You are on page 1of 14

THE JOURNAL OF BIOLOGICAL CHEMISTRY Vol. 278, No. 46, Issue of November 14, pp.

45255–45268, 2003
© 2003 by The American Society for Biochemistry and Molecular Biology, Inc. Printed in U.S.A.

Regulated Cell Surface Pro-EGF Ectodomain Shedding Is a


Zinc Metalloprotease-dependent Process*
Received for publication, July 17, 2003, and in revised form, August 21, 2003
Published, JBC Papers in Press, August 28, 2003, DOI 10.1074/jbc.M307745200

Sylvain M. Le Gall‡, Rodolphe Auger, Catherine Dreux, and Philippe Mauduit§


From the UMR 8619, Institut de Biochimie Biophysique Moléculaire et Cellulaire, Université Paris XI,
91405 Orsay Cedex, France

Epidermal growth factor receptor (EGFR) ligands are submaxillary glands by Cohen (1) based on its ability to stim-
synthesized as type I membrane protein precursors ex- ulate epithelial cell growth. This growth factor is now known to
posed at the cell surface. Shedding of the ectodomain of be part of a family of growth factors that includes transforming
these proteins is the way cells regulate the equilibrium growth factor-␣ (TGF-␣) (2), heparin binding EGF-like growth
between cell-associated and diffusible forms of these factor (HB-EGF) (3), amphiregulin (AR) (4), betacellulin (5),
growth factors. Whereas the regulated shedding of and epiregulin (6). These growth factors share the ability to
transforming growth factor-␣, HB-EGF, and amphiregu- bind the same receptor, i.e. the EGF receptor (EGFR) and
lin precursors have been clearly established, regulation activate its intrinsic tyrosine kinase activity that induces cel-
of full-length pro-EGF shedding has not been clearly lular differentiation and mitogenic effects (7, 8). Most growth
demonstrated. Here, using both wild-type and M2 mu- factors are produced as soluble inactive precursors that are
tant CHO-K1 as well as HeLa cell lines transiently trans-
activated by proteolysis in vesicular compartments of the se-
fected with epitope-tagged rat pro-EGF expression plas-
cretory pathway before being released in the extracellular me-
mid, we demonstrate that these cells synthesize EGF as
dium. Both cDNA and protein structure analysis clearly indi-
a high molecular weight membrane-associated precur-
sor glycoprotein expressed at the cell surface. All cell cate that EGFR ligands are also derived from precursor
lines are able to release the entire ectodomain of pro- molecules (9). However, they are not synthesized as soluble but
EGF in the extracellular medium following juxtamem- as nondiffusible, glycosylated membrane-associated precursors
brane cleavage of the precursor once it is present at the exposed at the cell surface.
cell surface. More significantly we clearly established In the case of EGF, the cDNA analysis from human, mouse,
that CHO-M2 and HeLa cells only constitutively release and rat species predicted very large precursor molecules (re-
low levels of pro-EGF. This shedding is a regulated phe- spectively 1207, 1217, and 1133 amino acids) (10 –13) with an
nomenon in wild-type CHO cells where it can be induced apparent molecular mass ranging from 150 to 185 kDa for the
by different agents such as phorbol 12-myristate 13-ac- mature glycosylated precursors. These precursor molecules are
etate (PMA), pervanadate, and serum but not by calcium predicted to be type I membrane proteins. In the case of rat
ionophores. Using specific inhibitors as well as protein pro-EGF, the 1035 N-terminal amino acids constitute the ex-
kinase C (PKC) depletion, PMA stimulation was shown tracellular domain that includes the EGF sequence in the jux-
to be completely dependent on PKC activation whereas tamembrane region and eight additional “EGF-like” modules, a
pervanadate and serum stimulation were not. Regulated 22-amino acid transmembrane domain, and a short C-terminal
ectodomain shedding involves the activity of a zinc met- intracellular part of 76 amino acids (13).
alloprotease as determined by inhibition with phenan- The biological importance of EGFR ligands underscores the
trolin and TAPI-2 and by the results obtained with the interest in the study of the enzymatic processing leading to
CHO-M2 shedding defective mutant cell line. Compari-
growth factor bioavailability (14, 15). The presence of EGF in
son of the ability of CHO and HeLa cell lines to shed
external biological fluids such as saliva, urine, milk, and tears
pro-EGF and pro-TNF-␣ upon stimulation greatly sug-
(16 –22) indicates that EGF secretion is mainly an exocrine
gests that TACE (ADAM 17) may not be the ectoprotease
involved in the secretion of pro-EGF ectodomain and secretory process. However, the cellular events leading from
that this protease, which remains to be identified, shows the high molecular weight precursor molecule to secretion of
a restricted cellular expression pattern. lower molecular weight soluble EGF into the external fluid are
currently not clearly identified.
Recent evidence suggests that proteolytic cleavage of cell
Epidermal growth factor (EGF)1 is a small (about 6 kDa) surface proteins, i.e. ectodomain shedding, is an important
diffusible polypeptide first discovered and purified from rodent mechanism whereby cells regulate the repertoire of proteins
expressed on their surface, shifting molecular species from
their membrane-bound forms to soluble ones with the potential
* The costs of publication of this article were defrayed in part by the modification of their biological properties (9, 23, 24). Several
payment of page charges. This article must therefore be hereby marked types of membrane proteins such as receptor ligands (among
“advertisement” in accordance with 18 U.S.C. Section 1734 solely to them EGFR ligands), receptors, cell-adhesion molecules, and
indicate this fact.
‡ Supported by a Doctoral contract from the Ministère de la jeunesse, ectoenzymes undergo ectodomain shedding (9, 23–26). With the
de l’éducation et de la recherche. important exception of the ␤-secretase activity (an aspartyl-
§ To whom correspondence should be addressed. Tel.: 33-1-69158033; protease) that sheds the prodomain of the ␤-amyloid precursor
Fax: 33-1-69853715; E-mail: philippe.mauduit@bbmpc.u-psud.fr.
1
protein (␤-APP) from the cell surface, cell surface proteolysis
The abbreviations used are: EGF, epidermal growth factor; EGFR,
EGF receptor; DMEM, Dulbecco’s modified Eagle’s medium; FCS, fetal
calf serum; PBS, phosphate-buffered saline; BSA, bovine serum albu- phorbol 12-myristate 13-acetate; CHO, chinese hamster ovary; PKC,
min; HA, hemagglutinin; RIA, radioimmunoassay; WT, wild type; PMA, protein kinase C; TGF-␣, transforming growth factor-␣.

This paper is available on line at http://www.jbc.org 45255

This is an Open Access article under the CC BY license.


