throbber
Regulation of mTOR function
`in response to hypoxia by REDD1
`and the TSC1/TSC2 tumor
`suppressor complex
`
`Iarnes Brugarolas} Kui Lei,‘°‘ Rebecca L. Hurley,3 Brendan D. Manning,‘ Ian H. Reiling,5
`Ernst Hafen,5 Lee A. Witters,3 Leif W. Ellisen,2 and William G. Kaelin ]r.1'6'7
`‘Dana-Farber Cancer Institute and Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts 02115,
`USA; 2Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, Massachusetts 02116, USA,-
`3Departments of Medicine and Biochemistry, Dartmouth Medical School and Department of Biological Sciences, Dartmouth
`College, Hanover, New Hampshire 03755, USA; “Department of Genetics & Complex Diseases, Harvard School of Public
`Health, Boston, Massachusetts 02115, USA; 5Zoologisches Institut, Uriiversitaet Zuerich, Winterthurerstr. 190, CH-8057
`Zuerich, Switzerland; ‘Howard Hughes Medical Institute, Chevy Chase, Maryland 20815, USA
`
`Mammalian target of rapamycin (mTOR) is a central regulator of protein synthesis whose activity is
`modulated by a variety of signals. Energy depletion and hypoxia result in mTOR inhibition. While energy
`depletion inhibits mTOR through a process involving the activation of AMP-activated protein kinase (AMI’Kl
`by LKB1 and subsequent phosphorylation of TSC2, the mechanism of mTOR inhibition byhypoxia is not
`known. Here we show that mTOR inhibition by hypoxia requires the TSC1/TSC2 tumor suppressor complex
`and the hypoxia-inducible gene REDD1/RTP801. Disruption of the TSC1/TSC2 complex through loss of TSC1
`or TSC2 blocks the effects of hypoxia on mTOR, as measured by changes in the mTOR targets SGK and
`4E-BP1, and results in abnormal accumulation of Hypoxia-inducible factor (HIP). In contrast to energy
`depletion, mTOR inhibition by hypoxia does not require AMPK or LKB1. Down-regulation of mTOR activity
`by hypoxia requires de novo mRNA synthesis and correlates with increased expression of the
`hypoxia-inducible REDD1 gene. Disruption of REDD1 abrogates the hypoxia-induced inhibition of mTOR, and
`REDD1 overexpression is sufficient to down-regulate SGK phosphorylation in a TSC1/TSC2-dependent
`manner. Inhibition of mTOR function by hypoxia is likely to be important for tumor suppression as
`TSC2-deficient cells maintain abnormally high levels of cell proliferation under hypoxia.
`
`[K85/'Words: Tuberous Sclerosis Complex; TSC1; TSC2; REDD1/RTP80l; mTOR; Hypoxia]
`Supplemental material is available at http://www.genesdev.org.
`Received August 31, 2004; revised version accepted October 6, 2004.
`
`Tuberous sclerosis complex, a disease characterized by
`benign tumors in multiple tissues, results from muta-
`tions in either Tuberous Sclerosis Complex 1 (TSC1) or 2
`(Tsc2) (Cheadle et al. 2000). Tscl (also called hamartin]
`and TscZ [also called tuberin) form a protein complex
`(van Slegtenhorst et al. 1998} that integrates signals from
`a variety of sources, including growth factors [Gao and
`Pan 2001; Potter et al. 2001; Tapon et al. 2001) and en-
`ergy stores (Inoki et al. 2003b}, with the protein transla-
`tion apparatus. Tscl functions as a GTPase-activating
`protein (GAP) toward the small G protein Rheb, which
`through a poorly understood mechanism controls mam-
`
`7Corresponding author.
`E-MAIL william_kaelin@dfci.l1arvard.edu; FAX (617) 632-4760.
`Article published onlinc ahead of print. Article and publication date are
`at http://www.genesdev.org/cgi/doi/10.I101/gad.125-6804.
`
`malian target of rapamycin (mTOR), a central regulator
`of protein translation (Castro et al. 2003; Gararni et al.
`2003; Inoki et al. 200321; Saucedo et al. 2003; Stocker et
`al. 2003; Tee et al. 2003; Y. Zhang et al. 2003).
`Regulation of mTOR by growth factors has been in-
`tensively studied. In response to growth factor stimula-
`tion, phosphatidylinositol 3-kinase (PI3K) is activated,
`leading to the generation of phosphaticlylinositol-3,4,5-
`triphosphate and the recruitment of Akt to the plasma
`membrane where it
`is activated by phosphorylation
`(Cantley 2002). Akt is a serine/threonine kinase that
`phosphorylates many effectors including Tsc2 (Dan et al.
`2002; Inoki et al. 2002; Manning et al. 2002; Potter et al.
`2002.). The mechanism whereby Akt phosphorylation
`regulates Tsc2 function is controversial, but it is thought
`ultimately to lead to its inactivation, thereby allowing
`the accumulation of Rheb-GTP and the activation of
`
`GENES 8:. DEVELOPMENT 182893-2904 © 2004 by Cold Spring Harbor Laboratory Press ISSN 0890-9369/04; www.genesdev.org
`
`2893
`
`Roxane Labs., Inc.
`Exhibit 1019
`
`Page 001
`
`Roxane Labs., Inc.
`Exhibit 1019
`Page 001
`
`

