ArticlePDF Available

c-Cb1/Sli-1 regulates endocytic sorting and ubiquitination of the epidermal growth factor receptor

Authors:

Abstract

Ligand-induced down-regulation of two growth factor receptors, EGF receptor (ErbB-1) and ErbB-3, correlates with differential ability to recruit c-Cbl, whose invertebrate orthologs are negative regulators of ErbB. We report that ligand-induced degradation of internalized ErbB-1, but not ErbB-3, is mediated by transient mobilization of a minor fraction of c-Cbl into ErbB-1-containing endosomes. This recruitment depends on the receptor's tyrosine kinase activity and an intact carboxy-terminal region. The alternative fate is recycling of internalized ErbBs to the cell surface. Cbl-mediated receptor sorting involves covalent attachment of ubiquitin molecules, and subsequent lysosomal and proteasomal degradation. The oncogenic viral form of Cbl inhibits down-regulation by shunting endocytosed receptors to the recycling pathway. These results reveal an endosomal sorting machinery capable of controlling the fate, and, hence, signaling potency, of growth factor receptors.
c-Cbl/Sli-1 regulates endocytic sorting
and ubiquitination of the epidermal
growth factor receptor
Gil Levkowitz,
1
Hadassa Waterman,
1,5
Eli Zamir,
2,5
Zvi Kam,
2
Shlomo Oved,
1
Wallace Y. Langdon,
3
Laura Beguinot,
4
Benjamin Geiger,
2
and Yosef Yarden
1,6
Departments of
1
Biological Regulation and
2
Molecular Cell Biology, The Weizmann Institute of Science, Rehovot 76100,
Israel;
3
Department of Pathology, The University of Western Australia, Queen Elizabeth II Medical Center, Nedlands,
Western Australia 6907, Australia;
4
Molecular Oncology Unit, DIBIT, and Instituto di Neuroscienze e Biommagini del CNR,
H.S. Raffaele, Milan 20132, Italy
Ligand-induced down-regulation of two growth factor receptors, EGF receptor (ErbB-1) and ErbB-3, correlates
with differential ability to recruit c-Cbl, whose invertebrate orthologs are negative regulators of ErbB. We
report that ligand-induced degradation of internalized ErbB-1, but not ErbB-3, is mediated by transient
mobilization of a minor fraction of c-Cbl into ErbB-1-containing endosomes. This recruitment depends on the
receptor’s tyrosine kinase activity and an intact carboxy-terminal region. The alternative fate is recycling of
internalized ErbBs to the cell surface. Cbl-mediated receptor sorting involves covalent attachment of ubiquitin
molecules, and subsequent lysosomal and proteasomal degradation. The oncogenic viral form of Cbl inhibits
down-regulation by shunting endocytosed receptors to the recycling pathway. These results reveal an
endosomal sorting machinery capable of controlling the fate, and, hence, signaling potency, of growth factor
receptors.
[Key Words: Endocytosis; ErbB/HER; protein degradation; signal transduction; tyrosine kinase]
Received June 15, 1998; revised version accepted October 6, 1998.
The ErbB family of growth factor receptors carrying an
intrinsic tyrosine kinase activity constitutes a layered
signaling network that includes a large family of EGF-
like ligands that bind to four transmembrane receptors
capable of forming ten homo- and heterodimeric combi-
nations (for review, see Alroy and Yarden 1997). Whereas
the EGF receptor (ErbB-1) binds several growth factors
whose prototype is EGF, both ErbB-3 and ErbB-4 bind all
isoforms of the Neu differentiation factor (NDF). The
third layer of the network includes a large group of sig-
naling molecules sharing one of several types of phos-
photyrosine-binding (PTB) domains (e.g., an SH2 do-
main). Although nonidentical sets of signaling proteins
are recruited, all ErbB receptors, like their invertebrate
orthologs (Perrimon and Perkins 1997), funnel their sig-
nals into the mitogen-activated protein kinase (MAPK)
pathway. The kinetics of MAPK activation and its rela-
tive potency, however, display remarkable differences
that correlate with the rate of ligand-induced endocyto-
sis of receptors, termed down-regulation (Pinkas-Kra-
marski et al. 1996), and recruitment of c-Cbl by the ac-
tivated receptor (Levkowitz et al. 1996).
c-Cbl isthemammalian ortholog of the Sli-1 protein of
Caenorhabditis elegans (Yoon et al. 1995). Genetic evi-
dence indicated that Sli-1 negatively regulates signaling
downstream of the single nematode ErbB protein
(Jongeward et al. 1995). c-Cbl is a major cellular substrate
of tyrosine phosphorylation: It undergoes increased
phosphorylation in response to ligand-induced stimula-
tion of a variety of surface receptors, including the EGF
receptor, lymphokine receptors, immunoglobulin recep-
tors, antigen receptors, and integrin receptors (Thien and
Langdon 1997, and references therein). Overexpression
of c-Cbl attenuates signaling down-stream of the immu-
noglobulin E receptor (Ota and Samelson 1997) and the
T-cell receptor (Boussiotis et al. 1997), yet the mecha-
nism of Cbl action remains unknown. Here we report
that c-Cbl can increase the rate of degradation of ErbB-1,
but not ErbB-3. The underlying mechanism involves
transient physical associations between c-Cbl and
ErbB-1 in endosomes, and subsequent ubiquitination of
the degradation-destined receptors. An oncogenic viral
Cbl appears to interfere with the sorting function of c-
Cbl, thereby directing incoming receptors to the recy-
cling pathway.
5
These authors contributed equally to this work.
6
Corresponding author.
E-MAIL liyarden@weizmann.weizmann.ac.il; FAX 972-8-9344116.
GENES & DEVELOPMENT 12:3663–3674 © 1998 by Cold Spring Harbor Laboratory Press ISSN 0890-9369/98 $5.00; www.genesdev.org 3663
Results
c-Cbl mediates selective degradation
of ligand-stimulated ErbB-1, but not ErbB-3
To investigate the possibility that EGF-driven ErbB-1 is
destined to lysosomal degradation because ErbB-1 can
interact with c-Cbl (Levkowitz et al. 1996), whereas
ErbB-3 is shunted tothe recycling pathway (Waterman et
al. 1998) because it cannot recruit c-Cbl, we transiently
overexpressed c-cbl and erbB-1 in Chinese hamster
ovary (CHO) cells. In addition to c-Cbl, we used two
deletion mutants that are schematically presented in
Figure 1A. These are a peptide-tagged amino-terminal
portion of c-Cbl analogous to the murine viral form, v-
Cbl, and the complementary deletion mutant, Cbl-C
(Fig. 1A). Cells were briefly stimulated with EGF and
tyrosine phosphorylation of the receptor analyzed by im-
munoblotting with anti-phosphotyrosine antibodies.
The results of this experiment indicated that ErbB-1 un-
derwent enhanced tyrosine phosphorylation in the pres-
ence of its ligand (10-fold), but c-Cbl overexpression
almost abolished this effect (Fig. 1B). Neither v-Cbl nor
Cbl-C were active, implying that the combination of
amino- and carboxy-terminal sequences is essential for
the effect of c-Cbl on receptor phosphorylation.
Reblotting with anti-ErbB-1 antibodies revealed that
c-Cbl overexpression led to degradation, rather than
catalytic inactivation, of ErbB-1 (Fig. 1B, bottom): prima-
rily the hyperphosphorylated form ofErbB-1, whose elec-
trophoretic mobility is retarded, was diminished (by
80%). This observation suggested that tyrosine phos-
phorylation of the surface-localized ErbB-1 molecules se-
lectively targets ErbB-1 to degradation through c-Cbl ac-
tion. Labeling of cell surface-exposed ErbB-1 molecules
by surface biotinylation confirmed this scenario (Fig.
1C): Overexpression of c-Cbl enhanced disappearance of
the hyperphosphorylated form of ErbB-1. Concomitant
with selective removal from the cell surface, signaling
downstream of ErbB-1 was down-regulated by c-Cbl.
This was exemplified by the ability of the ligand-stimu-
lated ErbB-1 to recruit one of its major substrates, the
Shc protein (Fig. 1C, bottom). In contrast with the EGF
receptor, the NDF-receptor (ErbB-3) was not affected by
c-Cbl (Fig. 1B). In conclusion, c-Cbl can down-regulate
ErbB-1 signaling, apparently by selective degradation of a
hyperphosphorylated form of the receptor.
Additional support for this model was provided by
analyses of ligand-induced redistribution of c-Cbl. Con-
sistent with the observation that EGF can induce physi-
cal association of its receptor with c-Cbl in living cells
(Bowtell and Langdon 1995; Galisteo et al. 1995; Meisner
et al. 1995; Tanaka et al. 1995), EGF treatment of cells
co-overexpressing ErbB-1 and ErbB-3 led to rapid changes
in the pattern of c-Cbl subcellular localization (Fig. 2A).
Whereas in untreated cells c-Cbl exhibited reticular/ve-
sicular distribution, it assumed a more punctate, vesicu-
lar-like, pattern in EGF-treated cells. In fact, some,
but not all, c-Cbl-bearing structures contained endocy-
tosed molecules of ErbB-1. No such redistribution of c-
Cbl, or ErbB-3, was noted on treatment with NDF (Fig.
Figure 1. c-Cbl, but not v-Cbl, in-
creases degradation of ErbB-1 by its re-
moval from the cell surface. (A) Shown
are the domain structures of c-Cbl and
three derivative proteins. The following
structural motifs are represented: A 7
residue-long histidine stretch (7 His), a
positively charged basic domain (Basic),
a ring-finger domain (RF), a proline-rich
domain (Pro-Rich), and a leucine-zipper
(LZ). An influenza virus hemagglutinin
(HA) epitope tag was added to the
amino-terminal end of each protein.
cDNAs corresponding to the three natu-
ral forms of Cbl were transiently trans-
fected into CHO cells. Forty-eight hours
after transfection, cells were lysed and
whole cell lysates subjected to immuno-
blotting (IB) with an anti-HA antibody.
(B)ErbB-1(left)orErbB-3(right) were
transiently expressed in CHO cells by
cotransfection with plasmids encoding
the indicated Cbl proteins, or with a
control empty plasmid (Cont.). Cells were incubated for 45 min at 37°C with EGF or NDF (each at 100 ng/ml). Thereafter, whole cell
lysates were subjected to immunoprecipitation (IP) and immunoblotting (IB) with the indicated antibodies. (C) CHO cells were
co-transfected with an ErbB-1 expression vector together with a vector directing expression of c-Cbl or a control empty vector (Cont.)
Cell surface-exposed proteins were covalently labeled with biotin at 4°C. Soluble biotin was then removed and cells incubated at 37°C
for 45 min in the presence or absence of EGF. Cell lysates were subjected to immunoprecipitation (IP) with an anti ErbB-1 antibody
and the electrophoretically resolved proteins were probed either with horseradish peroxidase-conjugated Streptavidin, or with an
anti-Shc antibody. The locations of molecular mass markers are indicated in kilodaltons. Note that the three Shc isoforms (p66, p52,
and p46) associate with ErbB-1.
Levkowitz et al.
3664 GENES & DEVELOPMENT
2A). This observation led us to compare the effects of
c-Cbl with the abilities of the two ligands to down-regu-
late their binding sites. To this end, we stably expressed
c-Cbl, or v-Cbl, in cells already overexpressing ErbB-1.
