Você está na página 1de 10

THE JOURNAL OF BIOLOGICAL CHEMISTRY VOL. 282, NO. 28, pp.

20534 –20543, July 13, 2007


© 2007 by The American Society for Biochemistry and Molecular Biology, Inc. Printed in the U.S.A.

Hypoxia-inducible Factor 1␣ Is Regulated by the Mammalian


Target of Rapamycin (mTOR) via an mTOR Signaling Motif*
Received for publication, December 22, 2006, and in revised form, May 10, 2007 Published, JBC Papers in Press, May 14, 2007, DOI 10.1074/jbc.M611782200
Stephen C. Land‡ and Andrew R. Tee§1
From the §Institute of Medical Genetics, Wales College of Medicine, Cardiff University, Heath Park, Cardiff CF14 4XN,
United Kingdom and the ‡Division of Maternal and Child Health Sciences, Ninewells Hospital and Medical School,
University of Dundee, Dundee DD1 9SY, United Kingdom

Tumors that form as a result of heightened mammalian target two key angiogenic factors; vascular endothelial growth fac-
of rapamycin (mTOR) signaling are highly vascularized. This tor (VEGF)-A (1) and angiopoietin-2 (Ang-2) (2). VEGF-A and
process of angiogenesis is regulated through hypoxia-inducible Ang-2 encourage angiogenesis by stimulating the formation of
factor (HIF)-mediated transcription of angiogenic factors. It is blood vessels that migrate into the tumor.
recognized that inhibition of mTOR with rapamycin can dimin- As well as vascularization, HIF is known to control the

Downloaded from www.jbc.org at Dundee University, MRC Unit on July 27, 2007
ish the process of angiogenesis. Our work shows that activation expression of ⬎70 genes involved in energy metabolism, apo-
of mTOR by Ras homologue enriched in brain (Rheb) overex- ptosis/survival, and metastasis (for reviews see Refs. 3, 4). Low
pression potently enhances the activity of HIF1␣ and vascular oxygen enhances the activity of HIF by stabilizing the ␣-subunit
endothelial growth factor (VEGF)-A secretion during hypoxia, of HIF. Oxygen-dependent prolyl hydroxylase domain (PHD)
which is reversed with rapamycin. Mutants of Rheb, which do proteins become active in the presence of oxygen that sequen-
not bind guanine nucleotide (D60K, D60V, N119I, and D122N) tially leads to hydroxylation of two proline residues within the
and are unable to activate mTOR, inhibit the activity of HIF oxygen-dependent degradation domain (ODDD) of the HIF
when overexpressed. We show that regulatory associated pro- ␣-subunit. Proline hydroxylation results in ubiquitin-mediated
tein of mTOR (Raptor) interacts with HIF1␣ and requires an degradation of the ␣-subunit that requires the Von Hippel-
mTOR signaling (TOS) motif located in the N terminus of Lindau (VHL) tumor suppressor protein, which is a component
HIF1␣. Furthermore, a mutant of HIF1␣ lacking this TOS motif of an E3 ubiquitin ligase complex (5). VHL syndrome, caused
dominantly impaired HIF activity during hypoxia and was through loss of function of this tumor suppressor, is a domi-
unable to bind to the co-activator CBP/p300. Rapamycin treat- nantly inherited familial syndrome that predisposes the patient
ments do not affect the stability of HIF1␣ and modulate HIF to renal cell carcinoma and cerebellar hemangioblastomas. In
activity via a Von Hippel-Lindau (VHL)-independent mecha- conditions of low oxygen tension, the HIF␣ protein is stable and
nism. We demonstrate that the high levels of HIF activity in cells binds to HIF␤ (also known as aryl hydrocarbon receptor
devoid of TSC2 can be reversed by treatments with rapamycin or nuclear translocator-ARNT). The HIF␣/HIF␤ dimer is then
the readdition of TSC2. Our work explains why human cancers rapidly translocated to the nucleus. As a dimer, they bind to
with aberrant mTOR signaling are prone to angiogenesis and RCGTG DNA sequences called hypoxia response elements.
suggests that inhibition of mTOR with rapamycin might be a Transcription is sequentially activated when the C terminus
suitable therapeutic strategy. transcriptional activation domain (C-TAD) of HIF␣ binds to the
co-activator CBP/p300 (for review see Ref. 6).
The tuberous sclerosis complex (TSC), which is a hamarto-
The supply of nutrients and oxygen is quickly depleted as mas syndrome, occurs through loss of function of the tumor
solid tumors grow in size. Nutritional and oxygen homeostasis suppressor proteins TSC1 (hamartin) and TSC2 (tuberin).
within solid tumors is reestablished by vascularization that Tumors that occur in TS patients are highly vascularized,
consequently results in tumor expansion. Oxygen sensing is implying that HIF might be involved and is regulated down-
known to control the mechanism of vasculogenesis through stream of TSC1/2. TSC is an autosomal genetic syndrome that
hypoxia-inducible factor (HIF)2-mediated transcription of occurs with an estimated prevalence of one in 6,000 newborns
and is characterized by slow growing benign tumors that form
* This research was supported by British Heart Foundation Intermediate in the heart, brain, kidneys, eyes, and skin (see Ref. 7 for review).
Research Fellowship No. FS/04/022 and Association of International Can- These tumors can lead to both renal and neurological compli-
cer Research Career Development Fellowship No 06-914/915 (to A. R. T.),
and funding from Tenovus (to S. L.). The costs of publication of this article
were defrayed in part by the payment of page charges. This article must human embryonic kidney; MEFs, mouse embryonic fibroblasts; mTOR,
therefore be hereby marked “advertisement” in accordance with 18 U.S.C. mammalian target of rapamycin; TOS, mTOR signaling; NF1, neurofibro-
Section 1734 solely to indicate this fact. min 1; PI3K, phosphoinositide 3-kinase; PHD, prolyl hydroxylases domain;
1
To whom correspondence should be addressed. Tel.: 029-2074-4055; Fax: PTEN, phosphatase and tensin homolog; Raptor, regulatory associated
029-2074-6551; E-mail: TeeA@cardiff.ac.uk. protein of mTOR; Redd1/2, regulated in development and DNA damage
2
The abbreviations used are: HIF, hypoxia-inducible factor; ARNT, aryl hydro- responses; Rheb, Ras homologue enriched in brain; Rictor, rapamycin-in-
carbon receptor nuclear translocator; CHAPS, 3-(3-cholamidopropyl)dim- sensitive companion of mTOR; S6K1, ribosomal protein S6 kinase 1; STK11,
ethylammonio-1-propanesulfonate; DFX, deferoxamine mesylate; DMOG, serine/threonine kinase 11; TSC, tuberous sclerosis complex; VEGF, vascu-
dimethyloxalylglycine; DMEM, Dulbecco’s modified Eagle’s medium; DSP, lar endothelial growth factor; VHL, Von Hippel-Lindau; HA, hemagglutinin;
dithiobis(succinimdyl propionate); 4E-BP1, eIF4E-binding protein 1; HEK, FITC, fluorescein isothiocyanate.

