced activity of somestress response factors like HIF-1 (163–165),Nrf2 (92, 166), or HSF (101, 103, 104). Also,mTOR hypomorphic mice have a lower in-cidence of spontaneous malignant tumors(167). Although this does not imply thatstronger mTOR signaling necessarily en-hances anti-stress capabilities in tumors, thecoexistence of elevated activities of mTOR

and various stress-responsive transcription factors in different tumorscan provide an interesting scenario to study how mTOR signaling maycontribute to cellular resilience—or fragility—under stress. These studiesmight also reveal an opportunistic weakness in tumors that are over-relianton sustained mTOR activity to survive stresses.

Elucidating how mTOR complexes influence the balance between re-sistance and enhanced sensitivity to stress might also aid understandingtheir role in aging. Reduction of mTOR activity by pharmacological orgenetic means increases life span in yeast, worms, flies, and mice, an ob-servation that opens exciting challenges regarding the possibility of slow-ing aging by manipulating mTOR (6, 168–171). The precise mechanismsunderlying the longevity phenotype in mice with reduced mTOR activityare still being elucidated, but it is already apparent that some processesbenefit more than others from lower mTOR activity (168). Mice withlow mTOR expression throughout their entire life, and therefore reducedactivity of both mTORC1 and mTORC2, show improved neuromotor andcognitive performance and a lower incidence of malignant tumors at oldage (167). Decreasing mTOR activity in old mice with rapamycin also im-proves self-renewal capacity of HSCs (172), which agrees with the findingthat overactive mTORC1 impairs HSC renewal (96, 154). However,complete lack of mTORC1 impairs HSC regeneration (173). It is also im-portant to note that mTOR-null organisms are not viable (174, 175), andthat decreasing global mTOR activity below a certain threshold can se-verely weaken immune defenses against infections (167), an observation thatunderscores extensive evidence of a fundamental role of mTOR complexesin immune responses (176–180).

Prolonging longevity in an organism requires a good coordination be-tween extending the life span of terminally differentiated cells, maintainingtheir proper function in tissues, and ensuring the replicative and differen-tiation potential of progenitor cells. These processes need not be equallysensitive to changes in mTOR activity. Because cells in any organism will

mTOR

mTOR

Cytoplasm

Nucleus

Inducible mTOR mutant(Rapamycin/Torin1-insensitive)

Inducible release from repressor

mTOR

Endogenous mTOR

RapamycinTorin1

Inducible promoter

mTOR

R

R

Fig. 3. Schematic outline of approaches to address mTOR function in stress responses. Potentially,

mTOR could be reversibly and quantifiably manipulated in cells engineered to express inducible re-combinant mTOR mutants resistant to either rapamycin or Torin1. Endogenous mTOR complexescould be inhibited with rapamycin or Torin1, and resistant mutants (represented with a red dot) couldbe either expressed from an inducible promoter or activated by release from a repressor (for instance,estrogen receptor) in a time-controlled manner. These approaches could use wild-type or TSC1/2-deficientcells, and one could design ectopic mTOR mutants that are capable of interacting only with Raptor orRictor, to selectively reactivate mTORC1 or mTORC2 functions.

www.SCIENCESIGNALING.org 1 July 2014 Vol 7 Issue 332 re2 6

R E V I E W

on July 2, 2014 stke.sciencem

ag.orgD

ownloaded from

endure different stressors to varying degrees throughout their life, it is pos-sible that cellular longevity may be influenced by the ability of mTOR com-plexes to regulate sensitivity and resistance to stress. Reduced mTORC1activity could attenuate cellular stress under aging-promoting conditions(such as DNA damage, perturbed proteostasis, or ROS) while simulta-neously being sufficient to enhance specific stress adaptation responses.However, this is but one among various possibilities and will need to beexperimentally tested.

Several conceptual and experimental questions arise regarding the rele-vance of mTOR-mediated regulation of stress responses and the mechan-isms involved. To know whether mTOR activity is predictive of a better orworse outcome under stress, it will be important to determine three things:(i) how cells quantitate stress and balance this information against mTORstimulatory inputs; (ii) which mTOR functions are more and less sensitive todefined stress conditions of varying intensity and duration; and (iii) howimportant it is for the cell to enhance specific response mechanisms in anmTOR-dependent manner to survive stress.

Identifying genes and proteins that are sensitive to varying degrees ofstimulation or inhibition of mTOR activity in different stress scenarios anddifferent cell types is technically feasible with current transcriptomic, ge-nomic, metabolomics, and proteomic tools (11, 15, 67, 181–183). It is alsopossible to identify and quantitate changes in posttranslational modifica-tions and the abundance of individual proteins associated with stress re-sponses that are regulated by mTOR (184). However, dissecting howmTOR’s contribution to stress responses may affect a cell’s behavior dur-ing and after stress has subsided poses several challenges. ManipulatingmTOR-sensitive stress-responsive genes or proteins could provide cluesabout how changes in their expression or activity would influence cellularbehavior under stress, but this could be difficult if the number of genesand proteins involved were relatively large, thus complicating conventionalapproaches that silence or overexpress candidate genes. However, this taskcould be simplified if mTOR-sensitive stress response networks could bedelineated, and their key nodes identified. Characterization of functionalnodes and links in these networks would enable the manipulation ofkey elements to dissect the contribution of specific pathways in mTOR-regulated stress responses. Also, devising tools that allowed reversible andquantitative modulation of mTOR activity at different stages along thestress response could be useful to understand how mTOR may influencecellular functions during and after stress. Alternating repression and acti-vation of mTOR in a time-controlled manner could be accomplished bycombining current chemical inhibitors, such as rapamycin or Torin1 (26),with inducible mTOR mutants engineered to be resistant to those inhibi-tors (Fig. 3). Rapamycin-resistant mTOR mutants already exist (185, 186),and the available structure of Torin1-bound mTOR (187) should facilitatethe design of Torin1-resistant mutants.

Concluding Remarks

The activity of mTOR complexes is sensitive to diverse forms of stress, whichin general means that cells under stress often suppress mTOR signaling.At least in the case of mTORC1, attenuating its activity can improve cell sur-vival under certain stress conditions, whereas abnormally increased mTORC1function during stress can be detrimental for the cell. On the other hand, bothmTORC1 and mTORC2 can also enhance cellular responses that supportadaptation and survival to stress, and stress-activated transcriptional regulatorssuch as HIF-1a, HSF1, NFAT5, p53, and FOXO are stimulated by mTORC1.Different findings suggest that the communication between mTOR and stressresponses may play an important role in cellular functions, tumor biology, andperhaps long-term fitness of the organism. Aside from identifying relevantmTOR targets in stress response pathways and the regulatory mechanisms

involved, it will be important to elucidate how these are regulated by the bal-ance between mTORC1 and mTORC2 activities. It will also be necessary todissect the role of mTOR complexes in stress separately from their otherfunctions, and to determine how the contribution of mTOR to stress responsesmay affect longer-term outcomes in cell survival, proliferative capacity, main-tenance of specialized functions, differentiation potential, or longevity.

REFERENCES AND NOTES1. D. C. Fingar, C. J. Richardson, A. R. Tee, L. Cheatham, C. Tsou, J. Blenis, mTOR

controls cell cycle progression through its cell growth effectors S6K1 and 4E-BP1/eukaryotic translation initiation factor 4E. Mol. Cell. Biol. 24, 200–216 (2004).