Although the introduced Cbl proteins were detectably
expressed (Fig. 2B, bottom), c-Cbl- overexpressing cells
displayed a significantly lower amount of the ErbB-1 pro-
tein (Fig. 2B, left) and a six-fold decreased number of
EGF-binding sites relative to control- or v-cbl-trans-
fected cells (Fig. 2B, right). This basal down-regulation of
ErbB-1 was displayed by several independently selected
clones, and severely reduced the sensitivity of receptor
down-regulation assays. Therefore, we used transient co-
expression of a receptor and c-Cbl. Because of a high
level of receptor expression, EGF treatment of ErbB-1-
expressing cells led to only a moderate down-regulation
effect (Fig. 2C). The extent of ligand-induced down-regu-
lation displayed variation that apparently correlates with
receptor expression, but in all experiments receptor dis-
appearance was significantly enhanced on overexpres-
sion of c-Cbl (Fig. 2C). This observation indicates that
the amount of c-Cbl limits down-regulation when
ErbB-1 is overexpressed. In contrast, NDF treatment of
ErbB-3-expressing cells did not result in receptor down-
regulation, and c-Cbl exerted no significant effect on the
status of NDF-binding sites (Fig. 2C). In conclusion, c-
Cbl can enhance down-regulation of the EGF-receptor,
but not the NDF receptor, and this involves redistribu-
tion of c-Cbl into endocytic ErbB-1-containing vesicles.
Transient ligand-induced colocalization of c-Cbl
and ErbB-1 in endosomes
Because partial colocalization of ErbB-1 and c-Cbl was
observed on a 20-min-long incubation of cells with EGF
(Fig. 2A), we analyzed the kinetics of the phenomenon.
The results presented in Figure 3A demonstrate redistri-
bution of ErbB-1 and c-Cbl from their initial sites (mem-
branal and reticular, respectively), and partial colocaliza-
tion that peaks at 10–20 min. Concomitant with redis-
tribution, the double-stained vesicular structures
exhibited an apparent increase in fluorescence intensity.
Figure 2. c-Cbl colocalizes with ErbB-1, but not with ErbB-3, and accelerates its down-regulation. (A) CHO cells that stably co-
overexpress ErbB-1 and ErbB-3 (Tzahar et al. 1996) were plated on cover-slips and treated at 37°C with EGF (top)orNDF(bottom)for
the indicated periods of time. After fixation and permeabilization, cover slips were simultaneously stained with a polyclonal anti-Cbl
antibody and a mAb to ErbB-1 or to ErbB-3, as indicated. Antibody detection by immunofluorescence was performed as described
(Materials and Methods). (B) Stable CHO transfections expressing c-Cbl or v-Cbl were established by cotransfection of Cbl constructs
together with the pBABE/puro plasmid into CHO cells overexpressing ErbB-1 (Tzahar et al. 1996). Individual clones were screened by
immunoblotting with antibodies to ErbB-1 and to the HA tag of Cbl proteins (left, the locations of c-Cbl and v-Cbl protein bands are
indicated by arrows). To assess cell surface expression of ErbB-1, clones expressing c-Cbl or v-Cbl, as well as the parental ErbB-1
overexpressing cell line (−), were incubated for 90 min at 4°C with radiolabeled EGF (10 ng/ml). Cell-bound radioactivity is shown as
the average and range (bars) of duplicate determinations. (C) CHO cells were cotransfected with pairs of two plasmids: an ErbB-
expression vector and either a control empty pcDNA3 plasmid (; ErbB-1; , ErbB-3), or a c-Cbl expression vector (, ErbB-1; ,
ErbB-3). Cell monolayers were subjected to a down-regulation assay 48 hr post-transfection. The results are expressed as the average
fraction (and range, bars) of original binding sites that remained on the cell surface after exposure to the nonlabeled ligand at 37°C.
Endosome sorting by Cbl
GENES & DEVELOPMENT 3665
This process was further analyzed by use of computer-
ized analysis of digital images. First, we defined the bor-
ders of each c-Cbl- containing particle, and then sepa-
rately determined the fluorescence intensity of c-Cbl and
ErbB-1 within the framed two-dimensional structure.
When this was repeatedly performed at different time
intervals, we obtained the results presented in Figure 3B,
in which each dot represents one vesicular structure. In
untreated cells, labeling for c-Cbl in individual particles
was very low, but the cells contained relatively large
numbers of small particles. Following 10 min of incuba-
tion, a coordinated increase in ErbB-1 and c-Cbl labeling
became evident and it displayed a linear correlation co-
efficient of 0.83. Colocalization was maximal after 20
min of incubation (r = 0.87) and then gradually declined.
Analysis of the total number of intracellular vesicles/
particles per cell (data not shown), as well as the extent
of colocalization (Fig. 3B, right) revealed a 25% decrease
in the average density of c-Cbl-positive particles at 10
and 20 min. The number of ErbB-1 vesicles gradually
increased and reached a maximal level at 10 min. Impor-
tantly, the maximal number of ErbB-1-positive vesicles
was much lower than the number of c-Cbl-positive
vesicles, suggesting that only a small subpopulation of
c-Cbl-rich vesicles contained endocytosed ErbB-1 mol-
ecules. This was confirmed by direct evaluation of the
extent of colocalization: Approximately 15% of c-Cbl-
positive vesicles also contained ErbB-1, implying that
nearly all of the ErbB-1-containing vesicles became as-
sociated with c-Cbl.
Biochemical analyses that involved labeling of surface
ErbB-1 molecules with biotin, or selective immunopre-
cipitation of the membranal EGF receptors, indicated
that only the endocytosed receptor fraction physically
recruited c-Cbl (data not shown), consistent with the en-
dosomal site of interaction implied by the immunofluo-
rescence results. Indeed, separation of the endosomal
fraction from CHO cells by use of the method described
previously (Wada et al. 1992) and Rab-5 as an endosome
marker revealed that c-Cbl is a resident protein of the
endosome (Fig. 3C). In addition, physical interaction be-
tween c-Cbl and ErbB-1 was detectable in the endosomal
fraction only if cells were treated with EGF prior to lysis
and subcellular fractionation. c-Cbl and ErbB-1 redistri-
Figure 3. Time dependence of EGF-induced ErbB-1 and c-Cbl co-localization in endosomes. (A) CHO cells that coexpress ErbB-1 and
ErbB-3 were either fixed (0 min) or first incubated with EGF for 5 min at 37°C. Thereafter, EGF was removed and incubation continued
for the indicated time intervals. Double staining of ErbB-1 and c-Cbl was performed and visualized as described in Materials and
Methods. (B, left) For each time point, digital images of three representative cells were segmented according to the labeling of Cbl. The
fluorescence of c-Cbl and ErbB-1 labeling was calculated for each segmented vesicle. The scatter plots (arbitrary units) present ErbB-1
fluorescence vs. the c-Cbl fluorescence in each segmented vesicle. The correlation coefficient (r) indicates the strength of a linear
correlation between ErbB-1 and c-Cbl fluorescence. (B, right) Cbl-segmented vesicles, which showed ErbB-1 positivity above a thresh-
old, were considered Cbl-vesicles with ErbB1. (Top) Percentage of those vesicles from the total number of Cbl-segmented vesicles at
each time point. (Bottom) Percentage of ErbB-1 vesicles with Cbl from the total number of ErbB1-segmented vesicles. (C) CHO cells
were cotransfected with ErbB-1 and c-Cbl expression vectors and cells were incubated for 15 min at 37°C with EGF. Control
monolayers were mock stimulated (−). Endosomes were prepared as described in Materials and Methods and solubilized (1% Triton
X-100) for 30 min at 4°C. Cell lysates were cleared and subjected to immunoprecipitation (IP) and immunoblotting (IB) with the
indicated antibodies. Both the endosomal marker protein Rab-5 and c-Cbl were significantly enriched in the isolated fraction relative
to other fractions that werecollected (bottom;data notshown). (D) CHOcells werecotransfected withErbB-1 andan expressionvector
encoding c-Cbl fused inframe to a green fluorescence protein(GFP-Cbl). Forty-eighthours after transfection, cells were incubatedwith
Texas-red-labeled EGF (0.5 µg/ml) for 30 min at 4°C and then either transferred to 37°C (right), or left at 4°C (left), for an additional
incubation of 15 min.
Levkowitz et al.
3666 GENES & DEVELOPMENT
bution was independently supported by use of a fluores-
cently labeled EGF, andc-Cbl fused to a GFP. The results
presented in Figure 3D confirmed that the ligand-occu-
pied receptor molecules colocalize with Cbl-containing
vesicles.
c-Cbl and v-Cbl differently affect sorting
of endocytosed ErbB-1 molecules
Determination of the rate of intracellular accumulation
of EGF revealed that neither c-Cbl nor v-Cbl affected the
rate of ligand uptake (Fig. 4A), implying that Cbl acts at
a step distal to receptor binding and coated pit-mediated
internalization. This conclusion was also supported by
short time (1–5 min) ligand uptake experiments, and
analyses of the rate of EGF degradation, which was not
affected by c-Cbl or v-Cbl (data not shown). On the other
hand, examination of the receptor’s fate, by use of a
down-regulation assay indicated that c-Cbl, unlike
v-Cbl, significantly accelerated the rate of receptor
down-regulation (Fig. 4B). A surprising effect of v-Cbl,
however, was revealed on the background of the exten-
sive c-Cbl-induced down-regulation (Fig. 4B). Following
a rapid phase of partial ligand-induced down-regulation,
v-Cbl caused an almost complete recovery of ErbB-1 to
its original surface level. This effect of v-Cbl was sensi-
tive to monensin (Fig. 4B, right), a carboxylic ionophore
that exerts diverse intracellular effects, including disrup-
tion of recycling of EGF-occupied ErbB-1 molecules after
their endocytosis (Basu et al. 1981). Conceivably,
whereas c-Cbl can direct ErbB-1-loaded endosomes to
degradation, v-Cbl antagonizes this sorting function, and
directs the receptors to the alternative recycling path-
way.
Kinase activity and an intact carboxy-terminal region
are obligatory for Cbl-induced down-regulation
of ErbB-1
If c-Cbl plays a causative role in receptor down-regula-
tion, as implied by our results, then its interaction with
ErbB-1 may depend on kinase activity and autophos-
phorylation sites. This question was addressed by coex-
pressing a series of ErbB-1 mutant proteins, together
with c-Cbl, in CHO cells. The mutant proteins, along
with a wild-type receptor, are schematically depicted in
Figure 5A. They include a kinase-defective form (Kin
), a
deletion mutant lacking 214 amino acids of the carboxy-
terminal region (denoted Dc-214), and a protein whose
five major tyrosine autophosphorylation sites were re-
placed by phenylalanines (denoted F5). In addition, we
analyzed a receptor mutated at tyrosine 974 (denoted
A974) because this site has been implicated in the inter-
actions of ErbB-1 with the AP-2 adaptor of clathrin-
coated pits (Sorkin et al. 1996). Likewise, a receptor mu-
tated at both tyrosine residues 974 and 992 (denoted
Y5,6A) was analyzed because these sites are homologous
to the reported Cbl-docking site of the ZAP-70 tyrosine
kinase (Lupher et al. 1997). Transient expression of the
mutated proteins in CHO cells resulted in receptor lev-
els similar to that of the wild-type ErbB-1 (Fig. 5A). The
following aspects of Cbl interaction with ErbB-1 were
analyzed by use of this cellular system: ligand-induced
physical association, as well as tyrosine phosphoryla-
tion, of c-Cbl, recruitment of Shc, and receptor down-
regulation. The resultsof these assays are summarized in
a table (Fig. 5A), and presented in Figure 5, B and C. The
results obtained with Y5,6A are presented only in the
table.