20534 JOURNAL OF BIOLOGICAL CHEMISTRY VOLUME 282 • NUMBER 28 • JULY 13, 2007
Regulation of HIF1␣ by mTOR
cations. TSC1 and TSC2 function as a heterodimer that inhibits enhances the transcriptional activity of HIF1␣ that does not
cell growth by impairing the Ser/Thr protein kinase called the involve increased HIF1␣ stabilization. Furthermore, mTOR
mammalian target of rapamycin (mTOR) (see review in Ref. 8). activation of HIF1␣ requires the interaction of HIF1␣ with Rap-
TSC1/TSC2 heterodimers function to inhibit mTOR, by acting tor. Given that Raptor functions as a scaffold protein that
as a GTPase-activating protein (GAP) toward the small G-pro- recruits downstream mTOR substrates with a TOS motif to
tein, Ras homologue enriched in brain (Rheb). Rheb has been mTOR (15), this work reveals that HIF1␣ is a downstream tar-
shown to interact with mTOR, and promotes signal transduc- get of mTOR.
tion when Rheb is in the active GTP-bound state (9). When in a
complex with TSC1, TSC2 inhibits Rheb-induced mTOR sig- EXPERIMENTAL PROCEDURES
naling by reverting Rheb to its inactive GDP-bound state (10). Chemicals and Materials—[3H]GTP, [3H]GDP, and [35S]me-
Therefore, when the normal function of TSC1/TSC2 het- thionine-radiolabeled reagents were purchased from Amer-
erodimers become compromised, Rheb becomes constitutively sham Biosciences (GE Healthcare UK Ltd.). Dimethyloxalylg-
GTP-bound and mTOR signaling is significantly enhanced and lycine was purchased from Frontier Scientific Europe Ltd
contributes to the pathology associated with TSC. (Lancashire, UK). Deferoxamine mesylate, MG-132 (carboben-
Within mammalian cells, mTOR functions as two distinct zoxy-L-leucyl-L-leucyl-L-leucinal), rapamycin, and LY294002
multi-protein kinase-signaling complexes. Rictor (rapamycin- were obtained from Merck Biosciences Ltd. (Nottingham, UK).
insensitive companion of mTOR) and LST8 (also referred to as CHAPS was ordered from Pierce. DSP (dithiobis(succinimdyl

Downloaded from www.jbc.org at Dundee University, MRC Unit on July 27, 2007
G-protein ␤-subunit-like protein (G␤L)) interact with mTOR propionate)) was purchased from Apollo Scientific Ltd. (Stock-
to form an mTOR/Rictor/LST8 protein kinase complex (11). port, UK). Rat anti-HA antibodies were purchased from Roche
mTOR also forms another multi-protein kinase complex with a Applied Science. Mouse anti-HA antibodies were kindly pro-
different scaffold protein called Raptor (regulatory-associated vided by M. Chou (University of Pennsylvania, Philadelphia,
protein of mTOR), where signaling from this (12, 13) promotes PA). Cell Signaling Technology anti-S6K1 and anti-S6K1 phos-
mTOR-mediated phosphorylation of distinct downstream sig- pho-Thr389 antibodies were purchased from New England Bio-
naling targets, such as the ribosomal S6 protein kinase 1 (S6K1) labs Ltd. (Hertfordshire, UK). Anti-HIF1␣ antibodies (clone:
and eukaryotic initiation factor 4E-binding protein 1 (4E-BP1), H1alpha67) were obtained from Novus Biologicals (Stratech
which is potently impaired by treatments with the immunosup- Scientific Ltd.). Anti-Coactivator p300 (C-20) antibodies were
pressant drug rapamycin. mTOR regulates cell growth and pro- bought from Santa Cruz Biotechnology, Inc. (Heidelberg,
liferation and is known to involve both S6K1 and 4E-BP1 (for Germany). Anti-FLAG antibodies (clone: M2), anti-Myc
review see Ref. 8). It is believed that Raptor functions as a scaf- (clone: 9E10) and all other reagents (unless stated) were
fold protein that recruits mTOR to its downstream substrates obtained from Sigma-Aldrich.
through their mTOR signaling (TOS) motifs. The TOS motif is Plasmids and Molecular Biology—N-terminal FLAG-tagged
a 5 amino acid peptide found within rapamycin-sensitive tar- pRK7/Rheb was generated as described in Ref. 10. Human HA-
gets of mTOR (FDL/IDL and FEMDI within S6K1 (14) and tagged 4E-BP1 was a kind gift from N. Sonenberg (McGill Uni-
4E-BP1 (15), respectively). Mutation of this TOS motif (espe- versity, Montreal, Canada). HIF1␣ was cloned into a modified
cially the highly conserved phenylalanine at position 1) pre- pRK7 vector so it expressed a triple N-terminal HA tag using
vents mTOR-directed phosphorylation of both S6K1 and human HIF1␣ cDNA as a template (obtained from Novus Bio-
4E-BP1 (14, 15). logicals (Stratech Scientific Ltd.)). Myc-tagged Raptor/pRK5
Interestingly, a common feature of hamartomas syndromes was a kind gift from D. M. Sabatini (Whitehead Institute for
caused by the loss of function of TSC1, TSC2 (16), PTEN (phos- Biomedical Research, Boston, MA). Human 4E-BP1 and
phatase and tensin homolog) (17), LKB1 (also known as STK11 human Rheb were subcloned into pGEX-2T/GST to generate
(serine/threonine kinase 11)) (18), or NF1 (neurofibromin 1) GST-tagged recombinant protein. Point mutations were gener-
(19) tumor suppressors is that signaling through mTOR is sig- ated as described in the QuikChange威 site-directed mutagene-
nificantly enhanced. Furthermore, VEGF levels are increased in sis kit (Stratagene). The N-terminal FLAG-tagged pRK7/TSC2
these syndromes (16, 20 –22). It is possible that increased VEGF construct was generated as previously described (26). The
secretion caused through loss of function of these tumor sup- pVHL-HA vector was kindly provided by P. Ratcliffe (Henry
pressors is mediated via an mTOR-dependent mechanism. Wellcome Building of Molecular Physiology, Oxford Univer-
Indeed, it has been shown that rapamycin has anti-angiogenic sity, UK) as described (27). The firefly luciferase reporter pGL2-
properties by reducing the expression of VEGF (23, 24). Fur- TK-HRE plasmid was generated by subcloning three copies of
thermore, VEGF expression within cells lacking TSC1 or TSC2 the hypoxia response element (HRE) (5⬘-GTGACTACGT-
is highly sensitive to treatments of rapamycin (16). The mech- GCTGCCTAG-3⬘) from the inducible nitric-oxide synthase
anism by how mTOR enhances VEGF expression has not been promoter into the promoter region of the pGL2-TK vector as
fully elucidated. It is postulated that HIF becomes more stable previously described (28) and was kindly provided by G. Melillo
by heightened mTOR activity, which causes enhanced VEGF (National Cancer Institute at Frederick, Maryland).
expression (25). We wanted to explore the mTOR-specific acti- Tissue Culture and Analysis of Cell Lysates—Human embry-
vation of HIF1␣ in greater detail. In this study, we potently onic kidney 293 (HEK293) cells were cultured (at 37 °C within
activate mTOR by overexpressing Rheb. We have previously 5% CO2) and maintained in Dulbecco’s modified Eagle’s
shown that Rheb can bind to and enhance mTOR signaling to medium (DMEM) supplemented with 10% fetal bovine serum.
downstream targets (9, 10). We show that mTOR directly DMEM and fetal bovine serum were purchased from Invitro-