2. D. H. Kim, D. D. Sarbassov, S. M. Ali, J. E. King, R. R. Latek, H. Erdjument-Bromage,P. Tempst, D. M. Sabatini, mTOR interacts with raptor to form a nutrient-sensitivecomplex that signals to the cell growth machinery. Cell 110, 163–175 (2002).

3. K. Hara, Y. Maruki, X. Long, K. Yoshino, N. Oshiro, S. Hidayat, C. Tokunaga, J. Avruch,K. Yonezawa, Raptor, a binding partner of target of rapamycin (TOR), mediates TORaction. Cell 110, 177–189 (2002).

4. D. D. Sarbassov, S. M. Ali, D. H. Kim, D. A. Guertin, R. R. Latek, H. Erdjument-Bromage,P. Tempst, D. M. Sabatini, Rictor, a novel binding partner of mTOR, defines a rapamycin-insensitive and raptor-independent pathway that regulates the cytoskeleton.Curr. Biol. 14,1296–1302 (2004).

5. Q. Yang, K. Inoki, T. Ikenoue, K. L. Guan, Identification of Sin1 as an essential TORC2component required for complex formation and kinase activity. Genes Dev. 20,2820–2832 (2006).

6. R. Zoncu, A. Efeyan, D. M. Sabatini, mTOR: From growth signal integration to cancer,diabetes and ageing. Nat. Rev. Mol. Cell Biol. 12, 21–35 (2011).

7. X. M. Ma, J. Blenis, Molecular mechanisms of mTOR-mediated translational control.Nat. Rev. Mol. Cell Biol. 10, 307–318 (2009).

8. K. K. Lee-Fruman, C. J. Kuo, J. Lippincott, N. Terada, J. Blenis, Characterization ofS6K2, a novel kinase homologous to S6K1. Oncogene 18, 5108–5114 (1999).

9. K. Hara, K. Yonezawa, M. T. Kozlowski, T. Sugimoto, K. Andrabi, Q. P.Weng, M. Kasuga,I. Nishimoto, J. Avruch, Regulation of eIF-4E BP1 phosphorylation by mTOR. J. Biol.Chem. 272, 26457–26463 (1997).

10. C. Mayer, I. Grummt, Ribosome biogenesis and cell growth: mTOR coordinates tran-scription by all three classes of nuclear RNA polymerases. Oncogene 25, 6384–6391(2006).

11. T. Peng, T. R. Golub, D. M. Sabatini, The immunosuppressant rapamycin mimics astarvation-like signal distinct from amino acid and glucose deprivation. Mol. Cell. Biol.22, 5575–5584 (2002).

12. A. Grolleau, J. Bowman, B. Pradet-Balade, E. Puravs, S. Hanash, J. A. Garcia-Sanz,L. Beretta, Global and specific translational control by rapamycin in T cells uncovered bymicroarrays and proteomics. J. Biol. Chem. 277, 22175–22184 (2002).

13. M. J. James, J. C. Zomerdijk, Phosphatidylinositol 3-kinase and mTOR signaling path-ways regulate RNA polymerase I transcription in response to IGF-1 and nutrients. J. Biol.Chem. 279, 8911–8918 (2004).

14. R. H. Jimenez, J. S. Lee, M. Francesconi, G. Castellani, N. Neretti, J. A. Sanders, J. Sedivy,P. A. Gruppuso, Regulation of gene expression in hepatic cells by the mammalian targetof rapamycin (mTOR). PLOS One 5, e9084 (2010).

15. K. Düvel, J. L. Yecies, S. Menon, P. Raman, A. I. Lipovsky, A. L. Souza, E. Triantafellow,Q. Ma, R. Gorski, S. Cleaver, M. G. Vander Heiden, J. P. MacKeigan, P.M. Finan, C. B. Clish,L. O. Murphy, B. D. Manning, Activation of a metabolic gene regulatory network down-stream of mTOR complex 1. Mol. Cell 39, 171–183 (2010).

16. M. Laplante, D. M. Sabatini, Regulation of mTORC1 and its impact on gene expressionat a glance. J. Cell Sci. 126, 1713–1719 (2013).

17. E. Jacinto, R. Loewith, A. Schmidt, S. Lin, M. A. Rüegg, A. Hall, M. N. Hall, MammalianTOR complex 2 controls the actin cytoskeleton and is rapamycin insensitive. Nat. CellBiol. 6, 1122–1128 (2004).

18. J. D. Powell, G. M. Delgoffe, The mammalian target of rapamycin: Linking T cell differ-entiation, function, and metabolism. Immunity 33, 301–311 (2010).

19. D. W. Lamming, D. M. Sabatini, A central role for mTOR in lipid homeostasis. Cell Metab.18, 465–469 (2013).

20. E. Jacinto, V. Facchinetti, D. Liu, N. Soto, S. Wei, S. Y. Jung, Q. Huang, J. Qin, B. Su,SIN1/MIP1 maintains rictor-mTOR complex integrity and regulates Akt phosphorylationand substrate specificity. Cell 127, 125–137 (2006).

21. W. J. Oh, C. C. Wu, S. J. Kim, V. Facchinetti, L. A. Julien, M. Finlan, P. P. Roux, B. Su,E. Jacinto, mTORC2 can associate with ribosomes to promote cotranslational phospho-rylation and stability of nascent Akt polypeptide. EMBO J. 29, 3939–3951 (2010).

22. N. C. Barbet, U. Schneider, S. B. Helliwell, I. Stansfield, M. F. Tuite, M. N. Hall, TOR con-trols translation initiation and early G1 progression in yeast. Mol. Biol. Cell 7, 25–42(1996).

23. L. Beretta, A. C. Gingras, Y. V. Svitkin, M. N. Hall, N. Sonenberg, Rapamycin blocksthe phosphorylation of 4E-BP1 and inhibits cap-dependent initiation of translation.EMBO J. 15, 658–664 (1996).

www.SCIENCESIGNALING.org 1 July 2014 Vol 7 Issue 332 re2 7

R E V I E W

on July 2, 2014 stke.sciencem

ag.orgD

ownloaded from

24. C. K. Yip, K. Murata, T. Walz, D. M. Sabatini, S. A. Kang, Structure of the humanmTOR complex I and its implications for rapamycin inhibition. Mol. Cell 38, 768–774(2010).

25. A. Y. Choo, S. O. Yoon, S. G. Kim, P. P. Roux, J. Blenis, Rapamycin differentially inhib-its S6Ks and 4E-BP1 to mediate cell-type-specific repression of mRNA translation.Proc. Natl. Acad. Sci. U.S.A. 105, 17414–17419 (2008).

26. C. C. Thoreen, S. A. Kang, J. W. Chang, Q. Liu, J. Zhang, Y. Gao, L. J. Reichling, T. Sim,D. M. Sabatini, N. S. Gray, An ATP-competitive mammalian target of rapamycin inhibitorreveals rapamycin-resistant functions of mTORC1. J. Biol. Chem. 284, 8023–8032(2009).

27. S. A. Kang, M. E. Pacold, C. L. Cervantes, D. Lim, H. J. Lou, K. Ottina, N. S. Gray,B. E. Turk, M. B. Yaffe, D. M. Sabatini, mTORC1 phosphorylation sites encode theirsensitivity to starvation and rapamycin. Science 341, 1236566 (2013).