Evidently, two features of ErbB-1 are essential for pro-
Figure 4. v-Cbl promotes receptor recycling,
whereas c-Cbl induces receptor down-regula-
tion. (A) Ligand internalization analyses. CHO
cells were cotransfected with an ErbB-1 vector
along with one of the following plasmids:
pcDNA3 (control, ), c-Cbl expression vector
(), or a plasmid directing v-Cbl expression
(). Cell monolayers were treated for 2 hr at
4°C with
125
I-labeled EGF (at 10 ng/ml) and
then transferred to 37°C for the indicated pe-
riods of time. The fraction of internalized li-
gand was determined by use of a low-pH wash.
Each data point represents the average ±
S.E.
(bars) of triplicate measurements. (B) CHO
cells were cotransfected withan ErbB-1-encod-
ing plasmid along with an expression vector
encoding v-Cbl (), c-Cbl (), both v- and c-
Cbl (), or with an empty vector (control, ).
Cells were rinsed and incubated at37°Cfor the
indicated periods of time with EGF (at 250 ng/
ml). Sister cultures were similarly treated, ex-
cept that monensin (100 µ
M) was added to the
medium. Down-regulation assays were per-
formed as described in Materials and Methods.
Endosome sorting by Cbl
GENES & DEVELOPMENT 3667
ductive interaction with c-Cbl. These are the intrinsic
tyrosine kinase activity, and the presence of an intact
carboxy-terminal region. The five autophosphorylation
sites of ErbB-1 arenot crucial for the interactionbetween
c-Cbl and ErbB-1 (Fig. 5), although they were essential for
physical association with another ErbB-1 substrate,
namely Shc (Fig. 5B). In accordance with their inability
to interact with c-Cbl, the Kin
and the Dc214 mutants
displayed markedly reduced ligand internalization and
down-regulation (Sorkin et al. 1992; Chang et al. 1993).
We note that in some experiments basal association be-
tween c-Cbl and akinase-defective mutant of ErbB-1 was
detectable (cf. Figs. 5B and 7B), but in no case was it
ligand dependent. Interestingly, the carboxy-terminally
deleted mutant of ErbB-1 mediated both prolonged acti-
vation of the MAPK pathway (data not shown) and en-
hanced tyrosine phosphorylation of c-Cbl (Fig. 5B, bot-
tom). Nevertheless, it was refractory to the c-Cbl-in-
duced down-regulation effect (Fig. 5C), implying that
c-Cbl phosphorylation is insufficient for degradation of
Figure 5. Structural determinants of ErbB-1 that are essential for functional interactions with c-Cbl. (A) Schematic representation of
ErbB-1 mutants and their interactions with c-Cbl. The domain structure of ErbB-1 is shown by boxes that correspond to the double
cysteine-rich domain of the extracellular (EC) region, the transmembrane domain (TM), the juxtamembrane domain (JM), the tyrosine
kinase (TK) domain, and the carboxy-terminal tail (CT). The five major tyrosine autophosphorylation sites, along with the -adaptin
tyrosine-based internalization signal [Y
974
; (Sorkin et al. 1996)] are indicated. The ATP-binding lysine residue (K
721
) was mutated to
an alanine residue in the kinase-defective mutant (Kin
). A carboxy-terminal deletion mutant (Dc214) lacking 214 carboxy-terminal
amino acids, and an ErbB-1 mutant in which the five major tyrosine phosphorylation sites were mutated to phenylalanine (F5) have
been described previously (Sorkin et al. 1996). A double tyrosine to alanine mutant (Y5,6) is also shown. A summary of the results
shown in B and C is presented in the table. The histogram presents the results of an assay that determined the binding of radioactive
EGF to the surface of cells transiently expressing the indicated mutants. (B) Monolayers of CHO cells were separately cotransfected
with plasmids encoding the indicated ErbB-1 mutants together with a c-Cbl-encoding vector. Sister plates were incubated for 15 min
at 37°C with or without EGF (at 100 ng/ml). Thereafter, whole cell lysates were subjected to immunoprecipitation (IP) and immu-
noblotting (IB) withthe indicated antibodies. (C) Theindicated ErbB-1 mutants were introduced into CHO cells by cotransfection with
a control vector () or a c-Cbl plasmid (). EGF-induced down-regulation assay was then performed.
Levkowitz et al.
3668 GENES & DEVELOPMENT
ErbB-1. On the other hand, complex formation between
c-Cbl and ErbB-1 correlated with down-regulation (Fig.
5A), suggesting that stable physical association of c-Cbl
with the receptor is critical for directing ErbB-1 to deg-
radation.
c-Cbl increases ligand-induced ubiquitination
of ErbB-1
Next, we addressed the effect of c-Cbl on receptor deg-
radation by using chloroquine, an inhibitor of prelyso-
somal/lysosomal proteolysis. Chloroquine exerted no ef-
fect on the limited ability of EGF to induce degradation
of an overexpressed ErbB-1 in CHO cells (20% of ErbB-1
molecules underwent degradation following EGF treat-
ment, regardless of chloroquine presence, Fig. 6A). Co-
overexpression of c-Cbl significantly enhanced ligand-
induced degradation of ErbB-1 (80% of ErbB-1 molecules
underwent degradation), but chloroquine was able to
partly attenuate this Cbl-mediated enhanced degrada-
tion (only 44% of ErbB-1 molecules underwent degrada-
tion). Conceivably, c-Cbl affects receptor processing up-
stream to the chloroquine-sensitive late endosomal step.
Because ligand-induced ubiquitination of ErbB-1 pre-
cedes its intracellular degradation, and it depends on en-
docytosis of the ligand-receptor complexes (Galcheva-
Gargova et al. 1995), we analyzed the ability of c-Cbl to
affect receptor ubiquitination. In line with previous re-
ports, we detected a ligand-induced increase in ErbB-1
ubiquitination (Fig. 6B). By overlapping anti-ErbB-1 im-
munoblots with the ubiquitin signals, we learned that
the ubiquitinated fraction of ErbB-1 was minor (<5%,
open arrow in Fig. 6B). Overexpression of c-Cbl dramati-
cally increased the amount of ubiquitin that underwent
covalent attachment to ErbB-1 (Fig. 6B), implying that
the cellular level of Cbl critically controls the extent of
receptor ubiquitination.
In experiments that are not presented, we found that
treatment of cells with an inhibitor of proteasomal ac-
tivity, MG132, increased the extent of receptor ubiqui-
tination in cellsoverexpressing c-Cbl. Therefore, we ana-
lyzed the effect of MG132 on the fraction of surface-
exposed receptors by using a down-regulation assay. The
presence of MG132 decreased not only the basal EGF-
induced disappearance of ligand binding sites, but also
the accelerated down-regulation that was promoted by
an overexpressed c-Cbl (Fig. 6C). Because inhibition of
either c-Cbl (Fig. 4B) or proteasomes (Fig. 6C) can en-
hance recycling of ErbB-1, butblocking lysosomal hydro-
lases allows no recycling (data not shown), it is likely
that c-Cbl acts upstream to the proteasomal and lyso-
somal degradation processes.
Ligand-dependent ubiquitination corresponds
to the down-regulation activity of mutant Cbl
and ErbB-1 proteins
The contention that c-Cbl canaccelerate the degradation
rate of receptor molecules by increasing their ubiquiti-
nation predicts that mutant Cbl proteins, which are un-
able to down-regulate ErbB-1, will be defective in induc-
ing receptor ubiquitination. The two oncogenic forms of
c-Cbl, v-Cbl, and 70Z-Cbl, a deletion mutant lacking 17
internal amino acids (Langdon et al. 1989), were either
unable to enhance receptor down-regulation and ubiqui-
tination or elevated receptor expression (Fig. 7A). Like-
Figure 6. c-Cbl-induced down-regulation in-
volves an increase in ErbB-1 ubiquitination.
(A) Chloroquine sensitivity. The wild-type
form of ErbB-1was expressed in CHO cells by
cotransfection of an erbB-1-encoding plasmid
together with either a c-Cbl-expression vec-
tor or an empty vector (Cont.). Cells were in-
cubated for 45 min at 37°C in the absence or
presence of EGF (at 100 ng/ml) and chloro-
quine (CQ, 0.1 m
M). Cell lysates were pre-
pared and subjected to immunoprecipitation
(IP) and immunoblotting (IB)with anti-ErbB-1
antibodies. (B) CHO cells were transfected
and treated as in A. Cell lysates were sub-
jected to immunoprecipitation (IP) with anti-
bodies to ErbB-1 and immunoblotting (IB)
with antibodies to either ubiquitin (Ub) or
ErbB-1. (Closed arrowheads) The major band
of ErbB-1; (open arrowheads) the minor frac-
tion that underwent ubiquitination. (C)
ErbB-1 was transiently expressed in CHO
cells by cotransfection with either an empty
expression vector (control, ) or a c-Cbl ex-
pression vector (). EGF-induced down-regu-
lation of ErbB-1 was determined in the pres-
ence or absence of the proteasomal inhibitor
MG132 (10 µ
M).
Endosome sorting by Cbl
GENES & DEVELOPMENT 3669
wise, mutant ErbB-1 proteins that cannot stably recruit
c-Cbl displayed no ubiquitination. These are a kinase-
defective receptor, and a carboxy-terminally deleted
ErbB-1 (Fig. 7B). However, a receptor whose five major
autophosphorylation sites were mutated (F5) retained
enhanced ubiquitination (data not shown), in line with
its ability to recruit c-Cbl (Fig. 5). Because the defective
mutants were unable to associate with c-Cbl and accel-
erate receptor down-regulation and degradation (Figs. 5
and 7B), we concluded that functional interaction with
c-Cbl is necessary for receptor degradation, down-regu-
lation, and ubiquitination. The relative order of these
processes and their presumed compartmentalized orga-
nization are discussed below and summarized in a model
(Fig. 8).
Discussion
Endocytosis of the ErbB-1 is one of the best-characterized
routes of induced internalization of ligand-receptor com-
plexes (for review, see Sorkin and Waters 1993; Trow-
bridge et al. 1993). The most rapid endocytic pathway,
which utilizes clathrin-coated pits and vesicles, is char-
acterized by saturability: The rate of internalization de-
creases with increasing receptor occupancy (Lund et al.
Figure 7. Effect of Cbl proteins on ubiq-
uitination of ErbB-1 and its mutants. (A)
CHO cells were cotransfected with a plas-
mid encoding ErbB-1 together with vectors
directing the expression of the indicated
Cbl proteins. An empty vector was used
for control (Cont.). Cell monolayers were
treated for 15 min at 37°C with EGF (100
ng/ml). Thereafter, we eitheranalyzed cell
lysates by immunoprecipitation (IP) and
immunoblotting (IB) with the indicated
antibodies (left, arrowheads are as in Fig. 6)
or performed a ligand-binding assay as de-
scribed in the legend to Fig. 2B. (B) Mono-
layers of CHO cells were transfected with
a plasmid expressing c-cbl or a control
empty vector (−) together with vectors en-
coding the wild-type form (WT) of ErbB-1,
or the indicated mutants. Cell monolayers
were treated with EGF as in A and their
whole lysates subjected to immunopre-
cipitation (IP) with an antibody directed to
the extracellular portion of ErbB-1. Immunoblotting (IB) was performed with an antiserum to ubiquitin, or with an antibody directed
to the most carboxy-terminal 14 amino acids of ErbB-1. To confirm expression of the Dc214 mutant of ErbB-1, which was not
recognized by the immunoblotting antibody, we performed a ligand-binding assay on living cells (data not shown).
