C. L. Scott, D. J. Walker, E. Cwiklinski, C. Tait, A. R. Tee and S. C. Land

You might also like

Download as pdf or txt
Download as pdf or txt
You are on page 1of 18

C. L. Scott, D. J. Walker, E. Cwiklinski, C. Tait, A. R. Tee and S. C.

Land
Am J Physiol Lung Cell Mol Physiol 299:455-471, 2010. First published Jul 9, 2010;
doi:10.1152/ajplung.00348.2009

You might find this additional information useful...

Supplemental material for this article can be found at:


http://ajplung.physiology.org/cgi/content/full/ajplung.00348.2009/DC1
This article cites 59 articles, 33 of which you can access free at:
http://ajplung.physiology.org/cgi/content/full/299/4/L455#BIBL
Updated information and services including high-resolution figures, can be found at:
http://ajplung.physiology.org/cgi/content/full/299/4/L455
Additional material and information about AJP - Lung Cellular and Molecular Physiology can be found
at:
http://www.the-aps.org/publications/ajplung

Downloaded from ajplung.physiology.org on September 30, 2010


This information is current as of September 30, 2010 .

AJP - Lung Cellular and Molecular Physiology publishes original research covering the broad scope of molecular, cellular, and
integrative aspects of normal and abnormal function of cells and components of the respiratory system. It is published 12 times a
year (monthly) by the American Physiological Society, 9650 Rockville Pike, Bethesda MD 20814-3991. Copyright © 2010 by the
American Physiological Society. ISSN: 1040-0605, ESSN: 1522-1504. Visit our website at http://www.the-aps.org/.
Am J Physiol Lung Cell Mol Physiol 299: L455–L471, 2010.
First published July 9, 2010; doi:10.1152/ajplung.00348.2009.

Control of HIF-1␣ and vascular signaling in fetal lung involves cross talk
between mTORC1 and the FGF-10/FGFR2b/Spry2 airway branching
periodicity clock
C. L. Scott,1 D. J. Walker,1 E. Cwiklinski,1 C. Tait,1 A. R. Tee,2 and S. C. Land1
1
Centre for Cardiovascular and Lung Biology, Division of Medical Sciences, Ninewells Hospital and Medical School,
University of Dundee, Dundee, Scotland; and 2Institute of Medical Genetics, Cardiff University, Heath Park, Cardiff, Wales,
United Kingdom
Submitted 5 October 2009; accepted in final form 1 July 2010

Scott CL, Walker DJ, Cwiklinski E, Tait C, Tee AR, Land SC. for hypoxia is unlikely, as branched airway growth occurs at
Control of HIF-1␣ and vascular signaling in fetal lung involves cross talk higher oxygen tensions (7, 37, 46, 49, 54). Vascular branching,
between mTORC1 and the FGF-10/FGFR2b/Spry2 airway branching by contrast, is almost completely dependent on the low PO2 of
periodicity clock. Am J Physiol Lung Cell Mol Physiol 299: L455–L471, the fetal lung and is abolished by experimental increases in
2010. First published July 9, 2010; doi:10.1152/ajplung.00348.2009.—

Downloaded from ajplung.physiology.org on September 30, 2010


oxygen tension (1, 22, 54). Despite these different sensitivities
Lung development requires coordinated signaling between airway and
vascular growth, but the link between these processes remains un-
to oxygen, airway and vascular systems develop in tandem to
clear. Mammalian target of rapamycin complex-1 (mTORC1) can form an interwoven tubular network with a shared semifractal
amplify hypoxia-inducible factor-1␣ (HIF-1␣) vasculogenic activity pattern of branching to the level of the respiratory acinus (57).
through an NH2-terminal mTOR binding (TOS) motif. We hypothe- Thus, although hypoxia is necessary for proper lung develop-
sized that this mechanism coordinates vasculogenesis with the fibro- ment, its effects are secondary to the signaling network that
blast growth factor (FGF)-10/FGF-receptor2b/Spry2 regulator of air- links airway and vascular growth together.
way branching. First, we tested if the HIF-1␣ TOS motif participated Airway branching morphogenesis begins by evagination of
in epithelial-mesenchymal vascular signaling. mTORC1 activation by the laryngo-tracheal groove into the surrounding mesenchyme
insulin significantly amplified HIF-1␣ activity at fetal PO2 (23 mmHg) tissue. After dividing into two distal buds, a series of sequential
in human bronchial epithelium (16HBE14o-) and induced vascular branching events occurs to form the conductive airways of the
traits (Flk1, sprouting) in cocultured human embryonic lung mesen- respiratory tree. Outgrowth of the airways is induced by
chyme (HEL-12469). This enhanced activation of HIF-1␣ by
fibroblast growth factor-10 (FGF-10), which is transiently
mTORC1 was abolished on expression of a HIF-1␣ (F99A) TOS-
mutant and also suppressed vascular differentiation of HEL-12469
expressed in the mesenchyme tissue surrounding each prospec-
cocultures. Next, we determined if vasculogenesis in fetal lung in- tive bud. This is believed to activate Ras/Raf/MEK1 signaling
volved regulation of mTORC1 by the FGF-10/FGFR2b/Spry2 path- to the extracellular-signal regulated kinases 1 and 2 (ERK1/2)
way. Fetal airway epithelium displayed distinct mTORC1 activity in through its receptor, FGFR2b, expressed in the airway epithe-
situ, and its hyperactivation by TSC1⫺/⫺ knockout induced wide- lium (7, 52, 53). Negative feedback of this pathway is
spread VEGF expression and disaggregation of Tie2-positive vascular mediated by Sprouty2 (Spry2), whose activity is induced as
bundles. FGF-10-coated beads grafted into fetal lung explants from an additional, but time-delayed, response to FGFR2b/
Tie2-LacZ transgenic mice induced localized vascular differentiation ERK1/2 activation (37, 53). Thus, the temporal, spatial, and
in the peripheral mesenchyme. In rat fetal distal lung epithelial concentration-dependent activation of FGFR2b by FGF-10
(FDLE) cells cultured at fetal PO2, FGF-10 induced mTORC1 and and inhibition by Spry2 control the length of each branch
amplified HIF-1␣ activity and VEGF secretion without induction of and the location of bifurcations. This mechanism acts as an
ERK1/2. This was accompanied by the formation of a complex
airway branching periodicity regulator or “clock” that de-
between Spry2, the cCBL ubiquitin ligase, and the mTOR repressor,
TSC2, which abolished GTPase activity directed against Rheb, the G fines the semifractal branching pattern of the conducting
protein inducer of mTORC1. Thus, mTORC1 links HIF-1␣-driven airways in the lung (40, 55).
vasculogenesis with the FGF-10/FGFR2b/Spry2 airway branching The mechanism that links vasculogenesis to this process
periodicity regulator. remains unknown. The differentiation of mesenchyme progen-
itor cells into vascular tissue is initiated by the secretion of
lung development; epithelium; mesenchyme; hypoxia; rheb; tuberous vascular endothelial growth factor (VEGF) from the airway
sclerosis complex
epithelium. VEGF expression is regulated by hypoxia-induc-
ible factors (HIFs), a family of transcription factors whose
HYPOXIA IS AN IMPORTANT PHYSIOLOGICAL context for fetal devel- hypoxia-activated ␣-subunit is required for transcriptional ac-
opment, but its role in organ morphogenesis is not well under- tivity. Three HIF-␣ isoforms are expressed in the fetal lung
stood. In the embryonic lung, low PO2 favors airway branching (21, 47). HIF-1␣ occurs in the branching epithelium, and its
and bud formation (18, 19, 54), yet a critical morphogenic role homozygous knockout in mice causes severe vascular defects
and death at embryonic day (E) 10.5 (29). HIF-2␣ occurs in the
epithelium and the interstitium. Its knockout does not affect the
Address for reprint requests and other correspondence: S. C. Land, Centre growth of the conducting vasculature but produces vascular
for Cardiovascular and Lung Biology, Division of Medical Sciences,
MACHS2 Level 4 Mailroom, Ninewells Hospital and Medical School, Univ.
defects during alveolar septation (10). HIF-3␣ is a truncated
of Dundee, Dundee, DD1 9SY Scotland, United Kingdom (e-mail: s.c.land isoform that lacks the COOH-terminal transactivation domain
@dundee.ac.uk). present in HIF-1␣ and 2␣ and so may act as a repressor of these
http://www.ajplung.org 1040-0605/10 Copyright © 2010 the American Physiological Society L455
L456 mTORC1 AND VASCULAR SIGNALING IN FETAL LUNG

transcription factors (6). Although it is present in the develop- action influenced vasculogenesis in the fetal lung in vivo and
ing lung, its function remains unknown. whether it was subject to regulation by airway branching
Of these, HIF-1␣ plays the major role in early pulmonary morphogens in primary cultures of fetal distal lung epithelium
vasculogenesis and so essentially defines the growth of the (FDLE). Our results demonstrate that FGF-10 induces
conducting vasculature (21, 29, 47). HIF-1␣ activity is regu- mTORC1 activity via Spry2 in fetal airway epithelium and that
lated by O2-dependent prolyl-hydroylase domain proteins this amplifies HIF-1␣-evoked vascular signaling beyond the
(PHD1, 2, 3) and the asparagine hydroxylase factor inhibiting level induced by exposure to fetal PO2 alone. We postulate a
HIF (FIH-1) (reviewed in Ref. 28). In normoxia, these en- model whereby mTORC1 coordinates HIF-1␣ vascular signal-
zymes silence HIF signaling by catalyzing the hydroxylation of ing with the FGF-10/FGFR2b/Spry2 airway branching period-
proline residues 402 and 564, and so allow the proteasomal icity clock.
degradation of HIF-1␣ by the von Hippel-Lindau E3 ubiquitin
ligase. In addition, FIH-1 catalyzes the hydroxylation of aspar- MATERIALS AND METHODS
agine 803 and blocks the interaction of the HIF-1␣ transacti-
vation domain with the p300 transcriptional coactivator. In Chemicals and reagents. Recombinant human FGF-10 was from
hypoxia, degradation of PHD proteins by another E3 ubiquitin R&D Systems (Abingdon, UK). Rapamycin, LY-294002, and U0126
were obtained from Merck Biosciences (Nottingham, UK). Antibod-
ligase, SIAH (seven in absentia homolog), allows rapid stabi- ies against Flk1 (A-3), -Flt-1, and VEGF (A-20) were from Santa
lization, nuclear translocation, and transactivation of HIF-1␣ Cruz Biotechnology; Tie2 (ab24859) and Spry2 (ab50317) were from
leading to the expression of hypoxia-regulated proteins (45). Abcam (Cambridge, UK); and S6K1, S6K1 phospho-T389, ERK1/2,

