You are on page 1of 18

Epidermal growth factor receptor endocytic traffic

perturbation by phosphatidate phosphohydrolase


inhibition: new strategy against cancer
Ronan Shaughnessy1,2, Claudio Retamal1,2, Claudia Oyanadel1,2, Andres Norambuena1,2,
 pez , Marcela Bravo-Zehnder , Fabian J. Montecino , Claudia Metz1,2,
Alejandro Lo 1,2 1,2 1,2

Andrea Soza1,2 and Alfonso Gonzalez1,2


lica de Chile, Santiago, Chile
1 Departamento de Inmunologıa Clınica y Reumatologıa, Facultad de Medicina, Pontificia Universidad Cato
2 Centro de Envejecimiento y Regeneracio n, Departamento de Biologıa Celular y Molecular, Facultad de Ciencias Biolo
gicas, Pontificia
lica de Chile, Santiago, Chile
Universidad Cato

Keywords Epidermal growth factor receptor (EGFR) exaggerated (oncogenic) func-


apoptosis; cancer; EGFR; endocytosis; tion is currently targeted in cancer treatment with drugs that block recep-
phosphatidic acid
tor ligand binding or tyrosine kinase activity. Because endocytic trafficking
is a crucial regulator of EGFR function, its pharmacological perturbation
Correspondence
A. González, Departamento de Inmunologı́a might provide a new anti-tumoral strategy. Inhibition of phosphatidic acid
Clı́nica y Reumatologı́a, Facultad de (PA) phosphohydrolase (PAP) activity has been shown to trigger PA sig-
Medicina, Marcoleta 367, Pontificia naling towards type 4 phosphodiesterase (PDE4) activation and protein
Universidad Católica de Chile, 8330025 kinase A inhibition, leading to internalization of empty/inactive EGFR.
Santiago, Chile Here, we used propranolol, its L- and D- isomers and desipramine as PAP
Fax: +56 2 2229995
inhibitors to further explore the effects of PAP inhibition on EGFR endo-
Tel: +56 2 26862713
cytic trafficking and its consequences on EGFR-dependent cancer cell line
E-mail: agonzara@med.puc.cl
models. PAP inhibition not only made EGFR inaccessible to stimuli but
(Received 12 July 2013, revised 2 February also prolonged the signaling lifetime of ligand-activated EGFR in recycling
2014, accepted 26 February 2014) endosomes. Strikingly, such endocytic perturbations applied in acute/inter-
mittent PAP inhibitor treatments selectively impaired cell proliferation/
doi:10.1111/febs.12770 viability sustained by an exaggerated EGFR function. Phospholipase D
inhibition with FIPI (5-fluoro-2-indolyl des-chlorohalopemide) and PDE4
inhibition with rolipram abrogated both the anti-tumoral and endocytic
effects of PAP inhibition. Prolonged treatments with a low concentration
of PAP inhibitors, although without detectable endocytic effects, still coun-
teracted cell proliferation, induced apoptosis and decreased anchorage-
independent growth of cells bearing EGFR oncogenic influences. Overall,
our results show that PAP inhibitors can counteract EGFR oncogenic
traits, including receptor overexpression or activating mutations resistant
to current tyrosine kinase inhibitors, perturbing EGFR endocytic traffick-
ing and perhaps other as yet unknown processes, depending on treatment
conditions. This puts PAP activity forward as a new suitable target against
EGFR-driven malignancy.

Abbreviations
DAG, diacylglycerol; EEA1, early endosome antigen 1; EGF, epidermal growth factor; EGFR, EGF receptor; GFP, green fluorescent protein;
LPP, lipid phosphate phosphatase; PA, phosphatidic acid; PAP, phosphatidic acid phosphohydrolase; PARP, poly ADP ribose polymerase;
PDE4, type 4 phosphodiesterases; PKA, protein kinase A; PLD, phospholipase D; TfR, transferrin receptor.