`
`Brugarolas at al.
`
`mTOR. In support of the idea that Tscl/Tsc2 plays a
`critical role in mTOR regulation by growth factors, cells
`deficient for Tscl or Tsc2 fail to down-regulate 1nTOR
`function in response to growth factor deprivation (Gao et
`al. 2002; Inoki et al. 2002; Iaeschlce et al. 2002; Kwiat-
`kowski et al, 2002; H. Zhang et al. 2003].
`Recent studies have also shed light on how Tscl/Tsc2
`regulates mTOR function in response to changes in en-
`ergy availability. AMP-activated protein kinase {AMPK}
`is a master regulator of energy metabolisrn that is acti-
`vated in response to energy deprivation (Carling 2004).
`AMPK functions as a serine/threonine kinase and di-
`rectly phosphorylates Tsc2 (Inoki et al. 2003b). Cells de-
`ficient for Tscl/Tsc2, or producing a TSC2 variant that
`can not be phosphorylated by AMPK, fail to down-regu-
`late mTOR in situations of energy deprivation (Inoki et
`al. 2003b). Signaling by energy depletion also involves
`the Lkbl tumor suppressor protein, which is inactivated
`in Peutz-Ieghers syndrome. Lkbl is a serine/threonine
`kinase that pbosphorylates a variety of substrates, in-
`cluding AMPK (Hawley et al. 2003; Woods et al. 2003;
`Shaw et al. 2004b). AMPK activation with consequent
`Tscl /Tsc2-mediated inhibition of mTOR by energy
`depletion requires Lkbl [Corradetti et al. 2004; Shaw et
`al. 2004b). Thus under conditions that are adverse for
`growth, such as in the presence of reduced energy stores,
`mTOR function is inhibited, thereby down-regulating
`protein synthesis and conserving energy.
`mTOR function is also regulated by amino acid avail-
`ability (Gingras et al. 2001}. Whereas mTOR regulation
`by growth factors and energy stores requires an intact
`Tscl /Tsc2 complex, amino acids seem to regulate
`mTOR function through both Tscl/Tsc2-dependent and
`independent pathways (Gao et al. 2002, H. Zhang et al.
`2003}.
`mTOR is a conserved serine/threonine kinase that
`phosphorylates a series of substrates involved in protein
`translation including 4E-BP1 and S6K (Gingras et al.
`2001). 4E—BPl binds the translation initiation factor eIF-
`4E, preventing its interaction with other members of the
`eIF-4 complex and thereby inhibiting translation initia-
`tion of 5’ cap (7-methyl GTP) mRNAs. 4E—BPl phos-
`phorylation by mTOR, as well as other kinases in the
`PI3K pathway, relieves this inhibition, thereby promot-
`ing mRNA translation (Gingras et al. 2001).
`In mammals, there are two 36K genes, 36K] and S6K2,
`and each has two different splice forms. While S6K2 and
`the long form (~85 l<Da) of S6Kl localize to the nucleus,
`the short form of S6Kl [~70 l<Da) is cytoplasmic. 86K is
`thought to exist in an inactive, closed conformation, re-
`sulting from an intramolecular interaction between the
`catalytic domain and a pseudosubstrate domain (Fingar
`and Blenis 2004). There are at least eight phosphoryla-
`tion sites in S6Kl and its activation requires a complex
`process of phosphorylation involving mTOR as well as
`other PIBK effectors. Phosphorylation in the linker re-
`gion, at Ser 371 and Thr 389, is essential for S6Kl acti-
`vation [Dufner and Thomas 1999). Thr 389 is the major
`rapamycin-sensitive site and can be phosphorylated by
`mTOR in vitro, suggesting that it is a direct mTOR tar-
`
`2894
`
`GENES EL DEVELOPMENT
`
`get (Dufner and Thomas 1999). Phosphorylation at this
`site has also been shown to be regulated in response to
`changes in Tscl/Tsc2 levels [Inoki et al. 2002, Iaeschke
`et al. 2002; Kwiatkowski et al. 2002, Manning et al.
`2002; H. Zhang et al. 2003).
`The best-characterized substrate of S6Kl is the ribo-
`sornal protein S6. S6 is an integral component of the 40S
`subunit that is required for cell proliferation but whose
`precise function is unclear (Volarevic et al. 2000). S6 is
`thought to be regulated primarily through successive
`phosphorylation events beginning at S236 and S235,
`Phosphorylation at these sites is rapamycin-sensitive
`and largely mediated by SGK [Dufner and Thomas 1999).
`Indeed, cells deficient for both S6Kl and S6K2 have pro-
`foundly reduccd levels of S6 S235/236 phosphorylation
`(Pende et al. 2004). Thus, S235/236 phosphorylation pro-
`vides an accurate readout for endogenous 56K activity.
`The I-Iypoxiadnducible factor
`[Hif) has also been
`shown to be regulated by mTOR (Hudson et al. 2002).
`Hif is a heterodimeric transcription factor composed of a
`stable [3 and a labile or subunit whose levels are con-
`trolled by oxygen tension {Semenza 2000). In the pres-
`ence of oxygen, Hif-or subunits are hydroxylated at spe-
`cific prolyl residues and targeted for degradation by an E3
`ubiquitin ligase that contains the von Hippel-Lindau tu-
`mor suppressor protein (pVHL) (Kaelin 2002). pVHL in-
`activation in patients with von Hippel-Lindau disease
`results in Hif up-regulation and the development of tu-
`mors. As in pVHL-deficient cells, Tsc2-deficient cells
`harbor increased levels of Hif-or relative to wild~type
`cells, especially under growth factor poor conditions
`[Brugarolas et al. 2003). Hif up—regulation in Tsc2-defi-
`cient cells is likely to result from increased mTOR ac-
`tivity as mTOR increases Hif stability and increased Hif
`levels in Tsc2-deficient cells can be normalized by treat-
`ment with rapamycin [Hudson et al. 2002, Brugarolas et
`al. 2003}.
`It was recently shown that mTOR function is regu-
`lated by hypoxia (Arsham et al. 2003). Hypoxia down-
`regulates 4E—BPl phosphorylation and increases 4E-BP1
`binding to eIF-4E at 5’ cap structures. Similarly, hypoxia
`down-regulates S6K phosphorylation at multiple sites
`including T389 and inhibits S6 phosphorylation. Hy-
`poxia-induced inhibition of mTOR is dominant over
`mTOR activating signals from growth factors and nutri-
`ents and occurs independently of Hif—loz [Arsham et al.
`2003). However, how mTOR function is regulated by
`hypoxia is not known.
`Here we show that mTOR inhibition by hypoxia re-
`quires an intact Tscl/Tsc2 complex. Furthermore,
`down-regulation of mTOR function by hypoxia requires
`de novo transcription and the expression of the hypoxia-
`inducible Redd] /RTP801 gene.
`
`Results
`
`Tscl/Tsc2 complex is required for mTOR regulatiozi
`by hypoxia
`
`To examine the contribution of the Tscl/Tsc2 complex
`to the regulation of mTOR function by hypoxia, the ef-
`
`Roxane Labs., Inc.
`Exhibit 1019
`
`Page 002
`
`Roxane Labs., Inc.
`Exhibit 1019
`Page 002
`
`