28. D. D. Sarbassov, S. M. Ali, S. Sengupta, J. H. Sheen, P. P. Hsu, A. F. Bagley, A. L. Markhard,D. M. Sabatini, Prolonged rapamycin treatment inhibits mTORC2 assembly and Akt/PKB.Mol. Cell 22, 159–168 (2006).

29. E. Caron, S. Ghosh, Y. Matsuoka, D. Ashton-Beaucage, M. Therrien, S. Lemieux,C. Perreault, P. P. Roux, H. Kitano, A comprehensive map of the mTOR signaling net-work. Mol. Syst. Biol. 6, 453 (2010).

30. M. N. Corradetti, K. L. Guan, Upstream of the mammalian target of rapamycin: Do allroads pass through mTOR? Oncogene 25, 6347–6360 (2006).

31. J. H. Reiling, D. M. Sabatini, Stress and mTORture signaling.Oncogene 25, 6373–6383(2006).

32. S. Sengupta, T. R. Peterson, D. M. Sabatini, Regulation of the mTOR complex 1 path-way by nutrients, growth factors, and stress. Mol. Cell 40, 310–322 (2010).

33. C. M. Hung, L. Garcia-Haro, C. A. Sparks, D. A. Guertin, mTOR-dependent cell survivalmechanisms. Cold Spring Harb. Perspect. Biol. 4, a008771 (2012).

34. J. Huang, B. D. Manning, A complex interplay between Akt, TSC2 and the twomTOR complexes. Biochem. Soc. Trans. 37, 217–222 (2009).

35. X. Wang, C. G. Proud, mTORC1 signaling: What we still don’t know. J. Mol. Cell Biol.3, 206–220 (2011).

36. A. Efeyan, D. M. Sabatini, Nutrients and growth factors in mTORC1 activation. Biochem.Soc. Trans. 41, 902–905 (2013).

37. Y. Sancak, L. Bar-Peled, R. Zoncu, A. L. Markhard, S. Nada, D. M. Sabatini, Ragulator-Rag complex targets mTORC1 to the lysosomal surface and is necessary for its acti-vation by amino acids. Cell 141, 290–303 (2010).

38. L. Bar-Peled, L. D. Schweitzer, R. Zoncu, D. M. Sabatini, Ragulator is a GEF for theRag GTPases that signal amino acid levels to mTORC1. Cell 150, 1196–1208 (2012).

39. S. Menon, C. C. Dibble, G. Talbott, G. Hoxhaj, A. J. Valvezan, H. Takahashi, L. C. Cantley,B. D. Manning, Spatial control of the TSC complex integrates insulin and nutrient reg-ulation of mTORC1 at the lysosome. Cell 156, 771–785 (2014).

40. K. Inoki, Y. Li, T. Xu, K. L. Guan, Rheb GTPase is a direct target of TSC2 GAP activityand regulates mTOR signaling. Genes Dev. 17, 1829–1834 (2003).

41. G. Benvenuto, S. Li, S. J. Brown, R. Braverman, W. C. Vass, J. P. Cheadle, D. J. Halley,J. R. Sampson, R. Wienecke, J. E. DeClue, The tuberous sclerosis-1 (TSC1) geneproduct hamartin suppresses cell growth and augments the expression of the TSC2product tuberin by inhibiting its ubiquitination. Oncogene 19, 6306–6316 (2000).

42. H. Chong-Kopera, K. Inoki, Y. Li, T. Zhu, F. R. Garcia-Gonzalo, J. L. Rosa, K. L. Guan,TSC1 stabilizes TSC2 by inhibiting the interaction between TSC2 and the HERC1ubiquitin ligase. J. Biol. Chem. 281, 8313–8316 (2006).

43. C. C. Dibble, W. Elis, S. Menon, W. Qin, J. Klekota, J. M. Asara, P. M. Finan,D. J. Kwiatkowski, L. O. Murphy, B. D. Manning, TBC1D7 is a third subunit of the TSC1-TSC2 complex upstream of mTORC1. Mol. Cell 47, 535–546 (2012).

44. K. Inoki, Y. Li, T. Zhu, J. Wu, K. L. Guan, TSC2 is phosphorylated and inhibited byAkt and suppresses mTOR signalling. Nat. Cell Biol. 4, 648–657 (2002).

45. P. P. Roux, B. A. Ballif, R. Anjum, S. P. Gygi, J. Blenis, Tumor-promoting phorbol estersand activated Ras inactivate the tuberous sclerosis tumor suppressor complex via p90ribosomal S6 kinase. Proc. Natl. Acad. Sci. U.S.A. 101, 13489–13494 (2004).

46. K. Inoki, H. Ouyang, T. Zhu, C. Lindvall, Y. Wang, X. Zhang, Q. Yang, C. Bennett,Y. Harada, K. Stankunas, C. Y. Wang, X. He, O. A. MacDougald, M. You, B. O. Williams,K. L. Guan, TSC2 integrates Wnt and energy signals via a coordinated phosphorylationby AMPK and GSK3 to regulate cell growth. Cell 126, 955–968 (2006).

47. K. Inoki, T. Zhu, K. L. Guan, TSC2 mediates cellular energy response to control cellgrowth and survival. Cell 115, 577–590 (2003).

48. A. Alexander, S. L. Cai, J. Kim, A. Nanez, M. Sahin, K. H. MacLean, K. Inoki, K. L. Guan,J. Shen, M. D. Person, D. Kusewitt, G. B. Mills, M. B. Kastan, C. L. Walker, ATM signalsto TSC2 in the cytoplasm to regulate mTORC1 in response to ROS. Proc. Natl. Acad.Sci. U.S.A. 107, 4153–4158 (2010).

49. J. S. Oakhill, R. Steel, Z. P. Chen, J. W. Scott, N. Ling, S. Tam, B. E. Kemp, AMPK isa direct adenylate charge-regulated protein kinase. Science 332, 1433–1435 (2011).

50. D. G. Hardie, D. Carling, S. J. Gamblin, AMP-activated protein kinase: Also regulated byADP? Trends Biochem. Sci. 36, 470–477 (2011).

51. D. G. Hardie, The AMP-activated protein kinase pathway—New players upstream anddownstream. J. Cell Sci. 117, 5479–5487 (2004).

52. M. Zheng, Y. H. Wang, X. N. Wu, S. Q. Wu, B. J. Lu, M. Q. Dong, H. Zhang, P. Sun,S. C. Lin, K. L. Guan, J. Han, Inactivation of Rheb by PRAK-mediated phosphorylationis essential for energy-depletion-induced suppression of mTORC1. Nat. Cell Biol. 13,263–272 (2011).

53. M. N. Corradetti, K. Inoki, N. Bardeesy, R. A. DePinho, K. L. Guan, Regulation of theTSC pathway by LKB1: Evidence of a molecular link between tuberous sclerosiscomplex and Peutz-Jeghers syndrome. Genes Dev. 18, 1533–1538 (2004).

54. M. P. DeYoung, P. Horak, A. Sofer, D. Sgroi, L. W. Ellisen, Hypoxia regulates TSC1/2–mTOR signaling and tumor suppression through REDD1-mediated 14–3–3 shuttling.Genes Dev. 22, 239–251 (2008).

55. A. V. Budanov, M. Karin, p53 target genes sestrin1 and sestrin2 connect genotoxicstress and mTOR signaling. Cell 134, 451–460 (2008).