2172 FEBS Journal 281 (2014) 2172–2189 ª 2014 FEBS


R. Shaughnessy et al. Targeting endocytosis blocks oncogenic EGFR

other signaling receptors, are intertwined with the


Introduction
mechanisms that regulate endocytic trafficking [6–
The epidermal growth factor receptor (EGFR) belongs 9,21], involving a variety of kinases from diverse
to the family of ErbB tyrosine kinase receptors that signaling pathways [22]. Therefore, exploring the
regulate processes of cell proliferation, differentiation, potential anti-tumoral actions of drugs previously
migration and survival, playing crucial roles in devel- reported to alter EGFR endocytic traffic, acting upon
opment, tissue repair and cancer [1,2]. An exaggerated protein kinase A (PKA) [23], seems quite attractive.
(oncogenic) activity due to EGFR overexpression or Inhibition of basal PKA activity not only delays
activating mutations is frequently found in tumor cells, sorting of ligand-activated EGFR to the degradation
contributing to their malignancy. A major challenge pathway but also induces internalization of empty/
for targeted anti-cancer treatments is to interfere with inactive EGFRs making them inaccessible to stimula-
such oncogenic EGFR function, which gives an oppor- tion [24]. These two endocytic perturbations might be
tunity to attack cancerous cells with minimal damage expected to exert deleterious effects on EGFR-depen-
to normal cells [2,3]. Current drugs have so far been dent malignant cells. PKA and its regulatory systems
directly targeted to the EGFR molecule, either to are intensively studied in the search for drug design
block ligand binding (antibodies) or to inhibit its tyro- strategies, which include targeting the synthesis and
sine kinase activity (small molecules), both demon- degradation of cAMP [25]. Indeed, the most known
strating therapeutic effectiveness against certain signaling pathway towards decreasing PKA activity is
EGFR-dependent cancers [2]. Limitations such as low the activation of Gai proteins by certain G-protein-
response rates and development of resistance prompt a coupled receptors, but an effective intervention
search for new strategies to counteract the malignant through this system would depend on which of these
influence of the EGFR [4]. An interesting, yet little receptors are expressed [26]. A more suitable approach
explored, alternative is to perturb EGFR endocytic would be to target the ubiquitous signaling pathway of
trafficking. phosphatidic acid (PA), which for a long time has
Endocytic trafficking has gained preponderance in been described to decrease PKA activity [27]. This PA/
EGFR function control [5], offering multiple opportu- PKA pathway can be easily triggered by cationic
nities to modify the intensity, location and duration of amphiphilic compounds currently used in clinical prac-
receptor signaling [5–9]. Upon ligand binding, EGFRs tice, such as propranolol and desipramine [28], and
undergo dimerization, tyrosine kinase activation, trans- has recently arisen as a new control system of EGFR
phosphorylation and ubiquitylation, altogether con- endocytosis [23].
tributing to define the endocytic trafficking of active PA is a phospholipid present in small amounts in bio-
EGFRs, which adds spatiotemporal dynamics to sig- logical membranes where its levels can be regulated by
naling [5,10–14]. Depending on ligand concentration several enzymes [29]. Besides serving as a lipid synthesis
and threshold-controlled ubiquitylation [11], activated precursor, PA is also involved in signal transduction,
EGFRs can follow a clathrin-dependent/recycling pH sensing and membrane bending, which are function-
route, which prolongs signaling activity, or a clathrin- ally projected in processes such as cell proliferation, sur-
independent/ubiquitination-dependent downregulation vival, apoptosis and migration [29], as well as in
pathway, which avoids excessive signaling [11,15]. membrane trafficking along the exocytic and endocytic
Downregulation involves ESCRT-mediated sorting routes [30]. PA has been involved in the endocytosis of
into intraluminal vesicles of multivesicular bodies that ligand-activated EGFR [31,32], as well as in PKA-regu-
then fuse with lysosomes [16]. Thus, activated EGFR lated endocytosis of empty/inactive EGFR [23]. Phos-
can remain signaling-competent for variable periods of pholipase D (PLD) is a main generator of PA in
time and at different cellular locations before degrada- response to different extracellular stimuli [33], while
tion, specifying different response outcomes [15]. phosphatidic acid phosphohydrolases (PAPs) are
Cancerous cells seem to exploit the endocytic traf- important PA downregulators, producing diacylglycerol
ficking machinery to increase EGFR oncogenic activity (DAG) [34]. PA signaling functions are mediated by
[17–19], and thus in principle might be more sensitive producing other signaling lipids, such as DAG, lyso-
than normal cells to endocytic perturbations. A recent phosphatidic acid and phospoinositides [29,33], and by
study [20] suggests that signaling imbalances with dele- recruiting and activating specific signaling proteins bear-
terious actions on EGFR-dependent cancer cells can ing PA binding domains [29,33]. An interesting example
be elicited by delaying the traffic of ligand-activated of such PA binding proteins are type 4 phosphodiester-
EGFRs into the lysosomal degradation pathway. The ases (PDE4) [27], which are the main regulators of
mechanisms that regulate EGFR signaling, as well as cAMP levels and compartmentalized cAMP signaling

FEBS Journal 281 (2014) 2172–2189 ª 2014 FEBS 2173


Targeting endocytosis blocks oncogenic EGFR R. Shaughnessy et al.