Downloaded from ajplung.physiology.org on September 30, 2010


PHD proteins critically regulate HIF-1–3␣ stability during ERK1/2 phospho-(T202/Y204, T183/Y185), TSC1, and TSC2 were
lung development, as their inhibition augments HIF target gene from Cell Signaling Technology (Danvers, MA). Antibodies against
expression and promotes blood vessel growth and formation of epitope tags and all other reagents (unless stated) were purchased
the blood gas barrier (2, 3, 22). This effect is so profound that from Sigma-Aldrich (Dorset, UK).
vascular signaling and growth can be sustained in explants Plasmids and molecular biology. NH2-terminal flag-tagged pRK7-
cultured in extreme hyperoxia [95% O2, (20)]. These studies Rheb, pRK7-HA-HIF-1␣, and pRK7-HA-HIF-1␣(F99A) and the
fail to explain, however, the temporal and spatial regulation of HIF-1␣ luciferase reporter gene, pHRE3-TK-GL2, were generated as
described (33). pRL incorporating the minimal thymidine kinase (TK)
HIF activity that would be necessary to form a branched promoter was from Promega. Kinase dead (D2357E) mTOR was
vascular and airway tubule network. One possibility is that kindly provided by Dr. Stuart Schreiber (Harvard Univ.), and pXJ40-
oxygen gradients radiating from the fetal pulmonary vascula- FLAG-human(h)Spry2, pXJ40-FLAG-Spry2 (Y55F), and pXJ40-HA-
ture may result in subtle differences in PHD1–3 activity that Rheb were kindly provided by P. Yusoff and G. Guy (Proteos,
coordinate HIF transcription between the two systems. In the Singapore).
fetal lung, however, any such gradient would be shallow [⬃15 Culture of cell lines. The diploid human embryonic lung mesen-
mmHg (20 ␮M)]: difference between the PO2 of the umbilical chymal fibroblast (HELMF) cell line, HEL-12469, was obtained from
vein (⬃30 mmHg) and the PO2 of the amniotic fluid (⬃15 the European Collection of Cell Cultures. An immortalized human
mmHg; Ref. 31) and ⬃10-fold below the Km O2 of the PHD bronchial airway epithelial cell line [16HBE14o-, termed HBE here
enzymes (230 –250 ␮M; Ref. 26). A second, more plausible, (12)] was obtained from Dr. D. Gruenert, California Pacific Medical
Center, San Francisco, CA. Both were routinely cultured in DMEM
scenario suggests that morphogens that are transiently ex- supplemented with Ham’s F-12 nutrient mix (DMEM/F-12) and 8.5%
pressed in the growing tip of the airway bud locally regulate FCS from Invitrogen (Paisley, UK) in a humidified atmosphere
HIF-1␣ and vascular signaling. Thus, the low PO2 of the fetal containing 21% O2 and 5% CO2. For coculture experiments, HELMF
lung maintains a baseline level of HIF-1␣ stability and activity were seeded into Costar six-well culture dishes at a density of 5 ⫻ 105
that is regionally amplified by growth-regulating signals. Such cells/ml⫺1 and allowed to establish for 4 days before each experiment.
events might be considered to confer contextual regulation HBE, seeded separately at a density of 2.5 ⫻ 105 cells·ml⫺1 onto
over HIF proteins, directing their activity subsequent to PHD 2.4-cm diameter Costar Transwell permeable supports, were other-
and FIH induction. wise treated identically. Sixteen hours before the start of each exper-
We have discovered that the mammalian target of rapamycin iment, the cells were placed into serum-free MEM (SFM), and then
(mTOR), a nutrient-sensitive regulator of growth and differen- the HBE and HELMF cells were combined under the experimental
conditions described in each figure by placing Transwell filters into
tiation, amplifies PHD/FIH-induced HIF-1␣ activity by inter- the wells containing the HELMF cells.
acting with a conserved mTOR signaling (TOS) motif within Primary culture of rodent fetal lung cells and organ explants.
the HIF-1␣ PAS-A domain (33). This interaction, which re- FDLE were isolated from gestation day 19 rat fetuses as described
quires the mTOR complex 1 (mTORC1) adaptor protein, previously (5, 32). All procedures conformed with the Animals
raptor, promotes the binding of the p300 transcription initiation (Scientific Procedures) Act, 1986, UK. FDLE were seeded in DMEM/
complex to the HIF-1␣ COOH-terminal transactivation domain F-12 containing 8.5% FCS onto Transwell permeable supports at a
(CTAD) and substantially amplifies HIF-1␣-evoked expres- density of 5 mg of packed cell wt·ml⫺1, whereas fetal mesenchymal
sion of vascular genes such as VEGF. HIF2␣ lacks this TOS lung fibroblasts (FMLF) obtained as a byproduct of the FDLE isola-
motif, and HIF-3␣ lacks the CTAD, thus the positive effect of tion procedure were seeded in the same media at a density of 1 ⫻ 106
this interaction on hypoxic gene expression is specific to cells/well in six-well plastic dishes. Twenty-four hours after isolation,
FDLE and FMLF were placed into DMEM/F-12 supplemented with
HIF-1␣. This complements a growing number of studies that 2% charcoal and ion-exchange resin-stripped serum (stripped serum)
show HIF-1␣ transactivation to be similarly regulated by other and placed at fetal PO2 until use.
kinases (9, 20, 41, 48). Here, we aimed to determine if Lung explants from gestation day 13 or 14 Sprague-Dawley rat
mTORC1 could, in principle, regulate HIF-1␣ and vasculo- fetuses or gestation day 12 Tie2-LacZ mouse fetuses were cultured in
genic signaling in a cell line culture model of the lung epithe- DMEM/F-12 supplemented with 2% stripped serum on Whatman
lial-mesenchymal interface. We then investigated if this inter- polycarbonate filter supports and cultured at fetal PO2. For bead

AJP-Lung Cell Mol Physiol • VOL 299 • OCTOBER 2010 • www.ajplung.org


mTORC1 AND VASCULAR SIGNALING IN FETAL LUNG L457
experiments, heparin-coated acrylic beads (Sigma) were prepared as dome. Images of spheroids were collected at 24-h intervals, and the
described in Supplemental Fig. S1 and grafted into an incision in the compound sprout length was calculated at 48 h. Spheroid images were
peripheral mesenchyme (Supplemental material for this article is digitally separated into quadrants of equal size, and sprout length was
available online at the Journal website). In all cases, digital images measured using Scion Image 4.0.2 software at five evenly spaced
were captured at the start and end of each experiment using a Leica intervals per quadrant. Measurements were pooled from four sphe-
DC 300F digital camera mounted on an MZ FLIII binocular micro- roids for each treatment, and each series of treatments was derived
scope under identical magnification settings (Leica Camera, Milton from independent experiments.
Keynes, UK). Measurement of airway surface complexity and epithelial cell
All cultures were maintained at 37°C in a humidified atmosphere height. Airway surface complexity [ASC; perimeter (mm)/公area
containing 5% CO2. The oxygen content of the atmosphere was (mm2)] was determined from digital images by tracing the dimensions
maintained at the level stated in each figure using a MACS VA500 of the airway perimeter as described (Ref. 46 and Refs. therein).
Microaerophilic Workstation (Don Whitley Scientific, Shipley, West Epithelial height was measured from images of hematoxylin and
Yorkshire, UK). eosin-stained sections generated from paraformaldehyde-fixed and
Transfection and cell lysis. HBE, HELMF, and FDLE were trans- paraffin wax-embedded gestation day 14 explants after 48 h of culture.
fected at 60 – 80% confluency using Lipofectamine 2000 (Invitrogen, Ten measurements were taken at even intervals around each airway
Paisley, UK) according to the manufacturer’s instructions. To mea- cross-section using an Openlab camera-calibrated micrometer. Fre-
sure transfection efficiency, HBE and HELMF cells were transfected quency distribution of epithelial cell height is represented as box plots
with a CMV-driven GFP reporter gene, and the number of GFP and are compared with equivalent measurements from gestation day
fluorescing cells was expressed as a proportion of DRAQ-5-stained 16 fetal rat lungs.
nuclei. Transfection efficiency was routinely ⬎95% for both HELMF Immunohistochemistry. Paraformaldehyde-fixed fetuses of wild-

Downloaded from ajplung.physiology.org on September 30, 2010


and HBE. pRL was used to assess even transfection of FDLE and was type (WT) and homozygous TSC1⫺/⫺ mice (E12.5) were kindly
not found to vary significantly between experiments. To create ly- provided by Prof. Jeremy Cheadle (Univ. of Cardiff, Wales, UK). A
sates, cells or explants were washed twice in PBS and then harvested Histostain Plus broad spectrum DAB kit was used to visualize anti-
in kinase lysis buffer containing Complete Protease Inhibitor Cocktail body-specific staining. Sections incubated in preimmune serum were
tablets (Roche) as described previously (33). treated as controls. Antigen retrieval and quenching of endogenous
Measurement of HIF-1␣ transcriptional activity. HIF-1␣ transcrip- peroxide activity were performed according to the manufacturer’s
tional activity in HBE was measured by luciferase assay as described instructions. Images were obtained using a Zeiss Axiovert microscope
previously (33). For HBE and HELMF cells, medium was replaced and Improvision 5 software.
after transfection with fresh SFM, and cells were allowed to acclima- Rheb-GAP assay. The ratio of GTP-to-GDP-bound Rheb was
tize to the respective experimental PO2 for 16 h. Subsequent treat- determined in FDLE as described by Tee et al. (51). Briefly, FDLE
ments with insulin or rapamycin were then performed for 6 h before transfected with FLAG-Rheb were incubated at fetal or ambient PO2
lysis. Experiments involving FGF-10 and/or Spry2 overexpression for 6 h in medium containing FGF-10 at the indicated concentrations
varied from this format as described in the legend to the relevant and 125 kBq/cm⫺2 [32P]O4 in DMEM containing 2% charcoal-
figures. In all cases, luciferase activity was corrected for protein stripped medium. After lysis, FLAG-Rheb was immunoprecipitated,
content and expressed as change relative to the control group stated in and the proportion of eluted GTP or GDP was determined by TLC on
the figure legend. HIF-1␣ transcriptional activity in FDLE was mea- PEI cellulose using 0.5 MKH2PO4 (pH 3.3) as the mobile phase. The
sured using the same procedure in conjunction with pRL (transfection migration position of radiolabeled spots (Rf; ratio of spot migration:
ratio of 8:1, pHRE3-TK-GL2:pRL) to control for potential variation in mobile phase migration) was visualized using a phosphorimager and
transfection efficiency between preparations. These results were ad- were 0.65 (GTP) and 0.81 (GDP), matching equivalent values calcu-
justed for protein content, normalized against Renilla expression, and lated from Tee et al. (51).
expressed as fold change relative to the stated control group. Statistics. All values are expressed as means ⫾ SE unless otherwise
Quantitative PCR. Detection of pluripotency marker genes in stated in the figure legend. The value of n for HBE/HELMF studies
HELMF (Oct4, Sox2, and NANOG) and FGFR2b and Spry2 in HBE was determined as the number of experimental repeats on cells of
was performed as described in Supplemental Figs. S2 and S4. different passage. For FDLEs and explants, n was determined as the
Immunoprecipitation and Western blotting. Total protein content of number of pregnant dams used to generate independent preparations
cell lysates was determined using the BioRad protein assay. For unless otherwise stated in the figure legend. In all cases, Student’s
immunoprecipitations (IP), 150 ␮g of total protein was precleared for t-test was used to assess significance between paired single treatments,
1 h at 4°C in nonconjugated protein G-sepharose. IP was then and one-way ANOVA with post hoc significance assessed with the
performed for a further 3 h using cCBL or FLAG antibodies conju- Tukey or Dunn test was used for multiple comparisons. A P value ⬍
gated to protein G-sepharose beads as appropriate. Beads were then 0.05 was considered to be statistically significant.
washed twice with buffer A [50 mM HEPES (pH 7.4), 100 mM NaCl,
10 mM MgCl2, 1 mg·ml⫺1 BSA, 1 mM dithiothreitol, and 1% Triton] RESULTS
and buffer B [50 mM HEPES (pH 7.4), 100 mM NaCl, 10 mM MgCl2,
and 0.1% Triton] with Complete Protease Inhibitor Cocktail (Roche) Part 1: Role of the HIF-1␣ TOS motif in vascular signaling.
included in all buffers. IP complexes eluting with the beads were The initial goal of our study was to establish a culture model
dissolved in Laemmli sample buffer and processed by Western blot- that could be used to explore the requirement for the HIF-1␣
ting. Western blots were performed on either cell lysates or IP samples TOS motif in vascular signaling. For ease of culture, transgene
by SDS-PAGE and transferred to nitrocellulose as described before expression, and relevance to the airway environment, we se-
(33). Even transfer onto nitrocellulose was confirmed using Ponceau lected HBE cells as a well-characterized model of the bronchial
S and by reference to actin. epithelium (12) and HELMF cells as a fetal lung-derived
VEGF ELISA. ELISA for human or rat VEGF-A was performed as
described before (29) using the DuoSet ELISA system from R&D
mesenchymal cell line with progenitor cell characteristics (con-
Systems. stitutive expression of the pluripotency markers, Oct4, Sox2,
Sprouting assay. HELMF were induced to form spheroids by and NANOG, under basal conditions. See Supplemental Fig.
nonadherent culture on soft agar. Spheroids of 400- to 600-␮m S2). A bicameral chamber was used to determine if HBE
diameter were placed into growth factor-reduced Matrigel (BD Bio- grown on a permeable support in the upper half of the chamber
sciences), and HBE-conditioned medium was laid above the collagen displayed vasculogenic activity (HIF-1␣ activity, VEGF re-