56. D. M. Gwinn, D. B. Shackelford, D. F. Egan, M. M. Mihaylova, A. Mery, D. S. Vasquez,B. E. Turk, R. J. Shaw, AMPK phosphorylation of raptor mediates a metabolic check-point. Mol. Cell 30, 214–226 (2008).

57. L. Li, K. L. Guan, Microtubule-associated protein/microtubule affinity-regulating kinase 4(MARK4) is a negative regulator of themammalian target of rapamycin complex 1 (mTORC1).J. Biol. Chem. 288, 703–708 (2013).

58. Z. Feng, A. J. Levine, The regulation of energy metabolism and the IGF-1/mTOR path-ways by the p53 protein. Trends Cell Biol. 20, 427–434 (2010).

59. A. Sofer, K. Lei, C. M. Johannessen, L. W. Ellisen, Regulation of mTOR and cell growthin response to energy stress by REDD1. Mol. Cell. Biol. 25, 5834–5845 (2005).

60. J. Brugarolas, K. Lei, R. L. Hurley, B. D. Manning, J. H. Reiling, E. Hafen, L. A. Witters,L. W. Ellisen, W. G. Kaelin Jr., Regulation of mTOR function in response to hypoxia byREDD1 and the TSC1/TSC2 tumor suppressor complex. Genes Dev. 18, 2893–2904(2004).

61. N. C. Wolff, S. Vega-Rubin-de-Celis, X. J. Xie, D. H. Castrillon, W. Kabbani, J. Brugarolas,Cell-type-dependent regulation of mTORC1 by REDD1 and the tumor suppressorsTSC1/TSC2 and LKB1 in response to hypoxia. Mol. Cell. Biol. 31, 1870–1884 (2011).

62. X. Gan, J. Wang, B. Su, D. Wu, Evidence for direct activation of mTORC2 kinase ac-tivity by phosphatidylinositol 3,4,5-trisphosphate. J. Biol. Chem. 286, 10998–11002(2011).

63. C. C. Dibble, J. M. Asara, B. D. Manning, Characterization of Rictor phosphorylation sitesreveals direct regulation of mTOR complex 2 by S6K1. Mol. Cell. Biol. 29, 5657–5670(2009).

64. C. Treins, P. H. Warne, M. A. Magnuson, M. Pende, J. Downward, Rictor is a novel targetof p70 S6 kinase-1. Oncogene 29, 1003–1016 (2010).

65. L. A. Julien, A. Carriere, J. Moreau, P. P. Roux, mTORC1-activated S6K1 phosphoryl-ates Rictor on threonine 1135 and regulates mTORC2 signaling. Mol. Cell. Biol. 30,908–921 (2010).

66. J. Huang, C. C. Dibble, M. Matsuzaki, B. D. Manning, The TSC1-TSC2 complex is re-quired for proper activation of mTOR complex 2. Mol. Cell. Biol. 28, 4104–4115 (2008).

67. M. C. Ortells, B. Morancho, K. Drews-Elger, B. Viollet, K. R. Laderoute, C. López-Rodríguez,J. Aramburu, Transcriptional regulation of gene expression during osmotic stress re-sponses by the mammalian target of rapamycin. Nucleic Acids Res. 40, 4368–4384(2012).

68. M. C. Brahimi-Horn, J. Pouysségur, HIF at a glance. J. Cell Sci. 122, 1055–1057 (2009).69. M. Y. Koh, G. Powis, Passing the baton: The HIF switch. Trends Biochem. Sci. 37,

364–372 (2012).70. A. Loboda, A. Jozkowicz, J. Dulak, HIF-1 versus HIF-2—Is one more important than

the other? Vascul. Pharmacol. 56, 245–251 (2012).71. W. G. Kaelin, Proline hydroxylation and gene expression. Annu. Rev. Biochem. 74,

115–128 (2005).72. W. G. Kaelin Jr., P. J. Ratcliffe, Oxygen sensing by metazoans: The central role of the

HIF hydroxylase pathway. Mol. Cell 30, 393–402 (2008).73. H. K. Eltzschig, P. Carmeliet, Hypoxia and inflammation. N. Engl. J. Med. 364, 656–665

(2011).74. G. L. Semenza, Oxygen sensing, homeostasis, and disease. N. Engl. J. Med. 365,

537–547 (2011).75. G. L. Semenza, Hypoxia-inducible factors in physiology and medicine. Cell 148,

399–408 (2012).76. R. J. DeBerardinis, J. J. Lum, G. Hatzivassiliou, C. B. Thompson, The biology of cancer:

Metabolic reprogramming fuels cell growth and proliferation. Cell Metab. 7, 11–20(2008).

77. L. Z. Shi, R. Wang, G. Huang, P. Vogel, G. Neale, D. R. Green, H. Chi, HIF1a-dependentglycolytic pathway orchestrates a metabolic checkpoint for the differentiation of TH17and Treg cells. J. Exp. Med. 208, 1367–1376 (2011).

78. J. B. Brugarolas, F. Vazquez, A. Reddy, W. R. Sellers, W. G. Kaelin Jr., TSC2 regulatesVEGF through mTOR-dependent and -independent pathways. Cancer Cell 4, 147–158(2003).

79. P. K. Majumder, P. G. Febbo, R. Bikoff, R. Berger, Q. Xue, L. M. McMahon, J. Manola,J. Brugarolas, T. J. McDonnell, T. R. Golub, M. Loda, H. A. Lane, W. R. Sellers, mTORinhibition reverses Akt-dependent prostate intraepithelial neoplasia through regulation ofapoptotic and HIF-1-dependent pathways. Nat. Med. 10, 594–601 (2004).

www.SCIENCESIGNALING.org 1 July 2014 Vol 7 Issue 332 re2 8

R E V I E W

on July 2, 2014 stke.sciencem

ag.orgD

ownloaded from

80. H. Nakamura, Y. Makino, K. Okamoto, L. Poellinger, K. Ohnuma, C. Morimoto, H. Tanaka,TCR engagement increases hypoxia-inducible factor-1a protein synthesis via rapamycin-sensitive pathway under hypoxic conditions in human peripheral T cells. J. Immunol.174, 7592–7599 (2005).

81. C. C. Hudson, M. Liu, G. G. Chiang, D. M. Otterness, D. C. Loomis, F. Kaper, A. J. Giaccia,R. T. Abraham, Regulation of hypoxia-inducible factor 1a expression and function by themammalian target of rapamycin. Mol. Cell. Biol. 22, 7004–7014 (2002).

82. G. Yuan, J. Nanduri, S. Khan, G. L. Semenza, N. R. Prabhakar, Induction of HIF-1a ex-pression by intermittent hypoxia: Involvement of NADPH oxidase, Ca2+ signaling, prolylhydroxylases, and mTOR. J. Cell. Physiol. 217, 674–685 (2008).

83. S. C. Land, A. R. Tee, Hypoxia-inducible factor 1a is regulated by the mammalian targetof rapamycin (mTOR) via an mTOR signaling motif. J. Biol. Chem. 282, 20534–20543(2007).

84. L. W. Ellisen, Growth control under stress: mTOR regulation through the REDD1-TSCpathway. Cell Cycle 4, 1500–1502 (2005).

85. K. Taguchi, H. Motohashi, M. Yamamoto, Molecular mechanisms of the Keap1–Nrf2pathway in stress response and cancer evolution. Genes Cells 16, 123–140 (2011).

