SlideShare a Scribd company logo
THE JOURNAL OF BIOLOGICAL CHEMISTRY Vol. 277, No. 23, Issue of June 7, pp. 20618–20624, 2002 
© 2002 by The American Society for Biochemistry and Molecular Biology, Inc. Printed in U.S.A. 
Herstatin, an Autoinhibitor of the Human Epidermal Growth Factor 
Receptor 2 Tyrosine Kinase, Modulates Epidermal Growth Factor 
Signaling Pathways Resulting in Growth Arrest* 
Received for publication, November 28, 2001, and in revised form, March 19, 2002 
Published, JBC Papers in Press, April 4, 2002, DOI 10.1074/jbc.M111359200 
Quincey A. Justman and Gail M. Clinton‡ 
From the Department of Biochemistry and Molecular Biology, Oregon Health & Sciences University, 
Portland, Oregon 97201 
Herstatin is an autoinhibitor of the ErbB family con-sisting 
of subdomains I and II of the human epidermal 
growth factor receptor 2 (ErbB-2) extracellular domain 
and a novel C-terminal domain encoded by an intron. 
Herstatin binds to human epidermal growth factor re-ceptor 
2 and to the epidermal growth factor receptor 
(EGFR), blocking receptor oligomerization and tyrosine 
phosphorylation. In this study, we characterized several 
early steps in EGFR activation and investigated down-stream 
signaling events induced by epidermal growth 
factor (EGF) and by transforming growth factor ! 
(TGF-!) in NIH3T3 cell lines expressing EGFR with and 
without herstatin. Herstatin expression decreased EGF-induced 
EGFR tyrosine phosphorylation and delayed 
receptor down-regulation despite receptor occupancy 
by ligand with normal binding affinity. Akt stimulation 
by EGF and TGF-!, but not by fibroblast growth factor 2, 
was almost completely blocked in the presence of her-statin. 
Surprisingly, EGF and TGF-! induced full activa-tion 
of MAPK in duration and intensity and stimulated 
association of the EGFR with Shc and Grb2. Although 
MAPK was fully stimulated, herstatin expression pre-vented 
TGF-!-induced DNA synthesis and EGF-induced 
proliferation. The herstatin-mediated uncoupling of 
MAPK from Akt activation was also observed in Chinese 
hamster ovary cells co-transfected with EGFR and her-statin. 
These findings show that herstatin expression 
alters EGF and TGF-! signaling profiles, culminating in 
inhibition of proliferation. 
The ErbB family of receptor tyrosine kinases includes the 
prototypical EGFR,1 human epidermal growth factor receptor 
(HER)-2, HER-3, and HER-4. The ErbB receptors consist of an 
extracellular ligand binding domain (ECD), a single transmem-brane 
domain, and a cytoplasmic tyrosine kinase domain (1–3). 
Several growth factors containing EGF-like domains bind with 
high affinity to each ErbB receptor except HER-2, which ap-pears 
to function solely as a transactivating co-receptor (4–6). 
Growth factor binding induces homomer and heteromer inter-actions 
between ErbB family members, which are required for 
kinase activation and receptor autophosphorylation in trans (2, 
7, 8). The tyrosine phosphorylation sites on ErbB receptors 
provide docking sites for signaling proteins that execute such 
diverse cellular responses as survival, proliferation, migration, 
differentiation, and apoptosis (9, 10). Given this wide range of 
action, regulation of ErbB signaling is critical, and misregula-tion 
has been implicated in the pathogenesis of many cancers 
(4 –10). 
The ErbB receptors transduce signals through the mitogen-activated 
protein kinase (MAPK) cascade and the phosphati-dylinositol 
3-kinase (PI3K)/Akt signaling pathway. Generally, 
EGF-like growth factors concomitantly activate these two path-ways 
(11–13), whereas several EGFR inhibitors suppress both 
signaling cascades (12, 14, 15). Although the MAPK and the 
PI3K/Akt pathways are commonly coupled, recent evidence 
indicates that Akt is more important in initiating proliferation 
and survival signals (11, 16). Targeted inactivation of Akt 
inhibits cell growth (17, 18), inducing arrest in G1 independent 
of MAPK activation by EGF (12). In addition, EGF induction of 
Akt activity protects against Fas-induced apoptosis by a 
MAPK-independent mechanism (19). Because activation of Akt 
protects against drug-induced death of human breast cancer 
cells (12, 18, 19), inhibitors that target the Akt pathway should 
be effective in enhancing tumor cell death. 
Interactions between ECDs are critical in ErbB receptor 
oligomerization and activation (20–22). Receptor interactions 
in vivo may also require a membrane anchor (21, 22), which 
increases the affinity between dimer partners !10,000-fold 
(23). Because HER-2 is the preferred heteromeric partner of 
ErbB receptors (24, 25), it has been hypothesized that domi-nant 
negative mutants containing the HER-2 ECD (21, 26) or 
subdomains from its ECD (27) could disrupt all combinations of 
ErbB receptor interactions. Indeed, a mutant missing most of 
the cytoplasmic domain of p185neu blocks formation of HER-2 
homodimers (28), HER-2/EGFR heteromers (15, 27, 28), and 
HER-2 association with HER-3 (29). This p185neu dominant 
negative mutant also suppresses EGF-mediated activation of 
both MAPK and PI3K/Akt (15, 28). 
ErbB splice variants that encode truncated ECDs have been 
suggested to modulate ErbB signaling (30) either by sequester-ing 
growth factors (31) or by altering receptor interactions (29, 
32). One of these, herstatin, is a secreted alternative product of 
the HER-2 gene containing ECD subdomains I and II followed 
by an intron-encoded 79-amino acid sequence (32). Herstatin 
has been shown to bind to EGFR and HER-2 and to block 
homomeric and heteromeric receptor interactions (29, 32). In 
* This study was supported by grants from the National Cancer 
Institute. The costs of publication of this article were defrayed in part 
by the payment of page charges. This article must therefore be hereby 
marked “advertisement” in accordance with 18 U.S.C. Section 1734 
solely to indicate this fact. 
‡ To whom correspondence should be addressed: Dept. of Biochemis-try 
and Molecular Biology, Oregon Health & Sciences University, 3181 
SW Sam Jackson Park Rd., Portland, OR 97201. Tel.: 503-494-2543; 
Fax: 503-494-8393; E-mail: clinton@ohsu.edu. 
1 The abbreviations used are: EGFR, epidermal growth factor recep-tor; 
EGF, epidermal growth factor; TGF-!, transforming growth factor 
!; MAPK, mitogen-activated protein kinase; HER, human epidermal 
growth factor receptor; ECD, extracellular ligand binding domain; 
PI3K, phosphatidylinositol 3-kinase; DMEM, Dulbecco’s modified Ea-gle’s 
medium; FBS, fetal bovine serum; CHO, Chinese hamster ovary; 
PBS, phosphate-buffered saline; FGF, fibroblast growth factor. 
20618 This paper is available on line at http://www.jbc.org 
Downloaded from http://www.jbc.org/ at Harvard Libraries on December 1, 2014
Herstatin Blocks EGF Activation of Akt and Growth 20619 
contrast to dominant negative mutants, herstatin does not 
require a membrane anchor to achieve complex formation and 
trans inhibition, suggesting that its novel C-terminal domain 
may confer high affinity binding to the receptors. Indeed, the 
intron-encoded domain, expressed as a recombinant peptide, 
binds to HER-2 and the EGFR (29, 32). Although herstatin 
inhibits the initial steps of receptor activation, its impact on 
ligand binding and intracellular signaling events has not been 
examined. In light of the novel structure and receptor binding 
properties of herstatin, determination of its effects on signaling 
is required to understand its mechanism of action and impact 
on the biology of the ErbB receptors. 
In this study, we demonstrate that herstatin selectively mod-ulates 
signaling cascades triggered by EGF and TGF-!. These 
results suggest that this naturally occurring, alternative 
HER-2 product provides a novel mechanism for generating 
signaling diversity by EGFs. 
EXPERIMENTAL PROCEDURES 
Cell Culture and Generation of Stable Herstatin EGFR3T3 Clones— 
EGFR3T3 cells were derived from NIH3T3 cells by transfection with 
human EGFR in mammalian expression vector pCDNA3.1 (Invitrogen). 
Stably transfected clones were selected in DMEM " 10% FBS supple-mented 
with 0.4 mg/ml G418. A clonal line expressing high levels of 
EGFR was transfected with human herstatin in pCDNA3.1/Hygro (In-vitrogen) 
using Superfect reagent (Qiagen) as per the manufacturer’s 
instructions. Control 3T3 cell lines were generated by transfection with 
herstatin alone or with the corresponding empty vector. Stable cell lines 
were selected with 0.1 mg/ml hygromycin B and maintained in DMEM 
" 10% FBS containing 0.4 mg/ml G418 and 0.1 mg/ml hygromycin B. 
Chinese hamster ovary (CHO) cells were grown in DMEM supple-mented 
with 10% fetal bovine serum. 
Antibodies—Herstatin polyclonal antibody was generated as de-scribed 
previously (32) and used at a dilution of 1:10,000. All antibodies 
were diluted into TBST (Tris-buffered saline plus 0.005% (v/v) Tween 
20). Herstatin monoclonal antibody was a generous gift from Upstate 
Biotechnology (Lake Placid, NY) and was used at a 1:1000 dilution. 
Antibodies to MAPK and Akt were obtained from Cell Signaling and 
used at a 1:1000 dilution. Phospho-specific polyclonal antibodies to 
MAPK (phosphorylated at T202 and Y204) and Akt (phosphorylated at 
S473) were also obtained from Cell Signaling and used at a 1:1000 
dilution. Rabbit polyclonal anti-EGFR antibody was obtained from 
Santa Cruz Biotechnology, Inc. and used at a 1:1000 dilution. Mono-clonal 
anti-Shc antibody was obtained from Santa Cruz Biotechnology, 
Inc. and used at a 1:1000 dilution. Rabbit polyclonal anti-Grb2 antibody 
was obtained from Santa Cruz Biotechnology, Inc. and used at a 1:1000 
dilution. Rabbit polyclonal anti-p185HER-2/neu antibody was charac-terized 
previously (33) and used at a 1:10,000 dilution. Anti-phospho-tyrosine 
monoclonal antibody was obtained from Sigma and used at a 
1:10,000 dilution. 
Transient Transfections—Cells were grown to !80% confluence in 
6-well plates and then the plasmid DNAs indicated in the figure legends 
were introduced using LipofectAMINE reagent (Invitrogen) as per the 
manufacturer’s instructions. Transfection efficiencies between samples 
were compared by co-transfection with fluorescent green protein ex-pression 
plasmid (Invitrogen) and inspection by fluorescence micros-copy. 
Proteins were analyzed at 40 h after DNA introduction. 
Receptor Internalization Assays—Cells were grown to 70% conflu-ence, 
serum-starved for 20 h in 0.5% FBS, washed twice in ice-cold PBS, 
and incubated with EGF (Upstate Biotechnology) at 100 ng/ml in cold 
DMEM for 2 h at 4 °C. Cells were then rinsed twice with PBS, placed in 
pre-warmed DMEM, and returned to 37 °C to allow receptor internal-ization. 
At various time points, cells were placed on ice and washed 
twice with ice-cold PBS, and then cell surface proteins were labeled 
with freshly dissolved EZ-link Sulfo-NHS-LC-Biotin (Pierce) at 0.5 
mg/ml in PBS for 30 min at room temperature. To quench the biotiny-lation 
reaction, cells were placed on ice and washed twice with cold PBS 
containing 0.2 mg/ml bovine serum albumin and twice with PBS. EGFR 
from lysed cells was immunoprecipitated (see below). Samples were 
resolved by SDS-PAGE in a 6% polyacrylamide gel, electrotransferred 
to nitrocellulose membrane, and overlaid with streptavidin-horseradish 
peroxidase at 1 "g/ml in TBST (Pierce). Biotinylated proteins were 
visualized by exposing blots to x-ray film (X-Omat; Eastman Kodak Co.) 
after treatment with Supersignal West Pico reagent (Pierce). 
Immunoprecipitations—Cells were washed in PBS and then lysed on 
ice in MTG (50 mM Tris, pH 8.0, 100 mM NaCl, 10% (v/v) glycerol, 1 (v/v) 
Nonidet P-40, and 2 mM sodium orthovanadate) containing protease 
and phosphatase inhibitor mixtures I and II (Sigma; used as per the 
manufacturer’s recommendations). Cell lysate was cleared by centrifu-gation, 
and protein concentrations were quantified by Bradford assay 
(Bio-Rad). EGFR from 150 "g of cell lysate was precipitated by over-night 
incubation with 1 "g of anti-EGFR at 4 °C. Signaling complexes 
from 500 "g of cell extract were precipitated by overnight incubation 
with 2 "g of anti-Grb2 at 4 °C. Immune complexes were bound to 25 "l 
of protein G-Sepharose (Amersham Biosciences) by co-incubation for 40 
min at 4 °C, centrifuged, and washed three times with 1 ml of ice-cold 
MTG (EGFR) or PBS (Grb2). Immune complexes were boiled in SDS-PAGE 
sample buffer for 5 min and analyzed as a Western blot. 
Western Blot Analysis—Western blotting was conducted as described 
previously (29). Briefly, cells were lysed, and protein concentrations 
were quantified as described for immunoprecipitations. Lysates were 
boiled in SDS-PAGE sample buffer and loaded onto polyacrylamide gels 
at 30 "g/lane. After electrophoresis, proteins were transferred onto 
nitrocellulose, stained with Ponceau S, incubated with antibody as 
described above, and visualized by exposure to x-ray film (X-Omat; 
Kodak) after treatment with SuperSignal West Pico reagent or Super- 
Signal West Dura reagent (Pierce). Blots were stripped with the Re-Blot 
Western blot recycling kit (Chemicon International, Inc.) as per the 
manufacturer’s instructions. 
[3H]Thymidine Incorporation Assay—Cells were grown to 70% con-fluence, 
starved for 24 h in DMEM with 0.5% FBS, and then treated 
with various concentrations of TGF-! for 18 h at 37 °C. Ligand was 
removed, and cells were incubated in the presence of [3H]thymidine (1 
"Ci/ml in DMEM) for 4 h at 37 °C. Cells were washed with cold PBS, 
incubated in 10% trichloroacetic acid at room temperature for 10 min, 
and washed twice with 5% trichloroacetic acid. DNA was precipitated 
with 100% ethanol and then solubilized by incubation in 0.2 N NaOH for 
10 min at room temperature. Samples were neutralized with 0.4 N HCl 
and counted in a scintillation counter. 
EGF Proliferation Assay—Cells were plated in quadruplicate, grown 
to confluence, serum-starved for 18 h in DMEM containing 0.5% FBS, 
and transferred to growth medium (DMEM containing 0.5% FBS and 
10 nM EGF). Three days later, growth medium was removed, and live 
cells were quantified with 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltet-razolium 
bromide (Sigma) as described previously (34). 
125I-EGF Binding—The 125I-EGF binding analysis was conducted as 
described previously (35). Briefly, cells were grown to 70% confluence, 
serum-starved for 24 h, and incubated at 4 °C for 2 h with 175 pM 
125I-EGF (NEN) and different amounts of unlabeled EGF (total EGF 
concentrations ranged from 175 pM to 10 nM) in binding buffer (DMEM 
plus 50 mM Hepes, pH 7.4, and 5% (w/v) bovine serum albumin). Cells 
were washed and then extracted in 0.1 N NaOH plus 0.1% (w/v) SDS, 
and bound 125I-EGF was quantified by gamma counting. 
RESULTS 
Herstatin Reduces EGF-stimulated Tyrosine Phosphoryla-tion 
and Down-regulation of the EGFR—Previous studies have 
shown that transient coexpression of herstatin with the EGFR 
in CHO cells results in diminished dimerization and tyrosine 
phosphorylation of the receptor in response to EGF (29). To 
further characterize the effects of herstatin on EGFR and to 
investigate its impact on EGF-induced intracellular signaling, 
stable cell lines that express different levels of herstatin were 
derived from NIH3T3 cells expressing human EGFR (termed 
EGFR3T3 cells). Efforts to stably express herstatin in several 
tumor cell lines that overexpress EGFR as well as HER-2 were 
thwarted, presumably because of an inhibitory effect on cell 
survival. EGFR3T3 cells were transfected with control or her-statin 
expression plasmid, and clonal populations were isolated 
by selection with hygromycin B. Varied levels of herstatin were 
expressed in herstatin-transfected cell lines but not in the 
control-transfected cells (Fig. 1A). 
To investigate the effects of herstatin on ligand-induced ac-tivation 
of the receptor, herstatin- and control-transfected 
EGFR3T3 cells were serum-starved for 20 h and treated with 
saturating concentrations (10 nM) of EGF for 20 min, and 
phosphotyrosine levels of the EGFR were assessed. Although 
clone 1 produced the greatest amount of herstatin, each of the 
three herstatin-expressing clones exhibited a similar reduction 
Downloaded from http://www.jbc.org/ at Harvard Libraries on December 1, 2014