[35,36]. Propranolol and desipramine used as PAP As previously reported for propranolol and PKA
inhibitors transiently increase PA levels leading to inhibitors [23,24], indirect immunofluorescence showed
PDE4 activation and the subsequent reduction of that all drugs induce EGFR redistribution from the
cAMP levels and PKA activity [27]. Recent evidence cell surface towards perinuclear endocytic compart-
shows that such a PA/PDE4/PKA signaling pathway ments (Fig. 1B). Therefore, propranolol and its iso-
induces endocytic removal of empty/inactive EGFRs mers can be used indistinctly in the endocytic
from the cell surface via both clathrin-dependent and experiments. In cell proliferation assays we used D-pro-
clathrin-independent routes, resulting in their reversible pranolol, which has 60–100-fold less b-adrenergic
accumulation in recycling endosomes [23]. The obvious blocker activity than L-propranolol [38,39].
remaining question is whether PAP inhibitors might eli-
cit endocytic perturbations with selective deleterious
PAP inhibition delays degradation and increases
consequences upon EGFR-dependent tumoral cells.
the pool of ligand-activated EGFR in endosomes
To study the effect of PAP inhibition on the degrada-
Results
tion kinetics and endocytic trafficking of ligand-acti-
vated EGFR, we used our ‘in-house’ HeLa cells as well
Various PAP inhibitors induce EGFR
as HeLa cells from the ATCC (HeLa-wt) permanently
internalization in the absence of ligand
transfected to overexpress EGFR-GFP (HeLa-EGFR)
Propranolol used as a PAP inhibitor induces internali- at almost the same level as our ‘in-house’ cells. HeLa-
zation of empty/inactive EGFR, making it inaccessible wt cells from the ATCC have been reported to express
to external stimuli [23]. This suggests the possibility of around 35 000–65 000 EGFRcell 1 [15] and therefore
interfering with EGFR oncogenic function using PAP are less convenient for assessing EGFR distribution.
inhibitors. However, specific PAP inhibitors are not Treatment of ‘in-house’ HeLa cells with epidermal
available and therefore the studies are based on cat- growth factor (EGF) (50 ngmL 1) induced a gradual
ionic amphiphilic compounds, such as propranolol and decrease in EGFR mass, with a half-life of about
desipramine, currently used in clinical practice for 100 min, reaching 70% degradation in 3 h. The pres-
other purposes [28,34,37]. Propranolol is a racemic ence of propranolol stabilized the initial slope of deg-
mixture of D- and L-propranolol isomers [38,39], which radation to about 30% within the first hour and was
are equivalent as inhibitors of PAP activity, as well as then maintained for the rest of the experiment (4 h)
inducers of the PA/PDE4/PKA pathway [27,37]. Simi- (Fig. 2A). Tyrosine phosphorylation reflecting EGFR
larly, desipramine inhibits PAP activity but to a signaling activity showed a prolonged lifetime under
greater extent than propranolol [37] and also triggers propranolol treatment (Fig. 2B). After 1 h of propran-
the PA/PDE4/PKA pathway [27]. Therefore, using our olol treatment we could detect an increment in the
previously characterized ‘in-house’ HeLa cells that pool of EGFR colocalizing with endosomes containing
express ~ 300 000 EGFRcell 1 [23,24], we first ana- either early endosome antigen 1 (EEA1) or transferrin
lyzed the potency of L- and D-propranolol isomers and receptor (TfR) (Fig. 2C).
desipramine upon EGFR internalization in the absence Taken together with the immunoblot analysis, these
of ligand. results indicate that PAP inhibition by propranolol not
HeLa cells were incubated with increasing concen- only induces EGFR internalization of empty/inactive
trations of each drug for 30 min at 37 °C and then receptors, as previously shown, but also delays sorting
radioligand binding was performed at 4 °C to assess of ligand-activated EGFR towards the lysosomal deg-
EGFRs remaining at the cell surface (Fig. 1A). D- and radation route. The increased pool of EGFRs in early/
L-propranolol were equivalent as inducers of EGFR recycling endosomes, mostly in perinuclear recycling
internalization, both displaying an EC50 of 75–100 lM, endosomes, probably include ligand-activated recep-
similar to that described for the propranolol racemic tors. This is shown later in the HeLa-EGFR cells
mixture [23]. Desipramine was more potent (EC50 of (Fig. 4, later).
20 lM) than propranolol at inducing EGFR internali-
zation, thus correlating with its higher potency as a
PAP inhibition has anti-proliferative/viability
PAP inhibitor [37]. Such EC50 concentrations are 10-
effects that require EGFR endocytosis
fold lower than those of propranolol used to com-
pletely inhibit PAP activity in cells [40,41]. Additive Recent studies have shown that induced increases in
effects were obtained with a mixture of 75 lM pro- the pool of active EGFRs in endosomal compart-
pranolol and 20 lM desipramine (Fig. 1A). ments can be deleterious to EGFR-dependent tumoral

2174 FEBS Journal 281 (2014) 2172–2189 ª 2014 FEBS


R. Shaughnessy et al. Targeting endocytosis blocks oncogenic EGFR

Fig. 1. PAP inhibitors induce EGFR


internalization in the absence of ligand. (A)
125
EGF binding was performed at 4 °C in
HeLa cells treated for 30 min with B
increasing concentrations of D-propranolol,
L-propranolol and desipramine (DMI). The

decrease in binding activity, which reflects


EGFR endocytosis [23], shows similar
EC50 (75–100 lM) for D- and L-propranolol
and lower EC50 (20–25 lM) for
desipramine. A combination of 75 lM of
racemic propranolol and 20 lM
desipramine show additive effects on
EGFR inaccessibility (internalization). (B)
Immunofluorescent pattern showing
internalized EGFR. HeLa cells were treated
with 100 lM racemic propranolol,
D-propranolol, L-propranolol or 20 lM
desipramine for 30 min.
Immunofluorescence with the monoclonal
HB8506 anti-EGFR shows similar
perinuclear location of EGFR with all
drugs.