86. K. Itoh, N. Wakabayashi, Y. Katoh, T. Ishii, K. Igarashi, J. D. Engel, M. Yamamoto, Keap1represses nuclear activation of antioxidant responsive elements by Nrf2 through bindingto the amino-terminal Neh2 domain. Genes Dev. 13, 76–86 (1999).

87. K. R. Sekhar, X. X. Yan, M. L. Freeman, Nrf2 degradation by the ubiquitin proteasomepathway is inhibited by KIAA0132, the human homolog to INrf2.Oncogene 21, 6829–6834(2002).

88. H. K. Bryan, A. Olayanju, C. E. Goldring, B. K. Park, The Nrf2 cell defence pathway:Keap1-dependent and -independent mechanisms of regulation. Biochem. Pharmacol.85, 705–717 (2013).

89. M. B. Sporn, K. T. Liby, NRF2 and cancer: The good, the bad and the importance ofcontext. Nat. Rev. Cancer 12, 564–571 (2012).

90. P. Shelton, A. K. Jaiswal, The transcription factor NF-E2-related factor 2 (Nrf2): Aprotooncogene? FASEB J. 27, 414–423 (2013).

91. K. Bray, R. Mathew, A. Lau, J. J. Kamphorst, J. Fan, J. Chen, H. Y. Chen, A. Ghavami,M. Stein, R. S. DiPaola, D. Zhang, J. D. Rabinowitz, E. White, Autophagy suppressesRIP kinase-dependent necrosis enabling survival to mTOR inhibition. PLOS One 7,e41831 (2012).

92. Y. Ichimura, S. Waguri, Y. S. Sou, S. Kageyama, J. Hasegawa, R. Ishimura, T. Saito,Y. Yang, T. Kouno, T. Fukutomi, T. Hoshii, A. Hirao, K. Takagi, T. Mizushima, H. Motohashi,M. S. Lee, T. Yoshimori, K. Tanaka, M. Yamamoto, M. Komatsu, Phosphorylation ofp62 activates the Keap1-Nrf2 pathway during selective autophagy. Mol. Cell 51,618–631 (2013).

93. K. P. Shay, A. J. Michels, W. Li, A. N. Kong, T. M. Hagen, Cap-independent Nrf2 tran-slation is part of a lipoic acid-stimulated detoxification stress response. Biochim. Biophys.Acta 1823, 1102–1109 (2012).

94. C. Lerner, A. Bitto, D. Pulliam, T. Nacarelli, M. Konigsberg, H. Van Remmen, C. Torres,C. Sell, Reduced mammalian target of rapamycin activity facilitates mitochondrialretrograde signaling and increases life span in normal human fibroblasts. Aging Cell12, 966–977 (2013).

95. R. Iglesias-Bartolome, V. Patel, A. Cotrim, K. Leelahavanichkul, A. A. Molinolo, J. B. Mitchell,J. S. Gutkind, mTOR inhibition prevents epithelial stem cell senescence and protectsfrom radiation-induced mucositis. Cell Stem Cell 11, 401–414 (2012).

96. C. Chen, Y. Liu, R. Liu, T. Ikenoue, K. L. Guan, Y. Liu, P. Zheng, TSC–mTORmaintainsquiescence and function of hematopoietic stem cells by repressing mitochondrialbiogenesis and reactive oxygen species. J. Exp. Med. 205, 2397–2408 (2008).

97. J. Anckar, L. Sistonen, Regulation of HSF1 function in the heat stress response: Impli-cations in aging and disease. Annu. Rev. Biochem. 80, 1089–1115 (2011).

98. M. Fujimoto, A. Nakai, The heat shock factor family and adaptation to proteotoxic stress.FEBS J. 277, 4112–4125 (2010).

99. Y. Shi, D. D. Mosser, R. I. Morimoto, Molecular chaperones as HSF1-specific tran-scriptional repressors. Genes Dev. 12, 654–666 (1998).

100. A. V. Gómez, D. Galleguillos, J. C. Maass, E. Battaglioli, M. Kukuljan, M. E. Andrés,CoREST represses the heat shock response mediated by HSF1.Mol. Cell 31, 222–231(2008).

101. M. L. Mendillo, S. Santagata, M. Koeva, G. W. Bell, R. Hu, R. M. Tamimi, E. Fraenkel,T. A. Ince, L. Whitesell, S. Lindquist, HSF1 drives a transcriptional program distinctfrom heat shock to support highly malignant human cancers. Cell 150, 549–562(2012).

102. D. A. Jurivich, J. Chung, J. Blenis, Heat shock induces two distinct S6 protein kinaseactivities in quiescent mammalian fibroblasts. J. Cell. Physiol. 148, 252–259 (1991).

103. S. D. Chou, T. Prince, J. Gong, S. K. Calderwood, mTOR is essential for the proteotoxicstress response, HSF1 activation and heat shock protein synthesis. PLOS One 7,e39679 (2012).

104. S. Santagata, M. L. Mendillo, Y. C. Tang, A. Subramanian, C. C. Perley, S. P. Roche,B. Wong, R. Narayan, H. Kwon, M. Koeva, A. Amon, T. R. Golub, J. A. Porco Jr., L.Whitesell,S. Lindquist, Tight coordination of protein translation and HSF1 activation supports theanabolic malignant state. Science 341, 1238303 (2013).

105. S. Bandhakavi, H. Xie, B. O’Callaghan, H. Sakurai, D. H. Kim, T. J. Griffin, Hsf1 acti-vation inhibits rapamycin resistance and TOR signaling in yeast revealed by combinedproteomic and genetic analysis. PLOS One 3, e1598 (2008).

106. C. Lopez-Rodriguez, J. Aramburu, A. S. Rakeman, A. Rao, NFAT5, a constitutively nu-clear NFAT protein that does not cooperate with Fos and Jun. Proc. Natl. Acad. Sci. U.S.A.96, 7214–7219 (1999).

107. H. Miyakawa, S. K. Woo, S. C. Dahl, J. S. Handler, H. M. Kwon, Tonicity-responsiveenhancer binding protein, a rel-like protein that stimulates transcription in response tohypertonicity. Proc. Natl. Acad. Sci. U.S.A. 96, 2538–2542 (1999).

108. J. Aramburu, K. Drews-Elger, A. Estrada-Gelonch, J. Minguillón, B. Morancho, V. Santiago,C. López-Rodríguez, Regulation of the hypertonic stress response and other cellularfunctions by the Rel-like transcription factor NFAT5. Biochem. Pharmacol. 72, 1597–1604(2006).

109. M. B. Burg, J. D. Ferraris, N. I. Dmitrieva, Cellular response to hyperosmotic stresses.Physiol. Rev. 87, 1441–1474 (2007).

110. L. A. Parrott, D. J. Templeton, Osmotic stress inhibits p70/85 S6 kinase through acti-vation of a protein phosphatase. J. Biol. Chem. 274, 24731–24736 (1999).

111. D. Chen, R. V. Fucini, A. L. Olson, B. A. Hemmings, J. E. Pessin, Osmotic shock inhibitsinsulin signaling by maintaining Akt/protein kinase B in an inactive dephosphorylatedstate. Mol. Cell. Biol. 19, 4684–4694 (1999).

112. S. Naegele, S. J. Morley, Molecular cross-talk betweenMEK1/2 and mTOR signaling duringrecovery of 293 cells from hypertonic stress. J. Biol. Chem. 279, 46023–46034 (2004).