JULY 13, 2007 • VOLUME 282 • NUMBER 28 JOURNAL OF BIOLOGICAL CHEMISTRY 20535
Regulation of HIF1␣ by mTOR
gen Ltd. (Paisley, UK). TSC2⫺/⫺ mouse embryonic fibroblasts O2 for 18 h on glass coverslips. Cells were then fixed in metha-
(MEFS) were a kind gift from D. J. Kwiatkowski (Harvard Med- nol at ⫺20 °C, quenched in 0.1% Na⫹ borohydrate in Tris-buff-
ical School, Boston, MA). The human renal carcinoma VHL⫺/⫺ ered saline (TBS; 50 mM Tris-Cl (pH 7.4), 150 mM NaCl) and
cells (786-O) were bought from ATCC. Cell transfections were then blocked for 1 h in TBS containing 10% goat serum. Mono-
carried out as described in the SuperFect威 manufacturer’s pro- clonal Anti-HA (clone HA-7) FITC conjugate (Sigma) was then
tocol (purchased from Qiagen Ltd. (West Sussex, UK)). applied to the cells in TBS containing 0.1% bovine serum albu-
HEK293 cells were transfected with ⬎95% efficiency, while min overnight at 4 °C and at 1:5000 dilution. After washing,
transfection of TSC2⫺/⫺ MEFS and human renal carcinoma coverslips were counterstained with DAPI antifade (Q-Bio-
VHL⫺/⫺ cells were ⬍10% efficient. Hypoxic treatments were gene) and mounted. The intracellular distribution of FITC and
carried out in a MACS VA500 Microaerophilic Work station DAPI fluorescence was observed using a Zeiss LSM 510 confo-
(Don Whitley Scientific, Shipley, West Yorkshire) containing a cal microscope.
humidified atmosphere equilibrated to 1% O2, 5% CO2, and Raptor Binding to mTOR Targets—To generate Raptor pro-
94% N2. To create cell lysates, cells were washed twice in phos-
tein to be used in the overlay assays, Myc-tagged Raptor was
phate-buffered saline, and then harvested with lysis buffer (10
overexpressed in HEK293 cells. These cells were lysed with 50
mM KH2PO4, 1 mM EDTA, 10 mM MgCl2, 50 mM ␤-glycero-
mM ␤-glycerophosphate, pH 7.4; 1 mM EDTA; 1 mM EGTA; 0.5
phosphate, 5 mM EGTA, 0.5% Nonidet P-40, 0.1% Brij 35, 1 mM
mM Na3VO4; 1 mM benzamidine hydrochloride; 1 mM dithio-
sodium orthovanadate, 40 mg/ml phenylsulfonyl fluoride, 10

Downloaded from www.jbc.org at Dundee University, MRC Unit on July 27, 2007
␮g/ml leupeptin, 5 ␮g pepstatin, pH 7.2). Cell lysates were son- threitol; 0.1 mM phenylmethane sulfonyl fluoride; 1% (v/v) Tri-
icated to help break down the nucleus and then spun at 14, 000 ton X-100, and 1 ␮g/ml each of pepstatin, antipain, and leupep-
rpm for 8 min at 4 °C to remove the cell debris. For Western blot tin. Proteins were transferred to Immobilon-P and blocked
analysis, these lysates were prepared as described for running with 5% (w/v) milk in Tris-buffered saline were incubated over-
NuPage威 Novex gels (bought from Invitrogen Ltd.) Proteins night at 4 °C with 1:10 diluted Myc-tagged Raptor lysate. Myc-
resolved on Novex gels were transferred to Millipore Immo- tagged Raptor that bound to proteins immobilized on Immo-
bilon-P (purchased from Upstate Ltd. (Hampshire, UK)) and bilon-P was then detected by incubating the blot with anti-Myc
blotted with the appropriate antibody followed by horseradish antibodies for 2 h in 2% (w/v) bovine serum albumin Tris-buff-
peroxidase-conjugated secondary antibodies. Enhanced ered saline followed by anti-mouse horseradish peroxidase-
chemiluminescence was carried out with Amersham Bio- conjugated secondary antibodies and ECL as described for
sciences ECLTM Western blotting detection reagents (pur- Western blotting. Raptor association with mTOR substrates
chased from GE Healthcare UK Ltd.). within cells was determined as previously described (15).
Immunoprecipitation—Proteins were immunoprecipitated Monitoring HIF1␣ Stability in Vivo—HEK293 cells trans-
using the relevant antibodies coupled to protein G-Sepharose fected with HA-HIF1␣ (grown on 6-cm2 plates) were incubated
(Amersham Biosciences). HIF1␣ immunoprecipitations were in methionine-free medium and labeled with 10 ␮Ci of
carried out overnight at 4 °C, and the protein G-Sepharose was [35S]methionine for 4 h in the hypoxic chamber set at 1% O2.
added for 1 h prior to being washed. Immunoprecipitates were The cells were washed twice in fresh media and then incubated
washed twice each with both buffer A (50 mM Hepes (pH 7.4), for 0, 0.5, 1, and 2 h in DMEM before cell lysates were gener-
100 mM NaCl, 10 mM MgCl2, 1 mg/ml bovine serum albumin, 1 ated. HA-HIF1␣ was immunoprecipitated for 1 h with anti-HA
mM dithiothreitol, 1% Triton) and buffer B (50 mM Hepes (pH antibodies bound to protein G-Sepharose. Immunoprecipi-
7.4), 100 mM NaCl, 10 mM MgCl2, 0.1% Triton) in the presence tated HIF1␣ was subjected to SDS-PAGE. The gel was stained,
of protease inhibitors. fixed, incubated with Amplify威 (Amersham Biosciences), dried,
Luciferase Reporter Assay—The Dual-Luciferase威 Reporter and then exposed to x-ray film.
Assay System (purchased from Promega UK Ltd. (Southamp-
Rheb Nucleotide Binding—To examine [3H]GTP and
ton UK)) was carried out using cell lysis buffer as recommended
[ H]GDP binding to Rheb in vitro, 2.5 ␮g of recombinant Rheb
3
by the manufacturer’s protocol with a Wallac Victor 2 1420
protein was incubated with either 1 ␮Ci of [3H]GTP or
Multi-label counter. We ran luciferase assays using an empty
[3H]GDP in 25 ␮l of loading buffer (50 mM HEPES, pH 7.5, 5
pGL3 vector (Promega) as a negative control.
mM EDTA, and 5 mg/ml bovine serum albumin). After 30 min,
VEGF Secretion—Secreted VEGF protein was detected using
the human VEGF DuoSet威 ELISA Development System (R&D 0.5 ␮l 1 M MgCl2 and 25 ␮l of cold 50 mM HEPES, pH 7.4, were
Systems, Abingdon, UK) according to the manufacturer’s added. The [3H]GTP- or [3H]GDP-loaded Rheb was then
instructions. After treatment, the cell medium was recovered applied to 0.2 ␮m cellulose nitrate membrane filter paper
following gentle agitation to maximize the recovery of secreted (Whatman) under suction in a vacuum manifold and washed
proteins and immediately frozen in liquid N2. The cells were three times with Rheb wash buffer (50 mM HEPES, pH 7.5, 0.5 M
then washed in phosphate-buffered saline and lysed for later NaCl, 0.1% Triton X-100, 5 mM MgCl2, 0.005% SDS plus prote-
analysis of protein content using Bradford reagent. The ase inhibitors).
quantity of VEGF secreted into the medium for each treat- Statistics—One-way analysis of variance with posthoc signif-
ment was expressed as a fraction of the respective cellular icance assessed with Tukey’s honestly significant difference test
protein content. using SigmaStat (version 2.0) was used on the data represented
Immunocytochemistry—Cells transfected with either empty by bar charts. Values are given as means with ⫾ S.D. A p value of
vector, HA-HIF1␣, or HA-HIF1␣ (F99A) were cultured at 1% ⬍0.05 was considered to be statistically significant.

20536 JOURNAL OF BIOLOGICAL CHEMISTRY VOLUME 282 • NUMBER 28 • JULY 13, 2007
Regulation of HIF1␣ by mTOR

FIGURE 1. mTOR activation enhances HIF1␣ transcription during treat-

Downloaded from www.jbc.org at Dundee University, MRC Unit on July 27, 2007
ment with DMOG. A, HEK293 cells transfected with a HIF-inducible luciferase
reporter and empty pRK7 or pRK7/FLAG-Rheb were serum-starved and where
indicted were treated with 1 mM DMOG and/or 50 nM rapamycin for 18 h.
Lysates prepared were analyzed for luciferase fluorescence. The HIF1␣ tran-
scriptional activity from the DMOG transfected without Rheb was standard-
ized to 100%. n ⫽ 6. *, p and **, p ⬍ 0.05 relative to activity obtained when
Rheb was overexpressed in the presence of DMOG. B, protein levels of FLAG-
Rheb, S6K1, and Thr389 phosphorylation of S6K1 were analyzed from the same
lysates. Endogenous HIF1␣ was immunoprecipitated from these lysates and
then subjected to Western blot analysis with HIF1␣ antibodies.