20620 Herstatin Blocks EGF Activation of Akt and Growth 
in tyrosine phosphorylation of the EGFR (Fig. 1B; see also Figs. 
4B and 6A), suggesting that maximal inhibition was achieved. 
The EGFR expression levels were comparable between paren-tal 
cells and the three different clones, showing that expression 
of herstatin did not down-regulate the EGFR (Fig. 1B). 
Because receptor tyrosine phosphorylation is required for 
endocytosis (36), we hypothesized that reduced EGFR auto-phosphorylation 
levels in the presence of herstatin alter EGF-mediated 
receptor down-regulation from the cell surface. Pa-rental 
and herstatin-expressing EGFR3T3 cells were saturated 
with EGF at 4 °C and then returned to 37 °C, a temperature 
permissive for internalization. After various incubation times, 
cell surface proteins were biotinylated, and EGFR was immu-noprecipitated 
and visualized by streptavidin-horseradish per-oxidase 
overlay. In the parental cell line, cell surface EGFR 
was reduced by 80% at 10 min and was undetectable by 20 min 
after the removal of the temperature block (Fig. 1C), in agree-ment 
with other studies (37, 38). In herstatin-expressing cells, 
however, cell surface EGFR was reduced only about 35% at 10 
min, and about one-third of the EGFR remained at the cell 
surface after 20 min of incubation at 37 °C. Delayed down-regulation 
is consistent with previous findings showing that 
EGFR degradation, a process contingent upon endosomal loca-tion 
of the receptor, is blocked in CHO cells that transiently 
express herstatin (29). 
Herstatin Inhibits EGF-stimulated Akt Phosphorylation, but 
MAPK Is Fully Activated—Signal transduction through the 
ErbB family includes both the MAPK and the PI3K/Akt signal-ing 
pathways, which are generally stimulated simultaneously 
by growth factors (11–13). To examine the role of herstatin in 
EGF signaling downstream from the receptor, we treated 
EGFR3T3 parental and herstatin-expressing cells with satu-rating 
amounts of EGF (16 nM) and observed the kinetics of 
MAPK and Akt phosphorylation over a 1-h time course. In the 
parental cell line, the highest level of phospho-Akt was de-tected 
at 20 min after EGF addition (Fig. 2A). In the herstatin-expressing 
cells, however, little phospho-Akt was observed in 
EGF-treated cells, with maximal levels reaching only 2% of the 
parental controls. Phosphorylation of Akt was almost com-pletely 
abolished in the presence of herstatin; interestingly, 
there was no reduction of phospho-MAPK levels. In parental 
and herstatin-expressing cells, the time course and extent of 
MAPK activation were identical; maximal activation was 
achieved by 5 min and declined at 20 min after the addition of 
EGF (Fig. 2A). To determine whether induction of MAPK in 
herstatin-expressing cells was due to ectopic overexpression of 
EGFR (39–41), we investigated EGF signaling through endog-enous 
receptors in 3T3 cells that express herstatin. As in the 
EGFR3T3 cells, we observed preferential EGF activation of 
MAPK and abrogation of Akt phosphorylation in the presence 
of herstatin (Fig. 2B). These data demonstrate that the signal-ing 
profile caused by herstatin expression is not affected by 
ectopic overexpression of EGFR. 
EGF Induces the Formation of a Signaling Complex Contain-ing 
EGFR, phospho-Shc, and Grb2 in Herstatin-expressing 
Cells—After EGF treatment, the adaptor protein Shc binds the 
autophosphorylation domain of EGFR, is tyrosine-phospho-rylated, 
and recruits the Grb2-Sos complex from the cytoplasm 
to the plasma membrane (42). To investigate whether EGF 
stimulation of MAPK in the presence of herstatin was induced 
by EGFR through Shc and Grb2, we immunoprecipitated Grb2 
and examined the immune complex by Western blotting. In 
both parental and herstatin-expressing cells, EGF-dependent 
association of EGFR and Shc with Grb2 was detected (Fig. 3). 
Furthermore, Shc was tyrosine-phosphorylated to a similar 
extent (Fig. 3), providing evidence for an active signaling com-plex. 
Because HER-2 is the preferred heterodimer partner of 
EGFR (4–6), endogenous HER-2 may be present in the EGFR-Shc- 
Grb2 signaling complex, contributing to EGF stimulation 
of the MAPK signaling cascade. However, HER-2 could not be 
detected in the signaling complex immunoprecipitated from 
500 "g of either parental or herstatin-expressing cells (data not 
shown), even though the high titer antibody used (33) detects 
p185HER-2 in 20 "g of 3T3 cell extract. These data suggest 
that Shc and Grb2 transduce the herstatin-mediated EGF sig-nal 
from EGFR to components of the MAPK cascade, with no 
evidence of HER-2 involvement. 
Characterization of the Effects of EGF Concentration on In-tracellular 
Signaling in the Presence and Absence of Hersta-tin— 
Previous studies have shown that at very low EGF con-centrations, 
MAPK activation occurs in the absence of Akt 
activation and EGFR tyrosine phosphorylation (43). We there-fore 
examined whether signaling in EGFR3T3 cells exhibits a 
similar sensitivity to EGF concentration. Maximal stimulation 
of MAPK occurred independently of Akt activation in 
EGFR3T3 cells treated with 0.01 nM EGF (Fig. 4A), suggesting 
that ectopic overexpression of EGFR did not eliminate sensi-tivity 
to very low concentrations of EGF. At 0.1 nM EGF, a 
subsaturating concentration, maximal stimulation of Akt was 
observed in parental cells, but stimulation of Akt was inhibited 
in the herstatin-expressing cells. Therefore, preferential inhi-bition 
of the Akt pathway by herstatin could reflect a quanti-tative 
reduction in effective EGF concentration by competitive 
FIG. 1. Herstatin expression, EGFR tyrosine phosphorylation, 
and receptor down-regulation in parental and herstatin-trans-fected 
EGFR3T3 cells. A, 30 "g of lysate from parental and herstatin-expressing 
EGFR3T3 cells was separated by 7.5% SDS-PAGE and 
subjected to Western blot analysis for herstatin as described under 
“Experimental Procedures.” B, herstatin- and mock-transfected 
EGFR3T3 cells were serum-starved for 20 h and then incubated with 10 
nM EGF for 20 min at 37 °C. Cell extracts were resolved by 6% SDS-PAGE 
and analyzed as a Western blot using anti-phosphotyrosine 
antibody. The blot was stripped as described under “Experimental 
Procedures” and reprobed with anti-EGFR. Results are representative 
of three independent experiments. C, herstatin (clone 1)- and mock-transfected 
EGFR3T3 cells were serum-starved as described above, 
incubated with 16 nM EGF at 4 °C for 2 h, and incubated at 37 °C for the 
durations indicated. Cell surface proteins were biotinylated as de-scribed 
under “Experimental Procedures.” EGFR was immunoprecipi-tated 
from 150 "g of lysate, and immune complexes were separated by 
6% SDS-PAGE, transferred to nitrocellulose, and then overlaid with 
streptavidin-horseradish peroxidase. Films were scanned by imaging 
densitometry (BioRad, model GS700) to quantitate streptavidin-horse-radish 
peroxidase signal. Similar results were observed in two inde-pendent 
experiments. 
Downloaded from http://www.jbc.org/ at Harvard Libraries on December 1, 2014
Herstatin Blocks EGF Activation of Akt and Growth 20621 
FIG. 2. EGF-induced signaling in parental and herstatin-expressing EGFR3T3 cells. A, herstatin (clone 1)- and mock-transfected 
EGFR3T3 cells were serum-starved and incubated with 16 nM EGF at 37 °C for the durations indicated. Cell lysates were separated by 7.5% and 
10% SDS-PAGE and then subjected to Western blot analysis using antibodies specific for phospho-Akt (phospho-Ser473) and phospho-p42/p44 
MAPK (phospho-T202 and Y204). Exposed films were scanned by imaging densitometry to quantitate phospho-Akt signal. Blots were stripped and 
probed with Akt antibody. Results are representative of six independent experiments. B, herstatin- and mock-transfected 3T3 cells were 
serum-starved and incubated with 10 nM EGF at 37 °C for the durations indicated. Cell lysates were separated by 7.5% and 10% SDS-PAGE and 
then subjected to Western blot analysis as described above. 
inhibition. However, after treatment with either 1 or 100 nM 
EGF (#100 times KD), EGFR phosphotyrosine levels were di-minished 
!10-fold, and Akt activation was reduced to the same 
extent (Fig. 4B). These data suggest that herstatin has a qual-itative 
impact on intracellular signaling that is independent of 
EGF concentration. 
Herstatin Expression Does Not Alter the Binding Affinity of 
EGF for EGR3T3 Cells—Previous studies demonstrated that 
herstatin binds to the extracellular domain of the HER-2 re-ceptor 
(32) and forms a stable complex with EGFR (29). Inhi-bition 
of EGFR by herstatin could be caused by interference 
with EGF binding. To examine this possibility, we character-ized 
the binding affinity of EGF to parental and herstatin-expressing 
clones of EGFR3T3 cells. The cells were incubated 
with subsaturating amounts (175 pM) of 125I-EGF, and its dis-placement 
by unlabeled EGF was measured (Fig. 5). The dis-placement 
curve exhibited by the parental cells was indistin-guishable 
from that of cell lines that expressed either high 
(clone 1) or low (clones 2 and 3) levels of herstatin (see Fig. 1A). 
Moreover, Scatchard analysis of the binding data, as described 
in the legend to Fig. 6, revealed the same apparent dissociation 
constant of !500 pM in the absence and presence of different 
levels of herstatin. These studies show that herstatin expres-sion 
does not prevent EGF binding or alter the EGF binding 
affinity, suggesting that herstatin is not a competitive inhibitor 
of EGF-mediated EGFR activation. Herstatin therefore modu-lates 
signaling of receptors that are complexed with EGF. 
Herstatin Inhibits TGF-!-induced Receptor Phosphorylation 
and Akt Phosphorylation, Whereas MAPK Activation Is Unaf-fected— 
TGF-! is an EGFR ligand that has increased mitogenic 
and transforming potency compared with EGF (44). Although 
these ligands compete for receptor binding, they exhibit subtly 
different binding properties to the EGFR (45) and show distinct 
co-receptor-dependent signal potentiation (37). Despite these 
differences, parental and herstatin-expressing EGFR3T3 cells 
treated with TGF-! displayed signaling profiles similar to 
those observed in response to EGF stimulation. Depression of 
EGFR tyrosine phosphorylation occurred in the herstatin-ex-pressing 
cells, particularly at high (100 nM) concentrations of 
TGF-! (Fig. 6A). Moreover, phospho-Akt levels were markedly 
decreased, whereas MAPK activation was unaffected at both 
low (1 nM) and high (100 nM) concentrations of TGF-! (Fig. 6A). 
Herstatin Expression Does Not Alter FGF-2 Stimulation of Akt— 
The strong suppression of EGF- and TGF-!-induced Akt phospho-rylation 
in the EGFR3T3 cells that stably express herstatin could 
be an indirect effect of herstatin expression or chronic ErbB recep-tor 
inhibition. To examine the integrity of the PI3K/Akt pathway, 
we monitored phospho-Akt levels after treatment with FGF-2, a 
growth factor that activates a heterologous receptor tyrosine ki-nase. 
Parental and herstatin-expressing EGFR3T3 cells were se-rum- 
starved, exactly as done before EGF treatment, and then cells 
were incubated with saturating amounts of FGF-2 (10 nM) for 20 
min. FGF-2 treatment stimulated the tyrosine phosphorylation of 
an 119-kDa protein, the approximate size of the FGF receptor (Fig. 
6B). Furthermore, FGF-2 increased phospho-Akt levels to an equiv-alent 
extent in the presence and absence of herstatin (Fig. 6B). 
These data demonstrate the functional integrity of the PI3K/Akt 
pathway and suggest that the reduction of phospho-Akt levels 
caused by herstatin expression is specific to EGF- and TGF-!- 
induced signaling. 
FIG. 3. EGF induces EGFR-Shc-Grb2 complex formation in 
herstatin-expressing EGFR3T3 cells. Herstatin (clone 1)- and 
mock-transfected EGFR3T3 cells were serum-starved and incubated 
with 10 nM EGF at 37 °C for 20 min. Proteins complexed to anti-Grb2 
were immunoprecipitated from 500 "g of cell lysate, separated by 8% 
and 12% SDS-PAGE, and then subjected to Western blot analysis using 
the antibodies indicated. The anti-phosphotyrosine blot was stripped 
and reprobed with anti-Shc antibody. Results are representative of two 
independent experiments. 
Downloaded from http://www.jbc.org/ at Harvard Libraries on December 1, 2014
20622 Herstatin Blocks EGF Activation of Akt and Growth 
Herstatin Uncouples MAPK from Akt Activation in Tran-siently 
Transfected CHO Cells—To evaluate whether the un-coupling 
of phospho-Akt from MAPK activation by herstatin 
was a feature confined to the 3T3 cell background, we exam-ined 
the EGF signaling profile in CHO cells, which do not 
express endogenous EGFR (25). The cells were transiently 
transfected with EGFR and different levels of herstatin expres-sion 
plasmids and then treated with EGF. Fig. 6C illustrates 
that EGF induction of MAPK phosphorylation were unaffected 
by expression of different amounts of herstatin. In contrast, 
Akt activation was inhibited in proportion to herstatin expres-sion 
levels. 
Herstatin Depresses Mitogenic Stimulation by EGF and 
TGF-!—To test whether herstatin affected the proliferative func-tions 
of the EGFR growth factors, TGF-!-induced mitogenesis 
was assessed by measuring DNA synthesis. Cells were first 
forced into quiescence by serum starvation for 40 h and then 
treated with different concentrations of TGF-!. The ligand was 
removed, and cells were incubated with [3H]thymidine to quan-tify 
DNA synthesis. In the presence of herstatin, we observed a 
striking decrease in TGF-!-induced [3H]thymidine incorporation 
that was not overcome at high ligand concentrations (Fig. 7A). 
The impact of herstatin on EGF-mediated cell proliferation was 
examined by quantitation of live cells by the 3-(4,5-dimethylthia-zol- 
2-yl)-2,5-diphenyltetrazolium bromide assay (34). Equal 
numbers of EGFR3T3 control and herstatin-transfected cells 
were plated, serum-starved, and then treated for 72 h with ve-hicle 
or EGF. Herstatin expression resulted in a significant re-duction 
in viable cells in the absence of EGF (p $ 0.007; Fig. 7B), 
which may reflect diminished survival under conditions of serum 
deprivation. Whereas EGF treatment significantly increased the 
control EGFR3T3 cells (p $ 0.006; Fig. 7B), cells expressing 
herstatin displayed no significant proliferation in response to 
EGF treatment. These results demonstrate that herstatin inter-rupts 
TGF-!- and EGF-mediated mitogenic signal transduction, 
resulting in inhibition of proliferation. 
DISCUSSION 
Investigating the mechanisms employed by ErbB inhibitors 
and their impact on signaling is critical to understanding the 
biology of these receptors and to developing anti-receptor tyro-sine 
kinase therapeutics. Whereas several inhibitors of the 
EGFR have been investigated, herstatin is distinguished by its 
novel structure, consisting of subdomains I and II of the HER-2 
ECD and an intron encoded-C-terminal domain (32). Further-more, 
herstatin is the only naturally occurring inhibitor of the 
EGFR in mammalian cells that exerts its action on the initial 
events in receptor activation: dimerization and autophospho-rylation 
(29, 32). In this study, we show that herstatin selec-tively 
modulates the intracellular signaling pathways stimu-lated 
by EGFR ligands. EGF binds to its receptor with normal 
affinity in the presence of herstatin, yet receptor tyrosine phos-phorylation 
and down-regulation are suppressed. Whereas her-statin 
allows full ligand stimulation of the MAPK pathway and 
its upstream effector, Shc, Akt phosphorylation is selectively 
blocked, resulting in suppression of cell growth. 
Herstatin is a secreted protein that binds to and inhibits the 
EGFR (29). Although the binding site has not been mapped, 
previous observations suggest that herstatin associates with 
the ECD of ErbB receptors (32). We therefore examined 
whether herstatin may interfere with binding of EGF. Our 
results demonstrate that neither the binding affinity nor the 
overall number of EGF binding sites was significantly altered 
in cells that expressed either low or very high levels of hersta-tin. 
Therefore, herstatin appears to inhibit EGFR that is occu-pied 