Figure 8. Proposed model of ligand-in-
duced endocytosis of ErbB-1. The model
summarizes the major steps of receptor
endocytosis and indicates their presumed
time scale. Ligand binding to ErbB-1 mol-
ecules, probably by elevating autophos-
phorylation (encircled P), induces their in-
teractions with clathrin-coated areas of
the plasma membrane, which rapidly in-
vaginate to form coated pits. c-Cbl may
not affect excision of the pit to form a
coated vesicle and the subsequent rapid
uncoating process. c-Cbl recruitment to
endosome-located ErbB-1 molecules tags
them for ubiquitination (Ub) and subse-
quent degradation through the combined
action of prelysosomal/lysosomal acid hy-
drolases, as well as by proteasomal pro-
teinases. v-Cbl shunts receptors to the de-
fault pathway, which involves recycling of
vesicles back to the cell surface. This step
is inhibitable by monensin.
Levkowitz et al.
3670 GENES & DEVELOPMENT
1990). Another limiting step is the sorting of internalized
receptors to lysosomal degradation (French et al. 1994).
On transient overexpression of ErbB-1 in CHO cells (2–
6×10
5
receptors/cell) the rapid endocytic pathway is
practically saturated and very low receptor down-regula-
tion occurs (Fig. 2C). Overexpression of c-Cbl on this
cellular background revealed a Cbl-dependent limiting
step that can regulate receptor degradation (Figs. 2C and
4B). Similarly, the negative regulation by c-Cbl of an-
other tyrosine kinase, Syk, was detectable only on Cbl
overexpression with recombinant vaccinia constructs
(Ota and Samelson 1997).
Down-regulation is the net result of receptor degrada-
tion and recycling. Because the rate of EGF internaliza-
tion was not detectably affected by c-Cbl overexpression
(Fig. 4A), but receptor down-regulation was accelerated
by c-Cbl (Fig. 4B), we propose that Cbl acts at a post-
coated pit step. In agreement with this difference, a ki-
nase-defective ErbB-1 mutant, which cannot interact
with Cbl, retained the ability to internalize EGF mol-
ecules and escaped degradation (Felder et al. 1990). The
observation that a co-overexpressed v-Cbl can reverse
c-Cbl’s action by shunting internalized receptors to a
monensin-sensitive pathway (Fig. 4B) led us to the propo-
sition that the recycling endosome (Trowbridge et al.
1993) is the site of Cbl’s action. In support of this possi-
bility, c-Cbl was localized to a vesicular compartment
(Figs. 2A and 3A) and it was fractionated with an endo-
somal fraction (Fig. 3C). Because receptor ubiquitination
was enhanced by c-Cbl (Fig. 6), and its structural require-
ments appear to reflect c-Cbl recruitment (cf. Figs. 7 and
5A), it is conceivable that the sorting function of c-Cbl
involves receptor ubiquitination. The scheme presented
in Figure 8 incorporates these conclusions into a model
that integrates data from the present and previous stud-
ies. According to this model, c-Cbl is not involved in the
entrapment of EGF receptors by coated regions of the
plasma membrane, and in the subsequent rapid invagi-
nation and scission that form coated pits and coated
vesicles, respectively. However, 1–2 min after binding
of EGF to thecell surface, c-Cbl becomes associatedwith
ErbB-1-containing vesicles (Fig. 3). On the basis of quan-
tification of fluorescent images, we assume that all ma-
ture ErbB-1-containing vesicles associate with c-Cbl, but
this recruitment engages only a small fraction of the cel-
lular pool of Cbl (Fig. 3B).
How exactly c-Cbl is recruited by ErbB-1 remains un-
clear. One possiblity that we have not addressed is the
involvement of several lysosome targeting motifs lo-
cated at the carboxy-terminal region of ErbB-1 (Opresko
et al. 1995). Although none of five tyrosine autophos-
phorylation sites located at this region appears essential
for Cbl-ErbB-1 interactions, we cannot exclude the pos-
sibility that tyrosine autophosphorylation of ErbB-1 also
plays a role in c-Cbl recruitment. For example, phos-
phorylation of tyrosine residues we have not mutated
may allow ErbB-1 to bind to c-Cbl either directly or in-
directly. Because c-Cbl isfound in living cells complexed
to several proteins, like Crk, phosphatidylinositol 3-ki-
nase, and the Grb2 adaptor protein (Meisner et al. 1995),
it is possible that c-Cbl/Grb2, or other complexes, asso-
ciate with the carboxy-terminal region of ErbB-1 through
compensatory tyrosine phosphorylation sites.
Biochemical and morphological studies that utilized a
kinase-defective mutant of ErbB-1 revealed causal rela-
tionships between receptor occupancy, kinase activity,
and the rate of internalization (Honegger et al. 1987;
Chen et al. 1989; Hopkins et al. 1990). However, the role
played by the catalytic activity of ErbB-1 in lysosomal
targeting is currently unclear. According to one possibil-
ity, targeting is independent of kinase activity (Wiley et
al. 1991), but an alternative model implies that the in-
trinsic kinase can interrupt receptor recycling (Felder et
al. 1990). According to this scenario, the early endocytic
pathways of wild-type and kinase-impaired receptors are
identical, but after 10–20 min the pathways diverge at
the multivesicular body (MVB): Wild-type ErbB-1, des-
tined for degradation, localizes to internal vesicles,
whereas kinase-defective ErbB-1, destined for recycling,
localizes to surface membranes of the MVBs. The coin-
cidental peak of c-Cbl recruitment by ErbB-1-containing
endosomes (Fig. 3), together with the ability of c-Cbl to
promote down-regulation of wild type, but not a kinase-
defective mutant of ErbB-1 (Fig. 5), strongly support the
identification of c-Cbl as the protein substrate whose
recruitment by internalized receptors allows transloca-
tion into internal vesicles of the MVB. Conceivably, ad-
ditional endosomal molecules are involvedin the sorting
mechanism. Examples ofcandidate sorting moleculesin-
clude annexin I (Futter et al. 1993), SNX1, a homolog of
a yeast vacuolar sorting protein that accelerates down-
regulation of ErbB-1 (Kurten et al. 1996), phosphatidyli-
nositol 3-kinase, (Joly et al. 1995), and Grb-2 (Wang and
Moran 1996).
The relationships between proteasomal and lysosomal
degradation processes is unclear at present. However, it
appears likely that ubiquitination occurs already in a
prelysosomal compartment. In accordance with this sce-
nario, we were unable to detect ubiquitinated ErbB-1
molecules on the surface of CHO cells by using biotin
labeling (data not shown). This observation is consistent
with reports that blocking endocytosis of either the
growth hormone receptor (Govers et al. 1997) or the EGF
receptor (Galcheva-Gargova et al. 1995), can prevent
ubiquitination. The situation is clearly different in yeast
cells: Ubiquitination of the Ste2p receptor, a G protein-
coupled receptor for the factor, is necessary for receptor
endocytosis (Hicke and Riezman 1996). In fact, transfer
of ErbB-1 to the lysosome may involve proteasomal ac-
tivity because inhibition at this step still allows some
receptor recycling (Fig. 6C). We note that degradation of
several other growth factor receptors such as the insulin-
like growth factor-1 receptor (Sepp Lorenzino et al.
1995), the PDGF-receptor (Mori et al. 1995), and the Met
receptor for the scatter factor (Jeffers et al. 1997), also
depend on proteasomal activity. Thus, complete degra-
dation of internalized ErbB-1 molecules probably in-
volves simultaneous proteolysis by hydrolases and pro-
teasomal proteinases at the late endosome and/or at the
lysosome.
Endosome sorting by Cbl
GENES & DEVELOPMENT 3671
Genetic analyses of mutant worms and flies indicated
that c-Cbl functions as a major negative regulator of in-
tercellular inductive processes controlling vulva
(Jongeward et al. 1995; Yoon et al. 1995) and eye (Meisner
et al. 1997) development, respectively. In both examples
growth factor signaling is funneled into the Ras-MAPK
pathway (for review, see Perrimon and Perkins 1997).
The analogous mammalian signaling machinery, which
is mediated by the ErbB network, also feeds into the
Ras-MAPK pathway (for review, see Alroy and Yarden
1997). However, unlike ErbB-1, which is strongly
coupled to c-Cbl, the two NDF receptors do not interact
with c-Cbl (Levkowitz et al.1996). Consequently, ErbB-3
and ErbB-4 undergono down-regulation following stimu-
lation with NDF (Figs. 1B and 2C; data not shown), and
no translocation of c-Cbl into receptor-containing endo-
somal vesicles occurs in response to NDF binding to
ErbB-3 (Fig 2A). These differences indicate that ErbB-1 is
subject to negative regulation by c-Cbl, whereas ErbB-3
and ErbB-4 escape Cbl-mediated attenuation. Conse-
quently, the mitogenic signal elicited by EGF is signifi-
cantly less potent than that induced by NDF binding to
ErbB-3 (Pinkas-Kramarski et al. 1996).
Our finding with ErbB-1 and ErbB-3 may be relevant to
the many other growth factor receptors that interact
with c-Cbl. Thus, it will be interesting to test the pre-
diction that most c-Cbl-coupled receptors will undergo
ligand-induced ubiquitination. In addition to ErbB-1, the
receptors for CSF-1 (Wang et al. 1996), PDGF (Mori et al.
1995), and the scatter factor (Jeffers et al. 1997) are also
candidates for c-Cbl-mediated down-regulation through
a ubiquitin-dependent process. Moreover, c-Cbl may
control also nonreceptor tyrosine kinases, such as Syk
(Ota and Samelson 1997), and Src (Tanaka et al. 1996). If
verified, this function may account for the ubiquitous
expression of c-Cbl, and for its general involvement in
apparently unrelated signaling pathways.
Materials and methods
Materials and antibodies
A recombinant form of NDF-
1177–246
was provided by Amgen
(Thousand Oaks, CA). Texas red-labeled EGF was from Molecu-
lar probes (Eugene, OR). Radioactive materials were purchased
from Amersham (Buckinghamshire, UK). Iodogen was from
Pierce. Biotin-X-NHS, MG-123, and lactacystin were from Cal-
biochem. Rabbit anti-c-Cbl (C-15) antibodies, as well as anti-
ErbB-3 antibodies, and a monoclonal antibody (mAb) to phos-
photyrosine antibody were from Santa-Cruz Biotechnology.
Murine mAbs to human ErbB-1 and ErbB-3 were from NeoMar-
kers (Fremont, CA). For immunoblot analysis of ErbB-1, Rab-5,
and Shc, we used antibodies from Transduction Laboratories
(Lexington, KY). Anti-ubiquitin antibody was kindly provided
by Drs. S. Yokota (Yamanashi Medical University, Japan) and
A. Amsterdam (Weizmann Institute, Rehovot, Israel). The anti-
hemagglutinin (HA) mAb was purchased from Boehringer
Mannheim.