AJP-Lung Cell Mol Physiol • VOL 299 • OCTOBER 2010 • www.ajplung.org


L458 mTORC1 AND VASCULAR SIGNALING IN FETAL LUNG

lease) when maintained at the PO2 of the fetal lung (23 mmHg). TOS mutant failed to express Flk-1 despite mTORC1 activa-
HELMF cells were maintained beneath the permeable support tion by Rheb. Reporter gene assays conducted in parallel
and probed for the appearance of vascular markers (Flk-1 showed that Rheb augmented HIF-1␣ activity in cells overex-
expression, sprouting) (Fig. 1A). HIF-1␣ activity was deter- pressing WT but not TOS-mutant HA-HIF-1␣.
mined by transfecting each culture with a HIF-driven reporter Sprouting assays were used to measure the potentiation of
gene, and mTOR-dependent modulation of this response was vascular outgrowth from HELMF spheroids induced by con-
determined by treating cells for 6 h with 20 nM insulin in ditioned medium from HBE treated as in Fig. 2A. Sprout
combination with the selective mTOR inhibitor, rapamycin. branch length was significantly elevated from spheroids ex-
LY-294002 (50 ␮M) was used to block the activity of PI3- posed to medium from HBE transfected with both WT-HIF-1␣
kinase, which lies upstream of the mTORC1 complex. Con- and Rheb (Fig. 2C). Exposure to medium from HBE express-
centration of insulin and LY-294002 were based on the effec- ing the HIF-1␣ TOS mutant suppressed sprouting, and, in both
tive doses shown to respectively induce and inhibit a PI3K- cases, was almost completely abolished by rapamycin.
regulated Na⫹ current in H441 human bronchial epithelial cells Part 2: mTOR-dependent regulation of vascular signaling in
(27). Choice of 100 nM rapamycin is based on the recom- the fetal lung. Experiments in Part 1 establish that HBE/
mended concentration for these compounds to achieve selec- HELMF cells can be used to explore vascular signaling in vitro
tive and complete inhibition of mTORC1 (4). Culture of and establish the principle that mTORC1 positively modulates
HELMF at fetal PO2 induced HIF-1␣ activity 2.5-fold above HIF-1␣-mediated vasculogenesis through the NH2-terminal
that at alveolar PO2. Insulin had no observable effect; however, TOS motif. We next explored the role of mTORC1 in fetal

Downloaded from ajplung.physiology.org on September 30, 2010


rapamycin and LY-294002 each produced a significant block- lung development and asked if it could act as a critical point of
ade of this endogenous activity (Fig. 1B). The scope for coordination between vascular signaling with the FGF-10/
HIF-1␣ activation at fetal PO2 (25-fold) was substantially FGFR2b/Spry2 airway branching periodicity regulator.
greater in HBE compared with HELMF and was inhibited by mTORC-1 is active in airway epithelium and participates in
⬃50% by either rapamycin or LY-294002. Insulin augmented airway morphogenesis. The pattern of mTORC1 activity in
this activity by a further fourfold. Rapamycin and LY-294002 gestation day 19 fetal rat lungs in situ was determined from the
suppressed this induced activity to the level observed without distribution of ␣S6K1-T389 phosphorylation. Nonimmune se-
insulin treatment (Fig. 1C). VEGF-A secretion was signifi- rum was used as a control. Intense staining was apparent in the
cantly induced by culture at fetal PO2 and was augmented nascent airway endodermal epithelium and in the mesothelium
further by insulin. Rapamycin and LY-294002 lowered the lining the lung periphery (Fig. 3). ␣S6K1-T389 phosphoryla-
insulin-evoked VEGF-A secretion to the spontaneous level tion was not uniform around the endodermal tubes appearing
induced by culture at fetal PO2 alone (Fig. 1D). more intense within the airway tip region.
The VEGF-A receptor, Flk1, promotes angioblast differen- These results suggest that endogenous factors induce
tiation and is a critical signaling component of epithelial- mTORC1 in the endodermal epithelium of the developing
endothelial cross talk, which is necessary for proper branching airway and that mTORC1 may participate in lung morphogen-
morphogenesis (14). We therefore examined whether HELMF esis. This was confirmed in isolated pseudoglandular stage
maintained in coculture with HBE displayed Flk-1 expression (gestation day 14) rat lung explants where inhibition of endog-
in response to culture at fetal PO2 or insulin (Fig. 1E). Both enous mTORC1 activity with rapamycin (0.1 ␮M) signifi-
conditions were found to induce Flk-1 protein expression in cantly augmented airway epithelial surface folding complexity
HELMF, and this corresponded with increased ␣S6K1 (T389) (ASC, a measure of branching tendency) and promoted epithe-
phosphorylation, a specific substrate of mTORC1. Rapamycin lial flattening (an indicator of epithelial cell maturation) at fetal
and LY-294002 abolished this activation and led to a reduction PO2 (Fig. 4, A–C). The rise in ASC appeared to be accompa-
in Flk-1 expression. HELMF cells subjected to the same nied by a loss of mesenchyme mass; therefore, we examined
treatment but in the absence of HBE did not express Flk-1 the effect of PO2 and mTOR activity on rat fetal mesenchymal
protein. lung fibroblast (FMLF) proliferation. Growth rate was greater
An intact HIF-1␣ TOS motif is required for vasculogenic at fetal compared with alveolar PO2 and was inhibited by either
trophic signaling between HBE and HELMF. mTORC1 can rapamycin or transfection with a kinase dead (D2357E) mutant
amplify HIF-1␣ activity by interacting with a mTOR signaling of mTOR (Fig. 4D) suggesting an additional role for both
(TOS) motif within the HIF-1␣ PAS-A domain. This effect is hypoxia and mTOR in regulating mesenchyme cell density.
independent of the prolyl hydroxylase regulation of HIF-1␣ but FGF-10 induces discrete expression of Tie2 in fetal
promotes the binding of the p300 translation initiation complex mouse lung explants. These experiments demonstrate that
to its COOH-terminal transactivation domain (33). We there- mTORC1 participates in lung morphogenesis, and our ex-
fore determined if mutation of the first phenylalanine residue of periments in HBE/HELMF predict that growth factor activa-
the TOS motif (F99A) interfered with vasculogenic signaling tion of mTORC1 will induce vasculogenic signaling in the fetal
from HBE to HELMF. Overexpression of the upstream acti- lung. We therefore explored whether FGF-10, a central inducer
vating G protein of mTOR, Rheb, together with rapamycin of airway outgrowth, induced vascular differentiation in mu-
treatment were used to further specify observed effects to the rine fetal lung organ explants. Beads soaked in FGF-10 (Sup-
mTOR pathway. Flk-1 expression and mTORC1 activity plemental Fig. S1) were grafted into the peripheral mesen-
(␣S6K1-T389 phosphorylation) was induced in HELMF ex- chyme of gestation day 12 fetal lung explants from mice
posed to conditioned medium from the HBE cells coexpressing bearing a Tie2-driven LacZ reporter gene. Explants were
WT HA-HIF-1␣ and Rheb (Fig. 2, A and B). Although this cultured at fetal PO2 for 48 h and then stained to reveal the
Flk-1 expression was insensitive to rapamycin, HELMF treated pattern of Tie2 gene activation. This expression was absent
with conditioned medium from HBE expressing the HIF-1␣ from the peripheral mesenchyme surrounding the distal airway

AJP-Lung Cell Mol Physiol • VOL 299 • OCTOBER 2010 • www.ajplung.org


mTORC1 AND VASCULAR SIGNALING IN FETAL LUNG L459

Downloaded from ajplung.physiology.org on September 30, 2010

Fig. 1. mTOR amplifies hypoxia-inducible factor-1␣ (HIF-1␣) activity and vasculogenic signaling in HBE-HELMF cocultures. A: the bicameral culture method
for examining epithelial-mesenchymal trophic signaling. Human bronchial epithelial cells (HBE) were cultured on permeable supports suspended above a
monolayer of HEL-12469 human embryonic lung fibroblast cells (HELMF) in serum-free MEM (SFM). Single culture was performed by culturing HBE and
HELMF in SFM separately. B: HIF-1␣-driven luciferase reporter gene expression in HELMF at alveolar (100 mmHg) or fetal (23 mmHg) PO2. Values are
expressed as fold change over untreated cells at alveolar PO2, n ⫽ 4, *P ⬍ 0.05. C: HIF-1␣ activity in HBE; n ⫽ 4, *P ⬍ 0.05. D: VEGF secretion in HBE
is elevated at fetal PO2 and is inhibited by rapamycin and LY-294002. VEGF production was undetectable from HELMF cells (data not shown); *P ⬍ 0.05, n ⫽
4. E: Flk-1 expression at fetal PO2 in HELMF cocultured with HBE (top) or alone (HELMF single culture, bottom). Representative of 3 independent experiments.