113. J. Van der Kaay, M. Beck, A. Gray, C. P. Downes, Distinct phosphatidylinositol 3-kinaselipid products accumulate upon oxidative and osmotic stress and lead to differentcellular responses. J. Biol. Chem. 274, 35963–35968 (1999).

114. D. Kwak, S. Choi, H. Jeong, J. H. Jang, Y. Lee, H. Jeon, M. N. Lee, J. Noh, K. Cho,J. S. Yoo, D. Hwang, P. G. Suh, S. H. Ryu, Osmotic stress regulates mammalian targetof rapamycin (mTOR) complex 1 via c-Jun N-terminal kinase (JNK)-mediated Raptorprotein phosphorylation. J. Biol. Chem. 287, 18398–18407 (2012).

115. M. J. Clemens, A. Elia, S. J. Morley, Requirement for the eIF4E binding proteins for thesynergistic down-regulation of protein synthesis by hypertonic conditions and mTORinhibition. PLOS One 8, e71138 (2013).

116. J. L. Crespo, K. Daicho, T. Ushimaru, M. N. Hall, The GATA transcription factors GLN3and GAT1 link TOR to salt stress in Saccharomyces cerevisiae. J. Biol. Chem. 276,34441–34444 (2001).

117. T. Yang, A. Zhang, M. Honeggar, D. E. Kohan, D. Mizel, K. Sanders, J. R. Hoidal,J. P. Briggs, J. B. Schnermann, Hypertonic induction of COX-2 in collecting duct cellsby reactive oxygen species of mitochondrial origin. J. Biol. Chem. 280, 34966–34973(2005).

118. X. Zhou, J. D. Ferraris, Q. Cai, A. Agarwal, M. B. Burg, Increased reactive oxygen spe-cies contribute to high NaCl-induced activation of the osmoregulatory transcription factorTonEBP/OREBP. Am. J. Physiol. Renal Physiol. 289, F377–F385 (2005).

119. L. Liu, D. R. Wise, J. A. Diehl, M. C. Simon, Hypoxic reactive oxygen species regulatethe integrated stress response and cell survival. J. Biol. Chem. 283, 31153–31162(2008).

120. P. Horak, A. R. Crawford, D. D. Vadysirisack, Z. M. Nash, M. P. DeYoung, D. Sgroi,L. W. Ellisen, Negative feedback control of HIF-1 through REDD1-regulated ROS sup-presses tumorigenesis. Proc. Natl. Acad. Sci. U.S.A. 107, 4675–4680 (2010).

121. W. A. Freed-Pastor, C. Prives, Mutant p53: One name, many proteins. Genes Dev. 26,1268–1286 (2012).

122. V. Marcel, M. L. Dichtel-Danjoy, C. Sagne, H. Hafsi, D. Ma, S. Ortiz-Cuaran, M. Olivier,J. Hall, B. Mollereau, P. Hainaut, J. C. Bourdon, Biological functions of p53 isoformsthrough evolution: Lessons from animal and cellular models. Cell Death Differ. 18,1815–1824 (2011).

123. A. V. Gudkov, E. A. Komarova, Pathologies associated with the p53 response. ColdSpring Harb. Perspect. Biol. 2, a001180 (2010).

124. R. G. Jones, D. R. Plas, S. Kubek, M. Buzzai, J. Mu, Y. Xu,M. J. Birnbaum, C. B. Thompson,AMP-activated protein kinase induces a p53-dependent metabolic checkpoint.Mol. Cell18, 283–293 (2005).

125. S. Matoba, J. G. Kang, W. D. Patino, A. Wragg, M. Boehm, O. Gavrilova, P. J. Hurley,F. Bunz, P. M. Hwang, p53 regulates mitochondrial respiration. Science 312, 1650–1653(2006).

126. A. A. Sablina, A. V. Budanov, G. V. Ilyinskaya, L. S. Agapova, J. E. Kravchenko,P. M. Chumakov, The antioxidant function of the p53 tumor suppressor. Nat. Med.11, 1306–1313 (2005).

127. I. A. Olovnikov, J. E. Kravchenko, P. M. Chumakov, Homeostatic functions of the p53tumor suppressor: Regulation of energy metabolism and antioxidant defense. Semin.Cancer Biol. 19, 32–41 (2009).

128. C. H. Lee, K. Inoki, M. Karbowniczek, E. Petroulakis, N. Sonenberg, E. P. Henske,K. L. Guan, Constitutive mTOR activation in TSC mutants sensitizes cells to energystarvation and genomic damage via p53. EMBO J. 26, 4812–4823 (2007).

129. D. D. Vadysirisack, F. Baenke, B. Ory, K. Lei, L. W. Ellisen, Feedback control of p53translation by REDD1 and mTORC1 limits the p53-dependent DNA damage response.Mol. Cell. Biol. 31, 4356–4365 (2011).

www.SCIENCESIGNALING.org 1 July 2014 Vol 7 Issue 332 re2 9

R E V I E W

on July 2, 2014 stke.sciencem

ag.orgD

ownloaded from

130. K. P. Lai, W. F. Leong, J. F. Chau, D. Jia, L. Zeng, H. Liu, L. He, A. Hao, H. Zhang,D. Meek, C. Velagapudi, S. L. Habib, B. Li, S6K1 is a multifaceted regulator of Mdm2that connects nutrient status and DNA damage response. EMBO J. 29, 2994–3006(2010).

131. A. P. Miceli, A. J. Saporita, J. D. Weber, Hypergrowth mTORC1 signals translation-ally activate the ARF tumor suppressor checkpoint. Mol. Cell. Biol. 32, 348–364(2012).

132. O. V. Leontieva, M. V. Blagosklonny, DNA damaging agents and p53 do not causesenescence in quiescent cells, while consecutive re-activation of mTOR is associatedwith conversion to senescence. Aging 2, 924–935 (2010).

133. M. V. Astle, K. M. Hannan, P. Y. Ng, R. S. Lee, A. J. George, A. K. Hsu, Y. Haupt,R. D. Hannan, R. B. Pearson, AKT induces senescence in human cells via mTORC1and p53 in the absence of DNA damage: Implications for targeting mTOR duringmalignancy. Oncogene 31, 1949–1962 (2012).

134. P. Hasty, Z. D. Sharp, T. J. Curiel, J. Campisi, mTORC1 and p53: Clash of the gods?Cell Cycle 12, 20–25 (2013).

135. A. Eijkelenboom, B. M. Burgering, FOXOs: Signalling integrators for homeostasismaintenance. Nat. Rev. Mol. Cell Biol. 14, 83–97 (2013).

136. M. A. Essers, S. Weijzen, A. M. de Vries-Smits, I. Saarloos, N. D. de Ruiter, J. L. Bos,B. M. Burgering, FOXO transcription factor activation by oxidative stress mediated bythe small GTPase Ral and JNK. EMBO J. 23, 4802–4812 (2004).

137. E. L. Greer, P. R. Oskoui, M. R. Banko, J. M. Maniar, M. P. Gygi, S. P. Gygi, A. Brunet,The energy sensor AMP-activated protein kinase directly regulates the mammalianFOXO3 transcription factor. J. Biol. Chem. 282, 30107–30119 (2007).

138. F. Wang, C. B. Marshall, K. Yamamoto, G. Y. Li, G. M. Gasmi-Seabrook, H. Okada,T. W. Mak, M. Ikura, Structures of KIX domain of CBP in complex with two FOXO3atransactivation domains reveal promiscuity and plasticity in coactivator recruitment.Proc. Natl. Acad. Sci. U.S.A. 109, 6078–6083 (2012).