Construction and transfection of expression vectors
To subclone c-cbl and 70Z-cbl into the pcDNA3 expression
vector (Invitrogen) containing the HA sequence tag (a gift from
Dr. Y. Haupt, Hebrew University, Jerusalem,Israel), we inserted
the cDNAs into the BamHI and XbaIorBamHI and XhoI sites
of pcDNA3, respectively. To generate thev-Cbl and Cbl-C trun-
cation mutants, we similarly subcloned cDNA fragments cor-
responding to amino acids 1–357 and 358–906, respectively. A
GFP–Cbl expression vector was generated by replacement of
Cbl’s stop codon with a SmaI site and insertion into the KpnI
and SmaI sites of pEGFP-N1 (Clontech). Expression vectors
(pcDNA3) containing the full-length cDNA of human ErbB-1
and ErbB-3 were described previously (Tzahar et al. 1996). The
ErbB-1 deletion mutant lacking 214 carboxy-terminal amino ac-
ids [Dc-214 (Sorkin et al. 1992)], as well as the F5 mutant in
which the five major tyrosine autophosphorylation sites were
mutated to phenylalanine residues (Soler et al. 1993), and the
A974 in which tyrosine 974 was replaced by an alanine (Sorkin
et al. 1996), have been described. All three mutants were sub-
cloned into pcDNA3. To generate a kinase-defective ErbB-1 (ly-
sine 721 mutated to alanine), and the double tyrosine to alanine
mutant Y5,6A (tyrosine residues 974 and 992 mutated to al-
anines), we used oligonucleotide-directed mutagenesis with T7-
DNA polymerase. Expression vectors were introduced to CHO
cells by the Lipofectamine transfection method (GIBCO-BRL).
The total amount of DNA in each transfection was normalized
with pcDNA3 plasmid. Twenty-four hours following transfec-
tion, cells were split and assayed 24 hr later.
Ligand binding, internalization analyses, and endosome
purification
Recombinant human ligands were labeled with Iodogen, and
their binding to cell monolayers and internalization were per-
formed as described (Waterman et al. 1998). An endosomal frac-
tion was prepared as described (Wada et al. 1992) with the fol-
lowing modifications: monolayers of CHO cells were scraped,
washed with PBS and resuspended in homogenization buffer
(250 m
M sucrose, 3 mM imidazole, 1 mM EDTA) containing
protease and phosphatase inhibitors. Cells were homogenized
(20 strokes, pestle B in a Dounce homogenizer) and centrifuged
for 10 min at 1500g. The sucrose concentration in the post-
nuclear supernatant was adjusted to 1.15
M, overlaid with 1.00
and 0.25
M sucrose cushions and centrifuged at 200,000g for 1.5
hr (Beckman SW41-Ti rotor). Endosomes were collected at the
0.25–1.00
M sucrose interface. The endosomal marker protein
Rab-5 was significantly enriched in this fraction relative to
other fractions of the sucrose gradient.
Receptor down-regulation and surface biotinylation assays
Ligand-induced receptor down-regulation was measured as fol-
lows: cells grown in 24-well plates were incubated at 37°C for
up to 2.5 hr with or without 250 ng/ml EGF or NDF in binding
buffer, and then rinsed with ice-cold binding buffer. Surface-
bound ligand molecules were removed by use of a low pH wash
(Pinkas-Kramarski et al. 1996). The number of ligand-binding
sites on the cell surface was then determined by incubating cells
at 4°C with the corresponding radiolabeled ligand for at least 1
hr. Biotinylation of cell surface proteins was performed as we
described previously (Waterman et al. 1998).
Immunoprecipitation and immunoblotting analyses
Subconfluent CHO cells were grown in 10-cm culture dishes,
washed briefly with PBS, and incubated for 10 min at 37°C with
the indicated recombinant growth factors (each at 100 ng/ml).
Levkowitz et al.
3672 GENES & DEVELOPMENT
To stop activation, cells were washed with ice-cold PBS and
kept on ice. Cell lysates were prepared at 4°C. Immunoprecipi-
tation, gel electrophoresis, and immunoblotting were per-
formed as we described previously (Levkowitz et al. 1996).
Immunofluorescence
Cells grown on cover-slips were rinsedwith serum-freemedium
and then treated in the absence or presence of ligands for 5 min
at 37°C. Thereafter, the medium was replaced and incubation
was continued for the indicated time intervals. Cells were fixed
for 15 min with 3% paraformaldehyde in PBS. For immunoflu-
orescent labeling, cells were permeabilized for 10 min at 22°C
with PBS containing 1% albumin and 0.2% Triton X-100. For
double labeling, cover-slips were incubated for 1 hr at room
temperature with mAb 111.1 or mAb 90 (anti-ErbB-1 or -3, re-
spectively) in combinationwith anti-Cbl antibodies. After wash
with PBS, the cover-slips were incubated with Cy3-conjugated
goat-anti-rabbit F(ab)
2
and FITC-conjugated goat-anti-mouse
F(ab)
2
-specific antibodies (Jackson ImmunoResearch Laborato-
ries) for an additional 1 hr. Finally, the coverslips were mounted
in Elvanol (Hoechst, Frankfurt).
Quantitative immunofluorescence microscopy
The system used for quantitative fluorescence microscopy and
image analysis will be described in detail elsewhere (E. Zamir,
B.-Z. Katz, K. Yamada, B. Geiger, and Z. Kam, in prep.). Speci-
mens were examined by use of a Zeiss Axioskope microscope
(Oberkochen, Germany) with a 100/1.3 plan-Neofluar objec-
tive. Images were acquired by a scientific, 12-bit, charged-
couple devise (CCD) camera (Model C220, Photometrics Co.).
Particles were segmented by the Water algorithm following
high-pass filter (subtracting from each pixel the average over
4 × 4 µm area around the particle). The parameters in Water
were adjusted to the dimension of the particles, and were kept
constant for all the analyses. To analyze the relationships be-
tween Cbl-labeled structures and ErbB-1-labeled vesicles,
vesicles were segmented separately and fluorescence intensities
measured for both Cbl- and ErbB1-segmented vesicles. Thetotal
number of Cbl- or ErbB1-containing vesicles was divided by the
total cell area to yield the average number of vesicles per 10
3
µm
2
. Cbl-segmented vesicles that had FITC fluorescent inten-
sity higher than a constant threshold were defined as Cbl-
vesicles with ErbB1, and their percentagefrom the total number
of Cbl-segmented vesicles was calculated. Similarly, the per-
centage of ErbB1-containing vesicles with Cbl from the total
number of ErbB1-segmented vesicles was obtained.
Acknowledgments
In cherished memory of Yarden Weinberg (1994–1998) who died
of rhabdomyosarcoma. We thank Drs. S. Yokota and A. Amster-
dam for anti-ubiquitin antibodies, Amar Sahay for constructing
the pcDNA3/F5 mutant, and Sara Lavi for technical help. This
work was supported by a grant from the National Institutes of
Health (CA72981).
The publication costs of this article were defrayed in part by
payment of page charges. This article must therefore be hereby
marked ‘advertisement’ in accordance with 18 USC section
1734 solely to indicate this fact.
Note added in proof
It has been reported very recently that Cbl enhances ubiquiti-
nation and degradation of the PDGF receptor [S. Miyake et al.
(1998) Proc. Natl. Acad. Sci. 95: 7927–7932].
References
Alroy, I. and Y. Yarden. 1997. The ErbB signaling network in
embryogenesis and oncogenesis: Signal diversification
through combinatorial ligand-receptor interactions. FEBS
Lett. 410: 83–86.
Basu, S.K., J.L. Goldstein, R.G.W. Anderson, and M.S. Brown.
1981. Monensin interrupts the recycling of low density lipo-
protein receptors in human fibroblasts. Cell 24: 493–502.
Boussiotis, V.A., G.J. Freeman, A. Berezovskaya, D.L. Barber,
and L.M. Nadler. 1997. Maintenance of humanT cell anergy:
Blocking of IL-2 gene transcription by activated Rap1. Sci-
ence 278: 124–128.
Bowtell, D.D. and W.Y. Langdon. 1995. The protein product of
the c-cbl oncogene rapidly complexes with the EGF receptor
and is tyrosine phosphorylated following EGF stimulation.
Oncogene 11: 1561–1567.
Chang, C.-P., C.S. Lazar, B.J. Walsh, M. Komuro, J.F. Collawn,
L.A. Kuhn, J.A. Tainer, I.S. Trowbridge, M.G. Farquhar,
M.G. Rosenfeld, H.S. Wiley, and G.N. Gill. 1993. Ligand-
induced internalization of the epidermal growth factor re-
ceptor is mediated by multiple endocytic codes analogous to
the tyrosine motif found in constitutively internalizing re-
ceptors. J. Biol. Chem. 268: 19312–19320.
Chen, W.S., C.S. Lazar, K.A. Lund, J.B. Welsh, C.-P. Chang,
G.M. Walton, C.J. der, H.S. Wiley, G.N. Gill, and M.G.
Rosenfeld. 1989. Functional independence of the epidermal
growth factor receptor from a domain required for ligand-
induced internalization and calcium regulation. Cell 59: 33–
43.
Felder, S., K.Miller, G. Moehren, A. Ullrich, J. Schlessinger, and
C.R. Hopkins. 1990. Kinase activity controls the sorting of
the epidermal growth factor receptor within the multive-
sicular body. Cell 61: 623–634.
French, A.R., G.P. Sudlow, H.S. Wiley, and D.A. Lauffenburger.
1994. Postendocytic trafficking of epidermal growth factor-
receptor complexes is mediated through saturable and spe-
cific endosomal interactions. J. Biol. Chem. 269: 15749–
15755.
Futter, C.E., S. Felder, J. Schlessinger, A. Ullrich, and C.R. Hop-
kins. 1993. Anexin I is phosphorylated in the multivesicular
body during the processing of the epidermal growth factor
receptor. J. Cell Biol. 120: 77–83.
Galcheva-Gargova, Z., S.J. Theroux, and R.J. Davis. 1995. The
epidermal growth factor receptor is covalently linked to
ubiquitin. Oncogene 11: 2649–2655.
Galisteo, M.L., I. Dikic, A.G. Batzer, W.Y. Langdon, and J.
Schlessinger. 1995. Tyrosine phosphorylation of the c-cbl
proto-oncogene protein product and association with epider-
mal growth factor (EGF) receptor upon EGF stimulation. J.
Biol. Chem. 270: 20242–20245.
Govers, R., P. van Kerkhof, A.L. Schwartz, andG.J. Strous. 1997.
Linkage of the ubiquitin-conjugating system and the endo-
cytic pathway in ligand-induced internalization of the
growth hormone receptor. EMBO J. 16: 4851–4858.
Hicke, L. and H. Riezman. 1996. Ubiquitination of a yeast
plasma membrane receptor signals its ligand-stimulated en-
docytosis. Cell 84: 277–287.
Honegger, A.M., T.J. Dull,S. Felder,E. vanObberghen, F. Bellot,
D. Szapary, A. Schmidt, A. Ullrich,and J. Schlessinger. 1987.
Point mutation at theATP bindingsite of EGF receptor abol-
ishes protein-tyrosine kinase activity and alters cellular
Endosome sorting by Cbl
GENES & DEVELOPMENT 3673
routing. Cell 51: 199–209.