AJP-Lung Cell Mol Physiol • VOL 299 • OCTOBER 2010 • www.ajplung.org


L460 mTORC1 AND VASCULAR SIGNALING IN FETAL LUNG

Downloaded from ajplung.physiology.org on September 30, 2010

Fig. 2. The HIF-1␣ F99A TOS mutation suppresses trophic signaling between HBE and HELMF. A, top blots: conditional phenotype of HBE cells used to
generate conditioned medium for HELMF culture. Rapamycin (100 nM) was used to suppress mTOR signaling. Bottom blots: expression of Flt1 and Flk1
[nascent (150 kDa) and glycosylated (200 kDa) forms shown] in HELMF following 18-h culture in HBE-conditioned medium. Blots are representative of 4
experiments. B: top histogram shows the actin-corrected change in Flk1 protein abundance in HELMF cultured in HBE-conditioned SFM. *P ⬍ 0.05, n ⫽ 5.
Bottom histogram shows the activity of HIF-1␣ in the HBE used to generate the conditioned medium. Activity is expressed as fold change over empty vector
(pRK7) control. *P ⬍ 0.05 relative to control group; n ⫽ 5. C: sprouting from HELMF spheroids in Matrigel following 48-h culture in medium obtained from
the HBE in A. Scale bar: 182 ␮m. Box plots at bottom show the data distribution of sprout length. (From bottom: 5th percentile, smallest nonoutlier, lower
quartile, median, top quartile, 95th percentile. Bars indicate the data range.) *P ⬍ 0.05 relative to wild-type transfected control group; **P ⬍ 0.001 relative to
wild-type rheb-transfected group; n ⫽ 4. One-way ANOVA on ranks with Dunn’s post hoc test.

AJP-Lung Cell Mol Physiol • VOL 299 • OCTOBER 2010 • www.ajplung.org


mTORC1 AND VASCULAR SIGNALING IN FETAL LUNG L461

Downloaded from ajplung.physiology.org on September 30, 2010


Fig. 3. mTORC1 activity in the pseudoglandular rat lung in vivo. Gestation day 19 fetal rat lungs were fixed and stained with a phospho-antibody against the
mTORC1 substrate, p70S6K1-T389. A: low-power (⫻40) image of fetal lung in situ. p70S6K-T389 phosphorylation is apparent in the nascent airway epithelium
(brown stain). B: control section treated with nonimmune IgG at same magnification. C: mid-power (⫻200) image showing airway (a), mesenchyme (m), and
endothelial tube (et) structures. Note apparent polarization of mTORC1 activity around the tube (white arrows). D: high-power (⫻400) image showing mTOR
activity extending laterally from the epithelium indicating activity within a possible branching node. Localized activity in cells of the mesenchyme is evident
(black arrows). E: high mTOR activity in the mesothelial lung lining (mth), a site of FGF9 expression (56). White arrow indicates regional activity in an epithelial
lateral branch node. F and G: ⫻200 and ⫻400 images of sections treated with nonimmune IgG.

tips but was evident around the proximal airway and in the (Fig. 6, A–C). TSC1⫺/⫺ mouse lungs also displayed apparently
nexus of airway branches. FGF-10-coated beads grafted into normal airway growth; however, mTORC1 phosphorylation of
the peripheral mesenchyme induced Tie2 gene activation ␣S6K1-T389 was homogenously distributed in the airway
around the bead, which was not observed with PBS-soaked epithelium. Similarly, VEGF expression was widely distrib-
control beads (Fig. 5). Note that the distortion of airway and uted throughout the lung and was accompanied by disaggre-
peripheral mesenchyme tissue around the bead position indi- gation of Tie2-positive vascular bundles (Fig. 6, D–F).
cates intimate contact between bead and tissue. This estab- FGF-10 regulates HIF-1␣ activity via mTORC1 in FDLE.
lishes the principle that a localized FGF-10 signal can promote Having established that endogenous factors sustain mTORC1
vascular differentiation of mesenchyme tissue at fetal PO2. activity in the fetal lung and that localized release of FGF-10
Homozygous knockout of TSC1⫺/⫺ causes aberrant vascular promotes vascular differentiation in mesenchyme, we next
growth in the fetal lung in situ. TSC1⫺/⫺ produces a hyperac- tested the hypothesis that FGF-10 regulates mTORC1-depen-
tive mTOR phenotype that is fatal in mice from gestation day dent HIF-1␣ and vascular signaling using primary cultures of
13.5 (58). To determine if mTOR hyperactivity produces a FDLE at either fetal or alveolar PO2. By culturing FDLE on
hypervascular lung, we performed immunohistochemistry on permeable supports, we aimed to maintain apical/basolateral
fixed sections of gestation day 12.5 from WT and TSC1⫺/⫺ polarization of these cells as shown before (5, 32, 42). FGF-10
mouse fetuses. As with pseudoglandular stage rat lung, produced a dose-dependent, rapamycin-sensitive activation of
mTORC1 phosphorylation of ␣S6K1-T389 in WT fetal lungs HIF-1␣ and VEGF secretion at fetal PO2, which was absent at
was distinct in airway epithelium and was particularly pro- alveolar PO2 (Fig. 7A). mTORC1 activity was dose depen-
nounced at the growing tips of airway buds. VEGF staining dently induced by FGF-10 at fetal PO2 but was not sustained at
was confined to airway epithelium, and Tie2 angiopoietin FGF-10 concentrations ⱖ0.01 ␮g·ml⫺1 at alveolar PO2 (Fig. 7,
receptor expression was confined to discrete regions of mes- B and C). As FGF-10 is reported to induce ERK1/2 signaling
enchymal tissue corresponding to nascent vascular bundles in adult murine airway epithelia (53) and can, in principle,

AJP-Lung Cell Mol Physiol • VOL 299 • OCTOBER 2010 • www.ajplung.org


L462 mTORC1 AND VASCULAR SIGNALING IN FETAL LUNG

Fig. 4. Endogenous mTOR activity is critical


for morphogenesis of fetal rat lung explants.
A: influence of culture PO2 and rapamycin on
airway surface complexity [ASC; perimeter/
公airway area (46)] in lung explants from
gestation day (G) 14 fetal rats. Circles, Con-
trol; triangles, 0.1 ␮M rapamycin. *P ⬍ 0.05
23 mmHg control vs. 100 mmHg control,
n ⫽ 15; †P ⬍ 0.05 rapamycin vs. control at
23 mmHg, n ⫽ 15. B: box plots showing
data distribution of epithelial thickness in

Downloaded from ajplung.physiology.org on September 30, 2010


G14 explants maintained for 48 h in the
presence or absence of rapamycin (0.1 ␮M)
at the indicated PO2. Measurements from
G16 lung in situ are included for comparison
(from bottom: 5th percentile, smallest non-
outlier, lower quartile, median, top quartile,
95th percentile. Bars indicate the data
range). *P ⬍ 0.05, n ⫽ 4. One-way ANOVA
on ranks with Dunn’s post hoc test. C: en-
dogenous activity of mTOR [␣S6K-(P)-
T389] in explants cultured in SFM for 48 h
at fetal or alveolar PO2. Insulin (20 nM) was
used as a positive control, and rapamycin
(0.1 ␮M) was used to block mTOR activity.
Representative of 4 replicates. D: growth
rate of fetal rat lung fibroblasts determined
over 48 h at fetal or alveolar PO2 in the
presence of rapamycin or following overex-
pression of a kinase dead (D2357E) mutant
of mTOR. *P ⬍ 0.05 relative to 23 mmHg
treatment, n ⫽ 6.

activate mTORC1 by repressing TSC1/2, we measured FGF-10 (Supplemental Fig. S4). In contrast to our findings in
ERK1/2 phosphorylation in parallel with mTORC1. Basal FDLE, the induction of mTORC1 by FGF-10 was accompa-
ERK1/2 phosphorylation was low in FDLE, and enhanced blot nied by a transient activation of ERK1/2 and was significantly
images showed it to be further suppressed by FGF-10 (Fig. 7, repressed by U0126 (Supplemental Fig. S5).
B and C). ERK1/2 activity was not required for mTORC1 FGF-10 destabilizes Spry2 and promotes interaction be-
activation by FGF-10 at fetal PO2 as S6K1-T389 phosphoryla- tween cCBL and TSC2 in FDLE. Current models of lung
tion was sustained in the presence of the MEK1 inhibitor, development suggest that FGF-10-FGFR2b signaling is antag-
U0126 (Fig. 7D). Notably, the low endogenous activity of onized by Spry2 and that this mechanism regulates airway
ERK1/2 in FDLE was echoed in perinatal rat lung, which branching periodicity. Given the lack of apparent control of
showed little measurable ERK1/2 activity in gestation days 19 mTORC1 by ERK1/2 in FDLE, we focused on the role of
and 21 homogenates but a significant activation during the Spry2 as a potential regulator of the mTORC1-HIF-1␣ inter-
early postnatal period (Supplemental Fig. S3). action. Spry2 was resolved in FDLE as a double band with
Parallel studies in HBE cells revealed a similar induction of masses of 38.9 (full length) and 35.6 kDa (truncated) using an
HIF-1␣ activity by FGF-10, which was significantly inhibited NH2-terminal epitope antibody (Abcam, ab50317). FGF-10
by rapamycin but declined at the highest concentrations of induced a dose-dependent degradation of full-length Spry2 at

AJP-Lung Cell Mol Physiol • VOL 299 • OCTOBER 2010 • www.ajplung.org


mTORC1 AND VASCULAR SIGNALING IN FETAL LUNG L463

Fig. 5. Vascular differentiation is induced by


FGF-10 in E12 fetal murine lung explants.
Beads coated in FGF-10 or PBS (control)
were grafted onto E12 murine fetal lung
explants from Tie2-LacZ transgenic mice.
Explants were maintained at fetal PO2 for 48
h, fixed, and then stained for LacZ reporter
expression. Grayscale images on left show
bead location and morphogenesis over 48 h;
color images on right show subsequent ex-
pression of Tie2-LacZ reporter gene with
bead excised. Bead position is indicated as: F
(FGF-10), P (PBS). Combined treatment
indicates PBS and FGF-10-coated beads