by growth factor. Two other classes of EGFR inhibitors 
compete with ligand binding, including the Drosophila ligand 
FIG. 4. Effects of EGF concentra-tion 
on signaling in parental and her-statin- 
expressing EGFR3T3 cells. A, 
herstatin (clone 1)- and mock-transfected 
EGFR3T3 cells were serum-starved, 
treated with 0, 0.01, and 10 nM EGF at 
37 °C for 20 min, and analyzed by West-ern 
blot as described in the Fig. 2 legend. 
Staining with Ponceau S confirms equal 
loading. B, herstatin (clone 1)- and mock-transfected 
EGFR3T3 cells were treated 
with l or 100 nM EGF as described in A. 
Exposed films were scanned by imaging 
densitometry to quantitate phosphoty-rosine 
signal. Results are representative 
of two independent experiments. 
FIG. 5. Displacement of 125I-EGF by unlabeled EGF in parental 
and herstatin-expressing EGFR3T3 cells. Herstatin and mock-transfected 
EGFR3T3 cells were plated at 5 % 104 cells/well in 24-well 
plates and serum-starved. Cells were incubated for 2 h at 4 °C with 175 
pM 125I-EGF and unlabeled EGF at total concentrations ranging from 
175 pM to 10 nM. Displacement of bound 125I-EGF by unlabeled EGF 
was plotted as the maximum percentage bound 125I-EGF. The dissoci-ation 
constants (KD) of EGF for herstatin- and mock-transfected 
EGFR3T3 clones were estimated by Scatchard analysis: bound/free 
versus bound was plotted using KaleidaGraph software (Synergy Anal-ysis, 
1997); the slopes of regression lines as calculated by the program 
provided the estimate of &1/KD. All regression line R2 values exceeded 
0.99. 
Downloaded from http://www.jbc.org/ at Harvard Libraries on December 1, 2014
Herstatin Blocks EGF Activation of Akt and Growth 20623 
FIG. 6. Herstatin effects on signaling induced by EGF, TGF-!, and FGF-2. A, herstatin (clone 1)- and mock-transfected EGFR3T3 cells 
were serum-starved and treated with l or 100 nM TGF-! at 37 °C for 20 min and analyzed as described in Fig. 4B. Results are representative of 
two independent experiments. B, herstatin (clone 1)- and mock-transfected EGFR3T3 cells were serum-starved and treated with 100 nM FGF-2 at 
37 °C for 20 min, and cell lysate was subjected to Western blot analysis using antibodies against phosphotyrosine and phospho-Akt. The blot was 
stripped and incubated with Akt antibody. C, CHO cells were grown to 80% confluence and transfected with 1.5 "g of EGFR and 0.5 or 1.5 "g of 
herstatin expression plasmids. 20 h after transfection, cells were serum-starved, treated with 10 nM EGF for 20 min at 37 °C, and analyzed for 
phospho-Akt and phospho-p42/p44 MAPK by Western blot. Equal loading was confirmed by Ponceau S staining. 
Argos (46, 47) and the EGFR monoclonal antibody C225 (48). 
Because herstatin inhibits ligand-occupied receptor, it may be 
effective even when growth factors are overproduced by tumors 
(49). 
In this study, we demonstrate that herstatin uncouples in-tracellular 
signaling pathways triggered by EGF and TGF-!. 
Whereas EGFR tyrosine phosphorylation and Akt activation 
are inhibited, Shc and MAPK are fully activated. This is in 
contrast to the intracellular signaling effects of several other 
inhibitors of EGFR including the quinazoline inhibitors (12), 
the p185neu dominant negative mutant (15), and the C225 
monoclonal antibody inhibitor (14), which suppress EGF-in-duced 
phosphorylation of both MAPK and Akt. Similar to her-statin, 
these inhibitors reduce EGFR tyrosine phosphorylation 
and cause prolonged retention of the EGFR at the cell surface. 
In agreement with previous studies (43), we also observed 
uncoupling of MAPK and Akt signaling cascades at very low 
concentrations of EGF (0.01 nM), suggesting that either limit-ing 
amounts of ligand or the presence of herstatin preferen-tially 
stimulated the MAPK cascade. The signaling effects of 
herstatin can not be explained by a reduction in the effective 
concentration of growth factor; the EGF binding affinity was 
unaffected by herstatin, and selective suppression of Akt sig-naling 
was observed at both subsaturating (0.1 nM) and very 
high ligand concentrations (100 nM). 
Because phosphorylation of specific tyrosine residues on ac-tivated 
receptors is responsible for the recruitment and activa-tion 
of distinct intracellular signaling molecules (51), inhibition 
of some, but not all, EGFR phosphorylation sites by herstatin 
may cause preferential recruitment of effector molecules (52). 
Evaluation of this possibility will require phosphopeptide map-ping 
of the EGFR from the herstatin-expressing cells. However, 
even in the absence of EGFR tyrosine phosphorylation, either 
by kinase-impaired EGFR (39–41) or EGFR missing its tyro-sine 
phosphorylation sites (53), MAPK is activated, whereas 
Akt is not stimulated by EGF (16, 54). In these cases (53, 55), 
as well as with herstatin, EGFR appears to be involved in 
activation of the MAPK cascade, as shown by its presence in a 
signaling complex containing tyrosine phosphorylated Shc and 
Grb2. An endogenous receptor tyrosine kinase or a cytoplasmic 
kinase such as src (56) may also participate in MAPK activa-tion 
by EGF. Endogenous HER-2, the preferred EGFR hetero-meric 
partner, is unlikely to be responsible for MAPK activa-tion 
because herstatin effectively disrupts HER-2 homomeric 
FIG. 7. TGF-!-induced DNA synthesis and EGF-induced prolif-eration 
of parental and herstatin-expressing EGFR3T3 cells. A, 
herstatin (clone 1)- and mock-transfected EGFR3T3 cells were plated at 
5 % 104 in triplicate, serum-starved, and treated with TGF-! at the 
concentrations indicated. At 18 h, cells were incubated with [3H]thymi-dine 
as described under “Experimental Procedures.” Levels of [3H]thy-midine 
incorporation are expressed as a percentage of the untreated 
control. Error bars indicate sample mean ' S.D. Results are represent-ative 
of three independent experiments. B, herstatin (clone 1)- and 
mock-transfected EGFR3T3 clones were grown to 90% confluence, se-rum- 
starved, and treated with 10 nM EGF. At 72 h, live cells were 
quantified by 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bro-mide 
assay as described under “Experimental Procedures.” Error bars 
indicate sample mean ' S.D. Results are representative of two inde-pendent 
experiments. 
Downloaded from http://www.jbc.org/ at Harvard Libraries on December 1, 2014
20624 Herstatin Blocks EGF Activation of Akt and Growth 
and heteromeric interactions (29). Although we were unable to 
detect HER-2 in the EGF-induced signaling complex, other 
endogenous tyrosine kinases may be involved in MAPK activa-tion 
in herstatin-expressing cells. 
A major question in signaling through receptor tyrosine ki-nases 
concerns the mechanism by which growth factors can 
stimulate diverse cellular responses (52, 57, 58). For the ErbB 
receptors, one level of diversity is achieved through generation 
of different ErbB dimer pairs that have been found to differen-tially 
stimulate intracellular signaling pathways (10, 51, 52, 
57). Alternatively, signaling by the same dimer pair may be 
altered in response to activation by different growth factors (8, 
22, 52). Additionally, EGF ligands have been shown to promote 
diverse cellular responses, depending on the cell type (59). The 
results presented here point to a novel mechanism of generat-ing 
diversity by which a single ligand, EGF, can achieve altered 
signal output within a given cell context. Our studies suggest 
that herstatin, a HER-2 receptor variant expressed in fetal 
kidney and liver cells, can selectively alter EGF signal output, 
resulting in growth arrest. 
The Ras/MAPK pathway was previously suggested to be the 
major mitogenic signaling pathway initiated by the EGFR (60). 
In the presence of herstatin, full stimulation of MAPK by 
TGF-! was insufficient to drive entry into S phase, as sug-gested 
by the suppression of DNA synthesis. Moreover, EGF 
stimulation of MAPK was insufficient to stimulate cell prolif-eration 
in the presence of herstatin. Because Akt activation 
was strongly inhibited, components of the PI3K signaling path-way 
that are required for growth factor-induced proliferation 
may not be activated in the presence of herstatin. This finding 
is in agreement with recent observations that interruption of 
the MAPK pathway by EGFR blockade with quinazoline inhib-itors 
is not the cause of G1 arrest, but rather interruption of 
PI3K function is required (12). 
Signaling through the EGF receptor is often enhanced in hu-man 
cancers through overexpression of the receptor and auto-crine 
stimulation by ligands produced by the tumor (49). En-hanced 
EGFR signaling in several carcinomas is directly coupled 
to inappropriate phospho-Akt survival signals, rendering many 
cancers resistant to apoptotic signals, including those activated 
by radiation and chemotherapies (12, 19). EGF stimulation of 
Akt kinase activity has been proposed as a major mechanism 
behind enhanced survival conferred by inappropriate EGF sig-naling. 
The activation of Akt appears to be both required and 
sufficient for the antiapoptotic function of EGF (19). Results 
presented here point to the effectiveness of herstatin in blockage 
of Akt activation and inhibition of proliferation stimulated by 
EGF. Previous studies also demonstrate the effectiveness of her-statin 
in blocking proliferation stimulated by HER-2 overexpres-sion, 
which often occurs in the same tumor cells that have en-hanced 
EGFR signaling. The results presented here further 
support the potential utility of herstatin in the development of 
therapeutics against cancers with ErbB receptor involvement. 
Acknowledgments—We thank L. S. Shamieh for helpful discussions 
and critical reading of the manuscript, M. E. Sommer for generating the 
EGFR3T3/herstatin cell lines, and M. C. Denton for constructing the 
herstatin and EGFR expression plasmids. 
REFERENCES 
1. Hanks, S. K., Quinn, A. M., and Hunter, T. (1988) Science 241, 42–52 
2. Schlessinger, J., Ullrich, A., Honegger, A. M., and Moolenaar, W. H. (1988) 
Cold Spring Harbor Symp. Quant. Biol. 53, 515–519 
3. Yarden, Y., and Ullrich, A. (1988) Annu. Rev. Biochem. 57, 443–478 
4. Dougall, W. C., Qian, X., Peterson, N. C., Miller, M. J., Samanta, A., and 
Greene, M. I. (1994) Oncogene 9, 2109–2123 
5. Hynes, N. E., and Stern, D. F. (1994) Biochim. Biophys. Acta 1198, 165–184 
6. Tzahar, E., and Yarden, Y. (1998) Biochim. Biophys. Acta 1377, M25&M37 
7. Heldin, C. H., and Ostman, A. (1996) Cytokine Growth Factor Rev. 7, 3–10 
8. Riese, D. J., II, and Stern, D. F. (1998) Bioessays 20, 41–48 
9. Burden, S., and Yarden, Y. (1997) Neuron 18, 847–855 
10. Olayioye, M. A., Neve, R. M., Lane, H. A., and Hynes, N. E. (2000) EMBO J. 19, 
3159–3167 
11. Blume-Jensen, P., and Hunter, T. (2001) Nature 411, 355–365 
12. Busse, D., Doughty, R. S., Ramsey, T. T., Russell, W. E., Price, J. O., Flanagan, 
W. M., Shawver, L. K., and Arteaga, C. L. (2000) J. Biol. Chem. 275, 
6987–6995 
13. Rommel, C., Clarke, B. A., Zimmermann, S., Nunez, L., Rossman, R., Reid, K., 
Moelling, K., Yancopoulos, G. D., and Glass, D. J. (1999) Science 286, 
1738–1741 
14. Liu, B., Fang, M., Lu, Y., Mendelsohn, J., and Fan, Z. (2001) Oncogene 20, 
1913–1922 
15. Qian, X., O’Rourke, D. M., Fei, Z., Zhang, H. T., Kao, C. C., and Greene, M. I. 
(1999) J. Biol. Chem. 274, 574–583 
16. Walker, F., Kato, A., Gonez, L. J., Hibbs, M. L., Pouliot, N., Levitzki, A., and 
Burgess, A. W. (1998) Mol. Cell. Biol. 18, 7192–7204 
17. Jones, S. M., and Kazlauskas, A. (2001) Nat. Cell. Biol. 3, 165–172 
18. Zhou, B. P., Liao, Y., Xia, W., Spohn, B., Lee, M. H., and Hung, M. C. (2001) 
Nat. Cell. Biol. 3, 245–252 
19. Gibson, S., Tu, S., Oyer, R., Anderson, S. M., and Johnson, G. L. (1999) J. Biol. 
Chem. 274, 17612–17618 
20. Lemmon, M. A., Bu, Z., Ladbury, J. E., Zhou, M., Pinchasi, D., Lax, I., 
Engelman, D. M., and Schlessinger, J. (1997) EMBO J. 16, 281–294 
21. O’Rourke, D. M., Qian, X., Zhang, H. T., Davis, J. G., Nute, E., Meinkoth, J., 
and Greene, M. I. (1997) Proc. Natl. Acad. Sci. U. S. A. 94, 3250–3255 
22. Tzahar, E., Pinkas-Kramarski, R., Moyer, J. D., Klapper, L. N., Alroy, I., 
Levkowitz, G., Shelly, M., Henis, S., Eisenstein, M., Ratzkin, B. J., Sela, M., 
Andrews, G. C., and Yarden, Y. (1997) EMBO J. 16, 4938–4950 
23. Tanner, K. G., and Kyte, J. (1999) J. Biol. Chem. 274, 35985–35990 
24. Graus-Porta, D., Beerli, R. R., Daly, J. M., and Hynes, N. E. (1997) EMBO J. 
16, 1647–1655 
25. Tzahar, E., Waterman, H., Chen, X., Levkowitz, G., Karunagaran, D., Lavi, S., 
Ratzkin, B. J., and Yarden, Y. (1996) Mol. Cell. Biol. 16, 5276–5287 
26. Qian, X., O’Rourke, D. M., Zhao, H., and Greene, M. I. (1996) Oncogene 13, 
2149–2157 
27. Kumagai, T., Davis, J. G., Horie, T., O’Rourke, D. M., and Greene, M. I. (2001) 
Proc. Natl. Acad. Sci. U. S. A. 98, 5526–5531 
28. O’Rourke, D. M., Nute, E. J., Davis, J. G., Wu, C., Lee, A., Murali, R., Zhang, 
H. T., Qian, X., Kao, C. C., and Greene, M. I. (1998) Oncogene 16, 1197–1207 
29. Azios, N. G., Romero, F. J., Denton, M. C., Doherty, J. K., and Clinton, G. M. 
(2001) Oncogene 20, 5199–5209L. J. 
30. Maihle, N. J., Baron, A. T., Barrette, B. A., Boardman, C. H., Christensen, 
T. A., Cora, E. M., Faupel-Badger, J. M., Greenwood, T., Juneja, S. C., 
Lafky, J. M., Lee, H., Reiter, J. L., and Podratz, K. C. (2002) Cancer 
Treatment Res. 107, 247–258 
31. Lee, H., Akita, R. W., Sliwkowski, M. X., and Maihle, N. J. (2001) Cancer Res. 
61, 4467–4473 
32. Doherty, J. K., Bond, C., Jardim, A., Adelman, J. P., and Clinton, G. M. (1999) 
Proc. Natl. Acad. Sci. U. S. A. 96, 10869–10874 
33. Christianson, T. A., Doherty, J. K., Lin, Y. J., Ramsey, E. E., Holmes, R., 
Keenan, E. J., and Clinton, G. M. (1998) Cancer Res. 58, 5123–5129 
34. Hansen, M. B., Nielsen, S. E., and Berg, K. (1989) J. Immunol. Methods 119, 
203–210 
35. Sliwkowski, M. X., Schaefer, G., Akita, R. W., Lofgren, J. A., Fitzpatrick, V. D., 
Nuijens, A., Fendly, B. M., Cerione, R. A., Vandlen, R. L., and Carraway, 
K. L., III. (1994) J. Biol. Chem. 269, 14661–14665 
36. Cadena, D. L., Chan, C. L., and Gill, G. N. (1994) J. Biol. Chem. 269, 260–265 
37. Lenferink, A. E., Pinkas-Kramarski, R., van de Poll, M. L., van Vugt, M. J., 
Klapper, L. N., Tzahar, E., Waterman, H., Sela, M., van Zoelen, E. J., and 
Yarden, Y. (1998) EMBO J. 17, 3385–3397 
38. Vieira, A. V., Lamaze, C., and Schmid, S. L. (1996) Science 274, 2086–2089 
39. Campos-Gonzalez, R., and Glenney, J. R., Jr. (1992) J. Biol. Chem. 267, 
14535–14538 
40. Hack, N., Sue, A. Q. A., Mills, G. B., and Skorecki, K. L. (1993) J. Biol. Chem. 
268, 26441–26446 
41. Selva, E., Raden, D. L., and Davis, R. J. (1993) J. Biol. Chem. 268, 2250–2254 
42. Ravichandran, K. S. (2001) Oncogene 20, 6322–6330 
43. Wennstrom, S., and Downward, J. (1999) Mol. Cell. Biol. 19, 4279–4288 
44. Salomon, D. S., Brandt, R., Ciardiello, F., and Normanno, N. (1995) Crit. Rev. 
Oncol. Hematol. 19, 183–232 
45. Winkler, M. E., O’Connor, L., Winget, M., and Fendly, B. (1989) Biochemistry 
28, 6373–6378 
46. Vinos, J., and Freeman, M. (2000) Oncogene 19, 3560–3562 
47. Zhao, D., and Bownes, M. (1999) Dev. Genet. 25, 375–386 
48. Mendelsohn, J. (1997) Clin. Cancer Res. 3, 2703–2707 
49. Klapper, L. N., Kirschbaum, M. H., Sela, M., and Yarden, Y. (2000) Adv. 
Cancer Res. 77, 25–79 
50. Deleted in proof 
51. Alroy, I., and Yarden, Y. (1997) FEBS Lett. 410, 83–86 
52. Sweeney, C., and Carraway, K. L., III. (2000) Oncogene 19, 5568–5573 
53. Gotoh, N., Tojo, A., Muroya, K., Hashimoto, Y., Hattori, S., Nakamura, S., 
Takenawa, T., Yazaki, Y., and Shibuya, M. (1994) Proc. Natl. Acad. Sci. 
U. S. A. 91, 167–171 
54. Deb, T. B., Su, L., Wong, L., Bonvini, E., Wells, A., David, M., and Johnson, 
G. R. (2001) J. Biol. Chem. 276, 15554–15560 
55. Walker, F., Hibbs, M. L., Zhang, H. H., Gonez, L. J., and Burgess, A. W. (1998) 
Growth Factors 16, 53–67 
56. Olayioye, M. A., Badache, A., Daly, J. M., and Hynes, N. E. (2001) Exp. Cell 
Res. 267, 81–87 
57. Carraway, K. L., III, and Cantley, L. C. (1994) Cell 78, 5–8 
58. Hunter, T. (2000) Cell 100, 113–127 
59. Yarden, Y., and Sliwkowski, M. X. (2001) Nat. Rev. Mol. Cell. Biol. 2, 127–137 
60. Malumbres, M., and Pellicer, A. (1998) Front. Biosci. 3, 887–912 
Downloaded from http://www.jbc.org/ at Harvard Libraries on December 1, 2014