RESULTS
We wanted to examine whether the HIF1␣ transcriptional
activity during treatment with dimethyloxalylglycine
(DMOG) could be modulated by mTOR. A HIF1␣-induced FIGURE 2. mTOR activation enhances HIF1␣ transcription during
luciferase reporter construct was co-transfected to allow us hypoxia. A, serum-starved HEK293 cells transfected with a HIF-inducible
luciferase reporter and empty pRK7 or pRK7/FLAG-Rheb were transferred to
to measure HIF1␣ transcriptional activities within these low oxygen (1%) or maintained at 21% O2, where indicated, for 18 h in the
cells. Cells were treated with 1 mM DMOG for 18 h to induce presence or absence of 50 nM rapamycin. Lysates prepared were analyzed for
the activity of HIF1␣ (Fig. 1A). Treatment with DMOG pre- luciferase fluorescence. The HIF1␣ transcriptional activity from the hypoxic-
treated cells transfected without Rheb was standardized to 100%. n ⫽ 6. *, p
vents proline hydroxylase-mediated proteasomal degrada- and **, p ⬍ 0.05 relative to activity obtained when Rheb was overexpressed
tion of HIF1␣ and thus increases the stability of HIF1␣ (29). during hypoxic treatment. B, protein levels of FLAG-Rheb, S6K1, and Thr389
To enhance the activity of mTOR we overexpressed Rheb phosphorylation of S6K1 was also analyzed. Total levels of endogenous HIF1␣
were analyzed after being immunoprecipitated with anti-HIF1␣ antibodies.
within HEK293 cells. Interestingly, Rheb enhanced the levels C, levels of VEGF-A secretion within the cell media was measured as pg/mg
of HIF1␣-mediated transcription during DMOG treatment cell protein⫺1. n ⫽ 6. *, p and **, p ⬍ 0.05 relative to levels of secretion
obtained when Rheb was overexpressed during hypoxic treatment.
(Fig. 1A). As a control, we show that Rheb potently stimu-
lates mTOR-dependent phosphorylation of S6K1 at Thr389
(Fig. 1B). Rapamycin, which specifically inhibits mTOR, We wanted to examine whether the activity of HIF1␣ was
markedly reduced the DMOG-induced transcriptional activ- enhanced by Rheb overexpression during hypoxic conditions.
ity of HIF1␣ upon Rheb overexpression (Fig. 1A) and Similar to the DMOG-treated cells (Fig. 1A), Rheb enhanced
blocked the phosphorylation of S6K1 at Thr389 (Fig. 1B). the activity of HIF1␣ during hypoxia, which was significantly
Other studies have suggested that mTOR could stabilize reduced with treatment with rapamycin (Fig. 2A). Increased
HIF1␣ (25). Therefore, we compared the total levels of activity of mTOR by Rheb overexpression, as observed by
HIF1␣ within these cells. In DMOG-treated cells without increased Thr389 phosphorylation of S6K1, did not enhance the
Rheb overexpression, we observed a reduction of HIF1␣ protein levels of endogenous HIF1␣ (Fig. 2B). We also observed
upon treatment with rapamycin, which confirms previous a similar pattern of HIF1␣ expression within hypoxic cells (Fig.
reports that inhibition of mTOR reduces the levels of endog- 2B), when compared with DMOG-treated cells (Fig. 1B). Given
enous HIF1␣ within cells (Fig. 1B) (30). Interestingly, we that Rheb induced HIF1␣ transcription without increasing its
observed that DMOG-treated cells expressing Rheb, which protein levels, we propose that mTOR targets the transcrip-
had the highest activity of HIF1␣ (Fig. 1A), also had the low- tional activity of HIF1␣ rather than its stability. To confirm that
est protein levels of HIF1␣ when compared with cells not Rheb overexpression was increasing the activity of HIF1␣
expressing Rheb (Fig. 1B, lower panel). Furthermore, rapa- within these cells, we analyzed the secreted protein levels of
mycin treatment in cells expressing Rheb, which signifi- VEGF-A, which is a well-characterized downstream gene target
cantly reduced the activity of HIF1␣, did not reduce the of HIF involved in angiogenesis (Fig. 2C). We observed that the
HIF1␣ protein amount. levels of VEGF-A secretion paralleled the activity of HIF during

JULY 13, 2007 • VOLUME 282 • NUMBER 28 JOURNAL OF BIOLOGICAL CHEMISTRY 20537
Regulation of HIF1␣ by mTOR

Downloaded from www.jbc.org at Dundee University, MRC Unit on July 27, 2007
FIGURE 4. HIF1␣ interacts with the Raptor and requires an mTOR signal-
ing motif. A, equal levels of recombinant GST-4E-BP1 and GST-4E-BP1(F115A)
were subjected to SDS-PAGE and a Raptor overlay assay (lower panel). Equal
levels of protein are shown using anti-GST antibodies (upper panel).
B, HEK293 cells were treated with 100 ␮M DFX for 24 h, where indicated.
Cells were treated with the MG-132 (50 ␮M) for 30 min prior to prevent
degradation of HIF1␣. Total lysates and purified HIF1␣ samples after
immunoprecipitation with anti-HIF1␣ antibodies were resolved on SDS-
PAGE and subjected to a Raptor overlay assay. The band that corresponds
to HIF1␣ is marked. C, HEK293 cells transfected with HA-HIF1␣ and Myc-
Raptor, where indicated, were lysed in the presence of reversible cross-linker
DSP and non-ionic detergent (CHAPS). HA-HIF1␣ was immunoprecipitated
FIGURE 3. Guanine nucleotide binding mutants of Rheb inhibit the tran- using anti-HA antibodies and associated Myc-Raptor was determined.
scription activity of HIF1␣. A, binding of both [3H]GTP and [3H]GDP to D, HEK293 cells were co-transfected with Myc-Raptor and either empty pRK7
recombinant GST-Rheb proteins (wild-type, D60V, D60K, N119I, and D121N) vector, HA-4E-BP1, HA-HIF1␣, or HA-HIF1␣(F99A), where indicated. Cells were
or GST control were analyzed as described under “Experimental Procedures.” lysed in the presence of reversible cross-linker DSP and non-ionic detergent
B, serum-starved HEK293 cells transfected with a HIF-inducible luciferase (CHAPS). Exogenous 4E-BP1 and HIF1␣ was immunoprecipitated using
reporter and either empty pRK7 or pRK7 vector with FLAG-Rheb (wild-type, anti-HA antibodies as shown. Co-purifed Myc-Raptor was analyzed.
D60V, D60K, N119I, or D121N mutants) were transferred to a hypoxic cham-
ber set at 1% O2 for 18 h. Lysates prepared in the hypoxic chamber were
analyzed for luciferase fluorescence. The HIF1␣ transcriptional activity from described previously (32)), but do possess some ability to impair
the empty pRK7 was standardized to 100%. n ⫽ 6. *, p ⬍ 0.05 relative to mTOR (33). We wanted to investigate whether these Rheb
activity of empty pRK7 vector control. C, protein levels of S6K1 and Thr389
phosphorylation to measure mTOR activation was analyzed. FLAG-Rheb mutants, which are deficient at binding guanine nucleotide,
expression was analyzed with anti-FLAG antibodies to show equal expression could alter the levels of HIF1␣ transcriptional activity during
of the Rheb mutants.
hypoxia. Interestingly, these Rheb mutants dominantly inhib-
ited the transcriptional activity of HIF1␣ (Fig. 3B), when
hypoxia. Interestingly, Rheb overexpression enhanced the lev- expressed to the same level as wild-type (Fig. 3C). This shows
els of VEGF-A secretion during conditions of normoxia that that these Rheb mutants can sufficiently impair the basal
was sensitive to rapamycin. mTOR-mediated activation of HIF1␣ within HEK293 cells.
Small G-protein mutants that do not bind guanine nucleo- Indeed, these mutants of Rheb were unable to induce Thr389
tide typically form inactive complexes with their upstream reg- phosphorylation of S6K1 (Fig. 3C).
ulators, i.e. the putative guanine exchange factor (GEF), which Well-characterized downstream targets of mTOR, eIF4E-
impedes small G-protein-mediated signal transduction. Stud- binding protein 1 (4E-BP1) and S6K1, are recruited to mTOR
ies in yeast identified dominant-negative mutants of Rheb through their association with Raptor. To examine Raptor
(D60V and D60K), which were unable to efficiently bind to interaction, we utilized a Raptor overlay assay to detect Raptor-
guanine nucleotides and impaired TOR signaling (31). We also interacting proteins resolved on SDS-PAGE. This Raptor overlay
generated two additional Rheb(N119I) and Rheb(D122N) assay specifically detects proteins containing a TOS motif. For
mutants where these two mutated residues lie within the instance, the Raptor-overlay assay only detected wild-type
NKXD nucleotide binding motif of Rheb. Guanine nucleotide 4E-BP1 and not a mutant of 4E-BP1 where the phenylalanine
binding assays reveal that these D60V, D60K, N119I, and within the TOS motif at position 115 was mutated to an alanine
D122N mutants of Rheb are unable to bind to guanine nucleo- (Fig. 4A). To determine whether Raptor interacted with HIF1␣,
tides in vitro (Fig. 3A). These guanine binding-deficient we carried out a Raptor overlay assay on immunoprecipitated
mutants of Rheb are unable to efficiently impair acute activa- HIF1␣. HEK293 cells were treated with 100 ␮M deferoxamine
tion of S6K1 by insulin treatment (data not shown, and mesylate (DFX) for 24 h and then incubated with the proteoso-