139. T. R. Kress, I. G. Cannell, A. B. Brenkman, B. Samans, M. Gaestel, P. Roepman,B. M. Burgering, M. Bushell, A. Rosenwald, M. Eilers, The MK5/PRAK kinase andMyc form a negative feedback loop that is disrupted during colorectal tumorigenesis.Mol. Cell 41, 445–457 (2011).

140. C. C. Chen, S. M. Jeon, P. T. Bhaskar, V. Nogueira, D. Sundararajan, I. Tonic, Y. Park,N. Hay, FoxOs inhibit mTORC1 and activate Akt by inducing the expression of Sestrin3and Rictor. Dev. Cell 18, 592–604 (2010).

141. D. R. Plas, C. B. Thompson, Akt activation promotes degradation of tuberin andFOXO3a via the proteasome. J. Biol. Chem. 278, 12361–12366 (2003).

142. C. Bouchard, J. Marquardt, A. Brás, R. H. Medema, M. Eilers, Myc-induced proliferationand transformation require Akt-mediated phosphorylation of FoxO proteins. EMBO J.23, 2830–2840 (2004).

143. A. Brunet, J. Park, H. Tran, L. S. Hu, B. A. Hemmings, M. E. Greenberg, Protein kinaseSGK mediates survival signals by phosphorylating the forkhead transcription factorFKHRL1 (FOXO3a). Mol. Cell. Biol. 21, 952–965 (2001).

144. J. M. García-Martínez, D. R. Alessi, mTOR complex 2 (mTORC2) controls hydrophobicmotif phosphorylation and activation of serum- and glucocorticoid-induced proteinkinase 1 (SGK1). Biochem. J. 416, 375–385 (2008).

145. K. Masui, K. Tanaka, D. Akhavan, I. Babic, B. Gini, T. Matsutani, A. Iwanami, F. Liu,G. R. Villa, Y. Gu, C. Campos, S. Zhu, H. Yang,W.H. Yong, T. F. Cloughesy, I. K. Mellinghoff,W. K. Cavenee, R. J. Shaw, P. S. Mischel, mTOR complex 2 controls glycolytic metab-olism in glioblastoma through FoxO acetylation and upregulation of c-Myc. Cell Metab.18, 726–739 (2013).

146. R. C. Hui, A. R. Gomes, D. Constantinidou, J. R. Costa, C. T. Karadedou, S. F. de Mattos,M. P. Wymann, J. J. Brosens, A. Schulze, E. W. Lam, The forkhead transcriptionfactor FOXO3a increases phosphoinositide-3 kinase/Akt activity in drug-resistantleukemic cells through induction of PIK3CA expression. Mol. Cell. Biol. 28, 5886–5898(2008).

147. K. Yang, G. Neale, D. R. Green, W. He, H. Chi, The tumor suppressor Tsc1 enforcesquiescence of naive T cells to promote immune homeostasis and function. Nat. Immunol.12, 888–897 (2011).

148. E. Abdelnour-Berchtold, Y. Cerantola, D. Roulin, A. Dormond-Meuwly, N. Demartines,O. Dormond, Rapamycin-mediated FOXO1 inactivation reduces the anticancer efficacyof rapamycin. Anticancer Res. 30, 799–804 (2010).

149. A. Y. Choo, S. G. Kim, M. G. Vander Heiden, S. J. Mahoney, H. Vu, S. O. Yoon,L. C. Cantley, J. Blenis, Glucose addiction of TSC null cells is caused by failedmTORC1-dependent balancing of metabolic demand with supply. Mol. Cell 38,487–499 (2010).

150. P. H. Patel, F. Tamanoi, Increased Rheb-TOR signaling enhances sensitivity of thewhole organism to oxidative stress. J. Cell Sci. 119, 4285–4292 (2006).

151. B. Ravikumar, C. Vacher, Z. Berger, J. E. Davies, S. Luo, L. G. Oroz, F. Scaravilli,D. F. Easton, R. Duden, C. J. O’Kane, D. C. Rubinsztein, Inhibition of mTOR inducesautophagy and reduces toxicity of polyglutamine expansions in fly and mouse models ofHuntington disease. Nat. Genet. 36, 585–595 (2004).

152. T. Wang, U. Lao, B. A. Edgar, TOR-mediated autophagy regulates cell death inDrosophila neurodegenerative disease. J. Cell Biol. 186, 703–711 (2009).

153. C. S. Conn, S. B. Qian, Nutrient signaling in protein homeostasis: An increase in quan-tity at the expense of quality. Sci. Signal. 6, ra24 (2013).

154. T. B. Campbell, S. Basu, G. Hangoc, W. Tao, H. E. Broxmeyer, Overexpression ofRheb2 enhances mouse hematopoietic progenitor cell growth while impairing stemcell repopulation. Blood 114, 3392–3401 (2009).

155. Y. J. Kang, M. K. Lu, K. L. Guan, The TSC1 and TSC2 tumor suppressors are requiredfor proper ER stress response and protect cells from ER stress-induced apoptosis. CellDeath Differ. 18, 133–144 (2011).

156. C. Appenzeller-Herzog, M. N. Hall, Bidirectional crosstalk between endoplasmic re-ticulum stress and mTOR signaling. Trends Cell Biol. 22, 274–282 (2012).

157. K. Shimada, I. Filipuzzi, M. Stahl, S. B. Helliwell, C. Studer, D. Hoepfner, A. Seeber,R. Loewith, N. R. Movva, S. M. Gasser, TORC2 signaling pathway guarantees genomestability in the face of DNA strand breaks. Mol. Cell 51, 829–839 (2013).

158. R. Weisman, M. Choder, The fission yeast TOR homolog, tor1+, is required for the re-sponse to starvation and other stresses via a conserved serine. J. Biol. Chem. 276,7027–7032 (2001).

159. K. Ikeda, S. Morigasaki, H. Tatebe, F. Tamanoi, K. Shiozaki, Fission yeast TOR com-plex 2 activates the AGC-family Gad8 kinase essential for stress resistance and cellcycle control. Cell Cycle 7, 358–364 (2008).

160. M. Schonbrun, D. Laor, L. López-Maury, J. Bähler, M. Kupiec, R. Weisman, TOR com-plex 2 controls gene silencing, telomere length maintenance, and survival under DNA-damaging conditions. Mol. Cell. Biol. 29, 4584–4594 (2009).

161. R. D. Read, T. R. Fenton, G. G. Gomez, J. Wykosky, S. R. Vandenberg, I. Babic,A. Iwanami, H. Yang, W. K. Cavenee, P. S. Mischel, F. B. Furnari, J. B. Thomas, Akinome-wide RNAi screen in Drosophila glia reveals that the RIO kinases mediate cellproliferation and survival through TORC2-Akt signaling in glioblastoma. PLOS Genet. 9,e1003253 (2013).

162. S. Wullschleger, R. Loewith, M. N. Hall, TOR signaling in growth and metabolism.Cell 124, 471–484 (2006).

163. H. Zhang, H. Li, H. S. Xi, S. Li, HIF1a is required for survival maintenance of chronicmyeloid leukemia stem cells. Blood 119, 2595–2607 (2012).

164. C. Muñoz-Pinedo, N. El Mjiyad, J. E. Ricci, Cancer metabolism: Current perspectivesand future directions. Cell Death Dis. 3, e248 (2012).