Downloaded from ajplung.physiology.org on September 30, 2010


grafted onto the same explant. Note lack of
Tie2-LacZ expression in the peripheral mes-
enchyme (PM) shown in bottom image.
Findings shown in these images were repli-
cated across explants isolated from 3 inde-
pendent litters.

both alveolar and fetal PO2 that correlated with the rise in FGF-10 and Sprouty2 regulate HIF-1␣ activity via
mTORC1 activation, HIF-1␣ reporter gene expression, and mTORC1 in HBE. The preceding results are perplexing be-
VEGF production (Figs. 7, A and B, and 8A). The cleavage of cause they suggest that Spry2 cleavage is necessary for
full-length Spry2 occurred by proteolysis as blockade of de mTORC1 activation but that FGF-10 promotes a stable inter-
novo synthesis using cycloheximide led to its clearance within action between Spry2, cCBL, and TSC2. To discriminate
3 h of addition of 0.1 ␮g/ml⫺1 FGF-10. This cleavage was between these outcomes, we reasoned that overexpression of
proteasomal as addition of MG132 abolished the truncated WT Spry2 would quench the cleavage of Spry2 by FGF-10 and
form of Spry2 and stabilized the full-length form in the thus abolish mTORC1 and its induction of HIF-1␣ and VEGF
presence of FGF-10 (Fig. 8, B and C). secretion. Conversely, overexpression of a mutant form of
To explain the association between Spry2 clearance and Spry2, which cannot interact with cCBL [Spry2(Y55F); Ref.
mTORC1 activation, we investigated whether repression of 17], would promote cCBL-mediated clearance of TSC2 and
mTORC1 by TSC1/2 was dependent on cCBL, an E3 ubiquitin thus Rheb-GTP binding, HIF-1␣ activity, and VEGF expres-
ligase that binds to the phosphorylated Y55 residue of Spry2 sion. FDLE transfected with empty vector (EV) alone dis-
(17). TSC2 consistently immunoprecipitated with cCBL, and played Spry2 clearance in response to 0.1 ␮g·ml⫺1 FGF-10,
this association increased significantly on exposure to 0.1 which corresponded with increased Rheb-GTP binding,
␮g/ml⫺1 FGF-10 and the proteasomal inhibitor, MG132. Sur- HIF-1␣ activation, and VEGF production (Fig. 10). Overex-
prisingly, Spry2 interaction with cCBL also increased with pression of WT-Spry2 sustained Spry2 abundance in the pres-
FGF-10 treatment, suggesting that TSC2 preferentially associ- ence of FGF-10, prevented Rheb-GTP binding, and did not
ates with the intact Spry2-cCBL complex (Fig. 9A). When in augment HIF-1␣ activity or VEGF secretion. Cells transfected
complex with TSC1, TSC2 inhibits mTORC1 assembly by with the Spry2(Y55F) mutant, however, showed marked in-
acting as a GTPase activating protein (GAP) towards Rheb; duction of Rheb-GTP binding, HIF-1␣ activation, and VEGF
therefore, we determined if the interaction between TSC2 and secretion. Densitometry revealed that although TSC2 abun-
cCBL/Spry2 corresponded with a decrease in Rheb-GAP ac- dance changed little with FGF-10 treatment in the EV-trans-
tivity and a subsequent rise in GTP-bound Rheb (Fig. 9B). fected cells, overexpression of WT-Spry2 significantly induced
FGF-10 tended to increase guanine nucleotide binding to TSC2 stability, whereas Spry2(Y55F) promoted TSC2 clear-
FLAG-Rheb expressed in FDLE at fetal PO2; however, the ance. These data provide independent confirmation that Spry2
proportion of radioactivity eluting with GTP was significantly regulates mTORC1 at the level of the TSC1/2 complex and that
greater at 0.1 and 1 ␮g·ml⫺1 FGF-10 compared with controls this regulation is mediated through the Y55 binding site for
and is consistent with decreased TSC1/2 GAP activity. cCBL. Moreover, our data illustrate that this effect is sufficient

AJP-Lung Cell Mol Physiol • VOL 299 • OCTOBER 2010 • www.ajplung.org


L464 mTORC1 AND VASCULAR SIGNALING IN FETAL LUNG

Fig. 6. Knockout of the upstream repressor of


mTOR, TSC1, alters vasculogenic signaling in
the murine lung. Wild-type (A–C) and TSC1⫺/⫺
(D–F) embryonic day (E) 12.5 murine fetuses
were sectioned and stained for mTOR activity
[S6K1 (T389) phosphorylation], VEGF, and
Tie2 expression. Arrows in A indicate raised
mTOR activity at sites of airway epithelial out-
growth in wild-type animals. Arrows in C indi-

Downloaded from ajplung.physiology.org on September 30, 2010


cate Tie2 receptor expression corresponding to
nascent vascular bundles. Images are represen-
tative of 4 fetuses from independent litters.
Bar ⫽ 200 ␮m.

to significantly alter vasculogenic signaling through HIF-1␣ cocultured mesenchyme cells (Figs. 1 and 2). In FDLE primary
and VEGF. cultures, we show that stimulating this interaction using FGF-
10, a critical initiator of airway budding, induces VEGF secre-
DISCUSSION tion and promotes localized differentiation of mesenchyme into
vascular tissue in fetal lung explants. Although our study did
mTOR complex 1 is a central regulator of protein synthesis
and cell growth and so acts as a hub where nutrient, metabolic, not demonstrate that endogenous FGF-10 induces mTORC1 in
hormonal, and cytokine signals are integrated towards a de- fetal airway epithelium in situ, we do show mTORC1 activity
fined pattern of growth. In mice, mTOR expression arises at is present in developing airway and that its hyperactivation
E8.5 in the mid-thoracic region, coinciding with the initiation strongly induces VEGF expression in fetal lung. Taken as a
of lung development. Its inhibition by mutation or rapamycin whole, these data establish that mTORC1 is a potent positive
treatment is lethal by E12.5 and is linked to forebrain defects, modulator of epithelial/mesenchymal vascular signaling that
loss of somites, and an inability to turn around the embryonic has the potential to coordinate HIF-1␣ activity with FGF-10, a
axis (25). Ontogenic studies of mTOR function in rat fetuses growth factor that controls airway branch outgrowth.
show its activity is high during the early stages of develop- These data imply that mTORC1 activity is insensitive to the
ment, declining towards term, but rebounds during the early low PO2 of the fetal environment. In tumor cell lines, hypoxic
neonatal period and corresponds with proportionate changes in induction of HIF-1␣ regulated genes such as Redd1, Redd2 (8,
translation efficiency during gestation and birth (42, 43). 11), and Bnip3 (35) repress mTORC1 activity resulting in a
Our study aimed to determine how the activity of HIF-1␣, a suppression of tumor growth. Of these genes, Redd1 is ex-
critical regulator of vasculogenesis, is coordinated with the pressed in fetal distal lung epithelial cells; however, it does not
signals that control airway morphogenesis. We show that in appear to mediate hypoxic repression of mTOR in these cells
both immortalized adult and primary fetal lung epithelia, ex- (42). We used immunohistochemistry to determine the distri-
posure to fetal PO2 induces the activity of HIF-1␣ and VEGF. bution pattern of ␣S6K1-T389 phosphorylation as a readout of
This is markedly amplified by growth-inducing factors such as mTORC1 activity in pseudoglandular stage fetal rat and mice
insulin and FGF-10 and is sensitive to mTORC1 inhibition by lungs in vivo. Our data show that mTORC1 activity was
rapamycin. In HBE cells, we demonstrate that mTORC1- evident in airway endodermal growth buds and in the mesothe-
dependent regulation of HIF-1␣ via the NH2-terminal TOS lium lining the periphery of the lung, a site of FGF-9 expres-
motif is sufficient to induce the appearance of vascular traits in sion (56) (Figs. 3 and 5). Experiments in isolated fetal rat lung

AJP-Lung Cell Mol Physiol • VOL 299 • OCTOBER 2010 • www.ajplung.org


mTORC1 AND VASCULAR SIGNALING IN FETAL LUNG L465

Fig. 7. FGF-10 positively modulates HIF-1␣


activity via mTORC1 in polarized, primary
FDLE cells. A: top graph shows HIF-1␣
activity measured from FDLE transfected
with pHRE3-TK-GL2 and pRL (8:1) and
cultured at fetal (23 mmHg; closed symbols)
or alveolar (100 mmHg; open symbols) PO2
for 6 h in the presence of increasing concen-
trations of FGF-10. Rapamycin (0.1 ␮M; tri-
angles) was added to a parallel set of cells to
determine mTORC1-sensitive HIF-1␣ activ-
ity. Values are normalized to 0 ␮g·ml⫺1
FGF-10 treatment at fetal PO2. Bottom graph
shows the level of VEGF secreted into the
medium beneath the permeable support from
the same cells. *P ⬍ 0.05 relative to control

Downloaded from ajplung.physiology.org on September 30, 2010


at fetal PO2; †P ⬍ 0.05 relative to paired
rapamycin treatment, n ⫽ 5. B: representa-
tive blots showing mTORC1 [␣S6K-(p)-
T389 phosphorylation] and ERK1/2 activa-
tion (T202/Y204 and T185/Y187 phosphor-
ylation) and Spry2 protein abundance
measured in samples from A. *Contrast is
enhanced separately from total ERK blot to
show decline in ERK1/2 phosphorylation
with FGF-10. C: densitometry of pooled
experimental blots represented in B. Open/
closed symbols are as described in A; trian-
gles represent ERK2 values. *P ⬍ 0.05 rel-
ative to control, n ⫽ 4. D: the MEK1 inhib-
itor U0126 (10 ␮M) does not prevent
mTORC1 activation (S6K1-T389 phosphor-
ylation) by 0.1 ␮g/ml⫺1 FGF-10 in FDLE at
fetal PO2. Rapamycin (0.1 ␮M) was added to
show inhibition of mTORC1. Histogram
(right) shows pooled densitometry data for
mTORC1 activity and ERK1/2 phosphory-
lation. *P ⬍ 0.05 relative to control; †not
significantly different from FGF-10 treat-
ment. ND, not detectable; n ⫽ 5.