`
`fects of hypoxia on MEFs derived from Tsc2—deficient
`embryos were analyzed. Loss of Tsc2 results in lethality
`at approximately day 10.5 of gestation when it is diffi-
`cult to obtain a large number of fibroblasts (Rennebeck
`et al. 1998). To extend the life span of the few fibroblasts
`that can be obtained from these embryos, mice were in-
`tercrossed with mice carrying mutations in p53, which
`enhances the life span of the limited number of MEFs
`that can be obtained (H. Zhang et al. 2005). In the ex-
`periments that follow, Tsc2‘/‘,«p53'/‘ MEFS were com-
`pared with Tsc2*/‘;p53“/‘ MEFs. For simplicity, these
`MEFs are referred to as ”Tsc2‘(‘” and ”Tsc2*/*,” respec-
`tively.
`Tscl/Tsc2 complex regulates mTOR in response to
`growth factors. As shown before, serum deprivation in-
`hibits mTOR, as evidenced by decreased phosphoryla-
`tion of the mTOR effector S6K (T389) (Fig, 1A). S6K
`T389 is the major rapamycin-sensitive site and is re-
`quired for S6K activity (Dufner and Thomas 1999). The
`down-regulation of 56K phosphorylation is associated
`with a decrease in S6 phosphorylation (S235/236). We
`also confirmed that mTOR inhibition by serum depri-
`vation requires an intact Tscl/Tsc2 complex and is
`abrogated in T302‘/' MEFS (Fig. 1A) (Iaeschke et al. 2002;
`H. Zhang et al. 2003). Rapamycin inhibits S6K phos-
`phorylation and activity regardless of Tse2 status, sup-
`porting the concept that Tscl/Tscl functions upstream
`of mTOR (Fig. IA),
`Tsc2*/’ MEFS down-regulated S6K phosphorylation
`and activity in response to hypoxia (Fig. IA), in keeping
`
`Hypoxia regulation of mTOR by REDDI and TSC1/TSC2
`
`with an earlier study using transformed human embry-
`onic kidney [HEK293) cells (Arsharn et al. 2003). Down-
`regulation of S6 phosphorylation in response to hy-
`poxia was also demonstrable in mice exposed to 6% oxy-
`gen, suggesting that this phenomenon is not restricted to
`tissue culture (Supplementary Fig. 1). In contrast, hy-
`poxia failed to down-regulate S6K and S6 phosphoryla-
`tion in Tsc2‘(‘ MEFs (Fig. IA) and Tscl‘/‘ mouse 3T3
`cells (Fig. 113), indicating that mTOR inhibition by hy-
`poxia requires an intact Tscl/Tsc2 complex. Likewise
`hypoxia promoted the binding of 4E—BP1 to 7—methyl
`GTP (7rnGTP) in T.sc2*/* cells but not Tsc2‘/” cells, con-
`sistent with Tsc2—dependent inhibition of 4E-BP1 phos-
`phorylation by mTOR in response to hypoxia (Fig. 1C).
`Taken together, these data indicate that mTOR inhibi-
`tion by hypoxia requires a functional Tscl/Tsc2 com-
`plex.
`We previously showed that Hif-oi (hereafter referred to
`as Hif) levels are regulated in response to growth factors
`through a mechanism that involves the Tscl/Tsc2 com-
`plex and inTOR (Brugarolas et al. 2003; see also Fig. 1A).
`As Tsc2‘/‘ MEFs failed to down-regulate mTOR activity
`in response to hypoxia, we postulated that Hif levels
`might also be affected. Tsc2‘/‘ MEFS were not impaired
`in the up-regulation of Hif in response to hypoxia. How-
`ever, whereas Tsc2*/* MEFs down-regulated Hif with
`prolonged hypoxia, Hif levels remained elevated in
`Tsc2’/' MEFs (Fig. 1D). Consistent with the idea that
`increased Hif levels in T502‘/‘ cells under prolonged hy-
`poxia result from increased mTOR activity, treatment
`
`A
`Rapamycin
`Scnmi
`
`M’-.29-7:~
`+
`+ +
`
`Hy‘ aoxia
`232-2-/A T‘)
`+
`r"‘£"‘
`352$
`+ 4?-
`(I3Yi9<7
`fr‘)
`
`B
`‘Hyyvxicx an
`
`T51’!-x-/+
`05$?
`
`T.\r~I-:.
`036$!
`
`C
`
`H35pivtTu'(h)
`
`
`HYJE
`
`Lysum
`l‘:‘¢'2+./+
`13:2.-z
`I)
`4
`u
`
`an era puiiaomz
`ms» T$u2«.
`
`
`
` E
`
`Sfifi-P
`
`
`D
`
`Xiyptzxin (hi
`Hif
`
`[I
`
`-
`
`.Ts'c,':.r'.
`2’.rc3»Ms
`3 8325125 0 3 813-
`
`.5-6,~§"
`
`Tuhulin
`
`;-‘~.-
`
`KAPAMYCIN
`7'm‘£'+/T+—.
`3 2; 132025 D
`
`3
`
`TsI:2-/-
`2:
`£23 2025
`
`
`
`:3
`
`Hypuxiamjx
`rut
`
`Tubulin
`
`Figure 1. Tsc2 regulates mTOR in response to hypoxia. (A) Western blot analysis of Tsc2*(* and Tsc2‘(’ MEFs. (Hif) Hif—l(1 and/or
`Hif-2a; (S6K-P) S6K phosphorylated T389; (S6-P) S6 phosphorylated on S235/236. Left panel shows MEF in 0.05% serum or following
`serum addition (10% serum for 45 min) pretreated or not with raparnycin (1.5 h prior to serum addition). Right panel shows MEFS
`exposed to hypoxia for the indicated periods of time. (B) Western blot analysis of Tsc1“/* and Tsc1‘(‘ mouse 3T3 cells treated with
`hypoxia for the indicated periods of time. (C) Western blot analysis of input (left) and 7mGTP—bound (right) proteins from extracts of
`7’sc2*(‘ and Tsc2’(’ MEFs exposed to hypoxia for the indicated periods of time. (D,E) Western blot analysis of extracts from Tsc2*(“ and
`Tsc2’(’ MEFS exposed to hypoxia for the indicated periods of time. In E all the cells were treated with rapamycin for 26 h prior to lysis
`regardless of the duration of hypoxia.
`
`GENES 8?. DEVELOPMENT
`
`2895
`
`Roxane Labs., Inc.
`Exhibit 1019
`
`Page 003
`
`Roxane Labs., Inc.
`Exhibit 1019
`Page 003
`
`