20538 JOURNAL OF BIOLOGICAL CHEMISTRY VOLUME 282 • NUMBER 28 • JULY 13, 2007
Regulation of HIF1␣ by mTOR
mal inhibitor MG-132 prior to lysis to increase the protein lev-
els of HIF1␣. DFX is an iron chelator and is known to stabilize
HIF1␣ (34). We detected the HIF1␣ band with the Raptor over-
lay assay (Fig. 4B). This data suggests that Raptor interacts with
HIF1␣ (Fig. 4B). To confirm this interaction, we investigated
whether HIF1␣ co-immunoprecipitated with Raptor. HEK293
expressing HA-HIF1␣ and Myc-Raptor, where indicated, were
treated with DMOG and then lysed in the presence of reversible
crosslinker DSP and non-ionic detergent (CHAPS). Immuno-
precipitated HA-HIF1␣ was observed to associate with Myc-
Raptor (Fig. 4C). We were unable to co-purify HA-HIF1␣ with
Myc-Raptor using immunoprecipitation buffers containing
Nonidet P-40 (data not shown).
Interestingly, HIF1␣ contains a potential conserved TOS
motif (FVMVL) within an area of unknown function of HIF1␣
that is immediately 3⬘ of the Period-ARNT-Sim conserved
domain-A (PAS-A). We mutated the phenylalanine at position

Downloaded from www.jbc.org at Dundee University, MRC Unit on July 27, 2007
99 within the TOS motif of HIF1␣ and examined whether this
FIGURE 5. The HIF1␣(F99A) TOS motif mutant dominantly inhibits HIF1␣
transcription during hypoxia. A, serum-starved HEK293 cells transfected prevented Raptor interaction (Fig. 4D). We also examined the
with a HIF-inducible luciferase reporter with either empty pRK7, HA-HIF1␣, or interaction of Raptor with a well-known Raptor-interacting
HA-HIF1␣(F99A) in the presence of absence of FLAG-Rheb were treated with protein, 4E-BP1. HIF1␣ interacted less well with raptor when
50 nM rapamycin, where indicated, and transferred to a hypoxic chamber set
at 1% O2 for 18 h. Lysates prepared in the hypoxic chamber were analyzed for compared with 4E-BP1, but the interaction we observed was
luciferase fluorescence. The HIF1␣ transcriptional activity of the cells trans- prevented when the TOS motif within HIF1␣ was mutated.
fected with empty pRK7 vector and FLAG-Rheb was standardized to 100%.
n ⫽ 6. *, p ⬍ 0.05 relative to activity of empty pRK7 vector control group. **,
To examine whether this motif is essential for mTOR-medi-
p ⬍ 0.01 relative to activity of wild-type HA-HIF1␣ vector control group. ated HIF1␣ transcriptional we compared the transcriptional
B, protein levels of HA-HIF1␣, FLAG-Rheb, S6K1, and Thr389 phosphorylation activity of HIF1␣(F99A) to wild-type within hypoxic HEK293
of S6K1 were analyzed.
cells (Fig. 5A). Mutation of the TOS motif rendered HIF1␣
transcriptionally inactive during
hypoxia. Furthermore, overexpres-
sion of HIF1␣(F99A) significantly
reduced the activity of endogenous
HIF1␣ showing that mutation of the
TOS motif yields a dominant nega-
tive HIF1␣ mutant. We show that
equal levels of wild-type HIF1␣ and
HIF1␣(F99A) are being expressed
and that Rheb over-expression is
sufficient to enhance mTOR-medi-
ated S6K1 phosphorylation at
Thr389 (Fig. 5B).
It was postulated that mTOR sig-
naling enhances the stability of
HIF1␣ (25). To examine whether
the stability of HIF1␣ is regulated
by mTOR, we pulse-chased cells
overexpressing either HA-HIF1␣
or HIF1␣(F99A) and Rheb with
[S35]methionine under hypoxia (Fig.
6). If mTOR enhanced the stability
of HIF1␣, we would expect the
HIF1␣(F99A) mutant to be less sta-
FIGURE 6. mTOR does not modulate the stability of HIF1␣. HEK293 cells co-expressing FLAG-Rheb with
either wild-type HA-HIF1␣ or HA-HIF1␣(F99A) were grown at 1% O2 in media containing [35S]methionine for ble. We would also expect that rap-
4 h, which was then replaced with unlabeled media in the presence or absence of rapamycin, where indicated. mycin would reduce the stability of
These cells were lysed after 0, 0.5, 1, and 2 h in the 1% O2 hypoxic chamber. Exogenous HA-HIF1␣ was immu-
noprecipitated with anti-HA antibodies and the relative level of [35S]methionine incorporation into HA-HIF1␣
HIF1␣. Instead, we found that the
was determined as described under “Experimental Procedures.” Protein levels of HA-HIF1␣, FLAG-Rheb, S6K1, protein half-life of the HIF1␣(F99A)
and Thr389 phosphorylation of S6K1 were also analyzed. The level of [35S]methionine-labeled HIF1␣ wild-type mutant was similar to that of the
and HIF1␣(F99A) at the zero time point was standardized at 100%. n ⫽ 3. There was no significant difference of
[35S]methionine incorporation when comparing wild-type HIF1␣ with the F99A mutant or when comparing wild-type and treatments with rapa-
cells treated with or without rapamycin at similar time point. mycin did not alter the stability of

JULY 13, 2007 • VOLUME 282 • NUMBER 28 JOURNAL OF BIOLOGICAL CHEMISTRY 20539
Regulation of HIF1␣ by mTOR

FIGURE 7. Rapamycin inhibits HIF transcription independently of VHL.