Hopkins, C.R., A. Gibson, M. Shipman, and K. Miller. 1990.
Movement of internalized ligand-receptor complexes along a
continuous endosomal reticulum. Nature 346: 335–339.
Jeffers, M., G.A. Taylor, K.M. Weidner, S. Omura, and G.F.
Vande Woude. 1997. Degradation of the Met tyrosine kinase
receptor by the ubiquitin-proteasome pathway. Mol. Cell.
Biol. 17: 799–808.
Joly, M., A. Kazlauskas, and S. Corvera. 1995. Phosphatidylino-
sitol 3-kinase activity is required at a postendocytic step in
platelet-derived growth factor receptor trafficking. J. Biol.
Chem. 270: 13225–13230.
Jongeward, G.D., T.R. Clandinin, and P.W. Sternberg. 1995. sli-
1, a negative regulator of let-23-mediated signaling in C. el-
egans. Genetics 139: 1553–1566.
Kurten, R.C., D.L. Cadena, and G.N. Gill. 1996. Enhanced deg-
radation of EGF receptors by a sorting nexin. Science
272: 1008–1010.
Langdon, W.Y., J.W. Hartley, S.P. Klinken, S.K. Ruscetti, and
H.C. Morse III. 1989. v-cbl, an oncogene from a dual-recom-
binant murine retrovirus that induces early B-lineage lym-
phomas. Proc. Natl. Acad. Sci. 86: 1168–1172.
Levkowitz, G., L.N. Klapper, E. Tzahar, A. Freywald, M. Sela,
and Y. Yarden. 1996. Coupling of the c-Cbl protooncogene
product to ErbB-1/EGF-receptor but not to other ErbB pro-
teins. Oncogene 12: 1117–1125.
Lund, K.A., L.K. Opresko, C. Starbuck, B.J. Walsh, and H.S.
Wiley. 1990. Quantitative analysis of the endocytic system
involved in hormone-induced receptor internalization. J.
Biol. Chem. 265: 15713–15723.
Lupher, M.L., Jr., Z. Songyang, S.E. Shoelson, L.C. Cantley, and
H. Band. 1997. The Cbl phosphotyrosine-binding domain se-
lects a D(N/D)XpY motif and binds to the Tyr292 negative
regulatory phosphorylation site of ZAP-70. J. Biol. Chem.
272: 33140–33144.
Meisner, H., B.R. Conway, D. Hartley, and M.P. Czech. 1995.
Interactions of Cbl with Grb2 and phosphatidylinositol 3-
kinase in activated Jurkat cells. Mol. Cell. Biol. 15: 3571–
3578.
Meisner, H., A. Daga, J. Buxton, B. Fernandez, A. Chawla, U.
Banerjee, and M.P. Czech. 1997. Interactions of Drosophila
Cbl with epidermal growth factor receptors and roleof Cblin
R7 photoreceptor cell development. Mol. Cell. Biol.
17: 2217–2225.
Mori, S., K. Tanaka, S. Omura, and Y. Saito. 1995. Degradation
process of ligand-stimulated platelet-derived growth factor
beta-receptor involves ubiquitin-proteasome proteolytic
pathway. J. Biol. Chem. 270: 29447–29452.
Opresko, L.K., C.P. Chang, B.H. Will, P.M. Burke,G.N.Gill, and
H.S. Wiley. 1995. Endocytosis and lysosomal targeting of
epidermal growth factor receptors are mediated by distinct
sequences independent of the tyrosine kinase domain. J.
Biol. Chem. 270: 4325–4333.
Ota, Y. and L.E. Samelson. 1997. The product of the proto-on-
cogene c-cbl: A negativeregulator of the Syk tyrosine kinase.
Science 276: 418–420.
Perrimon, N. and L.A. Perkins. 1997. There must be 50 ways to
rule the signal: The case of the Drosophila EGF receptor.
Cell 89: 13–16.
Pinkas-Kramarski, R., L. Soussan, H. Waterman, G. Levkowitz,
I. Alroy, L. Klapper, S.Lavi, R.Seger, B.Ratzkin, M.Sela, and
Y. Yarden. 1996. Diversification of Neudifferentiation factor
and epidermal growth factor signaling by combinatorial re-
ceptor interactions. EMBO J. 15: 2452–2467.
Sepp Lorenzino, L., Z. Ma, D.E. Lebwohl, A. Vinitsky, and N.
Rosen. 1995. Herbimycin A induces the 20 S proteasome-
and ubiquitin–dependent degradation of receptortyrosine ki-
nases. J. Biol. Chem. 270: 16580–16587.
Soler, C., L. Beguinot, A. Sorkin, and G. Carpenter. 1993. Tyro-
sine phosphorylation of ras GTPase-activating protein does
not require association with the epidermal growth factor re-
ceptor. J. Biol. Chem. 268: 22010–22019.
Sorkin, A. and C.M. Waters. 1993. Endocytosis of growth factor
receptors. BioEssays 15: 375–382.
Sorkin, A., K. Helin, C.M. Waters, G. Carpenter, and L. Begui-
not. 1992. Multiple autophosphorylation sites of the epider-
mal growth factor are essential for receptor kinase activity
and internalization. Contrasting significance of tyrosine 992
in the native and truncated receptors. J. Biol. Chem.
267: 8672–8678.
Sorkin, A., M. Mazzotti, T. Sorkina, L. Scotto, and L. Beguinot.
1996. Epidermal growth factor receptor interaction with cla-
thrin adaptors is mediated by the Tyr974-containing inter-
nalization motif. J. Biol. Chem. 271: 13377–13384.
Tanaka, S., L. Neff, R. Baron, and J.B. Levy. 1995. Tyrosine
phosphorylation and translocation of the c-Cbl protein after
activation of tyrosine kinase signaling pathways. J. Biol.
Chem. 270: 14347–14351.
Tanaka, S., M. Amling, L. Neff, A. Peyman, E. Uhlmann, J.B.
Levy, and R. Baron. 1996. c-Cbl is downstream of c-Src in a
signaling pathway necessary for bone resorption. Nature
383: 528–531.
Thien, C.B. and W.Y. Langdon. 1997. EGF receptor binding and
transformation by v-cbl is ablated by the introduction of a
loss-of-function mutation from the Caenorhabditis elegans
sli-1 gene. Oncogene 14: 2239–2249.
Trowbridge, I.S., J.F. Collawn, and C.R. Hopkins. 1993. Signal-
dependent protein trafficking in the endocytic pathway.
Annu. Rev. Cell Biol. 9: 129–161.
Tzahar, E., H. Waterman, X. Chen, G. Levkowitz, D. Karunaga-
ran, S. Lavi, B.J. Ratzkin,and Y. Yarden. 1996. A hierarchical
network of inter-receptor interactions determines signal
transduction by NDF/neuregulin and EGF. Mol. Cell Biol.
16: 5276–5287.
Wada, I., W.H. Lai, B.I. Posner, and J.M. Bergeron. 1992. Asso-
ciation of the tyrosine phosphorylated epidermal growth fac-
tor receptor with a 55-kD tyrosine phosphorylated protein at
the cell surface and in endosomes. J. Cell Biol. 116: 321–330.
Wang, Y., Y.G. Yeung, W.Y. Langdon, and E.R. Stanley. 1996.
c-Cbl is transiently tyrosine-phosphorylated, ubiquitinated,
and membrane-targeted following CSF-1 stimulation of
macrophages. J. Biol. Chem. 271: 17–20.
Wang, Z. and M.F. Moran. 1996. Requirement for the adapter
protein GRB2 in EGF receptor endocytosis. Science
272: 1935–1938.
Waterman, H., I. Sabanai, B. Geiger, and Y. Yarden. 1998. Al-
ternative intracellular routing of ErbB receptors may deter-
mine signaling potency. J. Biol. Chem. 273: 13819–13827.
Wiley, H.S., J.J. Herbst, B.J. Walsh, D.A. Lauffenburger, M.G.
Rosenfeld, and G.N. Gill. 1991. The role of tyrosine kinase
activity in endocytosis, compartmentation, and down-regu-
lation of the epidermal growth factorreceptor. J. Biol. Chem.
266: 11083–11094.
Yoon, C.H., J. Lee, G.D. Jongeward, and P.W. Sternberg. 1995.
Similarity of sli-1, a regulator of vulval development in C.
elegans, to the mammalian proto-oncogene c-Cbl. Science
269: 1102–1105.
Levkowitz et al.
3674 GENES & DEVELOPMENT
... Here, EGFR operates within a growth factor sensory system that can detect time-varying growth factor patterns by ligandinduced conversion of EGFR monomers to transient dimers (Verveer et al, 2000b;Reynolds et al, 2003;Chung et al, 2010;Low-Nam et al, 2011) that catalytically and autocatalytically generate an amplified phosphorylation response in EGFR monomers (Verveer et al, 2000b;Reynolds et al, 2003;Ichinose et al, 2004;Baumdick et al, 2018;Stanoev et al, 2018;Koseska & Bastiaens, 2020). These ligandless EGFR monomers are dynamically maintained at the plasma membrane (PM) by recycling through the recycling endosomes (REs; Baumdick et al, 2015) in contrast to EGF-bound and ubiquitinated EGFR complexes that uni-directionally traffic (Herbst et al, 1994;Baumdick et al, 2015) from early endosomes (EEs; Vieira et al, 1996) to late endosomes (LEs; Vanlandingham & Ceresa, 2009), to be degraded in lysosomes (Levkowitz et al, 1998;De Melker et al, 2001;Haglund et al, 2002;Baumdick et al, 2015;Koseska & Bastiaens, 2020). Because of this differential trafficking of liganded phosphorylated EGFR (EGF-EGFRp) and ligandless phosphorylated EGFR monomers (EGFRp), evolving growth factor patterns can generate contextual cellular responses by altering the dynamically established spatial distribution of phosphorylated EGFR between the PM and endosomal compartments Brü ggemann et al, 2021). ...
... This autocatalytic EGFR toggle-switch signaling system at the PM is distinct from the liganded EGF-EGFRp monomers that form the more stable homotypic complexes that accumulate in clathrin coated pits. Due to the orders of magnitude lower catalytic activity of RPTPγ on EGF-EGFRp with respect to EGFRp (Fig 5E), these EGF-EGFRp clusters are preferentially ubiquitinated by the ubiquitin ligase cbl (Levkowitz et al, 1998;Soubeyran et al, 2002;Baumdick et al, 2015), endocytosed and engage proliferative signaling from endosomes via the soluble Erk pool Brüggemann et al, 2021;Fig 7, green curved arrows). This branch of internalized proliferative EGFR signaling depletes the recycling EGFR pool in dependence on EGF concentration and EphR-mediated cell-cell contact signals that redistribute EGFR to EEs, enabling context dependent proliferative responses in an evolving tissue. ...