45256 Metalloprotease-dependent Regulated Pro-EGF Ectodomain Shedding
appears to be mediated by members of the metzincin super- pro-EGF shedding compared with other EGFR ligands. In or-
family of zinc-dependent proteases that include the matrix der to provide insights into pro-EGF shedding properties, we
metalloproteinases (MMPs) and ADAMs (for A Disintegrin And decided to study pro-EGF secretion in CHO-K1 (both wild type
Metalloproteinase) (24, 26 –28). The involvement of proteases and M2 mutant) and HeLa cell lines transiently transfected
such as ADAM 9 (MDC9/meltrin-␥) and more importantly with a full-length rat pro-EGF cDNA. The CHO-K1 cellular
ADAM 17 (TACE) in the regulation of EGFR ligand shedding model has been used previously to establish the mechanism of
as been proposed for TGF-␣ (24, 25, 28) HB-EGF (29, 30), and regulation of the secretion of other EGFR ligands (TGF-␣,
amphiregulin (29, 31). Unfortunately, the information concern- HB-EGF), and HeLa cells have also been used to study the
ing pro-EGF maturation and secretion does appear much shedding of various proteins including HB-EGF (54, 55). In the
more controversial. present study, we have been able to demonstrate that once
In rodents submaxillary glands, the precursor molecule is pro-EGF is exposed at the cell surface, it can be shed to release
not detectable and only the mature and diffusible 6 kDa form of its entire ectodomain in the extracellular medium. In CHO-K1,
EGF accumulates in the secretory granules of the regulated this ectodomain shedding can be stimulated by PMA, serum, or
exocrine secretory pathway (12, 32, 33). In this case, the release pervanadate. PMA acts through a PKC-dependent pathway
of EGF in the biological fluid (saliva) involves exocytosis, i.e. whereas serum and pervanadate act through PKC-independ-
fusion of the secretory granule membrane with the apical cel- ent pathway(s). The use of specific protease inhibitors as well
lular membrane. This phenomenon is highly regulated and as of the CHO-M2 mutant cell line that displays impaired
may be achieved at least through the activation of adrenergic constitutive and regulated pro-EGF ectodomain shedding dem-
receptors (34, 35). onstrated that shedding was achieved through the activation of
In other tissues studied so far such as kidney (12, 36 –39), a zinc-dependent metalloprotease activity. More important is
mammary (40), and lacrimal glands (41), the fully mature 6 the fact that, whereas the zinc-dependent metalloprotease
kDa EGF was undetectable, and EGF accumulated as its high ADAM 17 (TACE) has an important role in the regulation of
molecular weight precursor associated with the apical mem- EGFR ligands shedding such as TGF-␣, HB-EGF, and amphi-
brane of epithelial cells. The presence of EGF-containing pep- regulin (24, 25, 28, 29, 30, 31), our results suggest that TACE
tides of varying molecular masses (from 6 to 160 kDa) in urine may not be relevant in the shedding of pro-EGF since pro-EGF
(37, 42, 43) and milk (22) of different species indicates that shedding was not regulated in HeLa cells, which are known to
EGF can be released (shed) from its membrane-anchored pre- possess high amounts of TACE.
cursor. This occurs through proteolytic cleavage in the jux-
MATERIALS AND METHODS
tamembrane region. The full-length precursor was the major
Chemicals—Peroxidase-conjugated goat anti-mouse IgG, trypsin
substrate for shedding, leading to the initial release of the
(10000 BAEE units/mg), extravidin-peroxidase, soybean trypsin inhib-
entire ectodomain. In order to lead to the ultimate, mature itor (SBTI, 1 mg inhibits 10000 units trypsin), aprotinin, pepstatin A,
6-kDa form of the molecule, the ectodomain needs to be further E-64, phenantrolin, bisindolylmaleimide I (BIM I), EDTA, tunicamycin,
maturated in the extracellular medium. It has already been protein molecular weight markers, DMEM without methionine and
shown that, in vitro, mature EGF could be released from its cysteine, A23187, ionomycin, PMA, ampicillin, LB broth, LB agar, and
precursor through the action of different type of proteases; protein G-Sepharose were obtained from Sigma Chemical Co. QIAquick
Gel Extraction and PCR purification kits were obtained from Qiagen.
acidic protease such as pepsin (36) or serine-proteases such as BCA protein assay kit and Sulfo-NHS-LC-Biotin were from Perbio.
trypsin (22, 39, 41) and plasmin (44). Moreover, membrane Triton X-100 was obtained from MERCK. N- and O-deglycosylation as
fractions of both kidney (45, 46) and mammary gland (47) well as Quantum Prep Plasmid purification kits were from BioRad
contain a membrane-bound proteolytic activity that colocalized Laboratories. Desoxynucleotide trisphosphates, agarose, and DNA size
with pro-EGF and released soluble EGF from its membrane- markers were obtained from Amersham Biosciences. Upstream and
downstream oligonucleotide primers were synthesized by MWG-bio-
associated precursor. Based on its sensitivity to a set of inhib-
tech. pTargeTTM mammalian expression vector system, TNT-coupled
itors, it was shown to belong to the serine-protease family. This reticulocyte lysate systems and JM-109 or DH5␣-competent cells were
experimental evidence is in conflict with results obtained in from Promega. ExpandTM Reverse Transcriptase and ExpandTM High
two independent studies performed with cultured cells. In Fidelity PCR System were from Roche Applied Science. 125I-Na (100
these cells transfected either with a full-length (48) or trun- ␮Ci/ml, 3.7 MBq/ml) and Renaissance® Western blot Chemilumines-
cated (49) human pro-EGF cDNA, EGF release was sensitive to cence Reagent Plus were purchased from PerkinElmer Life Sciences.
TRAN35S-Label was from ICN. Ro-31-8220 (BIM IX; methanesulfon-
metalloprotease inhibitors (batimastat and BB-2116) and cat- ate), polyclonal anti-human TACE (AB1) antibody, TAPI-2 and TIMP1
ion chelators (EDTA and EGTA) and insensitive to non-metal- and 3 were from Calbiochem. Phosphate-buffered saline (PBS), NUT.
loprotease inhibitors (48). Another important feature concern- MIX.F12(HAM) Glutamax (HAM-F12), fetal calf serum (FCS),
ing EGFR ligand shedding is its potential regulation. It is now OptiMEM, Dulbecco’s modified Eagle’s medium (DMEM) Glutamax,
clearly established that secretion of TGF-␣, HB-EGF, and am- fungizone, penicillin-streptomycin solution, random primers,
PcDNA3.1/Myc-His mammalian expression vector and monoclonal anti-
phiregulin are regulated phenomena that can be triggered by
Myc antibody were from Invitrogen. Exgen 500 was from Euromedex.
PMA (through PKC activation), calcium ionophore (calcium Monoclonal anti-HA antibody was from Covance. Polyclonal rabbit anti-
influx), serum or pervanadate (tyrosine phosphatase inhibi- human TNF-␣ antibody was from Promocell and TNF-␣ from Prepro-
tion) (9, 23, 25, 31, 50 –53). Concerning EGF, its regulated Tech Inc.
shedding from cell surface is not so clearly established. Demp- Rat Pro-EGF cDNA Cloning and Construction of Epitope-tagged Pro-
sey et al. (48) working with MDCK cells transfected with the EGF—Total RNAs from Sprague Dawley rats were prepared as de-
scribed previously (56). Kidney RNAs were first subjected to reverse
full-length human pro-EGF cDNA failed to stimulate EGF transcription using random priming and ExpandTM Reverse Tran-
secretion with serum or PMA whereas Dong and Wiley (49) scriptase as described by the manufacturer’s protocol. PCR amplifica-
working with mammary cells (HB2) transfected with a prodo- tion of the cDNA was performed according to rat pro-EGF cDNA se-
main-truncated human pro-EGF cDNA also failed to stimulate quence as published by Saggi et al. (Ref. 13, GenBankTM accession
secretion with PMA but observed a stimulation of EGF release number: M63585). The 5⬘ sense primer, complementary to nucleotides
350 –368, carries a HindIII restriction site whereas 3⬘ antisense primer,
with both calcium ionophore and a tyrosine phosphatase
complementary to nucleotides 3881–3900, carries an EcoRI restriction
inhibitor. site. Amplification was performed using the ExpandTM high fidelity
From these studies, it appears that important discrepancies PCR system (Roche Applied Science). After an initial denaturation for 2
exist in the conclusions that can be drawn from the studies on min at 94 °C, cDNA was first subjected to 10 cycles of amplification with
Metalloprotease-dependent Regulated Pro-EGF Ectodomain Shedding 45257
the following conditions: denaturation for 15 s at 94 °C, annealing for molecular mass fully immunoreactive mature EGF from putative pre-
30 s at 55 °C, and extension for 2 min 30 s at 68 °C then followed by 25 cursor molecules.
cycles in the same conditions but with extension time incremented of 5 s Metabolic Labeling of Cellular Proteins—Plated CHO (both wild type
for each cycle. A 3550 bp cDNA was obtained, which was purified, and M2 mutant) cells grown in 6-well plates as described above were
A-tailed, inserted into pTargeTTM mammalian expression vector as transiently transfected with the pcD12-HA expression plasmid. 48 h
described by the manufacturer’s protocols (Promega) and finally cloned later, monolayers were washed once with PBS and then preincubated in
into JM-109 bacterial cells leading to the pT12 clone. The sequence of serum-free, methionine/cysteine-free DMEM (Met-Cys-free DMEM)
clone pT12 was confirmed by both restriction analysis and sequencing supplemented with 1% lipid-free BSA for 30 min at 37 °C under 5%
using selected primers. Its ability to drive the synthesis of prepro-EGF CO2. Cells were then labeled in the same medium containing 250
protein of high molecular weight was further confirmed by in vitro ␮Ci/ml for 2 h of TRAN35S Label at 37 °C under 5% CO2. Labeling was
transcription using TNT®-coupled reticulocyte lysate systems (Pro- stopped by washing the cells three times with PBS. Cells were then
mega). To introduce epitope tags into the cloned rat prepro-EGF cDNA further incubated in the chase medium (complete DMEM containing
sequence, the entire coding sequence (HindIII-EcoRI fragment) was either 1% BSA or 10% FCS) containing the appropriate drugs and for
first excised and subcloned into PcDNA3.1/Myc-His mammalian ex- the time specified in the figure legends.
pression vector (Invitrogen). The prepro-EGF/Myc-His construct was Biotinylation of Cell Surface Proteins—CHO (both wild type and M2
generated by suppressing the stop codon using a PCR-based strategy. mutant) or HeLa cells were grown on 6-well plates for 48 h after
An antisense oligonucleotides complementary to nucleotides 3768 – transient transfection with the appropriate expression plasmid. Prior to
3787 and carrying an ApaI restriction site was used, allowing the biotinylation, cells were washed three times with ice-cold PBS contain-
in-frame ligation of the prepro-EGF cDNA with both c-Myc and His6 ing 1 mM CaCl2 and 1 mM MgCl2 (PBS⫹), then biotinylated in the
epitopes sequences at the 3⬘-end of the cDNA. This cDNA was further presence of 1 mg/ml sulfo-NHS-LC-Biotin in the same buffer for 30 min
modified by inserting the HA1 epitope (YPYDVPDYA) of influenza on ice. Biotinylation was stopped by incubating cells for 10 min at 4 °C
virus hemagglutinin (57) between amino acids Ser975 and Asn976 of rat in the presence of 100 mM glycine in PBS⫹ and washed once with
prepro-EGF using PCR approach as described by Arribas and Massagué PBS⫹. Following biotinylation, cells were incubated at 37 °C with the
(51) for the insertion of the HA epitope into the rat TGF-␣ sequence. indicated chase medium (complete DMEM containing either 1% BSA or
This final modification supplies the pcD12-HA plasmid, which was 10% FCS) containing the appropriate drugs and for the time specified in
confirmed by DNA sequencing. the figure legends.
PCR-based strategy was also used to generate a cDNA allowing the Immunoprecipitation, Western Blotting Analysis, and Autoradiogra-
synthesis of a HA-tagged soluble form of EGF, which lacked most of its phy—All immunoprecipitation protocols were performed at 4 °C, unless
prodomain. Briefly, a stop codon was introduced in the extracellular otherwise stated. After incubating cells in the different conditions spec-
domain of pro-EGF-HA following the Lys1024 codon (i.e. the C-terminal ified in the legends of the figures, conditioned media and cell lysates
end of the mature urinary EGF sequence) and the prodomain deleted were prepared as described above. Equal relative amounts of cell ly-
between Leu32 and Val972 to generate the ⌬pro-⌬TM-EGF expression sates and conditioned media were incubated overnight with either
plasmid. This expression plasmid allows the synthesis of a secreted anti-rat EGF (rEGF2, 1/200), anti-HA (HA-11, 1/500) or anti-Myc (1/
soluble form of rat pro-EGF that conserved the signal peptide and the 500) antibodies. In order to precipitate immune complexes, 40 ␮l of a
first ten amino acids of the EGF prodomain (Met1 to Pro31) as well as 50% slurry of protein A (rEGF2) or protein G-Sepharose (HA-11 or Myc)
the arginine residue (Arg973) that preceded the mature EGF sequence were added for 2 h under constant shaking. Protein A- or G-Sepharose
(Asn974 to Lys1024) but lacks the downstream sequences encoding the was pelleted by centrifugation and immune complexes washed three
juxtamembrane, transmembrane, and intracellular domains. times with high salt lysis buffer (phosphate 50 mM, NaCl 300 mM, pH
Cell Culture and Transfection—The CHO-K1 and CHO-M2 cells 7.4; EDTA 5 mM, Triton X-100 1%).
were cultured in monolayers in HAM-F12 medium whereas HeLa cells Immunoprecipitated incubation medium or cell lysates were solubi-
were cultured in DMEM. Both medium were supplemented with 10% lized in SDS containing electrophoresis sample buffer and separated by
FCS, 100 ␮g/ml streptomycin, 100 units/ml penicillin, and 1.25 ␮g/ml SDS-PAGE on a 7.5% Tris-HCl or 16.5% Tris-Tricine slab gel. Following
amphothericin B (fungizone) at 37 °C under 5% CO2 and used within electrophoresis, proteins from non-labeled, 35S-labeled or biotinylated
ten passages. Cell monolayers were suspended by treatment with 5 mM samples were electrotransferred overnight to optitran BA-S 85 re-
EDTA in PBS (CHO) or trypsin (HeLa) and usually plated in 6-well inforced nitrocellulose membrane (0.45 ␮m, Schleicher & Schuell). For
plates (750,000 cells/plate) in culture medium the day before detection of 35S-radioactivity, membranes were air-dried and exposed to
transfection. photosensitive storage phosphorimaging plates in a STORM 840 instru-
CHO or HeLa cells (50 –70% confluence) were transiently transfected ment (Molecular Dynamics). Quantification of the radioactivity of the
with 2.5 ␮g of the appropriate expression plasmid containing either bands in each lane was performed using ImageQuant software.
native (pT12), HA-tagged pro-EGF (PcD12-HA), or HA-tagged soluble For non-labeled and biotinylated samples, membranes were blocked
pro-EGF cDNA (⌬pro-⌬TM-EGF) or else human pro-TNF-␣ cDNA in with 5% nonfat milk in TBS containing 0.2% Tween 20 (TBST) for 1 h
PcDNA 3 (a kind gift from Dr. F. Peiretti) using Exgen 500 reagent as at 37 °C. Blots of biotinylated samples were directly probed with horse-
described by the manufacturer. Following transfection, cells were radish peroxidase-conjugated streptavidin (1/2500 in 0.5% milk con-
switched in HAM-F12 or DMEM supplemented with 10% FCS and taining TBST) for 1 h at room temperature. To reveal specific HA or
antibiotics. Transfected cells were further incubated for 48 h to allow for Myc epitope-containing proteins, blots were immunostained with mono-
pro-EGF or pro-TNF-␣ expression and analyzed either directly or after clonal antibodies (anti-HA or anti-Myc, 1/5000 in 0.5% milk containing
additional incubations in varying conditions. At the end of these differ- TBST) for 3 h at room temperature and probed with 1/10000 dilution of
ent types of experiments, cells and conditioned media were collected. goat anti-mouse IgG antibody linked to horseradish peroxidase as de-
Conditioned medium was cleared of contaminating cells by centrifuga- scribed previously (56). Blots were probed in Renaissance® Western
tion (1000 ⫻ g/5 min). Cells monolayers were washed twice with PBS blot Chemiluminescence Reagent Plus according to manufacturer’s rec-
and lysed for 30 min in detergent-containing buffer (50 mM sodium ommendations and visualized by exposure to Amersham Biosciences
phosphate, pH 7.4 supplemented with 150 mM NaCl, 5 mM EDTA, and Hyperfilm-ECL.
1% Triton X-100). Cell lysates were cleared from insoluble material by Immunodetection of TACE (ADAM 17) in cell lysates from CHO and
centrifugation at 15,000 ⫻ g for 20 min. Both incubation media and cell HeLa cells was performed following 7.5% slab gel electophoresis. West-
lysates were used for EGF or TNF-␣ content analysis as described ern blot analysis was performed with the rabbit polyclonal anti-TACE
below. antibody (1/2000) exactly as described above.
EGF and TNF-␣ Radioimmunoassay—Rat EGF or human TNF-␣
was labeled with carrier free 125I-Na to a specific activity of 150 –200 RESULTS
␮Ci/␮g using the chloramine-T method. RIA analysis for human TNF-␣
contents was performed exactly as previously described for rat EGF In an attempt to gain insight into the mechanisms involved
(41). The IgG fraction of the rabbit polyclonal anti-rat EGF antibody in the fate of rat EGF secretion, we first decided to clone the
(rEGF2, 1/5000) or of the anti-human TNF-␣ antibody (1/1000) were entire rat pro-EGF cDNA. To facilitate analysis of membrane-
used as immunoprecipitating antibodies. Immunoreactive rat EGF anchored pro-EGF shedding, the rat EGF cDNA was modified
(irEGF) in both cells and conditioned media and human TNF-␣ in
in order to introduce epitope tags in both the ectodomain (HA
conditioned media were quantified 48 h after cells have been tran-
siently transfected. As discussed previously (41), in order to adequately epitope) and the cytoplasmic tail (Myc epitope) of the molecule.
quantify EGF amounts, aliquots from both conditioned media and cell The HA tag was introduced downstream of the prodomain,
lysates were subjected to trypsin hydrolysis in order to release low between Ser975 and Asn976, i.e. the second and third amino
45258 Metalloprotease-dependent Regulated Pro-EGF Ectodomain Shedding
TABLE I attempt to detect EGF-containing proteins by Western blot
RIA analysis of EGF synthesis and secretion analysis using rEGF2 antibody failed because this antibody
by transfected CHO-K1 cells
was not able to recognize the denatured and reduced forms of
CHO-K1 cells were grown in culture medium (HAM F12 supple-
mented with 10% FCS) to about 70% confluency in 90-mm dishes. EGF (data not shown). That is why we decided to introduce
Plated cells were either not transfected (WT) or transiently transfected epitope tags in both extracellular and intracellular parts of the
with unmodified (pT12) or epitope-tagged (pcD12-HA) prepro-EGF-con- precursor protein (see Fig. 1A for the schematic structure of
taining expression plasmids. Following transfection, cells were further
EGF constructs). Western blot analysis of anti-EGF immuno-
incubated in 5 ml of culture medium at 37 °C under 5% CO2. 48 h later,
conditioned media were harvested and cell lysates prepared. Aliquots of precipitates from pcD12-HA-transfected CHO cells show that
both cell lysates and conditioned media were subjected or not to trypsin pro-EGF is expressed as three high molecular weight species
hydrolysis before quantification of rat EGF by RIA as described previ- containing both HA and Myc epitopes (Fig. 1B, Cell), and we
ously (41). Total amounts of immunoreactive rat EGF in cell lysates and
did not observe any protein lacking one of the two epitopes. Size
conditioned media are reported as well as the percentage of secreted
EGF for each experimental condition. analysis showed a major species of 176 kDa and two minor ones
of 136 and 111 kDa. Three high molecular weight immunore-
Amount of EGF in % of EGF in
conditioned
active proteins, one major species of 165 kDa and two minor
Conditioned
medium Cell lysates medium species of 121 and 99 kDa were observed in the anti-EGF
immunoprecipitates from conditioned medium of pcD12 HA-
pmol
CHO WT ⫺ Trypsin NDa ND transfected cells (Fig. 1B, Medium). These proteins only con-
⫹ Trypsin ND ND tained the HA epitope and not the Myc epitope. Performing
CHO pT12 ⫺ Trypsin 2.3 2.6 46.9 electrophoresis with 16.5% Tris-Tricine acrylamide slab gels
⫹ Trypsin 6.8 9.5 41.5
did not allow us to detect any other lower molecular weight
CHO pcD12-HA ⫺ Trypsin 4 3.4 54.4
⫹ Trypsin 14 16.3 46.1 immunoreactive species, especially mature 6 kDa EGF, either
a
in cell lysates or in the conditioned medium (data not shown).
Not detected.
As could be predicted, no EGF-containing protein could be
detected either with anti-HA or anti-Myc antibodies in both cell
acids of mature EGF. The Myc and His6 tags were C-terminal
lysate and conditioned medium from pT12-transfected CHO
extension of the precursor molecule (see Fig. 1A for the sche-
cells.
matic representation of the epitope-tagged rat pro-EGF con-
The presence of both extracellular (HA) and intracellular
struct). Both unmodified (pT12) and epitope-tagged (pcD12-
(Myc) epitopes in anti-EGF immunoprecipitates from cell ly-
HA) expression plasmids encoding the EGF precursor as well
sates suggests that these species represent membrane-an-
as the derived expression plasmid encoding the prodomain-
chored forms of pro-EGF. The 176-kDa species would be the
truncated HA-tagged soluble EGF (⌬pro-⌬TM-EGF) were used
full-length pro-EGF whereas the two others may represent
to transiently transfect established cultured cells.
species shortened in the prodomain of the molecule. The loss of
RIA Analysis of the Synthesis and Secretion of Rat EGF
the intracellular C-terminal Myc tag in pro-EGF species from
Immunoreactive Materials in Transiently Transfected CHO-K1
the conditioned medium as well as the shift in molecular
Cells—We first wanted to determine if a heterologous protein,
weight led us to hypothesize that these proteins are derived
which contains the EGF epitope, was expressed in CHO-K1
cells transfected either with the pT12 or PcD12-HA plasmids. from the membrane-anchored pro-EGF through proteolytic
EGF contents under different experimental conditions were cleavage in the juxtamembrane domain, i.e. between EGF and
first determined by RIA. As reported in Table I, EGF was the transmembrane domain. Moreover, the similar shift in
undetectable in both lysate and conditioned medium of non- molecular mass (respectively 11, 15, and 12 kDa) suggests that
transfected cells both before and after trypsin treatment. In the different soluble pro-EGF species; 165, 121, and 99 kDa are
contrast, important and similar amounts of immunoreactive respectively derived from the 176, 136, and 111 cellular species
EGF could be detected in both lysate and conditioned medium through proteolytic cleavage in the juxtamembrane domain.
following transfection of cells with either the untagged pro- As shown on Fig. 1C (upper panel), labeling of cell surface
EGF (pT12) or the pro-EGF-HA/Myc-tagged encoding plasmid proteins with sulfo-NHS-LC-biotin demonstrated that all three
(pcD12-HA). In the experiments realized with the EGF-precur- different membrane-anchored pro-EGF species detected in Fig.
sor encoding plasmids, EGF immunoreactivity was increased 1B were exposed at the cell surface. In pcD12-HA transfected
by 3– 4.8-fold following trypsin treatment. These results indi- cells, biotinylated proteins of 176, 136, and 111 kDa could be
cate that CHO cells do not naturally synthesize nor secrete immunoprecipitated with the anti-EGF, anti-HA, or the anti-
detectable levels of EGF or that EGF synthesized by cells from Myc antibodies. These pro-EGF species were also observed in
Chinese hamster cannot be detected by our antibody, which is anti-EGF immunoprecipitates from pT12-transfected cells. In-
directed against rat EGF (41). The presence of immunoreactive cubation of biotinylated cells for 4 h in culture medium supple-
EGF in both cell lysates and conditioned media indicates that mented with 10% serum resulted in the release of biotinylated
cells transfected with either one of the two plasmids secreted pro-EGF species of 165, 121, and 99 kDa (Fig. 1C, lower panel).
EGF materials. Cells transfected with the two different pro- According to the results shown in Fig. 1B, these biotinylated
EGF plasmids did not only synthesize similar amounts of EGF proteins were immunoprecipitated with both anti-EGF and
but also secreted the same percentage of EGF (about 50% of anti-HA antibodies but not with the anti-Myc antibody. All
total). This points out that the cellular behavior of the encoded three soluble species were also observed in anti-EGF immuno-
proteins toward synthesis and secretion is very similar and precipitates from pT12-transfected cells. Since all three species
suggests that epitope tagging of pro-EGF does not have any (176, 136, and 111 kDa) of cellular pro-EGF could be biotiny-
significant effect. Moreover, as stated in a previous study (41), lated at the cell surface, this greatly suggests that the lower
the increase in EGF immunoreactivity following trypsin treat- molecular mass species (136 and 111 kDa) may not represent
ment may be an indication that EGF is synthesized and se- some non-glycosylated precursor forms of the 176 kDa species.
creted as high molecular weight EGF-containing precursor This hypothesis was confirmed by performing short pulse la-
proteins. beling experiments (5 min) of newly synthesized proteins with
35
Western Blot Analysis of Expression and Secretion of Rat S-methionine/cysteine followed by chase incubation (up to
pro-EGF in Transiently Transfected CHO-K1 Cells—Our initial 3 h). Following 35S labeling, the radioactive 176 kDa anti-HA
Metalloprotease-dependent Regulated Pro-EGF Ectodomain Shedding 45259