explants, cultured at the PO2 of the fetal lung, demonstrate that cin also abolishes transforming growth factor-␤-evoked in-
this activity was necessary for balanced airway growth because creases in cell size and migration/invasion during epithelial-
exposure to rapamcyin produced significant morphological to-mesenchymal transition in murine mammary epithelial cells
changes including accelerated airway branching (raised ASC in (30). In the developing lung, mTOR-directed translation is
Fig. 4), epithelial flattening, and mesenchymal thinning. Over- active during early and mid-stages of development and de-
all, these data show mTOR signaling participates in tissue- clines in late gestation, but peaks again on postnatal day 1 (42,
specific growth events despite the relative hypoxia of the fetal 43). This late-gestation decline in translation efficiency is
environment. accompanied by a switch to mTOR-independent transcript
This raises the question of whether mTOR is involved in expression involving genes linked to the development of im-
controlling cell pluripotency/differentiation in the developing munity, antioxidant defense, and the acquisition of a terminally
lung. In cancer, mTOR hyperactivation sustains cell prolifer- differentiated epithelial phenotype [e.g., SCGB3A1, (44)] and
ation and represses differentiation by positive induction of the may be necessary to prepare the lung for air breathing and
Notch signaling pathway (36). Elsewhere, the role of mTOR in exposure to airborne pathogens.
cell differentiation appears to be quite specific to cell type. For mTOR is recognized as an important inducer of vascular
example, rapamycin promotes terminal differentiation of vas- growth by modulating angiogenic signaling as well as endo-
cular smooth muscle cells (39) but maintains pluripotency of thelial cell proliferation and differentiation (reviewed in Ref.
bone marrow-derived mesenchymal stem cells (24). Rapamy- 34). To determine if mTOR signaling was necessary for vas-

AJP-Lung Cell Mol Physiol • VOL 299 • OCTOBER 2010 • www.ajplung.org


L466 mTORC1 AND VASCULAR SIGNALING IN FETAL LUNG

Fig. 7 —Conitnued

Downloaded from ajplung.physiology.org on September 30, 2010


culogenesis in the fetal lung in vivo, we examined how tar- proceeds up to at least E12.5 in TSC1⫺/⫺ fetal lungs and that
geted knockout of the upstream repressor of mTOR, TSC1, gross airway morphology (epithelial thickness, airway patency,
altered the distribution and expression of VEGF and the an- mesenchyme thickness) differs little from age-matched WT
giopoietin receptor, Tie2. Mice lacking the TSC1 allele display lungs (Fig. 6). However, mTOR activity appeared to be uni-
constitutively active mTOR activity, develop exencephaly and form in the airway epithelium of TSC1⫺/⫺ lungs and did not
abnormal vacuolarization of myocardial cells in utero, and fail show the distinct localization at growth tips observed in WT
to develop beyond E13 (58). We found that airway growth lungs. TSC1⫺/⫺ lungs also showed excessive staining for

Fig. 8. FGF-10 destabilizes Spry2 in FDLE.


A: abundance of Spry2 declines with in-
creasing FGF-10 concentration at fetal
(closed circles) and alveolar (open circles)
PO2. Densitometry performed on full-length
Spry2 in ratio to actin. *P ⬍ 0.05 relative to
control, n ⫽ 4. B: Spry2 becomes unstable in
the presence of FGF-10. FDLE were treated
with 100 ␮M cycloheximide (CHX) and 0.1
␮g·ml⫺1 FGF-10 as indicated at fetal PO2.
Representative blot is accompanied by graph
showing full-length Spry2 abundance mea-
sured in ratio to actin. Circles, control; tri-
angles, 0.1 ␮g·ml⫺1 FGF-10. *P ⬍ 0.05,
n ⫽ 4 relative to time 0. Error bars may be
within symbols. C: Spry2 cleavage is protea-
somal. FDLE were treated with 10 ␮M
MG132 and 0.1 ␮g·ml⫺1 FGF-10 at fetal
PO2 for 3 h as indicated. Proteins were sep-
arated by 10% SDS-PAGE to resolve both
full-length and cleaved Spry2 product. Blot
is representative of n ⫽ 3.

AJP-Lung Cell Mol Physiol • VOL 299 • OCTOBER 2010 • www.ajplung.org


mTORC1 AND VASCULAR SIGNALING IN FETAL LUNG L467

Fig. 9. Spry2 associates with cCBL and


TSC2. A: immunoprecipitation (IP) of cCBL
from FDLE at fetal PO2 treated with FGF-10
(0.1 ␮g·ml⫺1) or MG132 (10 ␮M) as indi-
cated. Immunoblots (IB) were performed to
detect TSC2, cCBL, and Spry2. Histograms
on right show ratio of TSC2 or Spry2 to
cCBL. *P ⬍ 0.05 relative to control, n ⫽ 5.
B: effect of FGF-10 on TSC1/2 GAP activity
towards the Rheb guanine nucleotide bind-
ing protein in FDLE at fetal or ambient PO2.
TLC phosphorimage shows resolution of
32
P-GTP and -GDP immunoprecipitating
with FLAG-Rheb in cells treated with FGF-

Downloaded from ajplung.physiology.org on September 30, 2010


10. Immunoblot beneath shows even recov-
ery of FLAG-Rheb by IP. Histogram shows
ratio of GTP:GDP as a percentage of total
radioactivity eluting with Rheb. The super-
imposed line graph shows fold change in
total [32P]-labeled guanine nucleotide phos-
phates eluting with Rheb, *P ⬍ 0.05 relative
to control at fetal PO2, n ⫽ 4.

VEGF and an absence of discretely organized Tie2-positive that mTOR activation amplifies HIF-1␣ transcriptional activity
vascular bundles around the airways. VEGF is a HIF-regulated and VEGF expression. The reason for the decline in coordi-
target gene, and its raised expression in TSC1 knockout mouse nated vasculogenesis, which follows, may arise from dysfunc-
lungs is consistent with our observations in Figs. 1, 2, and 7 tional regulation of Tie2 signaling caused by hyperexpression

Fig. 10. Overexpression of Spry2 in FDLE


abolishes FGF-10-evoked Rheb-GTP bind-
ing and is relieved by mutation of the Spry2
(Y55) cCBL docking site. A: TSC1/2 GAP
activity (ratio of GTP:GDP bound Rheb),
HIF-1␣ activity, and VEGF secretion in
FDLE transfected with empty vector, wild-
type, or Y55F Spry2. For GAP assays, the
transfection mix included FLAG-Rheb (ratio
of 1:4 Rheb to test vector), whereas for
HIF-1␣ activity assays and VEGF secretion,
the transfection mix included pHRE3-
TK-GL2 (1:4). All experiments were per-
formed at fetal PO2. *P ⬍ 0.05 relative to EV
control; n ⫽ 4 (Rheb GAP assay), n ⫽ 6
(HIF-1␣ luciferase and VEGF secretion).
B: representative blot showing TSC2, Spry2,
and actin expression levels. Note that the
Spry2 blot shows bands for the endogenous
full-length and cleaved forms of Spry2 in
EV lanes, which are superimposed by over-
expressed Spry2 in other lanes. Histogram
shows TSC2 abundance corrected for actin.
*P ⬍ 0.05 relative to respective EV and
Spry2 (Y55F) treatment, n ⫽ 6 independent
transfections.

AJP-Lung Cell Mol Physiol • VOL 299 • OCTOBER 2010 • www.ajplung.org


L468 mTORC1 AND VASCULAR SIGNALING IN FETAL LUNG

of VEGF. Tie2 and its ligands, angiopoietin 1 and 2 (Ang1 and tion would arise by ERK1/2-dependent phosphorylation and
2), control the proliferation and maturation of nascent blood subsequent inactivation of the TSC1/2 complex. We were
vessels. Proteolytic cleavage of Tie2 results in the shedding of therefore surprised to discover that FGF-10 actually suppressed
a truncated soluble form of this receptor, sTie2, which is ERK1/2 activity in FDLE cultured at fetal PO2 and that signal-
capable of binding Ang1 and 2 and so competitively inhibits ing through the mTORC1 pathway was unaltered by inhibition
the uncleaved membrane-bound Tie2 receptor. VEGF acceler- of the upstream kinase to ERK1/2, MEK1. Induction of
ates the cleavage and shedding of sTie2 and so its elevated ERK1/2 by FGF-10 was originally reported in an immortalized
expression decreases Tie-2-dependent blood vessel wall as- adult murine lung epithelial cell line (MLE15) that was cul-
sembly but promotes the proliferation of vascular endothelial tured, unpolarized, at ambient PO2 (53). We repeated these
cells (16). experiments using unpolarized, immortalized adult lung-de-
The final part of this study determined how mTORC1- rived HBE cells and observed transient induction of ERK1/2
dependent regulation of HIF-1␣ and VEGF secretion is linked by FGF-10, which was not sustained with time or increased
to the FGF-10/FGFR2b/Spry2 pathway, a central regulator of FGF-10 dosage (Supplemental Figs. S4 and S5). Although the
airway outgrowth and branching. FGF-10-coated beads pro- magnitude of ERK1/2 signaling was greater at alveolar com-
duced a localized increase in Tie2-promoted reporter gene pared with fetal PO2, mTORC1 induction by FGF-10 was,
expression in fetal murine lung explants. More detailed exam- nevertheless, ERK1/2 dependent, leading us to suggest that
ination of the link between mTORC1 and FGF-10 in FDLE ERK1/2 signaling induces, but does not sustain, mTORC1
(Figs. 7–10) and HBE (Supplemental Figs. S4 and S5) showed activity in HBE. These differences in FGF-10 signaling re-

Downloaded from ajplung.physiology.org on September 30, 2010


a clear induction of HIF-1␣ activity and VEGF secretion with sponses between FDLE and MLE15/HBE probably reflect
increasing FGF-10 dosage, which was suppressed to the basal variation in tyrosine kinase receptor expression linked to de-
hypoxic level by rapamycin. Given that FGF-10 has been velopment, culture, and oxygen exposure; however, our finding
reported to induce ERK1/2 activation and proliferation of that overall ERK1/2 activity is low in FDLE as well as in the
airway epithelium (53), we anticipated that mTORC1 activa- late-term fetal lung, but is augmented by oxygen in HBE and

Fig. 11. Model of integrated airway and vasculogenesis in the developing lung. Model is represented next to a late embryonic stage (E12) rat lung showing
formation of airway branches extending into the mesenchyme tissue surrounded by blood-filled vascular structures proximal to the airway tip. Foregut (Fg) is
shown displaced to left of lung. Zone A: FGF-10 expressed in the mesenchyme ahead of nascent airway buds induces Spry2 cleavage by an unknown ubiquitin
ligase (possibly SIAH2 or cCBL) in the progenitor epithelial cells of the airway tip. This cleavage event, although necessary for mTORC1 activation, is
accompanied by the formation of a complex between Spry2, cCBL, and TSC2 (Figs. 7 and 8), which dislocates the TSC1/2 complex through the proteolytic
cleavage of TSC2 (Fig. 10). This disrupts TSC1/2 GTPase activating protein (GAP) activity allowing GTP interaction with Rheb and activation of mTORC1.
Zones B and C: trophic signaling, involving VEGF release by airway epithelium into the mesenchyme and subsequent vasculogenesis, proceeds proximal to the
airway tip by binding of mTORC1 to the HIF-1␣ TOS motif and subsequent interaction with the p300 transcriptional initiation complex. This interaction locally
amplifies the transcriptional activity of HIF-1␣ causing increased release of VEGF and the appearance of vascular structures behind the growing tip of the airway.
Our model acknowledges that there are 2 oxygen-dependent points of control that are critical for proper lung morphogenesis. These are: 1) the proliferation of
mesenchyme tissue that requires low-oxygen tension and mTOR activity for sustained growth, and 2) hypoxic priming of HIF-1␣ transcriptional activity, which
can be positively modulated by locally expressed growth factors. The ERK1/2 activation by FGF-10 reported by Tefft et al. (53) and observed by us in HBE
(Supplementary Figs. S4 and S5) is represented by a dotted line. Our study shows that the magnitude of this response is also oxygen sensitive as it is suppressed
at fetal compared with alveolar PO2, leading us to speculate a secondary role for oxygen in regulating ERK1/2 signaling in the fetal lung. This phenomenon could
be linked to the conservation of progenitor cell populations during lung morphogenesis (50).