VHL⫺/⫺ MEFS were transiently transfected with a HIF-inducible luciferase
reporter and either empty pRK7 or VHL vector (where indicated). These cells
were maintained at 21% O2 for 18 h in the presence or absence of 50 nM
rapamycin. Lysates were analyzed for luciferase fluorescence and the HIF
transcriptional activity from the VHL⫺/⫺ cells was standardized to 100%. n ⫽
5. *, p ⬍ 0.05 relative to activity of the VHL⫺/⫺ control. To detect the presence
of HA-VHL, HA-VHL was immunoprecipitated from these lysates, then sub-
jected to Western blot analysis using anti-HA antibodies. FIGURE 8. HIF1␣ and HIF1␣ (F99A) localize to the nucleus in hypoxia. Cells

Downloaded from www.jbc.org at Dundee University, MRC Unit on July 27, 2007
were exposed to 1% O2 for 18 h following transfection and then processed for
immunocytochemistry. Anti HA-FITC conjugated antibody was used to
either the wild-type or the F99A mutant of HIF1␣. This exper- observe the distribution of HA-HIF1␣/HIF1␣(F99A) expression relative to the
DAPI-stained chromatin. The merged image illustrates that the HA-FITC anti-
iment supports our hypothesis that mTOR does not directly body was entirely nuclear in transfected cells. Images are representative of
modulate the stability of HIF1␣. four independent experiments.
It is known that VHL is required for the ubiquitin-mediated
degradation of the ␣-subunit of HIF (35). To verify that signal-
ing through mTOR does not modulate the stability of HIF␣
through a VHL-dependent mechanism, we analyzed HIF tran-
scription within human renal carcinoma VHL⫺/⫺ cells. In nor-
moxic conditions these VHL-null cells possess heightened
transcriptional activity of HIF (Fig. 7) showing that HIF is sta-
bilized and active in these cells during high oxygen tensions.
The transcriptional activity of HIF in these cells was due to the
loss of VHL as the levels of HIF activity was completely ablated
when VHL was transfected back. We observed that rapamycin
potently reduced the levels of HIF transcription in the absence
of VHL. This experiment shows that rapamycin inhibits HIF
transcription independently of VHL and again supports the FIGURE 9. Co-activator p300 interaction with HIF1␣ is impaired in the
notion that TOR does not influence the stability of HIF1␣. HIF1␣(F99A) mutant. Serum-starved HEK293 cells transfected with empty
pRK7, HA-HIF1␣, or HA-HIF1␣(F99A) where transferred to a hypoxic chamber
Given that mTOR does not appear to affect the stability of set at 1% O2 for 18 h before being lysed. Exogenous HA-HIF1␣ was immuno-
HIF1␣, the increased transcriptional activity of HIF1␣ by Rheb precipitated with anti-HA antibodies, and the amount of co-purified p300
must be either caused by enhanced nuclear translocation of was quantified by Western blot analysis followed by densitometry (Image J,
version 1.37V). Numbers below the panel indicate the ratios of the signals for
HIF1␣ or increased binding of HIF1␣ to other components of p300 relative to HIF.
the transcription activation complex. First of all we examined
whether the cellular distribution of the HIF1␣(F99A) mutant these cells during normoxic and hypoxic conditions (Fig. 10).
was different to that of wild-type HIF1␣. Confocal immunoflu- We observed that rapamycin robustly inhibited the hypoxia-
orescence studies showed that both wild-type and HIF1a(F99A) induced activation of HIF in cells without TSC2 by 70%. This
mutant was mainly nuclear (Fig. 8), as observed by co-localiza- rapamycin-sensitive level of HIF transcriptional activity during
tion with the nuclear stain, DAPI. We next examined if the hypoxia was caused by the loss of TSC2 because transient
HIF1␣(F99A) mutant was deficient at forming transcriptional expression of TSC2 also potently blocked the activity of HIF by
complexes. To examine this possibility, we measured the inter- 70%. This result demonstrates that the loss of function of TSC2
action of p300 with both the HIF1␣(F99A) mutant and wild- potently drives HIF-mediated transcription during conditions
type HIF1␣ (Fig. 9). We observed a significant loss of p300 bind- of low oxygen and is sensitive to treatments with rapamycin.
ing to the HIF1␣(F99A) mutant, which suggests that mTOR It is known that HIF-mediated gene expression is regulated
enhances HIF1␣ transcription through assembly of the HIF1␣ through PI3K- and mTOR-dependent mechanisms (35–37).
transcriptional machinery. To investigate PI3K and mTOR induced activation of HIF in
We wanted to examine the levels of HIF transcription within more detail, we treated HEK293 cells with insulin for 18 h in the
cells lacking TSC2. It is known that the loss of function of TSC2 presence and absence of LY294002 (to inhibit PI3K) and rapa-
potently enhances cell signaling through mTOR (8). We trans- mycin (to inhibit mTOR) during hypoxia. We investigated the
fected TSC2⫺/⫺ MEFS with the HIF reporter construct to activity of HIF (Fig. 11A) and the amount of VEGF-A protein
measure the relative levels of HIF transcriptional activity within that was secreted by these cells (Fig. 11B). Insulin increased the

20540 JOURNAL OF BIOLOGICAL CHEMISTRY VOLUME 282 • NUMBER 28 • JULY 13, 2007
Regulation of HIF1␣ by mTOR
sufficient to activate PI3K/mTOR signaling after 18 h of treat-
ment. Treatments with either LY294002 or rapamycin were
sufficient to block insulin-induced phosphorylation of S6K1
showing that we were inhibiting PI3K and mTOR-mediated
signaling in these cells. It is important to note that LY294002
also inhibits mTOR. Therefore, the difference of inhibition we
observe with rapamycin and LY294002 represents the level of
HIF transcriptional activity (Fig. 11A) and VEGF-A secretion
(Fig. 11B) that is dependent on PI3K. Upon insulin stimulation,
we observed a marked increased in the HIF transcriptional
activity within cells during hypoxia that was completely
blocked by treatments with LY294002 and significantly
impaired by rapamycin (Fig. 11A). Similarly, LY294002 blocked
FIGURE 10. Hypoxia-induced HIF transcription in TSC2-null cells are
highly sensitive to rapamycin. Serum-starved TSC2⫺/⫺ MEFS were tran-
insulin-induced VEGF-A secretion during hypoxia while
siently transfected with a HIF-inducible luciferase reporter and either empty VEGF-A secretion was significantly repressed by rapamycin
pRK7 or pRK7/FLAG-TSC2 (where indicated). These cells were serum-starved (Fig. 11B). These experiment shows that mTOR is necessary for
and were transferred to low oxygen (1%) or maintained at 21% O2, where