More Related Content

What's hot

Venters Molecular and Cellular Biology 2011 2253-2261
Venters Molecular and Cellular Biology 2011 2253-2261Venters Molecular and Cellular Biology 2011 2253-2261
Venters Molecular and Cellular Biology 2011 2253-2261Jordan Irvin
 
Receptor Tyrosine Kinases
Receptor Tyrosine KinasesReceptor Tyrosine Kinases
Receptor Tyrosine Kinasesfondas vakalis
 
Undergraduate Research Symposium Poster
Undergraduate Research Symposium PosterUndergraduate Research Symposium Poster
Undergraduate Research Symposium PosterTim Krueger
 
Stat family:stat 3 in skin disorders by yousry
Stat family:stat 3 in skin disorders  by yousryStat family:stat 3 in skin disorders  by yousry
Stat family:stat 3 in skin disorders by yousry
M.YOUSRY Abdel-Mawla
 
1-s2.0-S2211124715009225-main
1-s2.0-S2211124715009225-main1-s2.0-S2211124715009225-main
1-s2.0-S2211124715009225-mainAnirudh Prahallad
 
MED12 controls the response to multiple cancer drugs through regulation of TG...
MED12 controls the response to multiple cancer drugs through regulation of TG...MED12 controls the response to multiple cancer drugs through regulation of TG...
MED12 controls the response to multiple cancer drugs through regulation of TG...Anirudh Prahallad
 
Biology of mast cells
Biology of mast cellsBiology of mast cells
Lombardi valencia
Lombardi valenciaLombardi valencia
Lombardi valencia
Paolo Lombardi
 
Synthetic microbial communities : Microbial consortia engineering
Synthetic microbial communities : Microbial consortia engineeringSynthetic microbial communities : Microbial consortia engineering
Synthetic microbial communities : Microbial consortia engineering
Gaurav Bilolikar
 
Directed evolution
Directed evolutionDirected evolution
Directed evolution
Dr. K. P. Senthilkumar Naidu
 
J. Biol. Chem.-2008-Lin-15003-14
J. Biol. Chem.-2008-Lin-15003-14J. Biol. Chem.-2008-Lin-15003-14
J. Biol. Chem.-2008-Lin-15003-14Da-Wei Lin
 
Pyramiding of bacterial blight resistance genes in rice
Pyramiding of bacterial blight resistance genes in ricePyramiding of bacterial blight resistance genes in rice
Pyramiding of bacterial blight resistance genes in rice
awareswapnil1111
 

What's hot (19)

Venters Molecular and Cellular Biology 2011 2253-2261
Venters Molecular and Cellular Biology 2011 2253-2261Venters Molecular and Cellular Biology 2011 2253-2261
Venters Molecular and Cellular Biology 2011 2253-2261
 
Receptor Tyrosine Kinases
Receptor Tyrosine KinasesReceptor Tyrosine Kinases
Receptor Tyrosine Kinases
 
Undergraduate Research Symposium Poster
Undergraduate Research Symposium PosterUndergraduate Research Symposium Poster
Undergraduate Research Symposium Poster
 
Stat family:stat 3 in skin disorders by yousry
Stat family:stat 3 in skin disorders  by yousryStat family:stat 3 in skin disorders  by yousry
Stat family:stat 3 in skin disorders by yousry
 
Gpcr genetic variation
Gpcr genetic variationGpcr genetic variation
Gpcr genetic variation
 
1-s2.0-S2211124715009225-main
1-s2.0-S2211124715009225-main1-s2.0-S2211124715009225-main
1-s2.0-S2211124715009225-main
 
MED12 controls the response to multiple cancer drugs through regulation of TG...
MED12 controls the response to multiple cancer drugs through regulation of TG...MED12 controls the response to multiple cancer drugs through regulation of TG...
MED12 controls the response to multiple cancer drugs through regulation of TG...
 