FIG. 1. Epitope-tagged expression and shedding of pro-EGF in CHO-K1 cells. A, structure of epitope-tagged prepro-EGF constructs used
in this study. Location of HA, Myc, and His6 epitopes are as indicated. S, signal peptide; EGF, mature epidermal growth factor; TM, transmem-
brane domain; Cyt, cytoplasmic domain. B, characterization of the epitope-tagged EGF-containing proteins in cell lysate and conditioned medium.
CHO cells were transiently transfected with either unmodified prepro-EGF (pT12) or epitope-tagged prepro-EGF (pcD12-HA) containing expres-
sion plasmids and further incubated in conditioning medium (HAM F12 supplemented with 10% FCS). 48 h later, lysates and media were
harvested for immunoprecipitation (IP) with anti-rat EGF antibody. Immunoprecipitated EGF species were analyzed by Western blot (WB) using
the indicated antibodies as described under “Materials and Methods.” Apparent molecular masses of the different EGF species in lysate (°, 176 kDa;
°°, 136 kDa; °°°, 111 kDa) and incubation medium (*, 165 kDa; **, 121 kDa; ***, 99 kDa) were deduced from comigration with known standards.
C, characterization of cell surface expression and shedding of rat pro-EGF. CHO cells were transiently transfected with pT12 or pcD12-HA
expression plasmids. 48 h later, cell surface proteins were biotinylated using sulfo-NHS-LC-biotin as described under ‘‘Materials and Methods’’ and
further incubated for 4 h in conditioning medium as in B. Cell lysates and incubation media were harvested and immunoprecipitation (IP)
performed with the indicated antibody. Immunoprecipitated proteins were separated by SDS-PAGE, blotted to nitrocellulose and biotinylated
proteins revealed with peroxidase-conjugated streptavidin. D, expression and secretion of the HA-tagged prodomain-truncated soluble EGF. CHO
cells were transiently transfected with the ⌬pro-⌬TM-EGF expression plasmid. 48 h later, cells were washed and further incubated and processed
as described above (C). Anti-EGF immunoprecipitates were analyzed by electrophoresis on 16.5% Tris-Tricine slab gels, blotted to nitrocellulose
and analyzed by Western blot (WB) using the anti-HA (HA-11) antibody as described under ‘‘Materials and Methods.’’ Apparent molecular masses
of the different EGF species reported on the right side of the figures were deduced from comigration with known standards (left side).