cells [20]. To study the functional consequences of the trafficking perturbations, repeated twice a day with
endocytic perturbations induced by PAP inhibition we intervals of 6 h without drugs for 4 days. Under
compared HeLa-wt with permanently transfected these conditions, proliferation/viability significantly
HeLa-EGFR that mimic recently acquired overexpres- decreased in HeLa-EGFR cells, while HeLa cells
sion of EGFR as an oncogenic trait. HeLa-EGFR showed a slight decrease without reaching statistical
cells show higher rates of proliferation than their significance (Fig. 3D). This treatment was also ineffec-
parental HeLa-wt (ATCC) counterparts (Fig. 3A) and tive upon non-tumoral epithelial MDCK cells (not
express similar levels of EGFR to our ‘in-house’ shown), which express very low levels of EGFR and
HeLa cells (Fig. 3B). D-propranolol at the EC50 for represent conditions close to normal epithelial cells
EGFR internalization increased the levels of PA by [42]. Inhibition of PLD activity with FIPI (5-fluoro-
~ 60% in 20 min (Fig. 3C). Strikingly, these HeLa- 2-indolyl des-chlorohalopemide), which is a dual
EGFR cells showed higher sensitivity to acute and inhibitor of PLD-1 and PLD-2 generation of PA [43],
intermittent treatments with D-propranolol than counteracted the anti-proliferative/viability effect of D-
HeLa-wt and our ‘in-house’ HeLa cells (Fig. 3D). The propranolol (Fig. 3D), thus indicating dependence of
treatment consisted of incubations with 75 lM D-pro- D-propranolol effects on PLD-generated PA. These
pranolol for just 1 h, which, as shown, induces EGFR results by themselves suggest that perturbing EGFR

FEBS Journal 281 (2014) 2172–2189 ª 2014 FEBS 2175


Targeting endocytosis blocks oncogenic EGFR R. Shaughnessy et al.

A B

C D

Fig. 2. Propranolol reduces EGFR degradation and prolongs its signaling activity. HeLa cells were incubated with EGF (50 ngmL 1) in the
absence or presence of propranolol (100 lM) for the indicated times. (A) The mass of EGFR, assessed by immunoblot using the anti-EGFR-
968, decreases gradually to about 30% and propranolol prevents this decrease indicating a lower rate of degradation. (B) EGFR tyrosine
phosphorylation was analyzed by immunoblot with the monoclonal 4G10 anti-phosphotyrosine IgG after immunoprecipitation of the receptor
with the HB8506 antibody. Propranolol prolongs the lifetime of EGFR tyrosine phosphorylation that reflects an active receptor. (C)
Representative images of EGFR in early and recycling endosomes. After 1 h treatment with 50 ngmL 1 EGF in the absence or presence of
75 lM propranolol, HeLa cells were fixed and stained with anti-EGFR and anti-EEA1 or anti-TfR along with Hoescht. In control conditions the
receptor localizes mainly at the cell surface, whereas it is internalized upon EGF or EGF and propranolol incubation. (D) Quantified
colocalization with EEA1 and TfR, (*P < 0.05, **P < 0.01 Student’s t test).

2176 FEBS Journal 281 (2014) 2172–2189 ª 2014 FEBS


R. Shaughnessy et al. Targeting endocytosis blocks oncogenic EGFR

A B C

D E F

Fig. 3. Acute/intermittent PAP inhibition selectively reduces EGFR-driven proliferation/viability by endocytic- and PA-dependent mechanisms.
(A) Permanently transfected HeLa cells overexpressing EGFR (HeLa-EGFR) show higher EGFR mass, assessed by immunoblot with the
EGFR-968 antibody (the slower migrating band corresponds to the EGFR-GFP) and higher proliferation rates compared with their parental
HeLa cells (ATCC). Mean  SEM of three experiments, *P < 0.001, Student’s t-test. (B) 125EGF binding shows similar EGFR surface levels
in HeLa (in-house) and permanently transfected HeLa-EGFR cells. (C) D-propranolol (75 lM) treatment for 20 min increased PA levels by
~ 60% in HeLa-EGFR cells. Mean  SEM of three experiments. *P < 0.05, Student’s t-test. (D) Effect of PAP inhibition with D-propranolol
on proliferation/viability of the indicated HeLa cells. Cells were incubated for 1 h, twice a day, each 6 h, during three consecutive days with
75 lM propranolol. After each treatment, medium was replaced with fresh complete medium. HeLa-EGFR cells were also treated in the
absence or presence of the PLD inhibitor FIPI (750 nM). The number of live cells was assessed counting cells that exclude trypan blue.
Mean  SEM of three experiments. n.s., not significant; *P < 0.05, **P < 0.0005, one-way ANOVA for indicated comparisons. (E) EGFR
internalization induced by propranolol (75 lM) treatment for 1 h is inhibited by co-incubation with 30 lM rolipram. Indirect
immunofluorescence was performed on fixed and permeabilized cells with anti-GFP (bar 10 lm). (F) The anti-proliferative/viability effect of
D-propranolol is counteracted by 30 lM rolipram in the acute/intermittent treatment described in (D). Mean  SEM of two experiments.
*P < 0.05, **P < 0.005, one-way ANOVA for indicated comparisons.