165. J. Brugarolas, W. G. Kaelin Jr., Dysregulation of HIF and VEGF is a unifying featureof the familial hamartoma syndromes. Cancer Cell 6, 7–10 (2004).

166. T. Shibata, S. Saito, A. Kokubu, T. Suzuki, M. Yamamoto, S. Hirohashi, Global down-stream pathway analysis reveals a dependence of oncogenic NF-E2–related factor2 mutation on the mTOR growth signaling pathway. Cancer Res. 70, 9095–9105(2010).

167. J. J. Wu, J. Liu, E. B. Chen, J. J. Wang, L. Cao, N. Narayan, M. M. Fergusson, I. I. Rovira,M. Allen, D. A. Springer, C. U. Lago, S. Zhang,W. DuBois, T. Ward, R. deCabo, O. Gavrilova,B. Mock, T. Finkel, Increased mammalian lifespan and a segmental and tissue-specificslowing of aging after genetic reduction of mTOR expression. Cell Rep. 4, 913–920(2013).

168. D. W. Lamming, L. Ye, D. M. Sabatini, J. A. Baur, Rapalogs and mTOR inhibitors as anti-aging therapeutics. J. Clin. Invest. 123, 980–989 (2013).

169. S. C. Johnson, P. S. Rabinovitch, M. Kaeberlein, mTOR is a key modulator of ageingand age-related disease. Nature 493, 338–345 (2013).

170. M. V. Blagosklonny, TOR-driven aging: Speeding car without brakes. Cell Cycle 8,4055–4059 (2009).

171. M. V. Blagosklonny, Increasing healthy lifespan by suppressing aging in our lifetime:Preliminary proposal. Cell Cycle 9, 4788–4794 (2010).

172. C. Chen, Y. Liu, Y. Liu, P. Zheng, mTOR regulation and therapeutic rejuvenation ofaging hematopoietic stem cells. Sci. Signal. 2, ra75 (2009).

173. D. Kalaitzidis, S. M. Sykes, Z. Wang, N. Punt, Y. Tang, C. Ragu, A. U. Sinha, S. W. Lane,A. L. Souza, C. B. Clish, D. Anastasiou, D. G. Gilliland, D. T. Scadden, D. A. Guertin,S. A. Armstrong, mTOR complex 1 plays critical roles in hematopoiesis and Pten-loss-evoked leukemogenesis. Cell Stem Cell 11, 429–439 (2012).

174. M. Murakami, T. Ichisaka, M. Maeda, N. Oshiro, K. Hara, F. Edenhofer, H. Kiyama,K. Yonezawa, S. Yamanaka, mTOR is essential for growth and proliferation in earlymouse embryos and embryonic stem cells. Mol. Cell. Biol. 24, 6710–6718 (2004).

175. D. A. Guertin, D. M. Stevens, C. C. Thoreen, A. A. Burds, N. Y. Kalaany, J. Moffat,M. Brown, K. J. Fitzgerald, D. M. Sabatini, Ablation in mice of the mTORC componentsraptor, rictor, ormLST8 reveals that mTORC2 is required for signaling to Akt-FOXO andPKCa, but not S6K1. Dev. Cell 11, 859–871 (2006).

176. J. D. Powell, K. N. Pollizzi, E. B. Heikamp, M. R. Horton, Regulation of immune re-sponses by mTOR. Annu. Rev. Immunol. 30, 39–68 (2012).

177. C. Procaccini, M. Galgani, V. De Rosa, G. Matarese, Intracellular metabolic path-ways control immune tolerance. Trends Immunol. 33, 1–7 (2012).

178. K. Yang, H. Chi, mTOR and metabolic pathways in T cell quiescence and functionalactivation. Semin. Immunol 24, 421–428 (2012).

179. H. Chi, Regulation and function of mTOR signalling in T cell fate decisions. Nat. Rev.Immunol. 12, 325–338 (2012).

180. E. B. Heikamp, J. D. Powell, Sensing the immune microenvironment to coordinate Tcell metabolism, differentiation & function. Semin. Immunol. 24, 414–420 (2012).

www.SCIENCESIGNALING.org 1 July 2014 Vol 7 Issue 332 re2 10

R E V I E W

181. D. A. Guertin, K. V. Guntur, G. W. Bell, C. C. Thoreen, D. M. Sabatini, Functional ge-nomics identifies TOR-regulated genes that control growth and division. Curr. Biol. 16,958–970 (2006).

182. C. C. Thoreen, L. Chantranupong, H. R. Keys, T. Wang, N. S. Gray, D. M. Sabatini,A unifying model for mTORC1-mediated regulation of mRNA translation. Nature485, 109–113 (2012).

183. A. C. Hsieh, Y. Liu, M. P. Edlind, N. T. Ingolia, M. R. Janes, A. Sher, E. Y. Shi, C. R. Stumpf,C. Christensen, M. J. Bonham, S. Wang, P. Ren, M. Martin, K. Jessen, M. E. Feldman,J. S. Weissman, K. M. Shokat, C. Rommel, D. Ruggero, The translational landscape ofmTOR signalling steers cancer initiation and metastasis. Nature 485, 55–61 (2012).

184. P. P. Hsu, S. A. Kang, J. Rameseder, Y. Zhang, K. A. Ottina, D. Lim, T. R. Peterson,Y. Choi, N. S. Gray, M. B. Yaffe, J. A. Marto, D. M. Sabatini, The mTOR-regulated phos-phoproteome reveals a mechanism of mTORC1-mediated inhibition of growth factorsignaling. Science 332, 1317–1322 (2011).

185. M. C. Lorenz, J. Heitman, TOR mutations confer rapamycin resistance by preventinginteraction with FKBP12-rapamycin. J. Biol. Chem. 270, 27531–27537 (1995).

186. G. J. Brunn, C. C. Hudson, A. Sekulić, J. M. Williams, H. Hosoi, P. J. Houghton,J. C. Lawrence Jr., R. T. Abraham, Phosphorylation of the translational repressorPHAS-I by the mammalian target of rapamycin. Science 277, 99–101 (1997).

187. H. Yang, D. G. Rudge, J. D. Koos, B. Vaidialingam, H. J. Yang, N. P. Pavletich,mTOR kinase structure, mechanism and regulation. Nature 497, 217–223 (2013).

Acknowledgments: We thank the members of the J.A. and C.L.-R. groups for continuedand inspiring discussions. We also thank the editors and reviewers for insight and helpfulcomments. Our apologies go to those authors whose work was not cited because of ouroversight. Funding: J.A. and C.L.-R. are supported by grants from the Spanish Ministry ofEconomy and Competitiveness (SAF2011-24268 to J.A., SAF2012-36535 to C.L.-R.), byFundació la Marató TV3 (122530), and by Generalitat de Catalunya (2009 SGR601, 2014SGR1153). S.T. is supported by a predoctoral fellowship BES-2013-062670. Competing in-terests: The authors declare that they have no competing interests.

Submitted 31 March 2014Accepted 30 May 2014Final Publication 1 July 201410.1126/scisignal.2005326Citation: J. Aramburu,M.C.Ortells, S. Tejedor,M. Buxadé,C. López-Rodríguez, Transcriptionalregulation of the stress response by mTOR. Sci. Signal. 7, re2 (2014).

www.SCIENCESIGNALING.org 1 July 2014 Vol 7 Issue 332 re2 11

on July 2, 2014 stke.sciencem

ag.orgD

ownloaded from


Recommended