Biology of mast cells
Biology of mast cellsBiology of mast cells
Biology of mast cells
 
EASD 2009
EASD 2009EASD 2009
EASD 2009
 
emboj200920a
emboj200920aemboj200920a
emboj200920a
 
Lombardi valencia
Lombardi valenciaLombardi valencia
Lombardi valencia
 
PNS 0502002625
PNS 0502002625PNS 0502002625
PNS 0502002625
 
Synthetic microbial communities : Microbial consortia engineering
Synthetic microbial communities : Microbial consortia engineeringSynthetic microbial communities : Microbial consortia engineering
Synthetic microbial communities : Microbial consortia engineering
 
MCB
MCBMCB
MCB
 
Directed evolution
Directed evolutionDirected evolution
Directed evolution
 
J. Biol. Chem.-2008-Lin-15003-14
J. Biol. Chem.-2008-Lin-15003-14J. Biol. Chem.-2008-Lin-15003-14
J. Biol. Chem.-2008-Lin-15003-14
 
Pyramiding of bacterial blight resistance genes in rice
Pyramiding of bacterial blight resistance genes in ricePyramiding of bacterial blight resistance genes in rice
Pyramiding of bacterial blight resistance genes in rice
 
Tlr signaling 2013-lim-
Tlr signaling 2013-lim-Tlr signaling 2013-lim-
Tlr signaling 2013-lim-
 
Defense
DefenseDefense
Defense
 

Viewers also liked

Irata report 2013
Irata report 2013Irata report 2013
Irata report 2013
Jamshid Lodhi
 
"Никколо М"
"Никколо М""Никколо М"
"Никколо М"
Victoria Zakutskaya
 
Никколо М
Никколо МНикколо М
Никколо М
Victoria Zakutskaya
 
Раис Габитов
Раис Габитов Раис Габитов
Раис Габитов
Victoria Zakutskaya
 
Channelkit's first birthday
Channelkit's first birthdayChannelkit's first birthday
Channelkit's first birthday
Channelkit
 
Justman and Clinton 2002
Justman and Clinton 2002Justman and Clinton 2002
Justman and Clinton 2002Quincey Justman
 
презентация Никколо М
презентация Никколо Мпрезентация Никколо М
презентация Никколо М
Victoria Zakutskaya
 
Lенис Nерехов
Lенис NереховLенис Nерехов
Lенис Nерехов
Victoria Zakutskaya
 
Justman and Clinton 2002
Justman and Clinton 2002Justman and Clinton 2002
Justman and Clinton 2002Quincey Justman
 
Untitled Presentation
Untitled PresentationUntitled Presentation
Untitled Presentationblog15de14
 
Brujas, brujitas...
Brujas, brujitas...Brujas, brujitas...
Brujas, brujitas...
blog15de14
 
¿Cómo funcionan las cosas?
¿Cómo funcionan las cosas?¿Cómo funcionan las cosas?
¿Cómo funcionan las cosas?blog15de14
 
Jornada ESI
Jornada ESIJornada ESI
Jornada ESI
blog15de14
 

Viewers also liked (15)

Irata report 2013
Irata report 2013Irata report 2013
Irata report 2013
 
"Никколо М"
"Никколо М""Никколо М"
"Никколо М"
 
Compiled Thesis FINAL
Compiled Thesis FINALCompiled Thesis FINAL
Compiled Thesis FINAL
 
Никколо М
Никколо МНикколо М
Никколо М
 
Раис Габитов
Раис Габитов Раис Габитов
Раис Габитов
 
Channelkit's first birthday
Channelkit's first birthdayChannelkit's first birthday
Channelkit's first birthday
 
Justman and Clinton 2002
Justman and Clinton 2002Justman and Clinton 2002
Justman and Clinton 2002
 
презентация Никколо М
презентация Никколо Мпрезентация Никколо М
презентация Никколо М
 
Lенис Nерехов
Lенис NереховLенис Nерехов
Lенис Nерехов
 
Justman and Clinton 2002
Justman and Clinton 2002Justman and Clinton 2002
Justman and Clinton 2002
 
Untitled Presentation
Untitled PresentationUntitled Presentation
Untitled Presentation
 
Brujas, brujitas...
Brujas, brujitas...Brujas, brujitas...
Brujas, brujitas...
 
¿Cómo funcionan las cosas?
¿Cómo funcionan las cosas?¿Cómo funcionan las cosas?
¿Cómo funcionan las cosas?
 
Jornada ESI
Jornada ESIJornada ESI
Jornada ESI
 
CV_QAJ_2014
CV_QAJ_2014CV_QAJ_2014
CV_QAJ_2014
 

Similar to Justman and Clinton 2002

CHEM1110 Chemistry For The Life Sciences I.docx
CHEM1110 Chemistry For The Life Sciences I.docxCHEM1110 Chemistry For The Life Sciences I.docx
CHEM1110 Chemistry For The Life Sciences I.docx
write31
 
Role of normal and altered egfr
Role  of normal and altered egfrRole  of normal and altered egfr
Role of normal and altered egfr
EdreesEkr
 
Cell signaling her2 expression in breast cancer
Cell signaling her2 expression in breast cancerCell signaling her2 expression in breast cancer
Cell signaling her2 expression in breast cancer
Omid Yeganeh
 
Epidermal growth factor and its receptor tyrosine kinase
Epidermal growth factor and its receptor tyrosine kinaseEpidermal growth factor and its receptor tyrosine kinase
Epidermal growth factor and its receptor tyrosine kinase
Gedion Yilma
 
OSU-03012 sensitizes breast cancers to lapatinib-induced cell killing: a role...
OSU-03012 sensitizes breast cancers to lapatinib-induced cell killing: a role...OSU-03012 sensitizes breast cancers to lapatinib-induced cell killing: a role...
OSU-03012 sensitizes breast cancers to lapatinib-induced cell killing: a role...
Enrique Moreno Gonzalez
 
Cagle et al Final Print Copy
Cagle et al Final Print CopyCagle et al Final Print Copy
Cagle et al Final Print CopyPatrick Martin
 
Receptors with intrinsic protein kinase activity
Receptors with intrinsic protein kinase activityReceptors with intrinsic protein kinase activity
Receptors with intrinsic protein kinase activity
Dr.M.Prasad Naidu
 
cancer biology(ppt)
 cancer biology(ppt) cancer biology(ppt)
cancer biology(ppt)
Priyanka Jaikumar
 
Pres spring2009 john_leonard
Pres spring2009 john_leonardPres spring2009 john_leonard
Pres spring2009 john_leonardJohn Leonard
 
Cancer Signal Transduction
Cancer Signal TransductionCancer Signal Transduction
Cancer Signal TransductionDeepika Tripathi
 
Cancer signal-transduction
Cancer signal-transductionCancer signal-transduction
Cancer signal-transductionDeepika Tripathi
 
Evaluating the ability of anti-cancer drugs Etoposide and Staurosporine to in...
Evaluating the ability of anti-cancer drugs Etoposide and Staurosporine to in...Evaluating the ability of anti-cancer drugs Etoposide and Staurosporine to in...
Evaluating the ability of anti-cancer drugs Etoposide and Staurosporine to in...
Davient Bala
 
Tocolytic (Fartoks)
Tocolytic (Fartoks)Tocolytic (Fartoks)
Tocolytic (Fartoks)
Mirfaidah Nadjamuddin
 
Presentation1 fgfr
Presentation1 fgfrPresentation1 fgfr
Presentation1 fgfr
prashantkumbhaj
 
Recombinant growth factors
Recombinant growth factorsRecombinant growth factors
Recombinant growth factors
Mohamed Elasaly, PT, CKTP, MSc Biotech.
 
Examples Of Epigenetics
Examples Of EpigeneticsExamples Of Epigenetics
Examples Of Epigenetics
Lissette Hartman
 
1-s2.0-S0014480015000970-main
1-s2.0-S0014480015000970-main1-s2.0-S0014480015000970-main
1-s2.0-S0014480015000970-mainHelene Schulz
 

Similar to Justman and Clinton 2002 (20)

CHEM1110 Chemistry For The Life Sciences I.docx
CHEM1110 Chemistry For The Life Sciences I.docxCHEM1110 Chemistry For The Life Sciences I.docx
CHEM1110 Chemistry For The Life Sciences I.docx
 
Role of normal and altered egfr
Role  of normal and altered egfrRole  of normal and altered egfr
Role of normal and altered egfr
 
Cell signaling her2 expression in breast cancer
Cell signaling her2 expression in breast cancerCell signaling her2 expression in breast cancer
Cell signaling her2 expression in breast cancer
 
Epidermal growth factor and its receptor tyrosine kinase
Epidermal growth factor and its receptor tyrosine kinaseEpidermal growth factor and its receptor tyrosine kinase
Epidermal growth factor and its receptor tyrosine kinase
 
Ere
EreEre
Ere
 
OSU-03012 sensitizes breast cancers to lapatinib-induced cell killing: a role...
OSU-03012 sensitizes breast cancers to lapatinib-induced cell killing: a role...OSU-03012 sensitizes breast cancers to lapatinib-induced cell killing: a role...
OSU-03012 sensitizes breast cancers to lapatinib-induced cell killing: a role...
 
Cagle et al Final Print Copy
Cagle et al Final Print CopyCagle et al Final Print Copy
Cagle et al Final Print Copy
 
Receptors with intrinsic protein kinase activity
Receptors with intrinsic protein kinase activityReceptors with intrinsic protein kinase activity
Receptors with intrinsic protein kinase activity
 
1041
10411041
1041
 
cancer biology(ppt)
 cancer biology(ppt) cancer biology(ppt)
cancer biology(ppt)
 
Pres spring2009 john_leonard
Pres spring2009 john_leonardPres spring2009 john_leonard
Pres spring2009 john_leonard
 
Cancer Signal Transduction
Cancer Signal TransductionCancer Signal Transduction
Cancer Signal Transduction
 
Cancer signal-transduction
Cancer signal-transductionCancer signal-transduction
Cancer signal-transduction
 
Evaluating the ability of anti-cancer drugs Etoposide and Staurosporine to in...
Evaluating the ability of anti-cancer drugs Etoposide and Staurosporine to in...Evaluating the ability of anti-cancer drugs Etoposide and Staurosporine to in...
Evaluating the ability of anti-cancer drugs Etoposide and Staurosporine to in...
 
847673
847673847673
847673
 
Tocolytic (Fartoks)
Tocolytic (Fartoks)Tocolytic (Fartoks)
Tocolytic (Fartoks)
 
Presentation1 fgfr
Presentation1 fgfrPresentation1 fgfr
Presentation1 fgfr
 
Recombinant growth factors
Recombinant growth factorsRecombinant growth factors
Recombinant growth factors
 
Examples Of Epigenetics
Examples Of EpigeneticsExamples Of Epigenetics
Examples Of Epigenetics
 
1-s2.0-S0014480015000970-main
1-s2.0-S0014480015000970-main1-s2.0-S0014480015000970-main
1-s2.0-S0014480015000970-main
 