FEBS Journal 281 (2014) 2172–2189 ª 2014 FEBS 2177


Targeting endocytosis blocks oncogenic EGFR R. Shaughnessy et al.

endocytic trafficking by PAP inhibition can selectively (TfR) and late (Lamp1) endosomes showed a higher
exert deleterious actions upon EGFR-dependent tu- increase in recycling endosomes (Fig. 4C). This obser-
moral cells. vation agrees with the previous results showing
In our acute/intermittent treatment approach during decreased EGFR degradation (Figs 2A and 4B), indi-
three consecutive days, conditions currently used to cating that propranolol diverts activated EGFR from
inhibit endocytosis such as clathrin silencing with the lysosomal degradation route and lowers its recy-
small interfering RNA, cytosol acidification, sucrose cling. As suggested by the rolipram counteracting
hyperosmotic treatment and dynosore all reduced pro- effects, such endocytic perturbation seems to be
liferation by 50% or more (not shown), precluding related to the anti-proliferative/viability effects of pro-
their use to assess the role of endocytosis in the anti- pranolol.
proliferation/viability effects of D-propranolol. How-
ever, we could directly test the role of endocytosis
Prolonged treatment with lower concentrations
using the specific PDE4 inhibitor rolipram, which we
of D-propranolol and desipramine selectively
have previously shown to inhibit EGFR internaliza-
counteracts EGFR-dependent tumoral cell growth
tion induced by propranolol [23]. Rolipram treatment
for 1 h effectively abrogated endocytosis in HeLa- We also studied the effects of continuous and pro-
EGFR cells (Fig. 3E), and used in the acute/intermit- longed treatments (72 h) with 75 lM D-propranolol,
tent setting effectively decreased the anti-proliferative/ which was indiscriminately toxic to our model cells,
viability effect of D-propranolol (Fig. 3F). Therefore, including MDCK cells and our ‘in-house’ HeLa (not
endocytosis seems to be required for the effect of shown). Therefore, we searched for milder treatment
PAP inhibition on EGFR-dependent proliferation/via- conditions by testing combinations of concentrations
bility. more closely related to those reported in the blood of
Taken together with the previous results we can con- patients treated with D-propranolol and desipramine,
clude that an exaggerated PA signaling, such as that which would be safe for future trials in animal models.
triggered by D-propranolol, has the potential to coun- D-propranolol has been used experimentally in man to
teract the oncogenic influence of EGFR in cancerous treat arrhythmia independently of b-adrenergic block-
cells, involving endocytic traffic perturbations. age and has been found at concentrations up to 10 lM
measured in the blood several hours (twice a half-life)
after the last dose [44]. Desipramine reaches around
D-propranolol increases the pool of tyrosine-
1 lM in antidepressive treatments [45–47]. Therefore,
phosphorylated EGFR in recycling endosomes
we tested the effects of 10–30 lM D-propranolol alone
In the previous experiments we showed that PAP inhi- and in combination with 1 lM desipramine. At these
bition with propranolol decreased the rate of degrada- concentrations, D-propranolol still induced 10–20%
tion of total EGFR and increased the lifetime of EGFR internalization in 30 min experiments, whereas
tyrosine-phosphorylated EGFR (Fig. 2A). The high 1 lM desipramine produced negligible effects in this
sensitivity of HeLa-EGFR cell proliferation/viability assay (Fig. 1A). However, these conditions again were
to PAP inhibition prompted us to perform a more found to be highly selective against cells whose prolif-
detailed characterization of the endosomal distribution eration strongly depends on the EGFR. For instance,
of total and tyrosine-phosphorylated EGFR. After the proliferation/viability of HeLa-EGFR significantly
60 min of EGF (50 ngmL 1) treatment in the pres- decreased with a combination of 30 lM D-propranolol
ence of 75 lM propranolol, we detected an increment and 1 lM desipramine, contrasting with almost no
in the percentage of EGFR colocalizing with both effect on their parental untransfected HeLa (ATCC)
EEA1 and TfR positive endosomes, mainly distributed cells (Fig. 5A). Furthermore, both drugs alone or in
in a more perinuclear region than in control cells with combination were rather ineffective upon our ‘in-house’
EGF alone (Fig. 4A). The intracellular levels of tyro- HeLa cells (not shown), which as mentioned express
sine-phosphorylated EGFR (pEGFR), which reflect similar levels of EGFR to the transfected HeLa-EGFR.
ligand-activated EGFR, also increased and distributed MDCK cells were also not affected (Fig. 5A). The
mainly to perinuclear endosomes in propranolol trea- recently acquired high levels of EGFR expression of
ted cells (Fig. 4B). Activated pEGFR accumulated in the transfected cells seemed to provide a cellular con-
perinuclear recycling endosomes presumably due to text that promotes sensitivity to PAP inhibition.
recycling inhibition, as previously shown for empty/ Prolonged treatments of 24 or 48 h did not pro-
inactive EGFR and TfR [23]. In addition, the distribu- duce detectable changes in EGFR distribution
tion of pEGFR with respect to early (EEA1), recycling towards endosomal compartments (Fig. 5C). The

2178 FEBS Journal 281 (2014) 2172–2189 ª 2014 FEBS


R. Shaughnessy et al. Targeting endocytosis blocks oncogenic EGFR

Fig. 4. PAP inhibition by propranolol increases the pool of internalized ligand-activated EGFR. HeLa-EGFR cells seeded on glass coverslips
were incubated with 50 ngmL 1 EGF in the absence or presence of 75 lM propranolol for 30 and 60 min at 37 °C. The cells were then
fixed and co-stained with anti-GFP for total EGFR, anti-phospho-EGFR, to detect activated EGFR (pEGFR) and anti-EEA1, anti-TfR or anti-
Lamp1 for early, recycling and late endosomal localization, respectively. (A) Propranolol increases the percentage of total EGFR colocalizing
with the endosomal markers EEA1 and TfR, as shown by the quantitative analysis depicted in the graph. (B) The amount of intracellular
total EGFR and pEGFR increases in the presence of propranolol. (C) Mander’s coefficient indicates that propranolol (75 lM) increases the
amount of ligand-activated EGFR (pEGFR) in recycling endosomes and lowers it in late endosomes after 1 h treatment.