`
`Brugarolas at al.
`
`with rapamycin restored the down—regulation of Hif nor-
`mally observed after prolonged hypoxia (Fig. 1E).
`The importance of Tsc2 in the regulation of mTOR by
`hypoxia is further supported by reconstitution experi-
`ments. Tsc2"/‘ MEFs were infected with a retrovirus en-
`codingepitope-tagged human Tsc2. Retrovirally trans-
`duced Tsc2'/' MEFS achieved Tsc2 protein levels that
`were similar to endogenous levels in Tsc2*/* MEFS (Fig.
`2A}. Reintroduction of Tsc2 into Tsc2‘/‘ MEFs restored
`the down-regulation of S6 phosphorylation by hypoxia
`(Fig. 2A).
`To determine the generalizability of our observations,
`the role of Tscz in mTOR regulation by hypoxia was
`examined in other cell types. Tsc2 knockdown with two
`different Tsc2 siRNAs in HEK293 and HeLa cervical car-
`cinoma cells blocked the down—regulation of S6 phos-
`phorylation by hypoxia (Fig. 2B; Supplementary Fig. 2.),
`indicating that Tsc2 is required for mTOR regulation by
`hypoxia in multiple cell types.
`
`Tsc2 inactivation confers a proliferative advantage
`under hypoxia
`
`Taken together, these data establish the importance of
`Tscl/Tsc2 in the regulation of mTOR by hypoxia. As
`Tscl/Tsc2 functions as a tumor suppressor, we asked
`whether disruption of this complex would affect cell pro-
`liferation under hypoxic conditions. In keeping with pre-
`vious observations,
`the rates of cell proliferation of
`
`A
`
`4'
`
`TSC2
`.§".~c' pisuiic
`l'lypnxia (I1)
`03690369
`rscz. Fr 3
`l
`A
`
`56-!’
`
` Hit”
`
`Tubulin
`
`‘
`
`B
`
`In-paxin(k‘i
`TS(‘;2
`
`IILZK 2.93
`‘l‘SC2—(a)'|‘S(‘2{lv)
`.5 5 9 0 3 6' 9 n Ln: 9
`
`It
`
`VIIF
`
`$6-P
`
`Tubulin
`
`Figure 2. Tsc2 is both necessary and sufficient for the regula-
`tion of S6 phosphorylation by hypoxia. [A] Western blot analysis
`of T5c2”‘ MEFs retrovirally transduced with either a Tsc2 ex-
`pression vector or an empty vector and treated with hypoxia for
`the indicated periods of time. T552‘/* MEFS are included as con-
`trols. (B) Western blot analysis of HEK293 cells transfected with
`two different synthetic Tsc2 siRNAs (a and b) or a scrambled
`siRNA (Sc) and exposed to hypoxia for the indicated periods of
`time.
`
`2896
`
`GENES 5:. DEVELOPMENT
`
`Tsc2‘/‘ and Tsc2*/* MEFS were very similar under nor-
`moxie conditions for several days (Fig. 3A,- H. Zhang et
`al. 2003). After 4 d in culture the rate of proliferation of
`Tsc2*/* cells began to decline relative to T3132‘/“ cells,
`possibly due to depletion of nutrients and growth factors
`from the media. Consistent with this idea, T5132’/’ but
`not Tsc2*/* MEFs proliferate under conditions of serum
`deprivation (H. Zhang et al. 2003). We next measured the
`proliferation of Tsc2‘/‘ and Tsc2‘/* cells under hypoxic
`conditions. To avoid confounding effects from media
`depletion, the media was changed daily. Under hypoxic
`conditions Tsc.2'/’ MEFs exhibited a marked prolifera-
`tive advantage compared with T.sc2*/* MEFs (Fig. 3B).
`Increased proliferation of T502“/‘ MEFs under hypoxic
`conditions correlated with persistently elevated levels
`of S6 phosphorylation (Fig. 3C) and was abrogated by
`treatment with rapamycin (Fig. 3B). These data suggest
`that failure to down-regulate mTOR in response to hy-
`poxia in Tsc2‘/‘ MEFs confers a growth advantage that
`might contribute to tumor formation in TSC patients. It
`should be noted that the growth inhibitory effect of hy-
`poxia on Tsc2*/* MEFS in these assays is p53 indepen-
`dent since both the Tsc2*/* and Tsc2‘/“ MEFS used here
`lack p53.
`
`.
`
`Tscl /T362 regulation by hypoxia is AMPK
`and Lkbl independent
`
`Energy depletion results in Tscl/Tsc2-mediated mTOR
`inhibition through a mechanism that involves Lkbl and
`AMPK {lnoki et al. 2003b; Corradetti et al. 2004; Shaw et
`al. 2004a}. Since hypoxia might also affect cellular en-
`ergy stores, we sought to determine whether hypoxia
`regulated rnTOR through a similar pathway. AICAR (5-
`arninoomidazole-4-carboxyamide),
`a
`cell-permeable
`AMPK agonist, inhibited mTOR and down-regulated S6
`phosphorylation (Fig. 4A; Kirnura et al. 2003). As previ-
`ously reported (Shaw et al. 2004b}, Tsc2'/‘ cells failed to
`down-regulate S6 phosphorylation in response to AICAR
`despite robust activation of AMPK as determined by
`AMPK phosphorylation in the T loop and the phosphory-
`lation of its substrate acetyl-CoA carboxylase (ACC) (Fig.
`4A), thereby justifying the use of AICAR as an AMPK
`perturbant in the experiments described below.
`To ask whether hypoxia signals are transduccd
`through an energy depletion pathway in our system, we
`first examined the effects of hypoxia on AMPK activity.
`After 4 h of hypoxia, S6 phosphorylation was markedly
`down-regulated without a detectable increase in AMPK
`activity, as measured by ACC phosphorylation (Fig. 4B).
`The ability to detect ACC phosphorylation in response
`to AMPK activation was confirmed by studying cells
`treated with AICAR in parallel (Fig. 4B}. Furthermore, in
`vitro kinase assays with AMPK immunoprecipitates
`from the same lysates used for Figure 4B indicated that
`hypoxia, in contrast to AICAR, did not increase and may
`have decreased AMPK kinase activity (Fig. 4C). These
`results are consistent with a recent study of HEK293
`cells exposed to hypoxia for 30 min (Arsham et al. 2003].
`We next asked whether AMPK activity is necessary for
`
`Roxane Labs., Inc.
`Exhibit 1019
`
`Page 004
`
`Roxane Labs., Inc.
`Exhibit 1019
`Page 004
`
`