Article
Full-text available
Spatially organized reaction dynamics between proto-oncogenic epidermal growth factor receptor (EGFR) and protein tyrosine phosphatases determine EGFR phosphorylation dynamics in response to growth factors and thereby cellular behavior within developing tissues. We show that the reaction dynamics of mutual inhibition between RPTPγ phosphatase and autocatalytic ligandless EGFR phosphorylation enable highly sensitive promigratory EGFR signaling responses to subnanomolar EGF levels, when < 5% receptors are occupied by EGF. EGF thereby triggers an autocatalytic phospho-EGFR reaction by the initial production of small amounts of phospho-EGFR through transient, asymmetric EGF-EGFR2 dimers. Single cell RPTPγ oxidation imaging revealed that phospho-EGFR induces activation of NADPH oxidase, which in turn inhibits RPTPγ-mediated dephosphorylation of EGFR, tilting the autocatalytic RPTPγ/EGFR toggle switch reaction towards ligandless phosphorylated EGFR. Reversibility of this reaction to EGF is maintained by the constitutive phosphatase activity of endoplasmic reticulum-associated TCPTP. This RPTPγ/EGFR reaction at the plasma membrane causes promigratory signaling that is separated from proliferative signaling induced by accumulated, liganded, phosphorylated EGF-EGFR in endosomes. Accordingly, loss of RPTPγ results in constitutive promigratory signaling from phosphorylated EGFR monomers. RPTPγ is thus a suppressor of promigratory oncogenic but not of proliferative EGFR signaling.
... Interestingly EGFR-vIV displayed a complete lack of effector recruitment to EEs, despite being almost fully functional in recruiting the SH2 effectors to the PM (Fig. 7a). These observations may be linked to the fact that this mutant lacks the Y1045 phosphorylation site shown to promote receptor internalization and degradation through the recruitment of ubiquitin ligase c-CBL 37,38 . Alternatively, failure of the Y1045 mutant to recruit c-Cbl may indirectly impact EGFR trafficking by impairing ubiquitination (and subsequent activation) of (an) accessory protein(s) involved in receptor internalization as previously described 39 . ...
Article
Full-text available
Mutations of receptor tyrosine kinases (RTKs) are associated with the development of many cancers by modifying receptor signaling and contributing to drug resistance in clinical settings. We present enhanced bystander bioluminescence resonance energy transfer-based biosensors providing new insights into RTK biology and pharmacology critical for the development of more effective RTK-targeting drugs. Distinct SH2-specific effector biosensors allow for real-time and spatiotemporal monitoring of signal transduction pathways engaged upon RTK activation. Using EGFR as a model, we demonstrate the capacity of these biosensors to differentiate unique signaling signatures, with EGF and Epiregulin ligands displaying differences in efficacy, potency, and responses within different cellular compartments. We further demonstrate that EGFR single point mutations found in Glioblastoma or non-small cell lung cancer, impact the constitutive activity of EGFR and response to tyrosine kinase inhibitor. The BRET-based biosensors are compatible with microscopy, and more importantly characterize the next generation of therapeutics directed against RTKs.
... It is well-established that polyubiquitination regulates the stability and localization of signaling proteins that stimulate cell proliferation and survival. [35][36][37][38][39][40][41][42][43] Given the global reduction in polyubiquitination observed in the samples from patients with VEXAS syndrome, it is plausible that increased abundance or altered subcellular trafficking of these signaling proteins facilitates activation of pathways that enhance cell proliferation and/or survival. HSC proliferation and self-renewal capacity is also negatively regulated by specific E3 ubiquitin ligases, 42,[44][45][46][47][48] implying that HSC clonal expansion in the setting of impaired polyubiquitination could be due to diminished activity of a subset of E3 proteins. ...
Article
Full-text available
Somatic UBA1 mutations in hematopoietic cells are a hallmark of Vacuoles, E1 enzyme, X-linked, Autoinflammatory, Somatic (VEXAS) syndrome, which is a late-onset inflammatory disease associated with bone marrow failure and high mortality. The majority of UBA1 mutations in VEXAS syndrome comprise hemizygous mutations affecting methionine-41 (M41), leading to the expression of UBA1M41T, UBA1M41V, or UBA1M41L and globally reduced protein polyubiquitination. Here, we used CRISPR-Cas9 to engineer isogenic 32D mouse myeloid cell lines expressing hemizygous Uba1WT or Uba1M41L from the endogenous locus. Consistent with prior analyses of VEXAS syndrome patient samples, hemizygous Uba1M41L expression was associated with loss of the UBA1b protein isoform, gain of the UBA1c protein isoform, reduced polyubiquitination, abnormal cytoplasmic vacuoles, and increased production of interleukin-1β and inflammatory chemokines. Vacuoles in Uba1M41L cells contained a variety of endolysosomal membranes, including small vesicles, multivesicular bodies, and multilamellar lysosomes. Uba1M41L cells were more sensitive to the UBA1 inhibitor TAK243. TAK243 treatment promoted apoptosis in Uba1M41L cells and led to preferential loss of Uba1M41L cells in competition assays with Uba1WT cells. Knock-in of a TAK243-binding mutation, Uba1A580S, conferred TAK243 resistance. In addition, overexpression of catalytically active UBA1b in Uba1M41L cells restored polyubiquitination and increased TAK243 resistance. Altogether, these data indicate that loss of UBA1b underlies a key biochemical phenotype associated with VEXAS syndrome and renders cells with reduced UBA1 activity vulnerable to targeted UBA1 inhibition. Our Uba1M41L knock-in cell line is a useful model of VEXAS syndrome that will aid in the study of disease pathogenesis and the development of effective therapies.
... Immunofluorescence staining of EGFR and EEA1 revealed that GSDME knockdown promoted the colocalization of EGFR and EEA1, indicating that GSDME knockdown promoted the formation of early endosome-containing EGFR on the plasma membrane (Fig. 5D). c-Cbl is an E3 ubiquitin ligase that induces monoubiquitination and lysosome-mediated degradation of EGFR [8,37]. To test whether GSDME knockdown promotes EGFR ubiquitination and degradation by c-Cbl. ...
Article
Full-text available
Background: Lung cancer is the most lethal malignancy, with non-small cell lung cancer (NSCLC) being the most common type (~ 85%). Abnormal activation of epidermal growth factor receptor (EGFR) promotes the development of NSCLC. Chemoresistance to tyrosine kinase inhibitors, which is elicited by EGFR mutations, is a key challenge for NSCLC treatment. Therefore, more thorough understanding of EGFR expression and dynamics are needed. Methods: Human non-small cell lung cancer cells and HEK293FT cells were used to investigate the molecular mechanism of gasdermin E (GSDME) regulating EGFR stability by Western blot analysis, immunoprecipitation and immunofluorescence. GSDME and EGFR siRNAs or overexpression plasmids were used to characterize the functional role of GSDME and EGFR in vitro. EdU incorporation, CCK-8 and colony formation assays were used to determine the proliferation ability of non-small cell lung cancer cells. Results: GSDME depletion reduced the proliferation of non-small cell lung cancer cells in vitro. Importantly, both GSDME-full length (GSDME-FL) and GSDME-N fragment physically interacted with EGFR. GSDME interacted with cytoplasmic fragment of EGFR. GSDME knockdown inhibited EGFR dimerization and phosphorylation at tyrosine 1173 (EGFRY1173), which activated ERK1/2. GSDME knockdown also promoted phosphorylation of EGFR at tyrosine 1045 (EGFRY1045) and its degradation. Conclusion: These results indicate that GSDME-FL increases the stability of EGFR, while the GSDME N-terminal fragment induces EGFR degradation. The GSDME-EGFR interaction plays an important role in non-small cell lung cancer development, reveal a previously unrecognized link between GSDME and EGFR stability and offer new insight into cancer pathogenesis. Video abstract.
Preprint
Full-text available
ZNRF3 and RNF43 are closely related transmembrane E3 ubiquitin ligases with significant roles in development and cancer. Conventionally, their biological functions have been associated with regulating WNT signaling receptor ubiquitination and degradation. However, our proteogenomic studies have revealed EGFR as the most negatively correlated protein with ZNRF3/RNF43 mRNA levels in multiple human cancers. Through biochemical investigations, we demonstrate that ZNRF3/RNF43 interact with EGFR via their extracellular domains, leading to EGFR ubiquitination and subsequent degradation facilitated by the E3 ligase RING domain. Overexpression of ZNRF3 reduces EGFR levels and suppresses cancer cell growth in vitro and in vivo , whereas knockout of ZNRF3 / RNF43 stimulates cell growth and tumorigenesis through upregulated EGFR signaling. Together, these data highlight ZNRF3 and RNF43 as novel E3 ubiquitin ligases of EGFR and establish the inactivation of ZNRF3/RNF43 as a driver of increased EGFR signaling, ultimately promoting cancer progression. This discovery establishes a connection between two fundamental signaling pathways, EGFR and WNT, at the level of cytoplasmic membrane receptor, uncovering a novel mechanism underlying the frequent co-activation of EGFR and WNT signaling in development and cancer.
Article
Full-text available
Spontaneous dimerization of EGF receptors (EGFR) and dysregulation of EGFR signaling has been associated with the development of different cancers. Under normal physiological conditions and to maintain homeostatic cell growth, once EGFR signaling occurs, it needs to be attenuated. Activated EGFRs are rapidly internalized, sorted through early endosomes, and ultimately degraded in lysosomes by a process generally known as receptor down-regulation. Through alterations to EGFR trafficking, tumors develop resistance to current treatment strategies, thus highlighting the necessity for combination treatment strategies that target EGFR trafficking. This review covers EGFR structure, trafficking, and altered surface expression of EGFR receptors in cancer, with a focus on how therapy targeting EGFR trafficking may aid tyrosine kinase inhibitor treatment of cancer.
Article
Full-text available
The epidermal growth factor receptor (EGFR) plays crucial roles in several important biological functions such as embryogenesis, epithelial tissue development, and cellular regeneration. However, in multiple solid tumor types overexpression and/or activating mutations of the EGFR gene frequently occur, thus hijacking the EGFR signaling pathway to promote tumorigenesis. Non-small cell lung cancer (NSCLC) tumors in particular often contain prevalent and shared EGFR mutations that provide an ideal source for public neoantigens (NeoAg). Studies in both humans and animal models have confirmed the immunogenicity of some of these NeoAg peptides, suggesting that they may constitute viable targets for cancer immunotherapies. Peptide vaccines targeting mutated EGFR have been tested in multiple clinical trials, demonstrating an excellent safety profile and encouraging clinical efficacy. For example, the CDX-110 (rindopepimut) NeoAg peptide vaccine derived from the EGFRvIII deletion mutant in combination with temozolomide and radiotherapy has shown efficacy in treating EGFRvIII-harboring glioblastoma multiforme (GBM) patients undergone surgery in multiple Phase I and II clinical trials. Furthermore, pilot clinical trials that have administered personalized NeoAg peptides for treating advanced-stage NSCLC patients have shown this approach to be a feasible and safe method to increase antitumor immune responses. Amongst the vaccine peptides administered, EGFR mutation-targeting NeoAgs induced the strongest T cell-mediated immune responses in patients and were also associated with objective clinical responses, implying a promising future for NeoAg peptide vaccines for treating NSCLC patients with selected EGFR mutations. The efficacy of NeoAg-targeting peptide vaccines may be further improved by combining with other modalities such as tyrosine kinase or immune checkpoint inhibitor (ICI) therapy, which are currently being tested in animal models and clinical trials. Herein, we review the most current basic and clinical research progress on EGFR-targeted peptide vaccination for the treatment of NSCLC and other solid tumor types.