FEBS Journal 281 (2014) 2172–2189 ª 2014 FEBS 2179


Targeting endocytosis blocks oncogenic EGFR R. Shaughnessy et al.

A B

C D

Fig. 5. Continuous treatments with D-propranolol and desipramine in conditions that selectively reduce cell proliferation/viability of HeLa-
EGFR cells. (A) Proliferation of HeLa-wt (ATCC), HeLa-EGFR and (B) non-tumorigenic MDCK cells was assessed after 3 days under the
indicated treatments, which include the EGFR tyrosine kinase inhibitor AG1478 (0.1 lM), D-propranolol (D-prop) (10 and 30 lM) or
desipramine (DMI) (1 lM). While HeLa cells and MDCK cells are relatively insensitive to these drugs, HeLa-EGFR cell proliferation was
reduced especially by combined D-propranolol and desipramine treatment. Mean  SEM of three experiments. *P < 0.05; Student’s t-test.
(C) Immunofluorescence does not detect EGFR redistribution after treatments with the drugs for the indicated times. (D) EGFR mass is
maintained after prolonged/continuous treatments. Immunoblot was performed using anti-EGFR-968 after the indicated times of treatment
with 30 lM D-propranolol (P) and 1 lM desipramine (D).

EGFR mass showed no significant differences related to EGFR-driven growth seen with the acute/
between untreated or treated cells, indicating that the intermittent treatment is suggestive of at least par-
deleterious effects of prolonged treatment with 30 lM tially shared mechanisms. Actually, as mentioned,
D-propranolol and 1 lM desipramine cannot be 30 lM D-propranolol produced measurable endocyto-
explained by EGFR degradation (Fig. 5D). Subtle sis of EGFR within 30 min (Fig. 1A) and this in
alterations might still occur affecting EGFR endocytic combination with desipramine might have effects sim-
trafficking, yet undetectable to our analysis. Indeed, it ilar to those seen in acute/intermittent treatments.
cannot be discarded that other deleterious mechanism Desipramine probably contributes to the anti-prolifer-
(s) might become preponderant under continuous ation/viability effects by other mechanisms, as sug-
treatment with these lower concentrations of PAP gested by the high sensitivity of other model cells (see
inhibitors. However, the similarity of the selectiveness below).

2180 FEBS Journal 281 (2014) 2172–2189 ª 2014 FEBS


R. Shaughnessy et al. Targeting endocytosis blocks oncogenic EGFR

PAP inhibitors induce apoptosis and inhibit and desipramine compared with wild-type MKN45
anchorage-independent growth cells (Fig. 7A,C, respectively). These cells also internal-
ize EGFR under D-propranolol treatment (Fig. 7B).
We further characterized the effects of PAP inhibition
These results are consistent with the possibility that
on HeLa-EGFR cells, as these cells displayed selective
overexpression of EGFR as an acquired tumorigenic
sensitivity to PAP inhibition compared with their
trait increases the sensitivity to PAP inhibitors. Inter-
parental HeLa (ATCC) cells. Immunoblots against
estingly, MKN-EGFR cells were particular sensitive to
PARP, a substrate that is cleaved by activated cas-
1 lM desipramine alone, contrasting with the HeLa-
pase-3 [48], showed evidence of apoptosis induced by
EGFR cells. We do not yet know the source of such a
the combination of 30 lM D-propranolol and 1 lM
difference but very probably it involves another mech-
desipramine (Fig. 6A). Furthermore, D-propranolol
anism distinct from endocytic perturbation. The com-
also reduced the capability of HeLa-EGFR cells to
bined effects of D-propranolol and desipramine are
form colonies in soft agar (Fig. 6B), which reflects
higher in these cells.
anchorage-independent growth considered to be a hall-
We then tested these same treatment conditions on
mark of malignancy [49].
cells that have naturally acquired oncogenic EGFR
function. To this end we used widely characterized
D-propranol and desipramine are also effective H1975 cells from non-small-cell lung cancer cells,
against other EGFR-dependent cancerous cells which express a double mutant EGFRL858R/T790M and
are highly oncogenic and resistant to current receptor
To analyze how extensive the effects of continuous
tyrosine kinase inhibitors [51,52]. Binding assays and
treatment with D-propranolol and desipramine are to
immunofluorescence showed that these cells internalize
other model cells, we produced permanent transfec-
EGFR in response to propranolol (Fig. 7D,E). Strik-
tants of EGFR in the gastric cancer cell line MKN45,
ingly, their proliferation/viability decreased under
which do not express high levels of EGFR at the cell
prolonged treatment with a combination of 30 lM
surface [50]. Similar to HeLa-EGFR cells, MKN45-
D-propranolol and 1 lM desipramine (Fig. 7F).
EGFR cells showed increased expression of EGFR
Altogether, these results indicate that treatment with
(Fig. 7A, inset) that translated into a higher rate of
PAP inhibitors can be adjusted to selectively damage
proliferation and higher sensitivity to D-propranolol