`
`Nbrmtixiix.
`
`Hypoxia regulation of mTOR by REDD1 and TSCI/TSC2
`
`W
`
`3.a;=
`
`.5c
`
`'I's€?3-i-
`
`lixzpix
`4
`
`Figure 3. Tsc2 loss confers a proliferative advan-
`tags under hypoxic conditions.
`(A) Proliferation
`rates of Tsc2‘/‘ and Tsc2“(" MEFS under normoxic
`conditions.
`(BI Proliferation rates under hypoxic
`conditions of Tsc2‘/‘ and T3132”/‘ MEFs (treated or
`not with rapamycin). Error bars for A and B equal
`one standard deviation (n = 3]. Note different Y-axis
`scales in A and B.
`(C) Western blot analysis of
`Tsc2*(* and Tsc2"/“ MEFS cultured in parallel under
`hypoxic conditions for the indicated number of days.
`
`‘I25-L124‘-
`’Fsc2+z/+
`3401334
`“_\'p(I>.‘ia (day.-;)
` S6-I’
`
`Tulmlist
`
`the hypoxia—inducecl down-regulation of mTOR func-
`tion. Pharmacological inhibition of AMPK with com-
`pound C (Inoki et al. 2003b) at doses sufficient to block
`the down-regulation of S6 phosphorylation by AICAR
`did not block the down-regulation of S6 phosphorylation
`by hypoxia (Fig. 4D). Thus AMPK is required for AICAR
`signaling but not for hypoxia signaling.
`Next we examined the requirements for the AMPK
`kinase Lkbl. Consistent with previous studies, Lkb1‘/‘
`MEFs failed to down-regulate S6 phosphorylation in re-
`sponse to AICAR (Fig. 4E, Shaw et al. 2004b]. In contrast,
`we found that Lkbl loss did not affect the down-regula-
`tion of S6 phosphorylation by hypoxia (Fig. 4F). in addi-
`tion, HeLa cells, which are defective for Lkbl (Tiainen et
`al. 1999), also down-regulated S6 phosphorylation in re-
`sponse to hypoxia but not to AICAR (Supplementary Fig.
`3) indicating that Lkbl, like AMPK, is dispensable for
`hypoxia signaling. We noted, however, that ACC and
`AMPK do become phosphorylated in HeLa cells after
`prolonged hypoxia, possibly due to a non—Lkbl AMPK
`kinase (Supplementary Fig. 3). Nonetheless, inhibition of
`S6 phosphorylation occurred prior to appreciable AMPK
`activation in these cells, again in keeping with the idea
`that the acute down-regulation of mTOR in response to
`hypoxia is AMPK independent. It is possible that signal-
`ing to Tscl /Tsc2 in response to chronic hypoxia involves
`both Lkbl/AMPK-dependent and independent pathways
`since prolonged hypoxia would predictably lead to ATP
`depletion and accumulation of AMP.
`
`Regulation of mTOR by hypoxia requires
`Reddl induction
`
`To begin to unravel the signaling pathway whereby hy-
`poxia regulates Tscl/Tscl, we asked whether
`this
`mechanism required de novo transcription. Provoca-
`tively, inhibition of transcription with actinomycin D
`blocked the down-regulation of S6 phosphorylation by
`hypoxia, indicating that de novo transcription is neces-
`sary for hypoxia signaling (Fig. 5A). As a control actino-
`rnycin D alone did not affect baseline S6 phosphoryla-
`tion (Fig. 5A).
`
`Recently, the Drosophila ortholog of the hypoxia—in-
`ducible Redd] gene was isolated in a screen for suppres-
`sors of insulin signaling (Reiling and Hafen 2004). Redd]
`was originally identified by several groups as a gene that
`is induced at the mRNA level in response to stresses
`such as hypoxia or DNA damage (Ellisen et al. 2002,-
`Shoshani et al. 2002). Reddl encodes a protein with a
`predicted MW of 25 kDa that lacks any known func-
`tional domains (Ellisen et al. 2002; Shoshani et al. 2002).
`Reddl belongs to a family of highly conserved proteins
`that includes Reddl, which might also be regulated by
`hypoxia (Cuaz-Perolin et al. 2004}. Redd orthologs have
`been shown to inhibit insulin signaling in Drosophila
`and epistasis analysis suggests that they act upstream of
`Tscl/Tscl (Reiling and Hafen 2004).
`To ascertain whether Reddl was involved in the regu-
`lation of the mTOR by hypoxia, we first examined Reddl
`induction in response to hypoxia in MEFs. Consistent
`with previous data, Reddl mRNA levels were induced
`by hypoxia (Fig. 5B,- Shoshani et al. 2002). Redd2 expres-
`sion, however, was not detected in MEFs (data not shown).
`To ask whether Reddl
`is necessary for hypoxia—in—
`duced down—regulation of mTOR function, we obtained
`Reddl‘/‘ MEFS (K. Lei and L.W. Ellisen, in prep). In con-
`trast to Reddl‘/* MEFS, Reddl‘/’ MEFs failed to down-
`regulate phosphorylation of 86K and S6 in response to
`hypoxia (Fig. 5C). These data indicate that Reddl, like
`Tscl and Tsc2,
`is necessary for the hypoxia—induced
`down-regulation of mTOR signaling. This requirement ‘
`was specific because Reddl’/’ MEFS,
`in contrast
`to
`Tsc2‘/‘ MEFS, down—regulated S6 phosphorylation in re-
`sponse to serum deprivation (Fig. 6A,B). These data in-
`dicate that Reddl regulates mTOR signaling only in «re-
`sponse to certain stimuli, such as hypoxia.
`
`Reddl is sufficient to dowrmegulate
`S6K1 phospliorylation
`
`We next asked whether Reddl is sufficient to down-regu-
`late mTOR function. For this purpose,
`the effects of
`Reddl expression on S6K phosphorylation were deter-
`mined. HEK293 cells were transfected with HA-tagged
`
`GENES 8t DEVELOPMENT
`
`2897
`
`Roxane Labs., Inc.
`Exhibit 1019
`
`Page 005
`
`Roxane Labs., Inc.
`Exhibit 1019
`Page 005
`
`