AJP-Lung Cell Mol Physiol • VOL 299 • OCTOBER 2010 • www.ajplung.org


mTORC1 AND VASCULAR SIGNALING IN FETAL LUNG L469
increases in postnatal lung with the onset of breathing (Sup- We have incorporated these results into a model that links
plementary Data Figs. S3–S5), suggests that low oxygen ten- vascular signaling with the FGF-10/FGFR2b/Spry2 signaling
sions could have a role to play in constraining ERK1/2 signal- pathway in the developing fetal lung (Fig. 11). Our model
ing during fetal development. Numerous studies in embryonic differs from the classic view of branched airway morphogen-
stem cells have established that low oxygen tensions conserve esis in that we exclude a significant role for ERK1/2 and
pluripotency, and it is now shown that temporal activation of highlight, for the first time, the central importance of mTORC1
ERK1/2 induces differentiation to a defined phenotype (50). It as a coordinator of airway and vascular morphogenesis. This
is possible, then, that the constrained ERK1/2 activity we model also highlights the importance of the low oxygen tension
observed in fetal tissue may be critical to conserve populations of the fetal environment for priming key developmental events,
of progenitor cells in the fetal lung. particularly, establishing a basal activation of HIF-1␣ and the
Our studies thus highlight mTORC1, and not ERK1/2, as an maintenance of a mesenchymal cell population. By implica-
important downstream target of FGF-10 in FDLE. Given that tion, therapies that target the HIF signaling system to treat
Spry2 antagonism of the FGF-10 receptor, FGFR2b, is be- developmental vascular diseases such as bronchopulmonary
lieved to be an important regulator of airway branching peri- dysplasia need to take into account the role of the mTOR
odicity, it follows that mTORC1 activity and its regulation of signaling pathway as a major determinant of temporal and
HIF-1␣ should be closely governed by Spry2. We found the spatial vascular signaling.
activation of mTORC1 by FGF-10 was accompanied by three
key events: 1) proteasomal cleavage of Spry2 to a truncated ACKNOWLEDGMENTS

Downloaded from ajplung.physiology.org on September 30, 2010


form, 2) formation of a stable complex between Spry2, cCBL, We thank Claire Cockburn for assistance with qPCR determinations of
and TSC2, and 3) decreased TSC1/2 GAP activity towards pluripotency markers in HELMF, Prof. Jerry Cheadle and Dr. Cleo Bonett
Rheb as indicated by increased Rheb-GTP binding. Our Spry2 (Cardiff Univ.) for provision of fixed TSC1⫺/⫺ fetuses, and Drs. Graeme Guy
overexpression experiments demonstrated that enforced main- and Permeen Yusoff for provision of Spry2 vectors and related advice.
tenance of Spry2 levels during FGF-10 treatment sustained
TSC2 expression levels and suppressed Rheb-GTP binding, GRANTS
whereas overexpression of the Spry2 Y55F mutant, which This work was supported by grants to S. C. Land from The Wellcome Trust
lacks a functional cCBL binding domain (17), caused TSC2 (088032/z/08/z), TENOVUS (Scotland), and the Anonymous Trust. A. R. Tee
is supported by the Association of International Cancer Research through a
clearance, Rheb-GTP binding, and induced HIF-1␣ activity Career Development Fellowship (06-914/915).
and VEGF secretion. Together, these results suggest that Spry2
is a potent regulator of mTORC1 activation and that this DISCLOSURES
regulation depends on an interaction between cCBL and TSC2.
No conflicts of interest, financial or otherwise, are declared by the author(s).
Spry2 cleavage in hand with the formation of an apparently
stable Spry2-cCBL-TSC2 complex accords with several pro-
REFERENCES
posed physiological roles for Spry2 (reviewed in Ref. 23). One
scenario suggests that Spry2 sequesters cCBL activity. Cleav- 1. Acarregui MJ, Penisten ST, Goss KL, Ramirez K, Snyder JM. Vas-
age of Spry2 by another ubiquitin ligase (SIAH2), or cCBL cular endothelial growth factor gene expression in human fetal lung in
vitro. Am J Respir Cell Mol Biol 20: 14 –23, 1999.
itself, targets downstream elements of RTK signaling pathways 2. Asikainen TM, Chang LY, Coalson JJ, Schneider BK, Waleh NS,
for proteasomal clearance. Two variations on this theme sug- Ikegami M, Shannon JM, Winter VT, Grubb P, Clyman RI, Yoder
gest that Spry2 performs either a targeting or adaptor-protein BA, Crapo JD, White CW. Improved lung growth and function through
function that is necessary for cCBL to interact with its substrate hypoxia-inducible factor in primate chronic lung disease of prematurity.
proteins. Our finding that Spry2 was consistently cleaved by FASEB J 20: 1698 –1700, 2006.
3. Asikainen TM, Schneider BK, Waleh NS, Clyman RI, Ho WB, Flippin
FGF-10 treatment and that relief of the Spry2-cCBL interaction LA, Gunzler V, White CW. Activation of hypoxia-inducible factors in
by overexpression of the Spry2-Y55F mutant led to TSC2 hyperoxia through prolyl 4-hydroxylase blockade in cells and explants of
clearance and Rheb-GTP binding suggests that a cCBL seques- primate lung. Proc Natl Acad Sci USA 102: 10212–10217, 2005.
tering role for Spry2 is possible in unstimulated cells. How- 4. Bain J, Plater L, Elliott M, Shpiro N, Hastie CJ, McLauchlan H,
Klevernic I, Arthur JS, Alessi DR, Cohen P. The selectivity of protein
ever, immunoprecipitations performed against endogenous kinase inhibitors: a further update. Biochem J 408: 297–315, 2007.
cCBL showed that FGF-10 treatment does not lead to the 5. Baines DL, Ramminger SJ, Collett A, Haddad JJ, Best OG, Land SC,
complete cleavage of Spry2 with some remaining associated Olver RE, Wilson SM. Oxygen-evoked Na⫹ transport in rat fetal distal
with cCBL when in complex with TSC2, thus a targeting or lung epithelial cells. J Physiol 532: 105–113, 2001.
adaptor-protein role is also possible. Spry2 binds protein phos- 6. Bardos JI, Ashcroft M. Negative and positive regulation of HIF-1: a
complex network. Biochim Biophys Acta 1755: 107–120, 2005.
phatases (SHP2; PP2A) that can mediate its own dephosphor- 7. Bellusci Grindley SJ, Emoto H, Itoh N, Hogan BL. Fibroblast growth
ylation as well as that of other phosphoproteins. As TSC2 is a factor 10 (FGF10) and branching morphogenesis in the embryonic mouse
substrate of both Shp2 and PP2A (38, 59) and Spry2 has been lung. Development 124: 4867–4878, 1997.
shown to associate with Shp2 in fetal airway epithelium (52), 8. Brugarolas J, Lei K, Hurley RL, Manning BD, Reiling JH, Hafen E,
Witters LA, Ellisen LW, Kaelin WG Jr. Regulation of mTOR function
it is conceivable that the decline in Spry2 abundance with in response to hypoxia by REDD1 and the TSC1/TSC2 tumor suppressor
FGF-10 disrupts phosphatase activity directed towards TSC2 complex. Genes Dev 18: 2893–2904, 2004.
thereby promoting Rheb-GTP binding and mTORC1 activa- 9. Cho H, Ahn DR, Park H, Yang EG. Modulation of p300 binding by
tion (38, 59). Finally, we acknowledge that Spry2 can regulate posttranslational modifications of the C-terminal activation domain of
HIF-1␣ independently of RTKs by targeting prolyl hydroxy- hypoxia-inducible factor-1␣. FEBS Lett 581: 1542–1548, 2007.
10. Compernolle V, Brusselmans K, Acker T, Hoet P, Tjwa M, Beck H,
lases 1 and 3 for degradation via the Siah2 ubiquitin ligase Plaisance S, Dor Y, Keshet E, Lupu F, Nemery B, Dewerchin M, Van
(45); however, this mechanism was not investigated further Veldhoven P, Plate K, Moons L, Collen D, Carmeliet P. Loss of HIF-2␣
here. and inhibition of VEGF impair fetal lung maturation, whereas treatment

AJP-Lung Cell Mol Physiol • VOL 299 • OCTOBER 2010 • www.ajplung.org


L470 mTORC1 AND VASCULAR SIGNALING IN FETAL LUNG

with VEGF prevents fatal respiratory distress in premature mice. Nat Med 34. Li DF, Huang MC. All roads lead to mTOR: integrating inflammation
8: 702–710, 2002. and tumor angiogenesis. Cell Cycle 6: 3011–3014, 2007.
11. Corradetti MN, Inoki K, Guan KL. The stress-inducted proteins 35. Li Y, Wang Y, Kim E, Beemiller P, Wang CY, Swanson J, You M,
RTP801 and RTP801L are negative regulators of the mammalian target of Guan KL. Bnip3 mediates the hypoxia-induced inhibition on mammalian
rapamycin pathway. J Biol Chem 280: 9769 –9772, 2005. target of rapamycin by interacting with Rheb. J Biol Chem 282: 35803–
12. Cozens AL, Yezzi MJ, Kunzelmann K, Ohrui T, Chin L, Eng K, 35813, 2007.
Finkbeiner WE, Widdicombe JH, Gruenert DC. CFTR expression and 36. Ma J, Meng Y, Kwiatkowski DJ, Chen X, Peng H, Sun Q, Zha X,
chloride secretion in polarized immortal human bronchial epithelial cells. Wang F, Wang Y, Jing Y, Zhang S, Chen R, Wang L, Wu E, Cai G,
Am J Respir Cell Mol Biol 10: 38 –47, 1994. Malinowska-Kolodziej I, Liao Q, Liu Y, Zhao Y, Sun Q, Xu K, Dai J,
13. Davies DM, Johnson SR, Tattersfield AE, Kingswood JC, Cox JA, Han J, Wu L, Zhao RC, Shen H, Zhang H. Mammalian target of
McCartney DL, Doyle T, Elmslie F, Saggar A, de Vries PJ, Sampson rapamycin regulates murine and human cell differentiation through
JR. Sirolimus therapy in tuberous sclerosis or sporadic lymphangio- STAT3/p63/Jagged/Notch cascade. J Clin Invest 120: 103–114, 2010.
leiomyomatosis. N Engl J Med 10: 200 –203, 2008. 37. Mailleux AA, Tefft D, Ndiaye D, Itoh N, Thiery JP, Warburton D,
14. Del Moral PM, Sala FG, Tefft D, Shi W, Keshet E, Bellusci S, Bellusci S. Evidence that SPROUTY2 functions as an inhibitor of mouse
Warburton D. VEGF-A signaling through Flk-1 is a critical facilitator of embryonic lung growth and morphogenesis. Mech Dev 102: 81–94, 2001.
early embryonic lung epithelial to endothelial crosstalk and branching 38. Marin TM, Clemente CF, Santos AM, Picardi PK, Pascoal VD,
morphogenesis. Dev Biol 290: 177–188, 2006. Lopes-Cendes I, Saad MJ, Franchini KG. Shp2 negatively regulates
15. Dunlop EA, Tee AR. Mammalian target of rapamycin complex 1: growth in cardiomyocytes by controlling focal adhesion kinase/Src and
signalling inputs, substrates and feedback mechanisms. Cell Signal 21: mTOR pathways. Circ Res 103: 813–824, 2008.
827–835, 2009. 39. Martin KA, Merenick BL, Ding M, Fetalvero KM, Rzucidlo EM,
16. Findley CM, Cudmore MJ, Ahmed A, Kontos CD. VEGF induces Tie2 Kozul CD, Brown DJ, Chiu HY, Shyu M, Drapeau BL, Wagner RJ,
Powell RJ. Rapamycin promotes vascular smooth muscle cell differenti-