immunoprecipitated protein is instantaneously and highly de- pcD12-HA-transfected CHO cells contain one major (176 kDa)
tectable in the cell lysate. At any time tested, this major pro- and two minor (136 and 111 kDa) pro-EGF species that are
EGF showed the same high molecular weight. Lower molecular membrane-anchored proteins exposed at the cell surface. Once
mass proteins of 136 and 111 kDa were only faintly detected exposed at the cell surface, these pro-EGF species may undergo
later (data not shown), a result that argues against their role as proteolytic cleavage in their juxtamembrane domain in order to
precursor forms of the 176-kDa species. release soluble pro-EGF of 165, 121, and 99 kDa. The protein
Taken altogether, these results indicate that pT12- and pattern observed with CHO cells transfected with the epitope-
45260 Metalloprotease-dependent Regulated Pro-EGF Ectodomain Shedding
tagged pro-EGF encoding pcD12-HA plasmid is clearly not a
consequence of epitope tagging of the molecule. As a matter of
fact, the same pattern of proteins was observed in both cell
lysate and conditioned medium following biotinylation of un-
tagged pro-EGF synthesized by pT12-transfected CHO cells.
Since the calculated size of the full-length epitope-tagged
membrane-anchored EGF precursor, 128 kDa, as determined
from cDNA sequence, is far smaller than the estimated size
found by SDS-PAGE (176 kDa) we suggested that this protein
might be glycosylated. This was confirmed following treatment
of both secreted and cellular pro-EGF with different glycosi-
dases (N-glycosidase, O-glycosidase, and N-acetyl-neuramini-
dase) or incubation of cells with tunicamycin (an inhibitor of
endogenous glycosylation). These experiments show that both
soluble (165, 121, and 99 kDa) as well as membrane-anchored
pro-EGF (176, 136, and 111 kDa) were N-glycosylated proteins
(data not shown). However, the shift in molecular mass follow-
ing deglycosylation (15–20 kDa) was not sufficient to explain
the discrepancy between the one calculated from the peptide
backbone and the one measured on SDS-PAGE. Size analysis of
the in vitro transcription/translation product from the
pcD12-HA expression plasmid yielded only one product, whose
estimated molecular mass of 154 –159 kDa is very close to the
estimated size (160 kDa) of the deglycosylated 176 kDa mem-
brane-anchored pro-EGF. These results greatly favor the hy-
pothesis that an unusual electrophoretic behavior of the EGF FIG. 2. Induction of pro-EGF shedding by PMA, pervanadate,
precursor molecules may explain the overestimation of both and FCS. CHO cells were transiently transfected with pcD12-HA ex-
membrane-anchored and soluble high molecular weight pro- pression plasmid. 48 h later, cells were labeled for 2 h with TRAN35S
EGF found in transfected CHO cells but also in other cell types label. At the end of the labeling, cells were extensively washed three
times with PBS and chased at 37 °C under 5% CO2 for 2 h in DMEM
such as MDCK and HB2 cells. supplemented with 1% BSA and containing either no addition (medi-
Fig. 1D show the results obtained with CHO cells transiently um) or one of the following agents: 0.1% Me2SO (DMSO), 1 ␮M calcium
transfected with the HA-tagged prodomain-truncated soluble ionophore (A23187), 1 ␮M phorbol ester (PMA), 100 ␮M Pervanadate
EGF expression plasmid (⌬pro-⌬TM-EGF). As can be shown, (V2O7) or 10% serum (FCS). Conditioned media were immunoprecipi-
tated with the anti-HA antibody as described under ‘‘Material and
following an incubation of 4 h in serum-containing medium, Methods.’’ Immunoprecipitated proteins were further separated by
anti-EGF immunoprecipitates from both cell lysate and condi- SDS-PAGE, blotted to nitrocellulose, and visualized by phosphorimag-
tioned medium were shown to contain an anti-HA immunore- ing using a STORM 840 instrument (upper panel). Radioactivity asso-
active protein of 8.4 kDa. This molecular mass is greater than ciated with each secreted pro-EGF (165, 121, 99 kDa as indicated on the
figure) was quantified for each experimental condition (lower panel),
the one that could be predicted for fully mature HA-tagged and results are expressed as the percentage of the amount of that
EGF (6.9 kDa) but perfectly matches the one predicted for an 35
S-labeled pro-EGF present in the control medium.
expressed protein that lacks the signal peptide but was not
cleaved at the level of its residual prodomain. The identical be used to account for the properties of pro-EGF secretion.
molecular masses that are observed for both cellular and se- Thus, in the following experiments only results concerning the
creted EGF indicated that the last one is not derived from the major pro-EGF kilodalton species (pro-EGF165) will be reported
first one through proteolytic cleavage. Thus, as observed with and quantified.
the full-length pro-EGF-encoding plasmids (Fig. 1, A–C), se- We have demonstrated here that in contrast to previously
creted EGF does not appear to be processed in its ectodomain published results (48), the release of soluble rat pro-EGF may
and particularly at the Arg973 level that would have led to the be regulated at least in one cell type, the CHO-K1 cells. How-
fully mature HA-tagged soluble 6.9-kDa EGF species as ob- ever, with that kind of experiment, the increased release of
served in rat urine (58). The way by which the 8.4 kDa soluble soluble forms of pro-EGF may be due to the stimulation of
EGF is secreted appeared to be completely different from the intracellular protein traffic in the secretory pathway and/or an
one involved for full-length pro-EGF and must follow the se- increase in the shedding rate of the membrane precursor. In
cretory pathway for soluble proteins that do not involve cell order to test for an increased cleavage of the cell surface pro-
surface shedding. EGF, we analyzed pro-EGF secretion upon PMA stimulation
Shedding of Rat Membrane-anchored pro-EGF Is Regulated following both 35S labeling and cell surface protein biotinyla-
in CHO-K1 Cells—As shown in Fig. 2 (upper panel), pcD12-HA- tion. As shown in Fig. 3, compared with control experiments,
transfected 35S-labeled CHO cells show an increased release of PMA induced a time-dependent increase of both 35S-labeled
all three species of pro-EGF upon incubation in the presence of (upper panel) and biotinylated pro-EGF165 (middle panel) in
PMA, pervanadate (V2O7), or serum (FCS). No stimulation the conditioned medium, suggesting that the HA-tagged pro-
of pro-EGF release was detected when cells were incubated teins secreted came from a previously labeled intrinsic cell
with either Me2SO (vehicle), the calcium ionophores A23187 surface protein, i.e. the membrane-anchored pro-EGF. Quanti-
(see Fig. 2) or ionomycin (data not shown). Following quantifi- fication of the kinetics of 35S-labeled pro-EGF165 release indi-
cation by phosphorimaging, the amount of each [35S]pro-EGF cates that the effect of PMA was linear for up to 2 h (lower
species was expressed relative to the unstimulated condition panel). Stimulation rapidly decreases thereafter probably as a
(medium). As seen in Fig. 2 (lower panel), the stimulatory result of both desensitization of the PKC-dependent pathway
agents tested increased the release of the three pro-EGF to the and down-regulation of the PKC activity. From these results, it
same extent. This indicates that any of the soluble pro-EGF can is obvious that the rise in pro-EGF detected in the medium
Metalloprotease-dependent Regulated Pro-EGF Ectodomain Shedding 45261

FIG. 4. Regulated EGF secretion in ⌬pro-⌬TM-EGF versus


pcD12-HA transfected cells. CHO cells were transiently transfected
with either HA-tagged full-length pro-EGF (pcD12-HA) or with the
HA-tagged prodomain truncated soluble EGF (⌬pro-⌬TM-EGF) expres-
sion plasmid as indicated on the figure. 48 h later, cells were extensively
washed three times with PBS and chased at 37 °C under 5% CO2 for 2 h
in DMEM supplemented with 1% BSA and containing or not (medium)
one of the following agents: 1 ␮M phorbol ester (PMA), 100 ␮M pervana-
date (V2O7), or 10% serum (FCS). Immunoreactive EGF in both condi-
tioned medium and cell lysates was quantified by RIA following trypsin
FIG. 3. Kinetic of stimulation of cell surface pro-EGF secre- treatment of the different samples as described under ‘‘Materials and
tion. CHO cells were transiently transfected with pcD12-HA expres- Methods.’’ EGF secretion in the incubation medium was first calculated
sion plasmid. 48 h later, cells were labeled for 2 h with TRAN35S Label. as a percentage of total immunoreactive EGF in both medium and cell
At the end of the labeling, cells were extensively washed three times lysate then normalized to percentage of EGF secretion in the medium
with PBS and cell surface proteins biotinylated using sulfo-NHS-LC- alone. Results are the average ⫾ S.E. of triplicate experiments.
biotin as described under ‘‘Materials and Methods.’’ Labeled cells were
chased for up to 180 min, in the absence or presence of 1 ␮M PMA as
indicated on the figure. At each time point, conditioned media were ulation is due to the stimulation of membrane-anchored pre-
immunoprecipitated with the anti-HA antibody and immunoprecipi-
tated proteins separated by SDS-PAGE and blotted to nitrocellulose. cursor shedding.
35
S-labeled proteins were visualized by phosphorimaging using a Role of PKC in the Regulation of Membrane-anchored pro-
STORM 840 instrument (upper panel) and biotinylated proteins visu- EGF Shedding—Since PKC activation is often linked to protein
alized by ECL⫹ using peroxidase-conjugated streptavidin (middle secretion and was clearly involved in the PMA-dependent reg-
panel) as described under ‘‘Materials and Methods.’’ Radioactivity as-
sociated with 165 kDa [35S]pro-EGF was quantified for each time point
ulation of EGFR ligand shedding, we have tested its role in the
(lower panel), and results are expressed as the percentage of the control of rat pro-EGF secretion. Implication of phorbol ester
amount of 35S-labeled pro-EGF165 present in the incubation medium activable PKC in pro-EGF secretion was first demonstrated by
upon 180 min of stimulation with PMA. the use of specific PKC inhibitors such as Ro 31-8220 or BIM I.
As can be seen on Fig. 5A, preincubation of CHO cells in the
upon PMA stimulation is due to the stimulation of membrane- presence of PKC inhibitors either partially (Ro 31-8220) or
anchored precursor shedding. completely (BIM I) abolished a subsequent stimulation of pro-
This conclusion was further confirmed by the experiments EGF165 secretion by PMA. In the same conditions, basal secre-
performed with CHO cells transiently transfected with ⌬pro- tion as well as pervanadate and serum stimulation was unaf-
⌬TM-EGF expression plasmid. RIA analysis of EGF content in fected (data not shown). The involvement of PKC in the
both conditioned media and cell lysates show us that cells stimulatory effect of PMA was further confirmed following its
transfected with ⌬pro-⌬TM-EGF or pcD12-HA synthesized down-regulation prior to cells stimulation. PcD12-HA-trans-
equal amounts of immunoreactive EGF (data not shown), how- fected cells were first incubated with 1 ␮M PMA for 18 h prior
ever the rate of EGF secretion by cells expressing the truncated to 35S labeling. As shown in Fig. 5B, down-regulation of PKC
8.4 kDa soluble form of EGF (11.2 ⫾ 0.89% per h) was two completely abolished the stimulation of pro-EGF165 secretion
times that of cells expressing the full-length membrane-an- by PMA. This inhibition is not the consequence of a general and
chored EGF precursor (4.9 ⫾ 0.35% per h), suggesting that both nonspecific inhibitory effect of the treatment since pervanadate
EGF species do not follow the same pathway to be secreted into stimulation was only slightly affected and serum-induced se-
the extracellular medium. Moreover, compared with pro-EGF- cretion was not. Moreover, PMA pretreatment did not show any
transfected cells, incubation in the presence of PMA, serum, or inhibitory effect on the incorporation of 35S-amino acids into
pervanadate showed that secretion of the soluble form of EGF cellular pro-EGF, i.e. pro-EGF synthesis (data not shown).
could not be up-regulated whereas the one resulting from ex- Taken together, these results and those obtained with PKC
pression of the full-length membrane-anchored pro-EGF could inhibitors, clearly indicate that the stimulatory effect of PMA is
be (Fig. 4). This result, and the fact that no molecular weight completely dependent on PKC activities whereas both pervana-
shift between the cellular and secreted HA-EGF was detected date and serum stimulation are not.
(8.4 kDa, Fig. 1D) indicate that a general stimulation of intra- Pro-EGF Ectodomain Shedding in CHO-K1 Cells Is Associ-
cellular protein trafficking does not account for the stimulation ated with a Zinc Metalloprotease Activity—The following exper-
of pro-EGF ectodomain secretion and reinforces the hypothesis iments were performed in order to determine the type of pro-
that the rise in pro-EGF165 detected in the medium upon stim- tease activity involved in membrane-anchored pro-EGF
45262 Metalloprotease-dependent Regulated Pro-EGF Ectodomain Shedding