Downloaded from www.jbc.org at Dundee University, MRC Unit on July 27, 2007
indicated, for 18 h in the presence or absence of 50 nM rapamycin. Lysates the maximal activation of HIF-mediated transcription upon
prepared were analyzed for luciferase fluorescence and the HIF transcrip- insulin stimulation.
tional activity from the TSC2⫺/⫺ cells during hypoxia was standardized to
100%. n ⫽ 6. *, p and **, p ⬍ 0.05 relative to activity of the TSC2⫺/⫺ control.
Protein levels of FLAG-TSC2 were determined by Western blot analysis after DISCUSSION
immunoprecipitation with anti-FLAG antibodies. The asterisk indicates a non-
specific band that migrates above FLAG-TSC2. We show that mTOR positively enhances the level of HIF-
mediated transcription. Rheb-specific activation of mTOR
enhanced the transcriptional activity of HIF during condi-
tions that favored HIF1␣ stabilization, i.e. during hypoxia
(Fig. 2A) and treatments with DMOG (Fig. 1A). This
enhanced HIF activity was blocked by treatments with rapa-
mycin showing that Rheb-induced HIF activity was caused
by the heightened activity of mTOR. A potential function of
mTOR would be to regulate the expression levels of HIF1␣.
Dogma has it that stability is the rate-limiting factor that
determines the protein levels of HIF1␣. However, it is
important to appreciate that the regulation of HIF1␣ expres-
sion is multifaceted with additional inputs that function at
the level of transcription and translation (30, 35–37). Indeed,
it is known that the translation of HIF1␣ can be modulated
by mTOR through cap-dependent mechanisms that is driven
by eIF4E and repressed by the translation repressor, 4E-BP1
(30). The reduction of endogenous HIF1␣ that we observe
after rapamycin treatment in the presence of DMOG or
hypoxia conditions (Figs. 1B and 2B, respectively), could be
accountable by the reduced rates of HIF1␣ protein synthesis.
For added complexity, the translation of HIF1␣ can also be
maintained through an internal ribosomal entry site (IRES).
FIGURE 11. Insulin-induced HIF activation and VEGF-A secretion is partially
IRES-mediated translation does not require eIF4E and so
blocked by rapamycin. A, serum-starved HEK293 cells transfected with a HIF- confers a rapamycin-insensitive mechanism to promote
inducible luciferase reporter were transferred to low oxygen (1%) or maintained HIF1␣ translation during times when mTOR signaling is
at 21% O2. Where indicated, cells where stimulated with 100 nM insulin in the
presence or absence of either 50 nM rapamycin or 50 ␮M LY294002. After 18 h, the switched off (38). Given that rapamycin did not reduce the
cells were harvested, and the lysates were analyzed for luciferase fluorescence. levels of HIF1␣ protein in cells over-expressing Rheb (Figs. 1B
The HIF1␣ transcriptional activity from hypoxic unstimulated cells was standard-
ized to 100%. B, levels of VEGF-A secretion within the cell media was measured as
and 2B), the reduction of Rheb-induced HIF transcriptional
pg/mg cell protein⫺1. The level of VEGF-A secretion from hypoxic-unstimulated activity by rapamycin was not caused by a loss of the HIF1␣
cells was standardized to 100%. For both graphs; n ⫽ 6. *, p and **, p ⬍ 0.05 protein. This rules out the possibility that mTOR modulates the
relative to the levels of HIF activity or VEGF-A secretion obtained from stimulated
cells during hypoxia. C, S6K1 phosphorylation was analyzed by a mobility shift on stability of HIF1␣ and is supported by the observation that
SDS-PAGE. p70 and p80 isoforms of S6K1 are marked. rapamycin treatment did not alter the rates of protein break-
down of HIF1␣ (Fig. 6). Furthermore, rapamycin potently
phosphorylation of S6K1 (as observed by a mobility shift of the impaired the activity of HIF in VHL-null cells (Fig. 7). VHL is
p70 and p85 isoforms of S6K1 to the higher phosphorylated required for the ubiquitin-mediated degradation of the ␣-sub-
bands) (Fig. 11C), showing that treatment with 100 nM insulin is unit of HIF. Therefore, our data suggests that mTOR promotes

JULY 13, 2007 • VOLUME 282 • NUMBER 28 JOURNAL OF BIOLOGICAL CHEMISTRY 20541
Regulation of HIF1␣ by mTOR
the transcriptional activity of HIF1␣ and does not involve VHL- with each of these hamartomas syndromes are known to have
mediated degradation of HIF1␣. high levels of mTOR activation (see review in Ref. 8). Interest-
The mechanism by how mTOR modulates HIF within cells ingly, we observed high levels of HIF activity in cells lacking
has remained elusive to date. In this manuscript, we have iden- TSC2, which was reversed when we added back TSC2 or inhib-
tified a potential FVMVL TOS motif within a previously unde- ited mTOR with rapamycin (Fig. 9). Our work suggests that the
fined region of HIF1␣. This motif is similar to well known TOS high degree of vascularization observed in tumors arising from
motifs found in both 4E-BP1 and S6K1, which are FEMDI and these syndromes could be the direct consequence of high levels
FD(L/I)DL, respectively. Raptor is thought to bind to the TOS of mTOR activity. For instance, aberrant signaling through
motif within mTOR substrates. This interaction recruits these mTOR would enhance the activity of HIF and encourage the
substrates to mTOR for their optimal phosphorylation. For process of angiogenesis during hypoxia.
instance, mutation of the TOS motif by alanine substitution of It is known that a negative feedback loop, which is activated
the phenylalanine rendered both 4E-BP1 and S6K1 unrespon- by HIF, inhibits the mTOR pathway and HIF function. This
sive to mTOR (14, 15). We show that raptor interacts with feedback loop is regulated by Redd1/2 (Regulated in Develop-
HIF1␣ (Fig. 4, B and C) and this interaction is inhibited when ment and DNA damage responses), which are also referred to as
the phenylalanine within the TOS motif is mutated to an ala- RTP801/801L and are transcriptionally up-regulated by HIF
nine (Fig. 4D). We observed a lower level of raptor interaction (40). The inhibition of mTOR by either Redd1 or Redd2
with HIF1␣ when compared with 4E-BP1 (Fig. 4D) and this requires TSC2 (41) and suggests that Redd1/2 activates

Downloaded from www.jbc.org at Dundee University, MRC Unit on July 27, 2007
might be caused by the differences in the cellular distribution of TSC1/2. The mechanism by how Redd1/2 signals through
both HIF1␣ and 4E-BP1, i.e. 4E-BP1 is cytoplasmic while HIF1␣ TSC2 is currently undefined. This negative feedback loop
is mainly nuclear. We show that mutation of the TOS motif makes physiological sense, as it would be unfavorable for cellu-
renders HIF1␣ transcriptionally inactive and this TOS mutant lar HIF responses to be maintained for long periods of time. In
is unresponsive to enhanced mTOR signaling when Rheb is our experiments, we override this Redd1/2 negative feedback
overexpressed (Fig. 5A). The HIF1␣ TOS mutant was predom- loop by overexpressing Rheb, which is sufficient to potently
inantly nuclear (Fig. 8). These data suggest that raptor interac- enhance mTOR signaling during long term conditions of
tion with HIF1␣ does not influence the nuclear translocation of hypoxia (Fig. 2B). Cells lacking functional TSC1/TSC2 would
HIF␣, a process that requires the interaction of HIF␤ with also lack this hypoxic induced negative feedback loop through
HIF1␣. Interestingly, mutation of the TOS motif within HIF1␣, Redd1/2. The inability of Redd1/2 to activate the TSC1/TSC2
transforms HIF1␣ into a dominant negative mutant that inhib- heterodimer in TSC2-null cells might be the reason why we
its endogenous HIF activity (Fig. 5A). It is possible that the observe high levels of HIF activity that is potently suppressed by
HIF1␣(F99A) mutant sequesters the HIF␤ subunit to form treatments with rapamycin (Fig. 9). This study suggests that
inactive heterodimers, and thus competes with the interaction inhibition of mTOR might be a suitable strategy to treat har-
of HIF␤ with endogenous HIF1␣. Mutation of the TOS motif matomas syndromes to repress mTOR-mediated angiogenesis
significantly impaired the interaction of HIF1␣ with the co- through HIF.
activator CBP/p300 (Fig. 9) and supports our view that the
HIF1␣(F99A) mutant is unable to form functional transcription Acknowledgments—We thank Prof. Grahame Hardie and the Divi-
complexes. sion of Molecular Physiology for their support.
HIF1␣ is a reported phosphoprotein. It is possible that
mTOR directly phosphorylates HIF1␣, which is required to
promote HIF1␣ ability to function as a transcription factor. We REFERENCES
are currently pursuing this line of investigation to see whether 1. Ikeda, E., Achen, M. G., Breier, G., and Risau, W. (1995) J. Biol. Chem. 270,
there are any mTOR-dependent and rapamycin-sensitive phos- 19761–19766
phorylation sites within HIF1␣. It would be interesting to 2. Mandriota, S. J., and Pepper, M. S. (1998) Circ. Res. 83, 852– 859
examine whether a functional mTOR/raptor/LST8 complex 3. Pugh, C. W., and Ratcliffe, P. J. (2003) Nat. Med. 9, 677– 684
4. Carmeliet, P. (1998) Nature 394, 485– 490
could form within the nucleus. It has been reported that mTOR 5. Kim, W. Y., and Kaelin, W. G. (2004) J. Clin. Oncol. 22, 4991–5004
shuttles between the cytoplasm and the nucleus and that this 6. Brahimi-Horn, C., Mazure, N., and Pouyssegur, J. (2005) Cell Signal. 17,
cytoplasmic-nuclear interchange of mTOR is necessary for the 1–9
mTOR-dependent phosphorylation of 4E-BP1 and S6K1 (39). 7. Cheadle, J. P., Reeve, M. P., Sampson, J. R., and Kwiakowski, D. J. (2000)
Several hamartomas syndromes that are caused through the Hum. Genet. 107, 97–114
8. Tee, A. R., and Blenis, J. (2005) Semin. Cell. Dev. Biol. 16, 29 –37
loss of tumor suppressors have highly vascularized tumors.
9. Tee, A. R., Blenis, J., and Proud, C. G. (2005) FEBS Lett. 579, 4763– 4768
These include TSC, Cowden’s disease, Peutz-Jeghers syndrome 10. Tee, A. R., Manning, B. D., Roux, P. P., Cantley, L. C., and Blenis, J. (2003)
and Neurofibromatosis that are caused through mutations that Curr. Biol. 13, 1259 –1268
impair the normal tumor suppressor function of TSC1 or 11. Sarbassov, D. D., Ali, S. M., Kim, D. H., Guertin, D. A., Latek, R. R., and
TSC2, PTEN, LKB1, and NF1, respectively. It is known that Erdjument-Bromage, H. (2004) Curr. Biol. 14, 1296 –1302
VEGF secretion is enhanced when there is a loss of function of 12. Hara, K., Maruki, Y., Long, X., Yoshino, K., Oshiro, N., and Hidayat, S.
(2002) Cell 110, 177–189
either one of the TSC1, TSC2 (16), PTEN (20), LKB1 (21), or
13. Kim, D. H., Sarbassov, D. D., Ali, S. M., Latek, R. R., Guntur, K. V., and
NF1 (22) tumor suppressors. This suggests that the normal Erdjument-Bromage, H. (2003) Mol. Cell 11, 895–904
function of these tumor suppressors is to repress VEGF expres- 14. Schalm, S. S., and Blenis, J. (2002) Curr. Biol. 12, 632– 639
sion and this process likely involves HIF. Lesions associated 15. Schalm, S. S., Fingar, D. C., Sabatini, D. M., and Blenis, J. (2003) Curr. Biol.