`
`Brugamlas et al.
`
`A
`
`B
`
`smioun
`
`
` .kI(‘.AR 41:) 1)
`
`ADC» .
`AMPK-P
`
`
`
`st»? ‘;
`Tulml'm
`
`D
`
`,AMPKi
`AICAR iiyporm
`
`AICAR
`
`
`
`'.i\50.inf1n
`Jmuuin
`zixxmlm
`
`llilhflkli
`
`‘E’
`
`1|
`szuus
`
`Hours u3690.3o905s9
`
`E
`
`LkM~fv-’+
`
`3.501»/~
`
`"‘7'C'P
`M
`
`
`
`.m:.va(m 0 L5
`,
`SM, WW.
`
`3
`
`5
`u’ L5 3
`. WWMW
`
`5(,,p W VV
`
`-
`
`»~
`
`,
`
`Tnlmiin
`
`.’*M<'o»'~ vow mu.
`
`2...... W.»
`
`F
`liyposialli)
`
`LItlI].+/+
`3
`6
`
`0
`
`9
`
`0
`
`rm./.
`'5
`‘6
`
`9
`
`SH.
`
`.,».._a
`
`We ms» o.<~eaoé=s\\‘
`
`5%
`
`inTOR regulation by hypoxia is both AMPK and Lkbl independent. (A) Western blot analysis of extracts of Tsc2*/” and
`Figure 4.
`Tsc2‘/‘ MEFS treated with AICAR for the indicated periods of time. (ACCP) Acetyl-CoA carboxylase phosphorylated at 579; (AMPK—P)
`AMPK phosphorylated at T172. (B) Western blot analysis of Tsc2*/‘ MEFS after 4 h of treatment with either hypoxia or AICAR. (C) In
`vitro AMPK kinase assay of the same extracts used in B irnrnunoprecipitated in antibody excess with a polyclonal anti-AMPK (oz
`AMPK) antibody or normal rabbit IgG (1gG). Samples were normalized for protein concentration prior to immunoprecipitation. Error
`bars equal one standard deviation (:1 = 3). Also shown are background activities of immunoprecipitates incubated in the absence of
`substrate (SAMS peptide). (D) Western blot analysis of Tsc2“”' cells pretreated or not with the AMPK inhibitor compound C and
`exposed to either AICAR or hypoxia for the indicated periods of time. All cells treated with compound C were exposed to the drug for
`9.5 h. (E,F) Western blot of Lkb1*/‘ or LI<b1'/' MEFS treated with AICAR (E) or hypoxia (F) for the indicated periods of time.
`
`human Reddl (R1) or a mutant lacking its evolutionarily
`conserved central region (R1dC,~ amino acids 96-153 out
`of 232). To exclude signal from untransfected cells, and
`thereby increase signal/noise ratio, a plasmid encoding
`HA—S6K1 (SGK) was included in the transfection mix.
`HA—S6Kl was then recovered by anti-HA imInunopre—
`cipitation and immunoblotted with an antibody specific
`for pl1ospho~S6Kl. Wild~type Reddl, but not mutant
`Reddl, down-regulated S6K1 phosphorylation (Fig. 7A).
`As a control, S6Kl phosphorylation was abrogated in
`cells treated with rapaniycin (Fig. 7A). These data indi-
`cate that Reddl
`is sufficient
`to doWn—regulate S6Kl
`phosphorylation and that
`this function requires the
`Reddl central domain.
`‘
`Simultaneous overexpression of two Redd orthologs in
`Drosophila, Scylla and Charybdis, results in additive ef—
`fects with respect to cell size (Reiling and Hafen 2004).
`Since cell size in Drosophila is controlled by 36K, we
`asked whether Redd2 also down-regulated S6KI phos-
`phorylation, For this purpose, we repeated these experi-
`ments with HA—Redd2 (R2), alone or in combination
`with Reddl. Reddl, like Reddl, inhibited S6Kl phos-
`phorylation and the effects of Reddl and Reddl together
`were possibly additive (Fig. 7A). While the same epitope
`tag in Reddl (or 2) and S6Kl, combined with immuno—
`precipitation, could theoretically confound our results,
`qualitatively similar effects were observed when S6Kl
`
`2898
`
`GENES & DEVELOPMENT
`
`phosphorylation was analyzed directly from cell lysates
`(data not shown).
`
`Down-regulation of 86K phospliorylation by Reddl
`requires Tsc2
`
`To ask whether Reddl functions upstream of Tsc2, simi-
`lar assays were performed in HeLa cells in which Tsc2
`function was impaired using siRNA. The effect of Reddl
`and Redd2 on S6Kl phosphorylation in HeLa cells trans-
`fected with irrelevant siRNA (Sc; scrambled) mirrored
`those observed in HEK293 cells (Fig. 7, cf. B and A). In
`stark contrast, the effects of Reddl and Redd2, alone or
`in combination, on S6K1 phosphorylation were corn~
`pletely abrogated in cells treated with either of two dif»
`ferent Tsc2 siRNAs (a and b) (Fig. 7B).
`To evaluate the effects of Reddl on endogenous (rather
`than ectopic) S6K as well as S6 phosphorylation, and its
`dependency on Tscl/Tsc2 for signaling, we generated a
`HA-Reddl-inducible cell line (Fig. 7C). These cells were
`transfected with siRNA (as above; Sc, Tsc2 a and b) and
`subsequently placed in media with tetracycline to in-
`duce I-IA-Reddl. Reddl induction resulted in a profound
`down-regulation of endogenous 56K and S6 phosphory-
`lation unless Tscl was downregulated by siRNA (Fig.
`7C). Therefore downregulation of S6K phosphorylation
`by Reddl and Redd2 requires Tsc2. In addition, S6K
`
`Roxane Labs., Inc.
`Exhibit 1019
`
`Page 006
`
`Roxane Labs., Inc.
`Exhibit 1019
`Page 006
`
`