A B

Fig. 6. PAP inhibitors induce apoptosis and reduce anchorage-independent growth in HeLa-EGFR cells. (A) Immunoblot analysis of apoptosis
in HeLa-EGFR cells treated with staurosporine (positive control), or a combination of D-propranolol (D-prop) (30 lM) and desipramine (DMI)
(1 lM) for the indicated time points, in the presence of 10% fetal bovine serum. Medium was replaced every 24 h. A polyclonal antibody
that detects the full-length PARP protein and its 89 kDa cleaved fragment produced by caspase-3 activity indicates increased apoptosis after
48 h of treatment. Numbers are the percentage of cleaved versus full-length PARP. b-actin was used as a loading control. (B) Anchorage-
independent growth of HeLa-EGFR cells was reduced by D-propranolol. Cells were seeded in 12-well plates containing soft agar and after
24 h were either treated with D-propranolol (25 lM) or left untreated for 14 days. Formed colonies counted manually show a decreased
number under D-propranolol treatment.

FEBS Journal 281 (2014) 2172–2189 ª 2014 FEBS 2181


Targeting endocytosis blocks oncogenic EGFR R. Shaughnessy et al.

A B C

D E F

Fig. 7. EGFR-dependent proliferation/viability of MKN45-EGFR and H1975 cells is sensitive to prolonged/continuous treatment with
D-propranolol and desipramine. (A) Gastric cancer MKN45 cells permanently transfected to overexpress the EGFR show higher levels of

EGFR in immunoblot with EGFR-968 antibody (inset) and higher proliferation rates than their parental MKN45 cells. (B) D-propranolol (D-prop)
(75 lM) induces internalization of MKN45-EGFR cells, as shown by GFP fluorescence. (C) D-propranolol in combination with desipramine
selectively decreases proliferation/viability of MKN-EGFR cells, contrasting with parental MKN45 cells. MKN45 cells also display higher
sensitivity to the EGFR tyrosine kinase inhibitor AG1478 (0.1 lM). Mean  SEM of three experiments. *P < 0.001, one-way ANOVA for
indicated comparisons with control treatment. (D) Lung cancer cells H1975 expressing a double mutant EGFR, which is both oncogenic and
resistant to current tyrosine kinase activity, internalize the receptor in response to either propranolol (75 lM) or desipramine (20 lM) or both.
(E) H1975 immunofluorescence shows EGFR endocytosis under D-propranolol (D-prop) treatment. (F) Proliferation/viability of H1975 cells is
sensitive to inhibition by D-propranolol (D-prop; 30 lM) alone or in combination with desipramine (DMI; 1 lM). *P < 0.05, **P < 0.005 one-
way ANOVA for indicated comparisons with control treatment.