Justman and Clinton 2002

  • 1. THE JOURNAL OF BIOLOGICAL CHEMISTRY Vol. 277, No. 23, Issue of June 7, pp. 20618–20624, 2002 © 2002 by The American Society for Biochemistry and Molecular Biology, Inc. Printed in U.S.A. Herstatin, an Autoinhibitor of the Human Epidermal Growth Factor Receptor 2 Tyrosine Kinase, Modulates Epidermal Growth Factor Signaling Pathways Resulting in Growth Arrest* Received for publication, November 28, 2001, and in revised form, March 19, 2002 Published, JBC Papers in Press, April 4, 2002, DOI 10.1074/jbc.M111359200 Quincey A. Justman and Gail M. Clinton‡ From the Department of Biochemistry and Molecular Biology, Oregon Health & Sciences University, Portland, Oregon 97201 Herstatin is an autoinhibitor of the ErbB family con-sisting of subdomains I and II of the human epidermal growth factor receptor 2 (ErbB-2) extracellular domain and a novel C-terminal domain encoded by an intron. Herstatin binds to human epidermal growth factor re-ceptor 2 and to the epidermal growth factor receptor (EGFR), blocking receptor oligomerization and tyrosine phosphorylation. In this study, we characterized several early steps in EGFR activation and investigated down-stream signaling events induced by epidermal growth factor (EGF) and by transforming growth factor ! (TGF-!) in NIH3T3 cell lines expressing EGFR with and without herstatin. Herstatin expression decreased EGF-induced EGFR tyrosine phosphorylation and delayed receptor down-regulation despite receptor occupancy by ligand with normal binding affinity. Akt stimulation by EGF and TGF-!, but not by fibroblast growth factor 2, was almost completely blocked in the presence of her-statin. Surprisingly, EGF and TGF-! induced full activa-tion of MAPK in duration and intensity and stimulated association of the EGFR with Shc and Grb2. Although MAPK was fully stimulated, herstatin expression pre-vented TGF-!-induced DNA synthesis and EGF-induced proliferation. The herstatin-mediated uncoupling of MAPK from Akt activation was also observed in Chinese hamster ovary cells co-transfected with EGFR and her-statin. These findings show that herstatin expression alters EGF and TGF-! signaling profiles, culminating in inhibition of proliferation. The ErbB family of receptor tyrosine kinases includes the prototypical EGFR,1 human epidermal growth factor receptor (HER)-2, HER-3, and HER-4. The ErbB receptors consist of an extracellular ligand binding domain (ECD), a single transmem-brane domain, and a cytoplasmic tyrosine kinase domain (1–3). Several growth factors containing EGF-like domains bind with high affinity to each ErbB receptor except HER-2, which ap-pears to function solely as a transactivating co-receptor (4–6). Growth factor binding induces homomer and heteromer inter-actions between ErbB family members, which are required for kinase activation and receptor autophosphorylation in trans (2, 7, 8). The tyrosine phosphorylation sites on ErbB receptors provide docking sites for signaling proteins that execute such diverse cellular responses as survival, proliferation, migration, differentiation, and apoptosis (9, 10). Given this wide range of action, regulation of ErbB signaling is critical, and misregula-tion has been implicated in the pathogenesis of many cancers (4 –10). The ErbB receptors transduce signals through the mitogen-activated protein kinase (MAPK) cascade and the phosphati-dylinositol 3-kinase (PI3K)/Akt signaling pathway. Generally, EGF-like growth factors concomitantly activate these two path-ways (11–13), whereas several EGFR inhibitors suppress both signaling cascades (12, 14, 15). Although the MAPK and the PI3K/Akt pathways are commonly coupled, recent evidence indicates that Akt is more important in initiating proliferation and survival signals (11, 16). Targeted inactivation of Akt inhibits cell growth (17, 18), inducing arrest in G1 independent of MAPK activation by EGF (12). In addition, EGF induction of Akt activity protects against Fas-induced apoptosis by a MAPK-independent mechanism (19). Because activation of Akt protects against drug-induced death of human breast cancer cells (12, 18, 19), inhibitors that target the Akt pathway should be effective in enhancing tumor cell death. Interactions between ECDs are critical in ErbB receptor oligomerization and activation (20–22). Receptor interactions in vivo may also require a membrane anchor (21, 22), which increases the affinity between dimer partners !10,000-fold (23). Because HER-2 is the preferred heteromeric partner of ErbB receptors (24, 25), it has been hypothesized that domi-nant negative mutants containing the HER-2 ECD (21, 26) or subdomains from its ECD (27) could disrupt all combinations of ErbB receptor interactions. Indeed, a mutant missing most of the cytoplasmic domain of p185neu blocks formation of HER-2 homodimers (28), HER-2/EGFR heteromers (15, 27, 28), and HER-2 association with HER-3 (29). This p185neu dominant negative mutant also suppresses EGF-mediated activation of both MAPK and PI3K/Akt (15, 28). ErbB splice variants that encode truncated ECDs have been suggested to modulate ErbB signaling (30) either by sequester-ing growth factors (31) or by altering receptor interactions (29, 32). One of these, herstatin, is a secreted alternative product of the HER-2 gene containing ECD subdomains I and II followed by an intron-encoded 79-amino acid sequence (32). Herstatin has been shown to bind to EGFR and HER-2 and to block homomeric and heteromeric receptor interactions (29, 32). In * This study was supported by grants from the National Cancer Institute. The costs of publication of this article were defrayed in part by the payment of page charges. This article must therefore be hereby marked “advertisement” in accordance with 18 U.S.C. Section 1734 solely to indicate this fact. ‡ To whom correspondence should be addressed: Dept. of Biochemis-try and Molecular Biology, Oregon Health & Sciences University, 3181 SW Sam Jackson Park Rd., Portland, OR 97201. Tel.: 503-494-2543; Fax: 503-494-8393; E-mail: clinton@ohsu.edu. 1 The abbreviations used are: EGFR, epidermal growth factor recep-tor; EGF, epidermal growth factor; TGF-!, transforming growth factor !; MAPK, mitogen-activated protein kinase; HER, human epidermal growth factor receptor; ECD, extracellular ligand binding domain; PI3K, phosphatidylinositol 3-kinase; DMEM, Dulbecco’s modified Ea-gle’s medium; FBS, fetal bovine serum; CHO, Chinese hamster ovary; PBS, phosphate-buffered saline; FGF, fibroblast growth factor. 20618 This paper is available on line at http://www.jbc.org Downloaded from http://www.jbc.org/ at Harvard Libraries on December 1, 2014
  • 2. Herstatin Blocks EGF Activation of Akt and Growth 20619 contrast to dominant negative mutants, herstatin does not require a membrane anchor to achieve complex formation and trans inhibition, suggesting that its novel C-terminal domain may confer high affinity binding to the receptors. Indeed, the intron-encoded domain, expressed as a recombinant peptide, binds to HER-2 and the EGFR (29, 32). Although herstatin inhibits the initial steps of receptor activation, its impact on ligand binding and intracellular signaling events has not been examined. In light of the novel structure and receptor binding properties of herstatin, determination of its effects on signaling is required to understand its mechanism of action and impact on the biology of the ErbB receptors. In this study, we demonstrate that herstatin selectively mod-ulates signaling cascades triggered by EGF and TGF-!. These results suggest that this naturally occurring, alternative HER-2 product provides a novel mechanism for generating signaling diversity by EGFs. EXPERIMENTAL PROCEDURES Cell Culture and Generation of Stable Herstatin EGFR3T3 Clones— EGFR3T3 cells were derived from NIH3T3 cells by transfection with human EGFR in mammalian expression vector pCDNA3.1 (Invitrogen). Stably transfected clones were selected in DMEM " 10% FBS supple-mented with 0.4 mg/ml G418. A clonal line expressing high levels of EGFR was transfected with human herstatin in pCDNA3.1/Hygro (In-vitrogen) using Superfect reagent (Qiagen) as per the manufacturer’s instructions. Control 3T3 cell lines were generated by transfection with herstatin alone or with the corresponding empty vector. Stable cell lines were selected with 0.1 mg/ml hygromycin B and maintained in DMEM " 10% FBS containing 0.4 mg/ml G418 and 0.1 mg/ml hygromycin B. Chinese hamster ovary (CHO) cells were grown in DMEM supple-mented with 10% fetal bovine serum. Antibodies—Herstatin polyclonal antibody was generated as de-scribed previously (32) and used at a dilution of 1:10,000. All antibodies were diluted into TBST (Tris-buffered saline plus 0.005% (v/v) Tween 20). Herstatin monoclonal antibody was a generous gift from Upstate Biotechnology (Lake Placid, NY) and was used at a 1:1000 dilution. Antibodies to MAPK and Akt were obtained from Cell Signaling and used at a 1:1000 dilution. Phospho-specific polyclonal antibodies to MAPK (phosphorylated at T202 and Y204) and Akt (phosphorylated at S473) were also obtained from Cell Signaling and used at a 1:1000 dilution. Rabbit polyclonal anti-EGFR antibody was obtained from Santa Cruz Biotechnology, Inc. and used at a 1:1000 dilution. Mono-clonal anti-Shc antibody was obtained from Santa Cruz Biotechnology, Inc. and used at a 1:1000 dilution. Rabbit polyclonal anti-Grb2 antibody was obtained from Santa Cruz Biotechnology, Inc. and used at a 1:1000 dilution. Rabbit polyclonal anti-p185HER-2/neu antibody was charac-terized previously (33) and used at a 1:10,000 dilution. Anti-phospho-tyrosine monoclonal antibody was obtained from Sigma and used at a 1:10,000 dilution. Transient Transfections—Cells were grown to !80% confluence in 6-well plates and then the plasmid DNAs indicated in the figure legends were introduced using LipofectAMINE reagent (Invitrogen) as per the manufacturer’s instructions. Transfection efficiencies between samples were compared by co-transfection with fluorescent green protein ex-pression plasmid (Invitrogen) and inspection by fluorescence micros-copy. Proteins were analyzed at 40 h after DNA introduction. Receptor Internalization Assays—Cells were grown to 70% conflu-ence, serum-starved for 20 h in 0.5% FBS, washed twice in ice-cold PBS, and incubated with EGF (Upstate Biotechnology) at 100 ng/ml in cold DMEM for 2 h at 4 °C. Cells were then rinsed twice with PBS, placed in pre-warmed DMEM, and returned to 37 °C to allow receptor internal-ization. At various time points, cells were placed on ice and washed twice with ice-cold PBS, and then cell surface proteins were labeled with freshly dissolved EZ-link Sulfo-NHS-LC-Biotin (Pierce) at 0.5 mg/ml in PBS for 30 min at room temperature. To quench the biotiny-lation reaction, cells were placed on ice and washed twice with cold PBS containing 0.2 mg/ml bovine serum albumin and twice with PBS. EGFR from lysed cells was immunoprecipitated (see below). Samples were resolved by SDS-PAGE in a 6% polyacrylamide gel, electrotransferred to nitrocellulose membrane, and overlaid with streptavidin-horseradish peroxidase at 1 "g/ml in TBST (Pierce). Biotinylated proteins were visualized by exposing blots to x-ray film (X-Omat; Eastman Kodak Co.) after treatment with Supersignal West Pico reagent (Pierce). Immunoprecipitations—Cells were washed in PBS and then lysed on ice in MTG (50 mM Tris, pH 8.0, 100 mM NaCl, 10% (v/v) glycerol, 1 (v/v) Nonidet P-40, and 2 mM sodium orthovanadate) containing protease and phosphatase inhibitor mixtures I and II (Sigma; used as per the manufacturer’s recommendations). Cell lysate was cleared by centrifu-gation, and protein concentrations were quantified by Bradford assay (Bio-Rad). EGFR from 150 "g of cell lysate was precipitated by over-night incubation with 1 "g of anti-EGFR at 4 °C. Signaling complexes from 500 "g of cell extract were precipitated by overnight incubation with 2 "g of anti-Grb2 at 4 °C. Immune complexes were bound to 25 "l of protein G-Sepharose (Amersham Biosciences) by co-incubation for 40 min at 4 °C, centrifuged, and washed three times with 1 ml of ice-cold MTG (EGFR) or PBS (Grb2). Immune complexes were boiled in SDS-PAGE sample buffer for 5 min and analyzed as a Western blot. Western Blot Analysis—Western blotting was conducted as described previously (29). Briefly, cells were lysed, and protein concentrations were quantified as described for immunoprecipitations. Lysates were boiled in SDS-PAGE sample buffer and loaded onto polyacrylamide gels at 30 "g/lane. After electrophoresis, proteins were transferred onto nitrocellulose, stained with Ponceau S, incubated with antibody as described above, and visualized by exposure to x-ray film (X-Omat; Kodak) after treatment with SuperSignal West Pico reagent or Super- Signal West Dura reagent (Pierce). Blots were stripped with the Re-Blot Western blot recycling kit (Chemicon International, Inc.) as per the manufacturer’s instructions. [3H]Thymidine Incorporation Assay—Cells were grown to 70% con-fluence, starved for 24 h in DMEM with 0.5% FBS, and then treated with various concentrations of TGF-! for 18 h at 37 °C. Ligand was removed, and cells were incubated in the presence of [3H]thymidine (1 "Ci/ml in DMEM) for 4 h at 37 °C. Cells were washed with cold PBS, incubated in 10% trichloroacetic acid at room temperature for 10 min, and washed twice with 5% trichloroacetic acid. DNA was precipitated with 100% ethanol and then solubilized by incubation in 0.2 N NaOH for 10 min at room temperature. Samples were neutralized with 0.4 N HCl and counted in a scintillation counter. EGF Proliferation Assay—Cells were plated in quadruplicate, grown to confluence, serum-starved for 18 h in DMEM containing 0.5% FBS, and transferred to growth medium (DMEM containing 0.5% FBS and 10 nM EGF). Three days later, growth medium was removed, and live cells were quantified with 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltet-razolium bromide (Sigma) as described previously (34). 125I-EGF Binding—The 125I-EGF binding analysis was conducted as described previously (35). Briefly, cells were grown to 70% confluence, serum-starved for 24 h, and incubated at 4 °C for 2 h with 175 pM 125I-EGF (NEN) and different amounts of unlabeled EGF (total EGF concentrations ranged from 175 pM to 10 nM) in binding buffer (DMEM plus 50 mM Hepes, pH 7.4, and 5% (w/v) bovine serum albumin). Cells were washed and then extracted in 0.1 N NaOH plus 0.1% (w/v) SDS, and bound 125I-EGF was quantified by gamma counting. RESULTS Herstatin Reduces EGF-stimulated Tyrosine Phosphoryla-tion and Down-regulation of the EGFR—Previous studies have shown that transient coexpression of herstatin with the EGFR in CHO cells results in diminished dimerization and tyrosine phosphorylation of the receptor in response to EGF (29). To further characterize the effects of herstatin on EGFR and to investigate its impact on EGF-induced intracellular signaling, stable cell lines that express different levels of herstatin were derived from NIH3T3 cells expressing human EGFR (termed EGFR3T3 cells). Efforts to stably express herstatin in several tumor cell lines that overexpress EGFR as well as HER-2 were thwarted, presumably because of an inhibitory effect on cell survival. EGFR3T3 cells were transfected with control or her-statin expression plasmid, and clonal populations were isolated by selection with hygromycin B. Varied levels of herstatin were expressed in herstatin-transfected cell lines but not in the control-transfected cells (Fig. 1A). To investigate the effects of herstatin on ligand-induced ac-tivation of the receptor, herstatin- and control-transfected EGFR3T3 cells were serum-starved for 20 h and treated with saturating concentrations (10 nM) of EGF for 20 min, and phosphotyrosine levels of the EGFR were assessed. Although clone 1 produced the greatest amount of herstatin, each of the three herstatin-expressing clones exhibited a similar reduction Downloaded from http://www.jbc.org/ at Harvard Libraries on December 1, 2014