FIG. 5. Role of PKC in the regulation of pro-EGF shedding. A, effect of PKC inhibitors on PMA induced pro-EGF shedding. CHO cells were
transiently transfected with pcD12-HA expression plasmid. 48 h later, cells were labeled for 2 h with TRAN35S Label. At the end of the labeling,
cells were extensively washed three times with PBS and chased for 30 min in DMEM supplemented with 1% BSA in the absence or presence of
PKC inhibitors (5 ␮M BIM or 5 ␮M RO 31– 8220) before addition or not of PMA (1 ␮M final concentration) for an additional 2 h. B, effect of PKC
‘‘down-regulation’’ on the regulation of pro-EGF shedding. CHO cells were transiently transfected with pcD12-HA expression plasmid. 30 h later,
cells were washed and further incubated for 18 h in the absence or presence of 1 ␮M PMA in order to down-regulate PKCs. Cells were then labeled
for 2 h with TRAN35S Label and extensively washed three times with PBS. Following labeling, cells were chased for 2 h at 37 °C under 5% CO2
in DMEM supplemented with 1% BSA in the absence (medium) or presence of different agents: 1 ␮M PMA; 100 ␮M V2O7; 0,1% Me2SO (DMSO);
10% FCS, or 1 ␮M A23187 as mentioned on the figure. Following chase incubations, conditioned media were immunoprecipitated with anti-HA
antibody. Immunoprecipitated proteins were further separated by SDS-PAGE, and blotted to nitrocellulose. 35S-labeled proteins were visualized
by phosphorimaging using a STORM 840 instrument (upper panels A and B). Radioactivity associated with 165 kDa [35S]pro-EGF was quantified
for each condition, and results are expressed as the percentage of the amount of 35S-labeled pro-EGF165 present in the incubation medium alone.

shedding. Therefore, we tested the effects of different protease volved in pro-EGF cleavage is greatly dependent upon the
inhibitors on pro-EGF165 secretion from PcD12-HA transfected presence of some divalent cations and suggest that it might be
CHO-K1 cells. The inhibitory effect was assayed both on PMA- a Zn2⫹ metalloprotease. This hypothesis was further supported
stimulated and -unstimulated 35S-labeled cells. As shown in by the use of TAPI-2, a hydroxamate acid-based derivative,
Fig. 6A, none of the protease inhibitors tested had any signif- which inhibits zinc-dependent metalloprotease activities by
icant inhibitory effect on the unstimulated secretion. However, binding to the catalytic site of the enzyme. As shown in Fig. 6B,
the PMA-stimulated [35S]pro-EGF165 secretion was signifi- TAPI-2 inhibited both unstimulated and PMA-stimulated
cantly reduced by the metalloprotease inhibitor phenantrolin, a [35S]pro-EGF165 secretion at concentrations below 1 ␮M. Half-
zinc chelator molecule, and this inhibition can be overcome by maximal inhibition of the PMA stimulation was achieved for a
the addition of an excess of zinc in the incubation medium (data concentration of 0.1 ␮M and complete inhibition reached at a
not shown). Neither aprotinin nor SBTI (two serine protease concentration of 3 ␮M. The inhibitory effect of TAPI-2 was not
inhibitors), nor pepstatin (an aspartyl protease inhibitor), nor restricted to the PMA stimulation since a 2 ␮M concentration
E64 (a cysteine protease inhibitor) had any significant inhibi- completely inhibited the serum- and pervanadate-stimulated
tory effect either on basal or stimulated secretion. EGTA and [35S]pro-EGF165 secretion (data not shown). Tissue inhibitors
EDTA, two commonly used metalloprotease inhibitors, which of matrix metalloprotease (TIMP 1 and 3) were also tested at
preferentially chelate Mg2⫹ and Ca2⫹, were also inefficient concentrations up to 50 nM but failed to prevent the PMA
(data not shown). stimulation of immunoreactive EGF secretion (data not
These results indicate that the activity of the protease in- shown).
Metalloprotease-dependent Regulated Pro-EGF Ectodomain Shedding 45263