20542 JOURNAL OF BIOLOGICAL CHEMISTRY VOLUME 282 • NUMBER 28 • JULY 13, 2007
Regulation of HIF1␣ by mTOR
13, 797– 806 29. Chan, D. A., Sutphin, P. D., Denko, N. C., and Giaccia, A. J. (2002) J. Biol.
16. El-Hashemite, N., Walker, V., Zhang, H., and Kwiatkowski, D. J. (2003) Chem. 277, 40112– 40117
Cancer Res. 63, 5173–5177 30. Treins, C., Giorgetti-Peraldi, S., Murdaca, J., Semenza, G. L., and Van,
17. Manning, B. D., Tee, A. R., Logsdon, M. N., Blenis, J., and Cantley, L. C. O. E. (2002) J. Biol. Chem. 277, 27975–27981
(2002) Mol. Cell 10, 151–162 31. Tabancay, A. P. Jr., Gau, C. L., Machado, I. M., Uhlmann, E. J., Gutmann,
18. Inoki, K., Zhu, T., and Guan, K. L. (2003) Cell 115, 577–590 D. H., Guo, L., and Tamanoi, F. (2003) J. Biol. Chem. 278, 39921–39930
19. Johannessen, C. M., Reczek, E. E., James, M. F., Brems, H., Legius, E., and 32. Li, Y., Inoki, K., and Guan, K. L. (2004) Mol. Cell. Biol. 24, 7965–7975
Cichowski, K. (2005) Proc. Natl. Acad. Sci. U. S. A. 102, 8573– 8578 33. Long, X., Lin, Y., Ortiz-Vega, S., Yonezawa, K., and Avruch, J. (2005) Curr.
20. Gomez-Manzano, C., Fueyo, J., Jiang, H., Glass, T. L., Lee, H. Y., Hu, M., Biol. 15, 702–713
Liu, J. L., Jasti, S. L., Liu, T. J., Conrad, C. A., and Yung, W. K. (2003) Ann. 34. Woo, K. J., Lee, T. J., Park, J. W., and Kwon, T. K. (2006) Biochem. Biophys.
Neurol. 53, 109 –117 Res. Commun. 343, 8 –14
21. Ylikorkala, A., Rossi, D. J., Korsisaari, N., Luukko, K., Alitalo, K., Henke- 35. Baba, M., Hirai, S., Yamada-Okabe, H., Hamada, K., Tabuchi, H., Koba-
meyer, M., and Makela, T. P. (2001) Science 293, 1323–1326 yashi, K., Kondo, K., Yoshida, M., Yamashita, A., Kishida, T., Nakaigawa,
22. Kotsuji-Maruyama, T., Imakado, S., Kawachi, Y., and Otsuka, F. (2002) J. N., Nagashima, Y., Kubota, Y., Yao, M., and Ohno, S. (2003) Oncogene 22,
Dermatol. 29, 713–717 2728 –2738
23. Guba, M., von Breitenbuch, P., Steinbauer, M., Koehl, G., Flegel, S., Hor- 36. Hudson, C. C., Liu, M., Chiang, G. G., Otterness, D. M., Loomis, D. C.,
nung, M., Bruns, C. J., Zuelke, C., Farkas, S., Anthuber, M., Jauch, K. W., Kaper, F., Giaccia, A. J., and Abraham, R. T. (2002) Mol. Cell. Biol. 22,
and Geissler, E. K. (2002) Nat. Med. 8, 128 –135 7004 –7014
24. Treins, C., Giorgetti-Peraldi, S., Murdaca, J., Monthouel-Kartmann, 37. Mayerhofer, M., Valent, P., Sperr, W. R., Griffin, J. D., and Sillaber, C.

Downloaded from www.jbc.org at Dundee University, MRC Unit on July 27, 2007
M. N., and Van Obberghen, E. (2005) Mol. Endocrinol. 19, 1304 –1317 (2002) Blood 100, 3767–3775
25. Brugarolas, J., Lei, K., Hurley, R. L., Manning, B. D., Reiling, J. H., Hafen, E., 38. Lang, K. J., Kappel, A., and Goodall, G. J. (2002) Mol. Biol. Cell 13,
Witters, L. A., Ellisen, L. W., and Kaelin, W. G., Jr. (2004) Genes Dev. 18, 1792–1801
2893–2904 39. Kim, J. E., and Chen, J. (2000) Proc. Natl. Acad. Sci. U. S. A. 97,
26. Tee, A. R., Fingar, D. C., Manning, B. D., Kwiatkowski, D. J., Cantley, L. C., 14340 –14345
and Blenis, J. (2002) Proc. Natl. Acad. Sci. U. S. A. 99, 13571–13576 40. Shoshani, T., Faerman, A., Mett, I., Zelin, E., Tenne, T., Gorodin, S.,
27. Cockman, M. E., Masson, N., Mole, D. R., Jaakkola, P., Chang, G. W., Moshel, Y., Elbaz, S., Budanov, A., Chajut, A., Kalinski, H., Kamer, I.,
Clifford, S. C., Maher, E. R., Pugh, C. W., Ratcliffe, P. J., and Maxwell, P. H. Rozen, A., Mor, O., Keshet, E., Leshkowitz, D., Einat, P., Skaliter, R., and
(2000) J. Biol. Chem. 275, 25733–25741 Feinstein, E. (2002) Mol. Cell. Biol. 22, 2283–2293
28. Rapisarda, A., Uranchimeg, B., Scudiero, D. A., Selby, M., Sausville, E. A., 41. Corradetti, M. N., Inoki, K., and Guan, K. L. (2005) J. Biol. Chem. 280,
Shoemaker, R. H., and Melillo, G. (2002) Cancer Res. 62, 4316 – 4324 9769 –9772

JULY 13, 2007 • VOLUME 282 • NUMBER 28 JOURNAL OF BIOLOGICAL CHEMISTRY 20543

Você também pode gostar