  • 3. 20620 Herstatin Blocks EGF Activation of Akt and Growth in tyrosine phosphorylation of the EGFR (Fig. 1B; see also Figs. 4B and 6A), suggesting that maximal inhibition was achieved. The EGFR expression levels were comparable between paren-tal cells and the three different clones, showing that expression of herstatin did not down-regulate the EGFR (Fig. 1B). Because receptor tyrosine phosphorylation is required for endocytosis (36), we hypothesized that reduced EGFR auto-phosphorylation levels in the presence of herstatin alter EGF-mediated receptor down-regulation from the cell surface. Pa-rental and herstatin-expressing EGFR3T3 cells were saturated with EGF at 4 °C and then returned to 37 °C, a temperature permissive for internalization. After various incubation times, cell surface proteins were biotinylated, and EGFR was immu-noprecipitated and visualized by streptavidin-horseradish per-oxidase overlay. In the parental cell line, cell surface EGFR was reduced by 80% at 10 min and was undetectable by 20 min after the removal of the temperature block (Fig. 1C), in agree-ment with other studies (37, 38). In herstatin-expressing cells, however, cell surface EGFR was reduced only about 35% at 10 min, and about one-third of the EGFR remained at the cell surface after 20 min of incubation at 37 °C. Delayed down-regulation is consistent with previous findings showing that EGFR degradation, a process contingent upon endosomal loca-tion of the receptor, is blocked in CHO cells that transiently express herstatin (29). Herstatin Inhibits EGF-stimulated Akt Phosphorylation, but MAPK Is Fully Activated—Signal transduction through the ErbB family includes both the MAPK and the PI3K/Akt signal-ing pathways, which are generally stimulated simultaneously by growth factors (11–13). To examine the role of herstatin in EGF signaling downstream from the receptor, we treated EGFR3T3 parental and herstatin-expressing cells with satu-rating amounts of EGF (16 nM) and observed the kinetics of MAPK and Akt phosphorylation over a 1-h time course. In the parental cell line, the highest level of phospho-Akt was de-tected at 20 min after EGF addition (Fig. 2A). In the herstatin-expressing cells, however, little phospho-Akt was observed in EGF-treated cells, with maximal levels reaching only 2% of the parental controls. Phosphorylation of Akt was almost com-pletely abolished in the presence of herstatin; interestingly, there was no reduction of phospho-MAPK levels. In parental and herstatin-expressing cells, the time course and extent of MAPK activation were identical; maximal activation was achieved by 5 min and declined at 20 min after the addition of EGF (Fig. 2A). To determine whether induction of MAPK in herstatin-expressing cells was due to ectopic overexpression of EGFR (39–41), we investigated EGF signaling through endog-enous receptors in 3T3 cells that express herstatin. As in the EGFR3T3 cells, we observed preferential EGF activation of MAPK and abrogation of Akt phosphorylation in the presence of herstatin (Fig. 2B). These data demonstrate that the signal-ing profile caused by herstatin expression is not affected by ectopic overexpression of EGFR. EGF Induces the Formation of a Signaling Complex Contain-ing EGFR, phospho-Shc, and Grb2 in Herstatin-expressing Cells—After EGF treatment, the adaptor protein Shc binds the autophosphorylation domain of EGFR, is tyrosine-phospho-rylated, and recruits the Grb2-Sos complex from the cytoplasm to the plasma membrane (42). To investigate whether EGF stimulation of MAPK in the presence of herstatin was induced by EGFR through Shc and Grb2, we immunoprecipitated Grb2 and examined the immune complex by Western blotting. In both parental and herstatin-expressing cells, EGF-dependent association of EGFR and Shc with Grb2 was detected (Fig. 3). Furthermore, Shc was tyrosine-phosphorylated to a similar extent (Fig. 3), providing evidence for an active signaling com-plex. Because HER-2 is the preferred heterodimer partner of EGFR (4–6), endogenous HER-2 may be present in the EGFR-Shc- Grb2 signaling complex, contributing to EGF stimulation of the MAPK signaling cascade. However, HER-2 could not be detected in the signaling complex immunoprecipitated from 500 "g of either parental or herstatin-expressing cells (data not shown), even though the high titer antibody used (33) detects p185HER-2 in 20 "g of 3T3 cell extract. These data suggest that Shc and Grb2 transduce the herstatin-mediated EGF sig-nal from EGFR to components of the MAPK cascade, with no evidence of HER-2 involvement. Characterization of the Effects of EGF Concentration on In-tracellular Signaling in the Presence and Absence of Hersta-tin— Previous studies have shown that at very low EGF con-centrations, MAPK activation occurs in the absence of Akt activation and EGFR tyrosine phosphorylation (43). We there-fore examined whether signaling in EGFR3T3 cells exhibits a similar sensitivity to EGF concentration. Maximal stimulation of MAPK occurred independently of Akt activation in EGFR3T3 cells treated with 0.01 nM EGF (Fig. 4A), suggesting that ectopic overexpression of EGFR did not eliminate sensi-tivity to very low concentrations of EGF. At 0.1 nM EGF, a subsaturating concentration, maximal stimulation of Akt was observed in parental cells, but stimulation of Akt was inhibited in the herstatin-expressing cells. Therefore, preferential inhi-bition of the Akt pathway by herstatin could reflect a quanti-tative reduction in effective EGF concentration by competitive FIG. 1. Herstatin expression, EGFR tyrosine phosphorylation, and receptor down-regulation in parental and herstatin-trans-fected EGFR3T3 cells. A, 30 "g of lysate from parental and herstatin-expressing EGFR3T3 cells was separated by 7.5% SDS-PAGE and subjected to Western blot analysis for herstatin as described under “Experimental Procedures.” B, herstatin- and mock-transfected EGFR3T3 cells were serum-starved for 20 h and then incubated with 10 nM EGF for 20 min at 37 °C. Cell extracts were resolved by 6% SDS-PAGE and analyzed as a Western blot using anti-phosphotyrosine antibody. The blot was stripped as described under “Experimental Procedures” and reprobed with anti-EGFR. Results are representative of three independent experiments. C, herstatin (clone 1)- and mock-transfected EGFR3T3 cells were serum-starved as described above, incubated with 16 nM EGF at 4 °C for 2 h, and incubated at 37 °C for the durations indicated. Cell surface proteins were biotinylated as de-scribed under “Experimental Procedures.” EGFR was immunoprecipi-tated from 150 "g of lysate, and immune complexes were separated by 6% SDS-PAGE, transferred to nitrocellulose, and then overlaid with streptavidin-horseradish peroxidase. Films were scanned by imaging densitometry (BioRad, model GS700) to quantitate streptavidin-horse-radish peroxidase signal. Similar results were observed in two inde-pendent experiments. Downloaded from http://www.jbc.org/ at Harvard Libraries on December 1, 2014
  • 4. Herstatin Blocks EGF Activation of Akt and Growth 20621 FIG. 2. EGF-induced signaling in parental and herstatin-expressing EGFR3T3 cells. A, herstatin (clone 1)- and mock-transfected EGFR3T3 cells were serum-starved and incubated with 16 nM EGF at 37 °C for the durations indicated. Cell lysates were separated by 7.5% and 10% SDS-PAGE and then subjected to Western blot analysis using antibodies specific for phospho-Akt (phospho-Ser473) and phospho-p42/p44 MAPK (phospho-T202 and Y204). Exposed films were scanned by imaging densitometry to quantitate phospho-Akt signal. Blots were stripped and probed with Akt antibody. Results are representative of six independent experiments. B, herstatin- and mock-transfected 3T3 cells were serum-starved and incubated with 10 nM EGF at 37 °C for the durations indicated. Cell lysates were separated by 7.5% and 10% SDS-PAGE and then subjected to Western blot analysis as described above. inhibition. However, after treatment with either 1 or 100 nM EGF (#100 times KD), EGFR phosphotyrosine levels were di-minished !10-fold, and Akt activation was reduced to the same extent (Fig. 4B). These data suggest that herstatin has a qual-itative impact on intracellular signaling that is independent of EGF concentration. Herstatin Expression Does Not Alter the Binding Affinity of EGF for EGR3T3 Cells—Previous studies demonstrated that herstatin binds to the extracellular domain of the HER-2 re-ceptor (32) and forms a stable complex with EGFR (29). Inhi-bition of EGFR by herstatin could be caused by interference with EGF binding. To examine this possibility, we character-ized the binding affinity of EGF to parental and herstatin-expressing clones of EGFR3T3 cells. The cells were incubated with subsaturating amounts (175 pM) of 125I-EGF, and its dis-placement by unlabeled EGF was measured (Fig. 5). The dis-placement curve exhibited by the parental cells was indistin-guishable from that of cell lines that expressed either high (clone 1) or low (clones 2 and 3) levels of herstatin (see Fig. 1A). Moreover, Scatchard analysis of the binding data, as described in the legend to Fig. 6, revealed the same apparent dissociation constant of !500 pM in the absence and presence of different levels of herstatin. These studies show that herstatin expres-sion does not prevent EGF binding or alter the EGF binding affinity, suggesting that herstatin is not a competitive inhibitor of EGF-mediated EGFR activation. Herstatin therefore modu-lates signaling of receptors that are complexed with EGF. Herstatin Inhibits TGF-!-induced Receptor Phosphorylation and Akt Phosphorylation, Whereas MAPK Activation Is Unaf-fected— TGF-! is an EGFR ligand that has increased mitogenic and transforming potency compared with EGF (44). Although these ligands compete for receptor binding, they exhibit subtly different binding properties to the EGFR (45) and show distinct co-receptor-dependent signal potentiation (37). Despite these differences, parental and herstatin-expressing EGFR3T3 cells treated with TGF-! displayed signaling profiles similar to those observed in response to EGF stimulation. Depression of EGFR tyrosine phosphorylation occurred in the herstatin-ex-pressing cells, particularly at high (100 nM) concentrations of TGF-! (Fig. 6A). Moreover, phospho-Akt levels were markedly decreased, whereas MAPK activation was unaffected at both low (1 nM) and high (100 nM) concentrations of TGF-! (Fig. 6A). Herstatin Expression Does Not Alter FGF-2 Stimulation of Akt— The strong suppression of EGF- and TGF-!-induced Akt phospho-rylation in the EGFR3T3 cells that stably express herstatin could be an indirect effect of herstatin expression or chronic ErbB recep-tor inhibition. To examine the integrity of the PI3K/Akt pathway, we monitored phospho-Akt levels after treatment with FGF-2, a growth factor that activates a heterologous receptor tyrosine ki-nase. Parental and herstatin-expressing EGFR3T3 cells were se-rum- starved, exactly as done before EGF treatment, and then cells were incubated with saturating amounts of FGF-2 (10 nM) for 20 min. FGF-2 treatment stimulated the tyrosine phosphorylation of an 119-kDa protein, the approximate size of the FGF receptor (Fig. 6B). Furthermore, FGF-2 increased phospho-Akt levels to an equiv-alent extent in the presence and absence of herstatin (Fig. 6B). These data demonstrate the functional integrity of the PI3K/Akt pathway and suggest that the reduction of phospho-Akt levels caused by herstatin expression is specific to EGF- and TGF-!- induced signaling. FIG. 3. EGF induces EGFR-Shc-Grb2 complex formation in herstatin-expressing EGFR3T3 cells. Herstatin (clone 1)- and mock-transfected EGFR3T3 cells were serum-starved and incubated with 10 nM EGF at 37 °C for 20 min. Proteins complexed to anti-Grb2 were immunoprecipitated from 500 "g of cell lysate, separated by 8% and 12% SDS-PAGE, and then subjected to Western blot analysis using the antibodies indicated. The anti-phosphotyrosine blot was stripped and reprobed with anti-Shc antibody. Results are representative of two independent experiments. Downloaded from http://www.jbc.org/ at Harvard Libraries on December 1, 2014
  • 5. 20622 Herstatin Blocks EGF Activation of Akt and Growth Herstatin Uncouples MAPK from Akt Activation in Tran-siently Transfected CHO Cells—To evaluate whether the un-coupling of phospho-Akt from MAPK activation by herstatin was a feature confined to the 3T3 cell background, we exam-ined the EGF signaling profile in CHO cells, which do not express endogenous EGFR (25). The cells were transiently transfected with EGFR and different levels of herstatin expres-sion plasmids and then treated with EGF. Fig. 6C illustrates that EGF induction of MAPK phosphorylation were unaffected by expression of different amounts of herstatin. In contrast, Akt activation was inhibited in proportion to herstatin expres-sion levels. Herstatin Depresses Mitogenic Stimulation by EGF and TGF-!—To test whether herstatin affected the proliferative func-tions of the EGFR growth factors, TGF-!-induced mitogenesis was assessed by measuring DNA synthesis. Cells were first forced into quiescence by serum starvation for 40 h and then treated with different concentrations of TGF-!. The ligand was removed, and cells were incubated with [3H]thymidine to quan-tify DNA synthesis. In the presence of herstatin, we observed a striking decrease in TGF-!-induced [3H]thymidine incorporation that was not overcome at high ligand concentrations (Fig. 7A). The impact of herstatin on EGF-mediated cell proliferation was examined by quantitation of live cells by the 3-(4,5-dimethylthia-zol- 2-yl)-2,5-diphenyltetrazolium bromide assay (34). Equal numbers of EGFR3T3 control and herstatin-transfected cells were plated, serum-starved, and then treated for 72 h with ve-hicle or EGF. Herstatin expression resulted in a significant re-duction in viable cells in the absence of EGF (p $ 0.007; Fig. 7B), which may reflect diminished survival under conditions of serum deprivation. Whereas EGF treatment significantly increased the control EGFR3T3 cells (p $ 0.006; Fig. 7B), cells expressing herstatin displayed no significant proliferation in response to EGF treatment. These results demonstrate that herstatin inter-rupts TGF-!- and EGF-mediated mitogenic signal transduction, resulting in inhibition of proliferation. DISCUSSION Investigating the mechanisms employed by ErbB inhibitors and their impact on signaling is critical to understanding the biology of these receptors and to developing anti-receptor tyro-sine kinase therapeutics. Whereas several inhibitors of the EGFR have been investigated, herstatin is distinguished by its novel structure, consisting of subdomains I and II of the HER-2 ECD and an intron encoded-C-terminal domain (32). Further-more, herstatin is the only naturally occurring inhibitor of the EGFR in mammalian cells that exerts its action on the initial events in receptor activation: dimerization and autophospho-rylation (29, 32). In this study, we show that herstatin selec-tively modulates the intracellular signaling pathways stimu-lated by EGFR ligands. EGF binds to its receptor with normal affinity in the presence of herstatin, yet receptor tyrosine phos-phorylation and down-regulation are suppressed. Whereas her-statin allows full ligand stimulation of the MAPK pathway and its upstream effector, Shc, Akt phosphorylation is selectively blocked, resulting in suppression of cell growth. Herstatin is a secreted protein that binds to and inhibits the EGFR (29). Although the binding site has not been mapped, previous observations suggest that herstatin associates with the ECD of ErbB receptors (32). We therefore examined whether herstatin may interfere with binding of EGF. Our results demonstrate that neither the binding affinity nor the overall number of EGF binding sites was significantly altered in cells that expressed either low or very high levels of hersta-tin. Therefore, herstatin appears to inhibit EGFR that is occu-pied by growth factor. Two other classes of EGFR inhibitors compete with ligand binding, including the Drosophila ligand FIG. 4. Effects of EGF concentra-tion on signaling in parental and her-statin- expressing EGFR3T3 cells. A, herstatin (clone 1)- and mock-transfected EGFR3T3 cells were serum-starved, treated with 0, 0.01, and 10 nM EGF at 37 °C for 20 min, and analyzed by West-ern blot as described in the Fig. 2 legend. Staining with Ponceau S confirms equal loading. B, herstatin (clone 1)- and mock-transfected EGFR3T3 cells were treated with l or 100 nM EGF as described in A. Exposed films were scanned by imaging densitometry to quantitate phosphoty-rosine signal. Results are representative of two independent experiments. FIG. 5. Displacement of 125I-EGF by unlabeled EGF in parental and herstatin-expressing EGFR3T3 cells. Herstatin and mock-transfected EGFR3T3 cells were plated at 5 % 104 cells/well in 24-well plates and serum-starved. Cells were incubated for 2 h at 4 °C with 175 pM 125I-EGF and unlabeled EGF at total concentrations ranging from 175 pM to 10 nM. Displacement of bound 125I-EGF by unlabeled EGF was plotted as the maximum percentage bound 125I-EGF. The dissoci-ation constants (KD) of EGF for herstatin- and mock-transfected EGFR3T3 clones were estimated by Scatchard analysis: bound/free versus bound was plotted using KaleidaGraph software (Synergy Anal-ysis, 1997); the slopes of regression lines as calculated by the program provided the estimate of &1/KD. All regression line R2 values exceeded 0.99. Downloaded from http://www.jbc.org/ at Harvard Libraries on December 1, 2014