Downloaded from ajplung.physiology.org on September 30, 2010


shedding via a phosphoinositide 3-kinase/Akt dependent pathway to mod-
ulate Tie2 signaling. Arterioscler Thromb Vasc Biol 27: 2619 –2626, 2007. ation through insulin receptor substrate-1/phosphatidylinositol 3-kinase/
17. Fong CW, Leong HF, Wong ES, Lim J, Yusoff P, Guy GR. Tyrosine Akt2 feedback signaling. J Biol Chem 282: 36112–36120, 2007.
phosphorylation of Sprouty2 enhances its interaction with c-Cbl and is 40. Metzger RJ, Klein OD, Martin GR, Krasnow MA. The branching
crucial for its function. J Biol Chem 278: 33456 –33464, 2003. programme of mouse lung development. Nature 453: 745–750, 2008.
18. Gebb SA, Fox K, Vaughn J, McKean D, Jones PL. Fetal oxygen tension 41. Mylonis I, Chachami G, Samiotaki M, Panayotou G, Paraskeva E,
promotes tenascin-C-dependent lung branching morphogenesis. Dev Dyn Kalousi A, Georgatsou E, Bonanou S, Simos G. Identification of MAPK
234: 1–10, 2005. phosphorylation sites and their role in the localization and activity of
19. Gebb SA, Jones PL. Hypoxia and lung branching morphogenesis. Adv hypoxia-inducible factor-1␣. J Biol Chem 281: 33095–33106, 2006.
Exp Med Biol 543: 117–125, 2003. 42. Otulakowski G, Duan W, Gandhi S, O’Brodovich H. Steroid and
20. Gradin K, Takasaki C, Fujii-Kuriyama Y, Sogawa K. The transcrip- oxygen effects on eIF4F complex, mTOR, and ENaC translation in fetal
lung epithelia. Am J Respir Cell Mol Biol 37: 457–466, 2007.
tional activation function of the HIF-like factor requires phosphorylation
43. Otulakowski G, Duan W, O’Brodovich H. Global and gene-specific
at a conserved threonine. J Biol Chem 277: 23508 –23514, 2002.
translational regulation in rat lung development. Am J Respir Cell Mol
21. Groenman F, Rutter M, Caniggia I, Tibboel D, Post M. Hypoxia-
Biol 40: 555–567, 2009.
inducible factors in the first trimester human lung. J Histochem Cytochem
44. Porter D, Lahti-Domenici J, Torres-Arzayus M, Chin L, Polyak K.
55: 355–363, 2007.
Expression of high in normal-1 (HIN-1) and uteroglobin related protein-1
22. Groenman FA, Rutter M, Wang J, Caniggia I, Tibboel D, Post M.
(UGRP-1) in adult and developing tissues. Mech Dev 114: 201–204, 2002.
Effect of chemical stabilizers of hypoxia-inducible factors on early lung
45. Qi J, Nakayama K, Gaitonde S, Goydos JS, Krajewski S, Eroshkin A,
development. Am J Physiol Lung Cell Mol Physiol 293: L557–L567, 2007.
Bar-Sagi D, Bowtell D, Ronai Z. The ubiquitin ligase Siah2 regulates
23. Guy GR, Jackson RA, Yusoff P, Chow SY. Sprouty proteins: modified tumorigenesis and metastasis by HIF-dependent and -independent path-
modulators, matchmakers or missing links? J Endocrinol 203: 191–202, ways. Proc Natl Acad Sci USA 105: 16713–16718, 2008.
2009. 46. Rae C, Cherry JI, Land FM, Land SC. Endotoxin-induced nitric oxide
24. Hegner B, Weber M, Dragun D, Schulze-Lohoff E. Differential regu- production rescues airway growth and maturation in atrophic fetal rat lung
lation of smooth muscle markers in human bone marrow-derived mesen- explants. Biochem Biophys Res Commun 349: 416 –425, 2006.
chymal stem cells. J Hypertens 23: 1191–1202, 2005. 47. Rajatapiti P, van der Horst IW, de Rooij JD, Tran MG, Maxwell PH,
25. Hentges KE, Sirry B, Gingeras AC, Sarbassov D, Sonenberg N, Tibboel D, Rottier R, de Krijger RR. Expression of hypoxia-inducible
Sabatini D, Peterson AS. FRAP/mTOR is required for proliferation and factors in normal human lung development. Pediatr Dev Pathol 11:
patterning during embryonic development in the mouse. Proc Natl Acad 193–199, 2008.
Sci USA 98: 13796 –13801, 2001. 48. Sang N, Stiehl DP, Bohensky J, Leshchinsky I, Srinivas V, Caro J.
26. Hirsilä M, Koivunen P, Günzler V, Kivirikko KI, Myllyharju J. MAPK signalling upregulates the activity of hypoxia-inducible factors by
Characterization of the human prolyl 4-hydroxylases that modify the its effects on p300. J Biol Chem 278: 14013–14019, 2003.
hypoxia-inducible factor. J Biol Chem 278: 30772–30780, 2003. 49. Shinkai T, Shinkai M, Pirker ME, Montedonico S, Puri P. The role of
27. Inglis SK, Gallacher M, Brown SG, McTavish N, Getty J, Husband oxygen tension in the regulation of embryonic lung development. J
EM, Murray JT, Wilson SM. SGK1 activity in Na⫹ absorbing airway Pediatr Surg 40: 32–35, 2005.
epithelial cells monitored by assaying NDRG1-Thr346/356/366 phosphor- 50. Stavridis MP, Collins BJ, Storey KG. Retinoic acid orchestrates fibro-
ylation. Pflügers Arch 457: 1287–1301, 2009. blast growth factor signalling to drive embryonic stem cell differentiation.
28. Kenneth NS, Rocha S. Regulation of gene expression by hypoxia. Development 137: 881–890, 2010.
Biochem J 414: 19 –29, 2008. 51. Tee AR, Blenis J, Proud CG. Analysis of mTOR signaling by the small
29. Kotch LE, Iyer NV, Laughner E, Semenza GL. Defective vasculariza- G-proteins, Rheb and RhebL1. FEBS Lett 579: 4763–4768, 2005.
tion of HIF-1-null embryos is not associated with VEGF deficiency but 52. Tefft D, De Langhe SP, Del Moral PM, Sala F, Shi W, Bellusci S,
with mesenchymal cell death. Dev Biol 209: 254 –267, 1999. Warburton D. A novel function for the protein tyrosine phosphatase Shp2
30. Lamouille S, Derynck R. Cell size and invasion in TGF-beta-induced during lung branching morphogenesis. Dev Biol 282: 422–431, 2005.
epithelial to mesenchymal transition is regulated by activation of the 53. Tefft D, Lee M, Smith S, Crowe DL, Bellusci S, Warburton D.
mTOR pathway. J Cell Biol 178: 437–451, 2007. mSprouty2 inhibits FGF10-activated MAP kinase by differentially bind-
31. Land S. Oxygen-sensing patghways and the development of mammalian ing to upstream target proteins. Am J Physiol Lung Cell Mol Physiol 283:
gas exchange. Redox Report 8: 326 –340, 2003. L700 –L706, 2002.
32. Land SC, Collett A. Detection of Cl⫺ flux in the apical microenviron- 54. van Tuyl M, Liu J, Wang J, Kuliszewski M, Tibboel D, Post M. Role
ment of cultured foetal distal lung epithelial cells. J Exp Biol 204: of oxygen and vascular development in epithelial branching morphogen-
785–795, 2001. esis of the developing mouse lung. Am J Physiol Lung Cell Mol Physiol
33. Land SC, Tee AR. Hypoxia-inducible factor 1␣ is regulated by the 288: L167–L178, 2005.
mammalian target of rapamycin (mTOR) via an mTOR signaling motif. J 55. Warburton D. Developmental biology: order in the lung. Nature 453:
Biol Chem 282: 20534 –20543, 2007. 733–735, 2008.

AJP-Lung Cell Mol Physiol • VOL 299 • OCTOBER 2010 • www.ajplung.org


mTORC1 AND VASCULAR SIGNALING IN FETAL LUNG L471
56. Weaver M, Batts L, Hogan BL. Tissue interactions pattern the mesen- and metastatic renal cell carcinoma. Hum Mol Genet 14: 1839 –1850,
chyme of the embryonic mouse lung. Dev Biol 258: 169 –184, 2003. 2005.
57. Weibel ER. Fractal geometry: a design principle for living organisms. Am 59. Xia H, Nho R, Kleidon J, Kahm J, Henke CA. Polymerized collagen
J Physiol Lung Cell Mol Physiol 261: L361–L369, 1991. inhibits fibroblast proliferation via a mechanism involving the formation
58. Wilson C, Idziaszczyk S, Parry L, Guy C, Griffiths DF, Lazda E, of a beta1 integrin-protein phosphatase 2A-tuberous sclerosis complex 2
Bayne RA, Smith AJ, Sampson JR, Cheadle JP. A mouse model of complex that suppresses S6K1 activity. J Biol Chem 283: 20350 –20360,
tuberous sclerosis 1 showing background specific early postnatal mortality 2008.

Downloaded from ajplung.physiology.org on September 30, 2010

AJP-Lung Cell Mol Physiol • VOL 299 • OCTOBER 2010 • www.ajplung.org

You might also like