FIG. 7. Expression and regulation of pro-EGF secretion in


CHO-M2. Wild-type CHO (CHO-wt) and mutant CHO (CHO-M2) cells
were transiently transfected with the pcD12-HA expression plasmid.
48 h later, cells were processed for cell surface proteins biotinylation
using sulfo-NHS-LC-biotin (part A) or labeled for 2 h with TRAN35S
Label (part B) as described under ‘‘Materials and Methods.’’ A, biotiny-
lated cells were further incubated for 4 h in conditioning medium as in
Fig. 1B. Cell lysates and incubation media were harvested and immu-
noprecipitated with the anti-HA antibody (HA-11) as described under
‘‘Materials and Methods.’’ Immunoprecipitated proteins were separated
by SDS-PAGE on 7.5% slab gels, blotted to nitrocellulose and biotiny-
lated proteins revealed with peroxidase-conjugated streptavidin. B, at
the end of the labeling, cells were extensively washed three times with
PBS and chased at 37 °C under 5% CO2 for 2 h in DMEM supplemented
FIG. 6. Effect of different protease inhibitors on pro-EGF shed- with 1% BSA and in the presence or not of different agents as indicated
ding. CHO cells were transiently transfected with pcD12-HA expres- below the figure. Conditioned media were harvested and immunopre-
sion plasmid. 48 h later, cells were labeled for 2 h with TRAN35S Label, cipitated with the anti-HA antibody (HA-11) as described under ‘‘Ma-
extensively washed three times with PBS and chased at 37 °C under 5% terial and Methods.’’ Immune complex were further separated by SDS-
CO2 in DMEM supplemented with 1% BSA. A, effect of type-specific PAGE on a 7.5% slab gel, blotted to nitrocellulose and visualized by
proteases inhibitors on pro-EGF shedding. Chase was first performed phosphorimaging using a STORM 840 instrument. Radioactivity asso-
for 30 min in the absence (medium) or presence of different proteases ciated with the pro-EGF165 species was quantified for each experimen-
inhibitors (10 ␮M aprotinin, 20 ␮M SBTI, 100 ␮M pepstatin, 100 ␮M E64, tal condition, expressed as arbitrary units, and are the average ⫾ S.E.
and 1 mM phenantrolin) before the addition (lower panel) or not (upper of triplicate experiments.
panel) of PMA (1 ␮M final concentration) for 2 h. B, effect of the specific
metalloprotease inhibitor TAPI-2 on PMA induced pro-EGF secretion. Wild-type CHO (wt) and mutant (M2) cells were first tran-
Labeled cells were first chased for 30 min in the presence or not of
increasing concentrations of TAPI-2 before stimulation (right part) or siently transfected with the pcD12-HA expression plasmid, cell
not (left part) with PMA (1 ␮M final concentration) for 2 h. Following surface biotinylated, and analyzed for their ability to release
chase incubations, conditioned media were immunoprecipitated with pro-EGF upon incubation in a serum-containing medium. Bi-
anti-HA antibody. Immunoprecipitated proteins were further separated otinylated HA-tagged proteins in both cell lysate and incuba-
by SDS-PAGE, and blotted to nitrocellulose. 35S-labeled proteins were
visualized by phosphorimaging using a STORM 840 instrument (panel tion medium were further analyzed as described in Fig. 1. As
A and upper panel B). Radioactivity associated with 165 kDa [35S]pro- can be seen on Fig. 7A, the amount of soluble biotinylated
EGF as reported in upper panel B was further quantified for each pro-EGF165 released by the M2 cells was greatly reduced com-
conditions, and results are expressed as the percentage of the amount of pared with the wild-type cells. This reduction was associated to
35
S-labeled pro-EGF165 present in the incubation medium upon stimu-
lation with PMA alone (lower panel B).
a concomitant accumulation of pro-EGF at the cell surface as
shown by the increase of the biotinylated pro-EGF176 species in
the cell lysate from CHO-M2 compared with CHO-wt. Since we
The metalloprotease-dependent ectodomain shedding of pro- verified by RIA analysis that both cell lines synthesized equal
EGF was further confirmed by studying pro-EGF secretion in a amounts of pro-EGF (data not shown), this result clearly dem-
CHO cell line (CHO-M2) defective in the metalloprotease-de- onstrates that the mutant CHO cell line was also defective in
pendent shedding of many cell surface proteins (25, 28, 51). cell surface pro-EGF shedding.
45264 Metalloprotease-dependent Regulated Pro-EGF Ectodomain Shedding
CHO-wt and CHO-M2 cell were further compared for their
ability to up-regulate pro-EGF secretion upon stimulation.
PcD12-HA- transfected 35S-labeled CHO-wt and CHO-M2 were
incubated in the presence of PMA, serum or pervanadate, and
pro-EGF secretion analyzed as described in Fig. 2. Results
reported in Fig. 7B showed that these agents were unable to
increase pro-EGF shedding from the cell surface of CHO-M2
cells and demonstrate that both constitutive and regulated
pro-EGF secretion was completely impaired in the CHO-M2
cell line compared with CHO-wt.
Taken as a whole, the results obtained with protease inhib-
itors as well as CHO-M2 clearly established that, at least in
CHO cells, both constitutive and regulated EGF secretion in-
volve one or more zinc-dependent metalloprotease(s) activities.
This or these metalloprotease activities shed the ectodomain of
pro-EGF from the membrane-anchored pro-EGF once exposed
at the cell surface.
Lack of Regulated pro-EGF Shedding in HeLa Cells—The
zinc metalloprotease TACE (ADAM 17) is now known to be
involved in shedding of many different cell surface proteins and
among them three members of the EGF growth factor family,
i.e. TGF-␣ (24, 25, 28, 29), HB-EGF (29, 30), and amphiregulin
(29, 31). So, in order to determine its possible involvement in
the shedding of pro-EGF, we have used the established HeLa
cell line, which is well known to possess high amounts of TACE
(Ref. 59 and Fig. 8A) and was able to cleave one of the sub-
strates of TACE, the ␤-amyloid precursor protein (␤APP) (60).
Using an anti-TACE antibody directed against the cytoplas-
mic domain of the protease, we have first verified the presence
of the protease in both CHO-K1 and HeLa cell lines (Fig. 8A).
As can be shown, HeLa cells contained high amount of both the
inactive high molecular weight proenzyme and of 2 mature
forms with lower masses. In contrast, when equal amounts of
cellular proteins from our CHO-K1 cells were analyzed, they
appeared to contain only low amounts of mature enzymes.
Like CHO-K1 cells, HeLa cells transiently transfected with
the pcD12-HA expression plasmid synthesize EGF as its high
molecular weight precursor. This precursor is membrane-an-
chored and is expressed at the cell surface as shown by bioti-
nylation experiments. However, compared with CHO cells, the
presence of HA-tagged pro-EGF in conditioned medium follow-
ing incubation in the presence of serum was only barely detect- FIG. 8. Regulation of EGF and TNF-␣ secretion in CHO and
able (data not shown). This result was confirmed when immu- HeLa cell lines. A, cell lysates from both CHO and HeLa cell lines
were prepared as described under ‘‘Materials and Methods.’’ Equal
noreactive EGF secretion was measured by RIA. HeLa cells
amount of cellular proteins were separated by SDS-PAGE on 7.5% slab
synthesized two to three times less EGF than CHO cells and gel and analyzed by Western blot using an anti-human TACE antibody.
with a basal EGF secretion rate, which was also 2⫻ lower (2.2% Arrows on the right show the position of the unprocessed full-length
per hour compared with 4.9% for CHO cells). In contrast to TACE precursor (proTACE) as well as of the mature and active TACE
species (TACE). B, HeLa (upper part) and CHO (lower part) cells were
CHO, HeLa cells have EGF receptors (EGFR) expressed at the
transiently transfected with either HA-tagged full-length pro-EGF
cell surface (61). As reported for TGF-␣ secretion by MDCK (pcD12-HA, left side) or with the human pro-TNF-␣ (right side) expres-
cells (62), we tested the possibility that secreted EGF could be sion plasmids. 48 h later, cells were extensively washed three times
lowered by a rapid consumption through receptor binding and with PBS and chased at 37 °C under 5% CO2 for 30 min for cells to
measure TNF-␣ secretion or for 2 h to measure EGF secretion. Chase
internalization. However, this is clearly not the explanation in
incubation was performed in DMEM supplemented with 1% BSA and
HeLa since none of the manipulations intended to inhibit this containing either no addition (medium) or one of the following agents: 1
effect had any increasing effect on the amount of secreted EGF. ␮M phorbol ester (PMA), 100 ␮M pervanadate (V2O7) or 10% serum
The conditions tested were the following: incubation in the (FCS). Immunoreactive TNF-␣ in conditioned medium as well as EGF
in both conditioned medium, and cell lysates were quantified by RIA as
presence of an excess of human EGF or of an anti-EGFR anti-
described under ‘‘Materials and Methods.’’ TNF-␣ secretion was ex-
body (mAb 225) that blocked the binding of secreted EGF to the pressed as percentages relative to the amount of immunoreactive
EGFR as well as blockade of receptor kinase activity by TNF-␣ in conditioned medium of unstimulated cells (medium). EGF
AG1478 that inhibited EGFR internalization (data not shown). secretion in the incubation medium was first calculated as a percentage
of total immunoreactive EGF in both medium and cell lysate then
More importantly, in HeLa cells, EGF secretion did not show normalized to percentage of EGF secretion in medium alone. Results
any strong up-regulation upon incubation in the presence of are the average ⫾ S.E. of triplicate experiments.
either PMA, serum, or pervanadate (Fig. 8B, upper left) under
conditions that strongly increase EGF secretion in CHO cells regulated shedding of cell surface proteins. To test this hypoth-
(Fig. 8B, lower left). Only a small but significant increase could esis, HeLa and CHO cells were transiently transfected with a
be observed upon PMA stimulation. One simple explanation pro-TNF-␣-containing expression plasmid, and secretion of
may be that our HeLa cell line may be unable to achieve TNF-␣ in both cell lines was analyzed by RIA. Results obtained
Metalloprotease-dependent Regulated Pro-EGF Ectodomain Shedding 45265
allowed us to rule out this hypothesis since they clearly showed probably derived from the major 176 kDa full-length precursor
that in HeLa cells, shedding of pro-TNF-␣ was enhanced by the through cleavage in the N-terminal distal part of the prodo-
different treatments: PMA, FCS, or pervanadate (Fig. 8B, up- main. They have not been observed with NIH 3T3 and MDCK
per right). Moreover, both basal and PMA-stimulated shedding cells transfected with the human pro-EGF (48, 67) but we
could be inhibited by the zinc metalloprotease inhibitor TAPI-2 previously reported a similar maturation of the membrane-
(data not shown). In contrast, CHO cells show only a small associated pro-EGF in rat kidney (41). Nevertheless, from the
increase in pro-TNF-␣ shedding upon stimulation with PMA present experiments, this limited prodomain release does not
and serum (⬃140% of basal) when compared with the effect seems to have any consequences on EGF secretion since all
obtained on pro-EGF shedding in the same cell line (200 –500% species appeared to be equally sensitive to juxtamembrane
of basal) or to the release of TNF-␣ from HeLa cells (200 – 400% cleavage.
of basal). According to studies on MDCK (48) and NIH 3T3 (67) cells,
Taken together, these results greatly favor the hypothesis we did not observe any proteolytic maturation of pro-EGF that
that HeLa cells may not have the enzymatic machinery (ecto- would lead to the release of the entire EGF prodomain. These
protease) necessary to shed the EGF precursor. The ADAM 17 observations are quite different from those reported for the
protease (TACE) was first identified as a protease that specif-
maturation process of other EGFR ligands such as TGF-␣ (50)
ically cleaves full-length pro-TNF-␣ (63, 64). The fact that
and human HB-EGF (68) where the precursor prodomain ap-
HeLa cells, which possess a high amount of TACE, did not
pears to be cleaved very rapidly once it has reached the cell
efficiently shed pro-EGF, whereas these cells were able to
surface. However, our results are very similar to the ones
cleave pro-TNF-␣, questioned the role of TACE in the process-
obtained with mouse HB-EGF (68) and amphiregulin (31),
ing of pro-EGF. This conclusion is further corroborated by the
where the full-length precursor preferentially accumulates at
ability of CHO cells that contain low amounts of TACE to
efficiently shed pro-EGF while being unable to shed pro-TNF-␣ the cell surface. This may reflect some specific and intrinsic
from the cell surface. Thus, while both pro-TNF-␣ and pro-EGF properties of the EGF prodomain that would be independent
shedding are achieved by zinc-dependent metalloproteases, both from the origin of EGF and of the expressing cells. It is
pro-TNF-␣ shedding appears to be correlated to the amount of often thought that membrane-anchored EGFR ligands are able
TACE, while this correlation does not apply to pro-EGF to interact with the EGF receptor (EGFR) located on an adja-