  • 6. Herstatin Blocks EGF Activation of Akt and Growth 20623 FIG. 6. Herstatin effects on signaling induced by EGF, TGF-!, and FGF-2. A, herstatin (clone 1)- and mock-transfected EGFR3T3 cells were serum-starved and treated with l or 100 nM TGF-! at 37 °C for 20 min and analyzed as described in Fig. 4B. Results are representative of two independent experiments. B, herstatin (clone 1)- and mock-transfected EGFR3T3 cells were serum-starved and treated with 100 nM FGF-2 at 37 °C for 20 min, and cell lysate was subjected to Western blot analysis using antibodies against phosphotyrosine and phospho-Akt. The blot was stripped and incubated with Akt antibody. C, CHO cells were grown to 80% confluence and transfected with 1.5 "g of EGFR and 0.5 or 1.5 "g of herstatin expression plasmids. 20 h after transfection, cells were serum-starved, treated with 10 nM EGF for 20 min at 37 °C, and analyzed for phospho-Akt and phospho-p42/p44 MAPK by Western blot. Equal loading was confirmed by Ponceau S staining. Argos (46, 47) and the EGFR monoclonal antibody C225 (48). Because herstatin inhibits ligand-occupied receptor, it may be effective even when growth factors are overproduced by tumors (49). In this study, we demonstrate that herstatin uncouples in-tracellular signaling pathways triggered by EGF and TGF-!. Whereas EGFR tyrosine phosphorylation and Akt activation are inhibited, Shc and MAPK are fully activated. This is in contrast to the intracellular signaling effects of several other inhibitors of EGFR including the quinazoline inhibitors (12), the p185neu dominant negative mutant (15), and the C225 monoclonal antibody inhibitor (14), which suppress EGF-in-duced phosphorylation of both MAPK and Akt. Similar to her-statin, these inhibitors reduce EGFR tyrosine phosphorylation and cause prolonged retention of the EGFR at the cell surface. In agreement with previous studies (43), we also observed uncoupling of MAPK and Akt signaling cascades at very low concentrations of EGF (0.01 nM), suggesting that either limit-ing amounts of ligand or the presence of herstatin preferen-tially stimulated the MAPK cascade. The signaling effects of herstatin can not be explained by a reduction in the effective concentration of growth factor; the EGF binding affinity was unaffected by herstatin, and selective suppression of Akt sig-naling was observed at both subsaturating (0.1 nM) and very high ligand concentrations (100 nM). Because phosphorylation of specific tyrosine residues on ac-tivated receptors is responsible for the recruitment and activa-tion of distinct intracellular signaling molecules (51), inhibition of some, but not all, EGFR phosphorylation sites by herstatin may cause preferential recruitment of effector molecules (52). Evaluation of this possibility will require phosphopeptide map-ping of the EGFR from the herstatin-expressing cells. However, even in the absence of EGFR tyrosine phosphorylation, either by kinase-impaired EGFR (39–41) or EGFR missing its tyro-sine phosphorylation sites (53), MAPK is activated, whereas Akt is not stimulated by EGF (16, 54). In these cases (53, 55), as well as with herstatin, EGFR appears to be involved in activation of the MAPK cascade, as shown by its presence in a signaling complex containing tyrosine phosphorylated Shc and Grb2. An endogenous receptor tyrosine kinase or a cytoplasmic kinase such as src (56) may also participate in MAPK activa-tion by EGF. Endogenous HER-2, the preferred EGFR hetero-meric partner, is unlikely to be responsible for MAPK activa-tion because herstatin effectively disrupts HER-2 homomeric FIG. 7. TGF-!-induced DNA synthesis and EGF-induced prolif-eration of parental and herstatin-expressing EGFR3T3 cells. A, herstatin (clone 1)- and mock-transfected EGFR3T3 cells were plated at 5 % 104 in triplicate, serum-starved, and treated with TGF-! at the concentrations indicated. At 18 h, cells were incubated with [3H]thymi-dine as described under “Experimental Procedures.” Levels of [3H]thy-midine incorporation are expressed as a percentage of the untreated control. Error bars indicate sample mean ' S.D. Results are represent-ative of three independent experiments. B, herstatin (clone 1)- and mock-transfected EGFR3T3 clones were grown to 90% confluence, se-rum- starved, and treated with 10 nM EGF. At 72 h, live cells were quantified by 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bro-mide assay as described under “Experimental Procedures.” Error bars indicate sample mean ' S.D. Results are representative of two inde-pendent experiments. Downloaded from http://www.jbc.org/ at Harvard Libraries on December 1, 2014
  • 7. 20624 Herstatin Blocks EGF Activation of Akt and Growth and heteromeric interactions (29). Although we were unable to detect HER-2 in the EGF-induced signaling complex, other endogenous tyrosine kinases may be involved in MAPK activa-tion in herstatin-expressing cells. A major question in signaling through receptor tyrosine ki-nases concerns the mechanism by which growth factors can stimulate diverse cellular responses (52, 57, 58). For the ErbB receptors, one level of diversity is achieved through generation of different ErbB dimer pairs that have been found to differen-tially stimulate intracellular signaling pathways (10, 51, 52, 57). Alternatively, signaling by the same dimer pair may be altered in response to activation by different growth factors (8, 22, 52). Additionally, EGF ligands have been shown to promote diverse cellular responses, depending on the cell type (59). The results presented here point to a novel mechanism of generat-ing diversity by which a single ligand, EGF, can achieve altered signal output within a given cell context. Our studies suggest that herstatin, a HER-2 receptor variant expressed in fetal kidney and liver cells, can selectively alter EGF signal output, resulting in growth arrest. The Ras/MAPK pathway was previously suggested to be the major mitogenic signaling pathway initiated by the EGFR (60). In the presence of herstatin, full stimulation of MAPK by TGF-! was insufficient to drive entry into S phase, as sug-gested by the suppression of DNA synthesis. Moreover, EGF stimulation of MAPK was insufficient to stimulate cell prolif-eration in the presence of herstatin. Because Akt activation was strongly inhibited, components of the PI3K signaling path-way that are required for growth factor-induced proliferation may not be activated in the presence of herstatin. This finding is in agreement with recent observations that interruption of the MAPK pathway by EGFR blockade with quinazoline inhib-itors is not the cause of G1 arrest, but rather interruption of PI3K function is required (12). Signaling through the EGF receptor is often enhanced in hu-man cancers through overexpression of the receptor and auto-crine stimulation by ligands produced by the tumor (49). En-hanced EGFR signaling in several carcinomas is directly coupled to inappropriate phospho-Akt survival signals, rendering many cancers resistant to apoptotic signals, including those activated by radiation and chemotherapies (12, 19). EGF stimulation of Akt kinase activity has been proposed as a major mechanism behind enhanced survival conferred by inappropriate EGF sig-naling. The activation of Akt appears to be both required and sufficient for the antiapoptotic function of EGF (19). Results presented here point to the effectiveness of herstatin in blockage of Akt activation and inhibition of proliferation stimulated by EGF. Previous studies also demonstrate the effectiveness of her-statin in blocking proliferation stimulated by HER-2 overexpres-sion, which often occurs in the same tumor cells that have en-hanced EGFR signaling. The results presented here further support the potential utility of herstatin in the development of therapeutics against cancers with ErbB receptor involvement. Acknowledgments—We thank L. S. Shamieh for helpful discussions and critical reading of the manuscript, M. E. Sommer for generating the EGFR3T3/herstatin cell lines, and M. C. Denton for constructing the herstatin and EGFR expression plasmids. REFERENCES 1. Hanks, S. K., Quinn, A. M., and Hunter, T. (1988) Science 241, 42–52 2. Schlessinger, J., Ullrich, A., Honegger, A. M., and Moolenaar, W. H. (1988) Cold Spring Harbor Symp. Quant. Biol. 53, 515–519 3. Yarden, Y., and Ullrich, A. (1988) Annu. Rev. Biochem. 57, 443–478 4. Dougall, W. C., Qian, X., Peterson, N. C., Miller, M. J., Samanta, A., and Greene, M. I. (1994) Oncogene 9, 2109–2123 5. Hynes, N. E., and Stern, D. F. (1994) Biochim. Biophys. Acta 1198, 165–184 6. Tzahar, E., and Yarden, Y. (1998) Biochim. Biophys. Acta 1377, M25&M37 7. Heldin, C. H., and Ostman, A. (1996) Cytokine Growth Factor Rev. 7, 3–10 8. Riese, D. J., II, and Stern, D. F. (1998) Bioessays 20, 41–48 9. Burden, S., and Yarden, Y. (1997) Neuron 18, 847–855 10. Olayioye, M. A., Neve, R. M., Lane, H. A., and Hynes, N. E. (2000) EMBO J. 19, 3159–3167 11. Blume-Jensen, P., and Hunter, T. (2001) Nature 411, 355–365 12. Busse, D., Doughty, R. S., Ramsey, T. T., Russell, W. E., Price, J. O., Flanagan, W. M., Shawver, L. K., and Arteaga, C. L. (2000) J. Biol. Chem. 275, 6987–6995 13. Rommel, C., Clarke, B. A., Zimmermann, S., Nunez, L., Rossman, R., Reid, K., Moelling, K., Yancopoulos, G. D., and Glass, D. J. (1999) Science 286, 1738–1741 14. Liu, B., Fang, M., Lu, Y., Mendelsohn, J., and Fan, Z. (2001) Oncogene 20, 1913–1922 15. Qian, X., O’Rourke, D. M., Fei, Z., Zhang, H. T., Kao, C. C., and Greene, M. I. (1999) J. Biol. Chem. 274, 574–583 16. Walker, F., Kato, A., Gonez, L. J., Hibbs, M. L., Pouliot, N., Levitzki, A., and Burgess, A. W. (1998) Mol. Cell. Biol. 18, 7192–7204 17. Jones, S. M., and Kazlauskas, A. (2001) Nat. Cell. Biol. 3, 165–172 18. Zhou, B. P., Liao, Y., Xia, W., Spohn, B., Lee, M. H., and Hung, M. C. (2001) Nat. Cell. Biol. 3, 245–252 19. Gibson, S., Tu, S., Oyer, R., Anderson, S. M., and Johnson, G. L. (1999) J. Biol. Chem. 274, 17612–17618 20. Lemmon, M. A., Bu, Z., Ladbury, J. E., Zhou, M., Pinchasi, D., Lax, I., Engelman, D. M., and Schlessinger, J. (1997) EMBO J. 16, 281–294 21. O’Rourke, D. M., Qian, X., Zhang, H. T., Davis, J. G., Nute, E., Meinkoth, J., and Greene, M. I. (1997) Proc. Natl. Acad. Sci. U. S. A. 94, 3250–3255 22. Tzahar, E., Pinkas-Kramarski, R., Moyer, J. D., Klapper, L. N., Alroy, I., Levkowitz, G., Shelly, M., Henis, S., Eisenstein, M., Ratzkin, B. J., Sela, M., Andrews, G. C., and Yarden, Y. (1997) EMBO J. 16, 4938–4950 23. Tanner, K. G., and Kyte, J. (1999) J. Biol. Chem. 274, 35985–35990 24. Graus-Porta, D., Beerli, R. R., Daly, J. M., and Hynes, N. E. (1997) EMBO J. 16, 1647–1655 25. Tzahar, E., Waterman, H., Chen, X., Levkowitz, G., Karunagaran, D., Lavi, S., Ratzkin, B. J., and Yarden, Y. (1996) Mol. Cell. Biol. 16, 5276–5287 26. Qian, X., O’Rourke, D. M., Zhao, H., and Greene, M. I. (1996) Oncogene 13, 2149–2157 27. Kumagai, T., Davis, J. G., Horie, T., O’Rourke, D. M., and Greene, M. I. (2001) Proc. Natl. Acad. Sci. U. S. A. 98, 5526–5531 28. O’Rourke, D. M., Nute, E. J., Davis, J. G., Wu, C., Lee, A., Murali, R., Zhang, H. T., Qian, X., Kao, C. C., and Greene, M. I. (1998) Oncogene 16, 1197–1207 29. Azios, N. G., Romero, F. J., Denton, M. C., Doherty, J. K., and Clinton, G. M. (2001) Oncogene 20, 5199–5209L. J. 30. Maihle, N. J., Baron, A. T., Barrette, B. A., Boardman, C. H., Christensen, T. A., Cora, E. M., Faupel-Badger, J. M., Greenwood, T., Juneja, S. C., Lafky, J. M., Lee, H., Reiter, J. L., and Podratz, K. C. (2002) Cancer Treatment Res. 107, 247–258 31. Lee, H., Akita, R. W., Sliwkowski, M. X., and Maihle, N. J. (2001) Cancer Res. 61, 4467–4473 32. Doherty, J. K., Bond, C., Jardim, A., Adelman, J. P., and Clinton, G. M. (1999) Proc. Natl. Acad. Sci. U. S. A. 96, 10869–10874 33. Christianson, T. A., Doherty, J. K., Lin, Y. J., Ramsey, E. E., Holmes, R., Keenan, E. J., and Clinton, G. M. (1998) Cancer Res. 58, 5123–5129 34. Hansen, M. B., Nielsen, S. E., and Berg, K. (1989) J. Immunol. Methods 119, 203–210 35. Sliwkowski, M. X., Schaefer, G., Akita, R. W., Lofgren, J. A., Fitzpatrick, V. D., Nuijens, A., Fendly, B. M., Cerione, R. A., Vandlen, R. L., and Carraway, K. L., III. (1994) J. Biol. Chem. 269, 14661–14665 36. Cadena, D. L., Chan, C. L., and Gill, G. N. (1994) J. Biol. Chem. 269, 260–265 37. Lenferink, A. E., Pinkas-Kramarski, R., van de Poll, M. L., van Vugt, M. J., Klapper, L. N., Tzahar, E., Waterman, H., Sela, M., van Zoelen, E. J., and Yarden, Y. (1998) EMBO J. 17, 3385–3397 38. Vieira, A. V., Lamaze, C., and Schmid, S. L. (1996) Science 274, 2086–2089 39. Campos-Gonzalez, R., and Glenney, J. R., Jr. (1992) J. Biol. Chem. 267, 14535–14538 40. Hack, N., Sue, A. Q. A., Mills, G. B., and Skorecki, K. L. (1993) J. Biol. Chem. 268, 26441–26446 41. Selva, E., Raden, D. L., and Davis, R. J. (1993) J. Biol. Chem. 268, 2250–2254 42. Ravichandran, K. S. (2001) Oncogene 20, 6322–6330 43. Wennstrom, S., and Downward, J. (1999) Mol. Cell. Biol. 19, 4279–4288 44. Salomon, D. S., Brandt, R., Ciardiello, F., and Normanno, N. (1995) Crit. Rev. Oncol. Hematol. 19, 183–232 45. Winkler, M. E., O’Connor, L., Winget, M., and Fendly, B. (1989) Biochemistry 28, 6373–6378 46. Vinos, J., and Freeman, M. (2000) Oncogene 19, 3560–3562 47. Zhao, D., and Bownes, M. (1999) Dev. Genet. 25, 375–386 48. Mendelsohn, J. (1997) Clin. Cancer Res. 3, 2703–2707 49. Klapper, L. N., Kirschbaum, M. H., Sela, M., and Yarden, Y. (2000) Adv. Cancer Res. 77, 25–79 50. Deleted in proof 51. Alroy, I., and Yarden, Y. (1997) FEBS Lett. 410, 83–86 52. Sweeney, C., and Carraway, K. L., III. (2000) Oncogene 19, 5568–5573 53. Gotoh, N., Tojo, A., Muroya, K., Hashimoto, Y., Hattori, S., Nakamura, S., Takenawa, T., Yazaki, Y., and Shibuya, M. (1994) Proc. Natl. Acad. Sci. U. S. A. 91, 167–171 54. Deb, T. B., Su, L., Wong, L., Bonvini, E., Wells, A., David, M., and Johnson, G. R. (2001) J. Biol. Chem. 276, 15554–15560 55. Walker, F., Hibbs, M. L., Zhang, H. H., Gonez, L. J., and Burgess, A. W. (1998) Growth Factors 16, 53–67 56. Olayioye, M. A., Badache, A., Daly, J. M., and Hynes, N. E. (2001) Exp. Cell Res. 267, 81–87 57. Carraway, K. L., III, and Cantley, L. C. (1994) Cell 78, 5–8 58. Hunter, T. (2000) Cell 100, 113–127 59. Yarden, Y., and Sliwkowski, M. X. (2001) Nat. Rev. Mol. Cell. Biol. 2, 127–137 60. Malumbres, M., and Pellicer, A. (1998) Front. Biosci. 3, 887–912 Downloaded from http://www.jbc.org/ at Harvard Libraries on December 1, 2014