`
`A
`
`'“»"l"”‘v"-“
`'\
`E,
`55"’
`ya‘
`A‘cx'I)
`_~°
`g5‘
`‘
`_
`Hours use-3 331290469
`
`Hit‘
`
`36",
`so
`
`
`
`Roeiiif-L
`Raid:/.~’+
`
`
`
`
`at-441+.»
`3
`is
`
`9
`
`up
`
`R.»/I:/2.,.
`3
`‘tr
`
`0
`
`B
`
`.
`nnmm my
`Rudrfl
`
`GAHHI
`
`C
`iiypnmrlut
`S6}:-F‘
`
`$6--P
`
`
`
`Hypoxia regulation of mTOR by REDD1 and TSC1/TSC2
`
`Figure 5. Down-regulation of mTOR function by hypoxia
`requires Reddl. (A) Western blot analysis of Tsc2*/* MEFS
`pretreated (.30 min prior to initiation of the hypoxia time
`course) or not, with Actinorriycin D and exposed to hypoxia
`for the indicated periods of time. Shown in parallel, analysis
`of Tsc2*(* cells treated with Actinomycin D for the indi-
`cated periods of time. Northern blot (B) and Western blot (C)
`analysis of Reddl“ and Reddl‘/‘ MEFS treated with by-
`poxia for the indicated periods of time.
`
`phosphorylation in Reddl’/‘ cells was down-regulated
`by treatment with rapamycin (data not shown), in keep-
`ing with the idea that Reddl acts upstream of mTOR.
`While we cannot formally exclude that Reddl acts in a
`parallel pathway, our data suggest that Reddl acts up-
`stream of
`the Tscl/TscZ complex to down-regulate
`mTOR function in response to hypoxia.
`
`Discussion
`
`The data presented here indicate that the down-regula-
`tion of mTOR function by hypoxia requires Tscl/Tsc2.
`Under conditions of hypoxia, Tsc2-deficient cells fail to
`down-regulate 86K phosphorylation (and activity) and
`accumulate Hif abnormally. Disruption of the Tscl/
`Tsc2 complex results in increased cell proliferation un-
`der hypoxic conditions. Hypoxia signaling requires nei-
`ther AMPK nor Lkbl but does require de novo transcrip-
`tion and the expression of Reddl. Reddl is necessary and
`sufficient for the down-regulation of mTOR by hypoxia.
`Our data reveal that mTOR regulation by hypoxia re-
`quires Tscl/Tsc2 in multiple cell types. Tsc2 was essen-
`tial for hypoxia-induced mTOR down-regulation in three
`
`A
`9.<'}5%Sz>.rum(S1') 0
`
`gt.-gm
`,3
`:3
`
`7:92./.
`9 area’:
`
`
`
`B
`o.rns<;~:.sm.muz)
`T9122
`
`R«rlIl)+.f+
`3
`r.’
`
`9‘
`
`0
`
`RL’fl'a‘U-1-
`3
`a
`
`1:
`
`9
`
`56-)’
`
`is required for growth factor
`Figure 6. Tsc2 but not Reddl
`signaling. Western blot analysis of Tsc2*(* and Tsc2‘/’ MEFs (A)
`or Reddl‘/“ and Redd1"’ MEFs (B) after scrum deprivation for
`the indicated periods of time.
`
`different cell types, MEFS, HEK293 cells, and HeLa cells.
`The requirement of Tsc2 for the down-regulation of
`mTOR in multiple diverse cell
`types suggests that
`mTOR regulation by hypoxia universally requires Tsc2.
`The reg