shedding. cent cell, thus acting as a juxtacrine growth factor (9, 71). This
property of EGFR ligands has been clearly demonstrated for
DISCUSSION both TGF-␣ (69 –71) and human HB-EGF (3, 68), whose mem-
In the course of a recent study (41) we have developed an brane-anchored precursors are known to be rapidly subjected
efficient polyclonal antibody (rEGF2) directed against the na- to prodomain release (50, 66). In contrast, the juxtacrine activ-
tive form of rat EGF. This antibody allowed us to demonstrate ity of the membrane-anchored mouse HB-EGF precursor is
the presence of immunoreactive EGF in the exocrine rat lacri- greatly reduced because of a decreased maturation of its prodo-
mal gland. In this tissue EGF was shown to be stored as its main (68). The physiological significance of the N-terminal
high molecular weight membrane-anchored precursor as al- processing still remains unknown. However, prodomain re-
ready shown in kidney (12, 36, 37, 39). Immunoreactive rat lease leads to the exposure of the growth factor moiety to the
EGF is also present in corresponding secretory fluids, i.e. tears cellular environment, thus potentially facilitating its interac-
and urine (18 –20, 37, 42, 43). However, the nature of the tion with the EGFR on an adjacent cell. Under these condi-
regulatory pathways as well as the proteases involved in the tions, the observation that both human (48, 67) and rat mem-
shedding of membrane-anchored pro-EGF that may account for brane-anchored pro-EGF (this study) do not undergo any
the release of soluble EGF are very poorly understood. The significant cleavage of their prodomain led us to question
reasons may be that there was neither simple physiological whether it would act as a juxtacrine growth factor. The hypoth-
model nor any immortalized cell line available to study the esis that the EGF precursor may have juxtacrine potency has
endogenous synthesis and secretion of EGF. not been deduced from co-culture experiments but was inferred
Since the tight regulation of shedding of EGFR ligands such from the ability of both soluble (secreted) pro-EGF (43, 67) and
as TGF-␣ (50, 53, 65) and HB-EGF (66) have been previously
detergent-solubilized membrane pro-EGF (36) to bind and ac-
demonstrated in the CHO cell line, we were then interested in
tivate the EGF receptor. However, under these two conditions
determining the possible regulation of rat pro-EGF secretion in
EGF is freed from the environmental constraint (steric hin-
this cellular model. In this way, we first cloned rat prepro-EGF
drance) of the membrane. This would be sufficient to allow a
cDNA and introduced epitope tags in both the extracellular and
low affinity interaction between the EGF moiety and the EGFR
intracellular domain of the molecule. This was done in order to
(high molecular mass pro-EGF is less biologically active than
conveniently follow the different parts of the precursor mole-
mature 6 kDa EGF) but it does not necessarily imply that the
cule and also because our anti-rat EGF antibody does not
recognize the denatured and reduced form of EGF, precluding native membrane-anchored pro-EGF has this property. To
any analysis by SDS-PAGE. RIA analysis of cell lysate and more definitely answer this question, we think that the results
conditioned media indicates that CHO cells transfected with that could be obtained from co-culture experiments with CHO
pro-EGF-containing expression plasmids efficiently synthesize cells expressing or not the membrane-anchored rat EGF pre-
and secrete EGF immunoreactive molecules. cursor and EGFR-expressing cells will be valuable.
Cellular pro-EGF was found to be expressed as three differ- We did not observe any precursor form to the 176-kDa rat
ent species that are all present at the cell surface. These pro- pro-EGF species, and in contrast to the results obtained in
teins that contain EGF as well as the intracellular and extra- MDCK cells, cell surface biotinylation experiments led to the
cellular epitopes, certainly represent the membrane-anchored efficient labeling of the 176-kDa pro-EGF (as well as the 136-
pro-EGF. The size of the major pro-EGF species (176 kDa) is and 111-kDa species). This favors the hypothesis that they
very similar to the one of the human pro-EGF expressed in represent mature pro-EGF species that have reached the cell
MDCK (48) and in NIH 3T3 (67) cell lines (respectively, 185 surface after trafficking through the cell. Thus, in CHO as in
and 170 kDa) and may be the full size rat pro-EGF. The lower NIH 3T3 cells, intracellular post-translational maturations
and minor molecular mass species (136 and 121 kDa) are and trafficking of EGF must be much more rapid than in
45266 Metalloprotease-dependent Regulated Pro-EGF Ectodomain Shedding
MDCK, leading to an increased steady-state level of the mature 50), we did not find any stimulatory effect using two different
precursor at the cell surface. calcium ionophores, namely A23187 and ionomycin. Addition-
As found previously with NIH 3T3 and MDCK cells, pro- ally, we found that, as well as for TGF-␣, both constitutive and
EGF-expressing CHO cells release high molecular mass EGF regulated pro-EGF ectodomain shedding was sensitive to zinc-
into the extracellular medium. Secreted pro-EGF species (one dependent metalloprotease inhibitors (phenantrolin and TAPI-
major 165 kDa and minor 121 and 99 kDa species) are shorter 2). Moreover, the PMA-stimulated pro-EGF ectodomain shed-
in size and appear to directly derive from their membrane ding was very sensitive to inhibition by the metalloprotease
counterpart (respectively, the major 176 kDa and minor 136 inhibitor TAPI-2. The IC50 value found in our study (0.1 ␮M)
and 111 kDa form). Because of the great similarity in the was 10 –100 times less than those currently reported for the
molecular size of cellular and secreted pro-EGF in NIH 3T3, inhibition of membrane protein shedding (23, 25). The key role
Mroczkowsky et al. (67) suggested that soluble pro-EGF might played by a metalloprotease in both constitutive and regulated
be the consequence of an inefficient stop-transfer sequence. pro-EGF shedding was also deduced from secretory experi-
Under these conditions, the non-inserted precursor may repre- ments performed with the mutant CHO-M2 cell line. CHO-M2
sent the secreted form. However, we can rule out this hypoth- were initially selected for their defect in TGF-␣ shedding but
esis since, at least in CHO cells, secreted pro-EGF lacks the were subsequently shown to be defective in the metalloprotease-
intracellular Myc epitope indicating that it has been cleaved at dependent shedding of many cell surface proteins (25, 28, 51).
its C-terminal domain. These shorter soluble species do not A defect that can now be extended to EGF since both constitu-
appear to be synthesized in this state since they were not tive and regulated pro-EGF secretion was completely impaired
observed in cell lysates. Nevertheless, we cannot exclude that in the CHO-M2 cell line compared with CHO-wt.
intracellular processing of the C-terminal domain might lead to We do not have any clear explanation for the inability of
pro-EGF species immediately released into the extracellular MDCK cells to secrete pro-EGF in a regulated manner (48).
medium. However, the fact that following cell surface biotiny- However, since intracellular maturation and/or traffic ap-
lation, pro-EGF released into the extracellular medium is bi- peared to be limiting steps in the course of EGF secretion, an
otinylated does not favor such an interpretation. To our knowl- explanation could be that a too low amount of steady state level
edge, this is the first direct evidence that part if not all of the in cell surface pro-EGF would preclude any detection of cell
secreted pro-EGF arise from the proteolytic cleavage of mem- surface precursor cleavage regulation. In addition, the consti-
brane-anchored pro-EGF exposed at the cell surface. The de- tutive metalloprotease activity of the basolateral membrane
termination of the exact cleavage site was not in the scope of that prevents accumulation of pro-EGF in this cellular domain
this study. However, the size decrease between the different may be already too high to be further increased. At last, the
cellular and soluble pro-EGF species was comprised between possibility that the regulated metalloprotease that cleaves pro-
11 and 15 kDa. This is very close to the predicted value (13–14 EGF in CHO cells may not be present or correctly located
kDa) when making the hypothesis that cleavage of cellular nearby its substrate in MDCK cells cannot be ruled out. This
pro-EGF occurred in the extracellular juxtamembrane domain. hypothesis may also be applied to HB2 cells since they ap-
Thus, our experimental results, if they do not directly demon- peared to have different possibility to regulate pro-EGF shed-
strate the juxtamembrane cleavage of membrane-anchored ding (49). As was stated by the authors, based on a differential
pro-EGF, significantly favors this hypothesis. sensitivity of proHB-EGF and pro-EGF shedding to PMA and
The regulated ectodomain shedding of EGFR ligands such as calcium ionophore, different metalloproteases may be involved
TGF-␣, HB-EGF, and amphiregulin is now clearly established. in the cleavage of EGF versus HB-EGF. Our results demon-
It can be triggered by different agents such as PMA (through strate that CHO cells cleave pro-EGF in a PMA but not calcium-
PKC activation), calcium ionophore (calcium influx), serum, or dependent manner whereas HB2 cells exhibit the opposite reg-
pervanadate (tyrosine phosphatase inhibition) (9, 23, 25, 31, ulation. As stated above, the difference between the two cell
50 –53) and inhibited by zinc-dependent metalloprotease inhib- lines may come from the involvement of different proteases.
itors (24, 25, 28 –31). Concerning EGF, its regulated shedding This discrepancy may not necessarily imply different protease
from cell surface is not so clearly established and experimental populations in the two cell lines, but could be the result of
results are sparse and controversial. To our knowledge, only differential recruitment of specific proteases because of the
two studies have addressed this question. In an extensive study different type of substrates involved in the shedding event. The
performed with MDCK cell line transfected with a full-length deletion of the prodomain would allow the interaction of the
human pro-EGF cDNA-encoding plasmid, the authors failed to substrate with one type of metalloprotease that can be acti-
observe any stimulation of pro-EGF ectodomain cleavage and vated by the calcium-dependent pathway whereas the full-
particularly upon phorbol ester (PMA) or serum treatments length substrate would interact with a metalloprotease acti-
(48). However, in this cellular model, the constitutive basolat- vated through the PMA/PKC pathway. So, in addition to the
eral and apical secretion of pro-EGF is partially sensible to the role that can be played by the stalk region, comparison of the
metalloprotease inhibitor batimastat. In a second study per- results of both studies suggests that the prodomain of pro-EGF
formed with a human mammary gland cell line (HB2) trans- may contribute to the specificity of the interaction between the
fected with a truncated human pro-EGF cDNA construct lack- substrate and the protease(s).
ing most of the prodomain region, EGF secretion was found to An increasing amount of experimental evidence suggests
be stimulated by a calcium ionophore (A23187) and a tyrosine that the ADAM protease TACE may be the sheddase of TGF-␣
phosphatase inhibitor (phenylarsine oxide) but not by phorbol (24, 25, 28), HB-EGF (29), and amphiregulin (29, 31). However,
esters (49). The metalloprotease inhibitor BB-2116 inhibited the recent demonstration of the, in vitro, inability for purified
the calcium ionophore-stimulated EGF release. TACE to cleave synthetic peptide presenting the cleavages
With regard to the above results, our finding that full-length sites of human pro-EGF (both N- and C-terminal) (29) would
rat pro-EGF ectodomain shedding was regulated in CHO-K1 argue against a role for TACE as the pro-EGF sheddase and
was surprising. As for other EGFR ligands, PMA, serum, and a would favor the possibility that different metalloproteases
tyrosine phosphatase inhibitor stimulated shedding. However, could be involved in the release of EGF versus TGF-␣. Our
whereas CHO cells expressing TGF-␣ showed an increased demonstration of the inability of HeLa cells to shed EGF from
shedding upon calcium ionophore treatment (A23187, see Ref. the cell surface in a regulated manner, whereas this cell line
Metalloprotease-dependent Regulated Pro-EGF Ectodomain Shedding 45267
appears to contain high amounts of TACE and secrete TNF-␣ membrane bound proteolytic activity that colocalizes with pro-
also argue against a role for TACE in regulated pro-EGF se- EGF. However, this activity, which is able to release soluble
cretion. This conclusion was reinforced by the observation that EGF from its membrane-associated precursor, belongs to the