Article
Breast cancer (BC) is the leading cause of cancer death in women. This disease is heterogeneous, with clinical subtypes being estrogen receptor-α (ER-α) positive, having human epidermal growth factor receptor 2 (HER2) overexpression, or being triple-negative for ER-α, progesterone receptor, and HER2 (TNBC). The ER-α positive and HER2 overexpressing tumors can be treated with agents targeting these proteins, including tamoxifen and pertuzumab, respectively. Despite these treatments, resistance and metastasis are problematic, while TNBC is challenging to treat due to the lack of suitable targets. Many studies examining BC and other tumors indicate a role for N-myc downstream-regulated gene-1 (NDRG1) as a metastasis suppressor. The ability of NDRG1 to inhibit metastasis is due, in part, to the inhibition of the initial step in metastasis, namely the epithelial-to-mesenchymal transition. Paradoxically, there are also reports of NDRG1 playing a pro-oncogenic role in BC pathogenesis. The oncogenic effects of NDRG1 in BC have been reported to relate to lipid metabolism or the mTOR signaling pathway. The molecular mechanism(s) of how NDRG1 regulates the activity of multiple signaling pathways remains unclear. Therapeutic strategies that up-regulate NDRG1 have been developed and include agents of the di-2-pyridylketone thiosemicarbazone class. These compounds target oncogenic drivers in BC cells, suppressing the expression of multiple key hormone receptors including ER-α, progesterone receptor, androgen receptor, and prolactin receptor, and can also overcome tamoxifen resistance. Considering the varying role of NDRG1 in BC pathogenesis, further studies are required to examine what subset of BC patients would benefit from pharmacopeia that up-regulate NDRG1.
Article
Full-text available
The role of epidermal growth factor (EGF) receptor autophosphorylation sites in the regulation of receptor functions has been studied using cells transfected with mutant EGF receptors. Simultaneous point mutation of 4 tyrosines (Y1068, Y1086, Y1148, Y1173) to phenylalanine, as well as removal of these sites by truncation of the carboxyl-terminal 123 amino acid residues, resulted in reduced receptor phosphorylation of an in vivo specific substrate phospholipase C-gamma 1 to less than 50% compared to the wild-type receptor. The internalization rate constant Ke was also significantly lower in these mutants (0.15/min) compared to cells transfected with wild-type receptor (0.27/min). Additional mutation of tyrosine 992 to phenylalanine in the truncated receptor mutant (Dc-123F) further decreased the receptor internalization rate to a minimal level (ke = 0.07-0.10/min), equivalent to the ke measured for cells expressing kinase-negative receptor (A721). Moreover, tyrosine kinase activity of the Dc-123F receptor toward phospholipase C-gamma 1, compared to wild-type receptor, was reduced by 90%. Taken together, these results show that EGF receptor lacking five autophosphorylation sites functions similar to a kinase-negative receptor. Mutation of tyrosine residue Y992 alone in the context of full length EGF receptor, however, did not affect receptor internalization or kinase activity toward phospholipase C-gamma 1. These data indicate that tyrosine 992 is critical for substrate phosphorylation and internalization only in the context of the truncated receptor, and that minor autophosphorylation sites, such as Y992, may act as compensatory regulatory sties in the absence of the major EGF receptor autophosphorylation sites.
Article
Full-text available
After the intraportal injection of EGF, the EGF receptor (EGFR) is rapidly internalized into hepatic endosomes where it remains largely receptor bound (Lai et al., 1989. J. Cell Biol. 109:2751-2760). In the present study, we evaluated the phosphotyrosine content of EGFRs at the cell surface and in endosomes in order to assess the consequences of internalization. Quantitative estimates of specific radioactivity of the EGFR in these two compartments revealed that tyrosine phosphorylation of the EGFR was observed at the cell surface within 30 s of ligand administration. However, the EGFR was also highly phosphorylated in endosomes reaching levels of tyrosine phosphorylation significantly higher than those of the cell surface receptor at 5 and 15 min after EGF injection. A 55-kD tyrosine phosphorylated polypeptide (pyp55) was observed in association with the EGFR at the cell surface within 30 s of EGF injection. The protein was also found in association with the EGFR in endosomes as evidenced by coprecipitation studies using a mAb to the EGFR as well as by coelution with the EGR in gel permeation chromatography. Limited proteolysis of isolated endosomes indicated that the tyrosine phosphorylated domains of the EGFR and associated pyp55 were cytosolically oriented while internalized EGF was intraluminal. The identification of pyp55 in association with EGFR in both hepatic plasma membranes and endosomes may be relevant to EGFR function and/or trafficking of the EGFR.
Article
Full-text available
Occupancy-induced down-regulation of cell surface epidermal growth factor (EGF) receptors attenuates signal transduction. To define mechanisms through which down-regulation of this class of growth factor receptors occurs, we have investigated the relative roles of ligand-induced internalization and recycling in this process. Occupied, kinase-active EGF receptors were internalized through a high affinity, saturable endocytic system at rates up to 10-fold faster than empty receptors. In contrast, full length EGF receptors lacking tyrosine kinase activity underwent internalization at a rate independent of occupancy. This "kinase-independent" internalization rate appeared to reflect constitutive receptor internalization since it was similar to the internalization rate of both receptors lacking a cytoplasmic domain and of antibodies bound to empty receptors. EGF internalized by either kinase-active or kinase-inactive receptors was efficiently recycled and was found within endosomes containing recycling transferrin receptors. However, targeting of internalized receptors to lysosomes did not require receptor kinase activity. All receptors that displayed ligand-induced internalization also underwent down-regulation, indicating that the proximal cause of down-regulation is occupancy-induced endocytosis. Tyrosine kinase activity greatly enhances this process by stabilizing receptor association with the endocytic apparatus.
Article
Full-text available
We have developed a quantitative method to evaluate the interaction between cell surface receptors and the endocytic apparatus. This method exploits occupancy-dependent changes in internalization rates that occur in cells expressing high numbers of receptors. We found that constitutive internalization of the transferrin receptor behaves as a simple, first order process that is unaltered by ligand. Internalization of the epidermal growth factor (EGF) receptor, however, behaves as a saturable, second order process that is induced by receptor occupancy. Internalization of EGF receptors occurs through at least two distinct pathways: a low capacity pathway that has a relatively high affinity for occupied receptors, and a low affinity pathway that has a much higher capacity. The high affinity pathway was observed in all cells having receptors with intrinsic tyrosine kinase activity. Mutant EGF receptors lacking kinase activity could not utilize the high affinity pathway and were internalized only through the low affinity one. Mutated receptors with decreased affinity for kinase substrates were also internalized at decreased rates through the high affinity, inducible pathway. In the case of vitellogenin receptors in Xenopus oocytes, occupied receptors competed more efficiently for internalization than empty ones. Insulin increased the endocytic capacity of oocytes for vitellogenin receptors. Similarly, serum increased the capacity of the inducible pathway for EGF receptors in mammalian cells. These data are consistent with a model of internalization in which occupied receptors bind to specific cellular components that mediate rapid internalization. Ligand-induced internalization results from an increase in the affinity of occupied receptors for the endocytic apparatus. Hormones can also indirectly regulate endocytosis by increasing the number of coated pits or their rate of internalization. The ability to dissect receptor-specific effects from cell-specific ones should be very useful in investigating the molecular mechanisms of receptor mediated endocytosis.
Article
Full-text available
Cas NS-1 is an acutely transforming murine retrovirus that induces pre-B and pro-B cell lymphomas. Molecular cloning showed it was generated from the ecotropic Cas-Br-M virus by sequential recombinations with endogenous retroviral sequences and a cellular oncogene. The oncogene sequence shows no homology with known oncogenes but some similarity to the yeast transcriptional activator GCN4. A 100-kDa gag-cbl fusion protein, with no detectable kinase activity, is responsible for the cellular transformation. The cellular homologue of v-cbl, present in mouse and human DNA, is expressed in a range of hemopoietic lineages.
Article
Ligand-induced internalization of the epidermal growth factor receptor (EGFR) leads to accelerated receptor degradation, Two models have been proposed to explain this. In the first model, induced internalization expands the intracellular pool of receptors, leading to enhanced lysosomal targeting. The second model proposes that activation of intrinsic receptor kinase activity induces inward vesiculation of endosomes, thus interrupting receptor recycling. To test these models, we created EGFR mutants that lack the conserved tyrosine kinase domain, but retain different parts of the distal carboxyl terminus regulatory region. Mutants lacking all distal regulatory sequences underwent slow internalization (0.02 min(-1)) and turnover (t(1/2) similar to 24 h), similar to unoccupied, holo-EGFR. Mutant receptors that lacked the kinase domain, but retained the entire distal regulatory domain, were constitutively internalized and targeted to lysosomes, even in the absence of EGF. The turnover of these receptors (t(1/2) similar to 11 h) was similar to that of occupied, kinase-active holo-EGFR (t(1/2) similar to 9.5 h). These results show that receptor tyrosine kinase activity is not required for the targeting of EGFR to lysosomes, Receptor mutants which expressed previously identified endocytic sequences underwent rapid internalization. Unexpectedly, enhanced turnover of EGFR mutants required additional sequences located between residues 945 and 991 in the holo-EGFR. Thus, internalization and lysosomal targeting of EGFR are separate processes mediated by distinct sequences. Our results indicate that induced internalization is necessary, but not sufficient, for enhanced EGFR degradation. Instead, down-regulation requires exposure of previously cryptic internalization and lysosomal targeting sequences. Occupied EGFR thus appear to be handled by the endocytic machinery in the same fashion as other constitutively internalized or lysosomally targeted receptors.
Article
Vulval induction during Caenorhabditis elegans development is mediated by LET-23, a homolog of the mammalian epidermal growth factor receptor tyrosine kinase. The sli-1 gene is a negative regulator of LET-23 and is shown here to encode a protein similar to c-Cbl, a mammalian proto-oncoprotein. SLI-1 and c-Cbl share approximately 55 percent amino acid identity over a stretch of 390 residues, which includes a C_3HC_4 zinc-binding motif known as the RING finger, and multiple consensus binding sites for Src homology 3 (SH3) domains. SLI-1 and c-Cbl may define a new class of proteins that modify receptor tyrosine kinase-mediated signal transduction.
Article
We compared the internalization and intracellular sorting of epidermal growth factor receptor (EGF-R) and point mutant kinase-negative EGF-R separately expressed in NIH 3T3 cells lacking endogenous receptor. Both EGF-Rs internalized rapidly, but kinase-negative receptor was surface down-regulated only with monensin or at 20 degrees C. Furthermore, EGF internalized by mutant receptor alone was, in significant proportion, returned to the cell surface undegraded. Hence unlike wild-type receptor, kinase-negative EGF-R recycles. By electron microscopy the early pathways of endocytosis for the two receptors were identical; however, after 10-20 min the pathways diverged at the multivesicular body (MVB). Wild-type EGF-R, destined for degradation, localized to internal vesicles, while kinase-negative EGF-R, destined for recycling, localized to surface membranes of the MVBs and moved to small tubulovesicles. We conclude that sorting of internalized receptor for degradation or recycling can occur through spatial segregation within the MVB, and sorting of EGF-R is controlled by tyrosine kinase activity.
Article
Complexes of cell-surface receptors and their ligands are commonly internalized by endocytosis and enter a prelysosomal endosomal pathway for further processing. Fluorescence microscopy and video recording of living cells to trace the passage of ligand-receptor complexes has identified the endosomal compartment as an extensive network of tubular cisternae. Endocytosed material entering this reticulum enters discrete swellings, identified as multivesicular bodies by electron microscopy, which move along the reticulum towards the pericentriolar area.