This document is available on Docket Alarm but you must sign up to view it.


Or .

Accessing this document will incur an additional charge of $.

After purchase, you can access this document again without charge.

Accept $ Charge
throbber

Still Working On It

This document is taking longer than usual to download. This can happen if we need to contact the court directly to obtain the document and their servers are running slowly.

Give it another minute or two to complete, and then try the refresh button.

throbber

A few More Minutes ... Still Working

It can take up to 5 minutes for us to download a document if the court servers are running slowly.

Thank you for your continued patience.

This document could not be displayed.

We could not find this document within its docket. Please go back to the docket page and check the link. If that does not work, go back to the docket and refresh it to pull the newest information.

Your account does not support viewing this document.

You need a Paid Account to view this document. Click here to change your account type.

Your account does not support viewing this document.

Set your membership status to view this document.

With a Docket Alarm membership, you'll get a whole lot more, including:

  • Up-to-date information for this case.
  • Email alerts whenever there is an update.
  • Full text search for other cases.
  • Get email alerts whenever a new case matches your search.

Become a Member

One Moment Please

The filing “” is large (MB) and is being downloaded.

Please refresh this page in a few minutes to see if the filing has been downloaded. The filing will also be emailed to you when the download completes.

Your document is on its way!

If you do not receive the document in five minutes, contact support at support@docketalarm.com.

Sealed Document

We are unable to display this document, it may be under a court ordered seal.

If you have proper credentials to access the file, you may proceed directly to the court's system using your government issued username and password.


Access Government Site

We are redirecting you
to a mobile optimized page.





Document Unreadable or Corrupt

Refresh this Document
Go to the Docket

We are unable to display this document.

Refresh this Document
Go to the Docket