wild-type CHO cells we used in this study do efficiently regu- serine protease family and not to the metalloprotease one. In
late pro-EGF ectodomain shedding whereas they contain low order to resolve such a discrepancy, the establishment of a
amounts of TACE and are not able to regulate TNF-␣ secretion, more physiologically relevant cells line from organ cells, which
and that TIMP-3, which is known to be a potent inhibitor of naturally secrete EGF appears necessary and is now under
TACE (72), failed to inhibit pro-EGF secretion. Borroto et al. investigation.
(73) recently showed that the defect in ectodomain shedding of
Acknowledgments—We thank Dr. F. Peiretti from INSERM (EPI
the mutant CHO-M2 cells was the consequence of a defective 99 –36), Marseille, France for providing us with the human TNF-␣
removal of the prodomain that keeps TACE in an inactive form expression plasmid, Dr. Helene Jammes, from INRA Jouy en Josas,
in the early secretory pathway. Thus, our results obtained with France for providing us with the CHO-K1 cell line, Dr. K. Benihoud
this mutant CHO cells are in favor of a pro-EGF shedding from CNRS Orsay, France for the HeLa cell line, and Dr. J. Arribas
from Barcelona, Spain for the mutant CHO-M2 cell line. We particu-
controlled by TACE. However, despite their control experi- larly thank Jocelyne Dujancourt for expert technical assistance.
ments that show a normal processing for some other metallo-
REFERENCES
proteases involved in activated ectodomain shedding, it is pos-
1. Cohen, S. (1962) J. Biol. Chem. 237, 1555–1561
sible that the M2 cells are not only deficient in TACE activity 2. Derynck, R. (1992) Adv. Cancer Res. 58, 27–52
but also in an other unsuspected ADAM or zinc metallopro- 3. Higashiyama, S., Iwamoto, R., Goishi, K., Raab, G., Tanigushi, N., Klagsburn,
tease-related activity, which will be responsible for the con- M., and Mekada, E. (1995) J. Cell Biol. 128, 929 –938
4. Shoyab, M., Plowman, G. D., McDonald, V. L., Bradley, J. G., and Todaro, G. J.
trolled shedding of pro-EGF. We tried to obtain more direct (1989) Science 269, 234 –238
evidences about the involvement of TACE in pro-EGF shedding 5. Shing, Y., Christofori, G., Hanahan, D., Ono, Y., Sasada, R., Igarashi, K., and
Folkman, J. (1993) Science 259, 1604 –1607
using fibroblasts expressing an inactive form of TACE (Tace 6. Toyoda, H., Komurasaki, T., Ikeda, Y., Yoshimoto, M., and Morimoto, S. (1995)
⫺/⫺ cells). Unfortunately, we failed to obtain any expression of FEBS Lett. 377, 403– 407
pro-EGF in this cell line despite the fact that we tested many 7. Carpenter, G. (1987) Annu. Rev. Biochem. 56, 881–914
8. Fantl, W. J., Jonhson, D. E., and Williams, L. T. (1993) Annu. Rev. Biochem.
different transfection protocols and were able to transfect a 62, 453– 481
GFP-containing expression plasmid. 9. Massagué, J., and Pandiella, A. (1993) Annu. Rev. Biochem. 62, 515–541
10. Scott, J., Urdea, M., Sanchez-Pescador, M. Q. R., Fong, N., Rutter, M. S. W. J.,
As was demonstrated previously, shedding of TGF-␣, HB- and Bell, G. I. (1983) Science 221, 236 –240
EGF, and amphiregulin precursors are regulated phenomena 11. Gray, A., Thomas, J. D., and Ullrich, A. (1983) Nature 303, 722–725
12. Rall, L. B., Scott, J., Crawford, J. R., Penshow, D., Niall, H. D., Coghlan, J. P.,
that can be triggered by PMA (through PKC activation), cal- and Bell, G. I. (1985) Nature 313, 228 –231
cium ionophore (calcium influx), serum, or pervanadate (tyro- 13. Saggi, S. S. J., Safirstein, R., and Price, P. M. (1992) DNA Cell Biol. 11,
sine phosphatase inhibition) (9, 23, 25, 31, 50 –53). We have 481– 487
14. Fisher, D. A., Salido, A. C., and Barajas L. (1989) Annu. Rev. Physiol. 51,
been able to clearly demonstrate that pro-EGF shedding is also 67– 80
increased upon PMA, serum, and pervanadate treatment. 15. Hammerman, M. R., O’Shea, M., and Miller, S. B. (1993) Annu. Rev. Physiol.
55, 305–321
However, comparison of the effect of specific PKC inhibitors 16. Beardmore, J. M., and Richards, R. C. (1983) J. Endocrinol. 96, 287–292
and PKC depletion on the different ways of stimulation indi- 17. Carpenter, G. (1985) J. Cell Sci. 3, (suppl.) 1–9
cates that different intracellular pathways may be involved. In 18. Tsutsumi, O., Tsutsumi, A., and Oka, T. (1988) J. Clin. Investig. 81, 1067–1071
19. Ohashi, Y., Mtokura, M., Kinoshita, Y., Mano, T., Watanabe, H., Kinoshita, S.,
fact, the PMA stimulation is completely dependent on the ac- Manabe, R., Oshiden, K., and Yanaihara, C. (1989) Investig. Ophtalmol.
tivation of some PKC activities whereas both serum and per- Vis. Sci. 30, 1879 –1882
20. Van Setten, G. B., Viinikka, L., Tervo, T., Pesonen, K., Tarkkanen, K., and
vanadate stimulation are not. The fact that a specific PKC can Perheentupa, J. (1989) Graefes Arch. Exp. Ophtalmol. 227, 82– 87
be involved in the PMA stimulation of pro-EGF shedding can- 21. Fisher, D. A., and Lakshmanan, L. (1990) Endocrinol. Rev. 11, 418 – 442
22. Schaudies, R. P., Grimes, J., Wray, H. G., and Koldovsky, O. (1990) Am. J.
not be evidenced here but a study on proHB-EGF suggests that Physiol. 259, 1056 –1061
PKC-␦ was the target for the phorbol ester-induced proHB- 23. Hooper, N. M., Karran, E. H., and Turner, A. J. (1997) Biochem. J. 321,
EGF shedding (30). It has been recently suggested that the 265–279
24. Peschon, J. J., Slack, J. L., Reddy, P., Stocking, K. L., Sunnarborg, S. W., Lee,
MAP kinase signaling cascade could be involved in the regula- D. C., Russel, W. E., Castner, B. J., Johnson, R. S., Fitzner, J. N., Boyce,
tion of the ectodomain shedding of both TGF-␣ (53) and HB- R. W., Nelson, N., Kozloski, C. J., Wolfson, M. F., Rauch, C. T., Cerretti,
D. P., Paxton, R. J., March, C. J., and Black, R. A. (1998) Science 282,
EGF (66). Our preliminary results suggest that it may be also 1281–1284
involved in the regulation of pro-EGF shedding activated by 25. Arribas, J., Coodly, L., Vollmer, P., Kishimoto, T. K., Rose-John, S., and
Massague, J. (1996) J. Biol. Chem. 271, 11376 –11382
PMA, serum, and pervanadate. Since both PKC-dependent and 26. Blobel, C. P. (2000) Curr. Opinion Cell Biol. 12, 606 – 612
PKC-independent pathways can activate the MAP kinase path- 27. Black, R. A., and White, J. M. (1998) Curr. Opin. Cell Biol. 10, 654 – 659
way, MAP kinases such as ERK1/2 may be the target of these 28. Merlos-Suarez, A., Fernadez-Larrea, J., Reddy, P., Baselga, J., and Arribas, J.
(1998) J. Biol. Chem. 273, 24955–24962
different types of stimulation. The recent discovery that ERK 29. Sunnarborg, S. W., Hinkle, C. L., Stevenson, M., Russell, W. E., Raska, C. S.,
was able to phosphorylate the cytoplasmic tail of TACE at a Peschon, J. J., Castner, B. J., Gerhart, M. J., Paxton, R. J., Black, R. A., and
Lee, D. C. (2002) J. Biol. Chem. 277, 12838 –12845
specific site strongly suggests that it may modulate TACE 30. Izumi, Y., Hirata, M., Hasuwa, H., Iwamoto, R., Umata, T., Miyado, K., Tamai,
activity through serine phosphorylation (74), and potentially Y., Kurisaki, T., Sehara-Fujisawa, A., Ohno, S., and Mekada., E. (1998)
act in the same way to regulate other metalloproteases. EMBO J. 17, 7260 –7272
31. Brown, C. L., Meise, K. S., Plowman, G. D., Coffey, R. J., and Dempsey, P. J.
In this study, we have clearly demonstrated that as other (1998) J. Biol. Chem. 273, 17258 –17268
members of the EGFR ligand family, when expressed in CHO 32. Pasquini, F., Petris, A., Sbaraglia, G., Scopelliti, R., Cenci, G., and Frati, L.
(1974) Exp. Cell Res. 86, 233–236
cells, EGF is synthesized as a membrane-anchored precursor 33. Scott, J., Patterson, S., Rall, L., Bell, G. I., Crawford, R., Penshow, J., Niall, H.,
present at the cell surface. The entire ectodomain of the pre- and Goghlan, J. (1985) J. Cell Sci. 3, (suppl.) 19 –28
34. Byyny, R. L., Orth, D. N., Cohen, S., and Doyne, E. S. (1974) Endocrinology 95,
cursor can be shed from the cell through a proteolytic cleavage 776 –782
that involved a zinc-dependent metalloprotease activity that 35. Roberts, M. L. (1978) Biochim. Biophys. Acta 540, 246 –252
appears not to be TACE but can be activated by both PKC-de- 36. Breyer, J. A., and Cohen, S. (1990) J. Biol. Chem. 265, 16565–16570
37. Lakshmanan, J., Salido, E. C., Lam, R., Barajas, L., and Fisher, D. A. (1990)
pendent and -independent pathways. However, these experi- Biochem. Biophys. Res. Commun. 173, 902–911
mental results obtained with cultured cells, are in conflict with 38. Salido, E. C., Lakshmanan, J., Fisher, D. A., Shapiro, L. J., and Barajas, L.
(1991) Histochemistry 96, 65–72
those obtained with membrane fractions of both kidney (45, 46) 39. Schaudies, R. P., and Johnson, J. P. (1993) Am. J. Physiol. 264, F523-F531
and mammary gland (47). Indeed, these preparations contain a 40. Brown, C. F., Teng, C. T., Pencost, B. T., and Diaugustine, R. P. (1989) Mol.
45268 Metalloprotease-dependent Regulated Pro-EGF Ectodomain Shedding
Endocrinol. 3, 1077–1083 Chem. 276, 48510 – 48517
41. Maréchal, H., Jammes, H., Rossignol, B., and Mauduit, P. (1999) Am. J. 61. Lee, L. S. (1981) Proc. Natl. Acad. Sci. U. S. A. 78, 1042–1046
Physiol. 276, C734-C746 62. Dempsey, P. J., and Coffey, R. J. (1994) J. Biol. Chem. 269, 16878 –16889
42. Lakshmanan, J., Salido, E. C., Lam, R., and Fisher, D. A. (1992) Am. J. 63. Black, R. A., Rauch, C. T., Kozlosky, C. J., Peschon, J. J., Slack, J. L., Wolfson,
Physiol. 263, E142–E150 M. F., Castner, B. J., Stocking, K. L., Reddy, P., Srinivasan, S., Nelson, N.,
43. Parries, G., Chen, K., Misono, K., and Cohen, S. (1995) J. Biol. Chem. 270, Boiani, N., Schooley, K. A., Gerhart, M., Davis, R., Fitzner, J. N., Johnson,
27954 –27960 R. S., Paxton, R. J., March, C. J., and Cerretti, D. P. (1997) Nature 385,
44. Mauduit, P, Maréchal, H., Van Setten, G., and Rossignol, B. (1998) Proceed- 729 –733
ings of the XIII International Congress of Eye Research, Paris, 1998 64. Moss, M. L., Jin, S. LC, Milla, M. E., Burkhart, W., Carter, H. L., Chen, W. J.,
45. Jorgensen, P. E., Nexo, E., and Poulsen, S. S. (1991) Biochim. Biophys. Acta Clay, W. C., and Didsbury, J. R., Hassler, D., Hoffman, C. R., Kost, T. A.,
1074, 284 –288 Lambert, M. H., Leesnitzer, M. A., McCauley, P., McGeehan, G., Mitchell,
46. Journe, F., Wattiez, R., Piron, A., Carion, M., Laurent, G., Heuson-Stiennon, J., Moyer, M., Pahel, G., Rocque, W., Overton, L. K., Schoenen, F., Seaton,
J., and Falmagne, P. (1997) Biochim. Biophys. Acta 1357, 18 –30 T., Su, J. L., and Becherer, J. D. et al. (1997) Nature 385, 733–736
47. Jahnke, G. D., Chao, J., Walker, M. P., and Diaugustine, R. P. (1994) Endo- 65. Pandiella, A, and Massagué, J. (1991) Proc. Natl. Acad. Sci. U. S. A. 88,
crinology 135, 2022–2029 1726 –1730
48. Dempsey, P. J., Meise, K. S., Yoshitake, Y., Nishikawa, K., and Coffey, R. J. 66. Gechtman, Z, Alonso, J. L., Raab, G., Ingber, D. E., and Klagsbrun, M. (1999)
(1997) J. Cell Biol. 138, 747–758 J. Biol. Chem. 274, 28828 –28835
49. Dong, J., and Wiley, H. S. (2000) J. Biol. Chem. 275, 557–564 67. Mroczkowski, B., Reich, M., Chen, K., Bell, G. I., and Cohen, S. (1989) Mol. Cell
50. Pandiella, A., and Massagué, J. (1991) J. Biol. Chem. 266, 5769 –5773 Biol. 9, 2771–2778
51. Arribas, J., and Massagué, J. (1995) J. Cell Biol. 128, 433– 441 68. Nakagawa, T., Higashiyama, S., Mitamura, T., Mekada, E., and Taniguchi, N.
52. Vecchi, M., Rudolph-Owen, L. A., Brown, C. L., Dempsey, P. J., and Carpenter, (1996) J. Biol. Chem. 271, 30858 –30863
G. (1998) J. Biol. Chem. 273, 20589 –20595 69. Brachman, R., Lindquist, P. B., Nagashima, M., Korh, W., Lipari, T., Napier,
53. Fan, H., and Derynck, R. (1999) EMBO J. 18, 6962– 6972 M., and Derynck, R. (1989) Cell 56, 691–700
54. Davis-Fleische, K. M., Brigstock, D. R., and Besner, G. E. (2001) Growth 70. Wong, S. T., Winchell, L. F., McCune, B. K., Earp, H. S., Teixido, J., Massagué,
Factors, 19, 127–143 J., Herman, B., and Lee, D. C. (1989) Cell 56, 495–506
55. Nishi, E., Prat, A., Hospita, l V., Elenius, K., and Klagsbrun, M. (2001) EMBO 71. Anklesarie, P., Teixido, J., Laiho, M., Pierce, J. H., Greenberger, J. S., and
J. 20, 3342–3350 Massague, J. (1990) Proc. Natl. Acad. Sci. U. S. A. 87, 3289 –3293
56. Maréchal, H., Jammes, H., Rossignol, B., and Mauduit, P. (1996) Am. J. 72. Amour, A., Knight, C. G., English, W. R., Webster, A., Slocombe, P. M.,
Physiol. 270, C1164-C1174 Knäuper, V., Docherty, A. J. P., Becherer, J. D., Blobel, C. P., and Murphy,
57. Meloche, S., Pagčs, G., and Pouysségur, J. (1992) Mol. Cell Biol. 3, 63–71 G. (2002) FEBS Lett. 524, 154 –158
58. Nexo, E., Jorgensen, P. E., Thim L., and Roepstorff, P. (1990) Biochim. Bio- 73. Borroto, A., Ruiz-Paz, S., de la Torre, T. V., Borell-Pagčs, M., Merlos-Suarez,
phys. Acta 1037, 388 –393 A., Pandiella, A., Blobel, C. P., Baselga, J., and Arribas, J. (2003) J. Biol.
59. Borrell-Pagčs, M., Rojo, F., Albanell, J., Baselga, J., and Arribas, J. (2003) Chem. 278, 25933–25939
EMBO J. 22, 1114 –1124 74. Diaz-Rodriguez, E, Montero, J. C., Esparis-Ogando, A., Yuste, L, and Pan-
60. Merlos-Suarez, A., Ruiz-Paz, S., Baselga, J., and Arribas, J. (2001) J. Biol. diella, A. (2002) Mol. Biol. Cell 13, 2031–2044

You might also like