2182 FEBS Journal 281 (2014) 2172–2189 ª 2014 FEBS


R. Shaughnessy et al. Targeting endocytosis blocks oncogenic EGFR

cancerous cells whose proliferation depends on EGFR Previous work [23] showed that propranolol used as
overexpression or activating mutations as acquired a PAP inhibitor induces endocytosis of empty/inactive
oncogenic traits. EGFR through both clathrin-dependent and clathrin-
independent pathways, and also decreases recycling of
the receptor. As a result of these two perturbations the
Discussion
EGFR accumulates mainly at recycling endosomes, in
This work provides novel evidence to consider PAP a reversible manner, returning to the cell surface after
activity as a suitable target against cancer. In contrast discontinuing the treatment. The mechanism involves a
to current drugs that directly target the EGFR mole- signaling pathway initiated by transient PA elevations
cule, PAP inhibitors have the potential to counteract that activates PDE4 and decreases cAMP and PKA
EGFR-dependent cell proliferation/viability of cancer- activity. Thus, EGFR internalization induced by pro-
ous cells through a mechanism involving PA-mediated pranolol can be effectively reduced by inhibiting PLD
endocytic regulation. Our results indicate that PAP with FIPI or PDE4 with rolipram [23]. Here we add
inhibition can selectively reduce cell proliferation and that propranolol reduces the degradation of ligand-
induce apoptosis in tumoral cells that have acquired activated EGFRs and increases their pool at recycling
oncogenic EGFR dysfunction by overexpression or endosomes, mimicking previously reported effects of
oncogenic mutation. PAP inhibition makes unoccu- PKA inhibitors [24].
pied EGFR inaccessible to stimulation due to inter- Endosomal accumulation of activated EGFRs
nalization and prolongs the lifetime of ligand- leading to apoptosis has recently been described in
activated EGFRs at recycling endosomes, diverging MDA-MB-468 cells expressing extremely high levels of
them from the degradation pathway. Cell prolifera- EGFRs (around 106 EGFRscell 1), which saturate
tion/viability becomes impaired by PAP inhibition, the endocytic machinery, as well as in HeLa cells
unless EGFR endocytosis or PLD generation of PA is expressing physiological levels of EGFR and treated
concomitantly blocked. Therefore, endocytic traffick- with monensin to accumulate activated EGFR in early
ing perturbations derived from exaggerated PA signal- endosomes [20]. Because PAP inhibition delays degra-
ing can have deleterious effects upon EGFR- dation and increases the endosomal pool of ligand-
dependent tumoral cells. Besides their relevance in activated EGFR, while empty/inactive EGFRs can
cancer, these results also highlight the role of endocy- undergo internalization and thus become inaccessible
tic trafficking as a critical modulator of EGFR-medi- to further stimulation, it seems likely that these two
ated cell responses. endocytic perturbations contribute to lower EGFR-
PLD-generated PA has previously been proposed as dependent cell proliferation. Such a possibility is sup-
a target to counteract cancerous behavior but focusing ported by the effect of rolipram, which inhibits EGFR
on the inhibition of PLD activity [33,53–56]. PLD endocytosis and the decrease in proliferation/viability
activity is increased in response to mitogenic signals induced by propranolol.
and activated oncogenes, and its expression is fre- We show that L- and D-propranolol, which are equiv-
quently found to be elevated in cancerous cells, seem- alent in PDE4 activation derived from PAP inhibition
ingly contributing to cell transformation, tumoral [27], are also equivalent in inducing EGFR internaliza-
growth and malignancy [54,55]. PLD overproduction tion. Desipramine is a more potent PAP inhibitor than
of PA might contribute to cell malignancy by increas- propranolol [37] and also triggers the PA/PDE4/PKA
ing the activity of the MAPK pathway through PA- pathway [27]. Correlating with these properties, we
mediated recruitment of SOS and Raf [56–60] and also show that desipramine induces EGFR internalization
by PAP-mediated rapid generation of DAG [54], all displaying lower EC50 (20 lM) than propranolol
important elements in cell proliferation and cancer (75 lM). Propranolol and desipramine used at their cor-
[61,62]. Accordingly, inhibitors of PLD have been aris- responding EC50, far below the concentrations currently
ing and have proved to be effective against tumoral used to completely inhibit PAP activity [40,41], have
cell proliferation [33,53], migration and metastasis [55]. additive effects on EGFR internalization. Therefore,
Thus, the alternative of inhibiting PAP activity to propranolol or its isomers can be used interchangeably
exaggerate PA signaling indeed appears at odds with as PAP inhibitors, either alone or in combination with
the proposed role of PLD-generated PA in cancer. desipramine, to perturb EGFR endocytosis.
However, our present evidence strongly suggests that Oncogenic signaling due to overexpression or muta-
acute increments in PA signaling might be beneficial at tion of EGFR is frequently found in epithelial
least in the context of oncogenic EGFR dysfunction, cell derived cancers [1,3] and is currently targeted in
acting through endocytic traffic perturbation. anti-tumoral therapies, expecting to produce less delete-

FEBS Journal 281 (2014) 2172–2189 ª 2014 FEBS 2183


Targeting endocytosis blocks oncogenic EGFR R. Shaughnessy et al.

rious effects to normal cells than conventional chemo- express a double mutation of EGFR, which is not only
therapy [2,3]. PAP inhibition might meet the criterion highly oncogenic but also resistant to current receptor
of selectivity against EGFR-dependent cancer traits. tyrosine kinase inhibitors [51,52], are also highly sensi-
Our permanently transfected model cells resemble an tive. As shown in HeLa-EGFR cells, this prolonged
acquired trait of EGFR overexpression leading to low dose treatment can also induce apoptosis and
increased proliferation. Strikingly, intermittent treat- reduce anchorage-independent growth, a hallmark of
ments with D-propranolol at the EC50 for EGFR inter- malignancy [49]. However, after 24 or 48 h under this
nalization, applied twice a day for 1 h and leaving treatment, we could not detect changes in EGFR cellu-
intervals of 6 h without the drug, is enough to decrease lar distribution and the mass of EGFRs is maintained,
proliferation/viability in HeLa-EGFR, but not in discarding downregulation as a deleterious mechanism.
HeLa-wt cells or ‘in-house’ HeLa cells expressing The concentration-dependent analysis shows that D-
similar EGFR levels. MDCK cells exemplifying normal propranolol at 10–30 lM induces 15–20% EGFR inter-
epithelial cells [42] are also resistant to such treatment nalization within 30 min (see Fig. 1A), while at 1 lM
(not shown). It seems that recently acquired overexpres- desipramine endocytosis is almost negligible. Because
sion of EGFR provides sensitivity to PAP inhibitors. we noticed that daily changes in medium with fresh
To test the role of endocytosis we used rolipram, drug are necessary to see the anti-proliferation/viability
which blocks both clathrin-dependent and clathrin- effects, it is possible that D-propranolol contributes to
independent endocytosis of EGFR internalization reduce proliferation/viability through acute endocytic
induced by propranolol [23]. We show that rolipram perturbations similar to those seen under acute/inter-
alone does not affect cell proliferation in the acute/ mittent treatments. The high sensitivity of MKN45
intermittent treatment protocol, but inhibits EGFR cells to 1 lM desipramine alone suggests a different yet
internalization and the anti-proliferative/viability unknown mechanism, which might operate in combi-
effects of D-propranolol. Furthermore, inhibition of nation with D-propranolol. Subtle changes in the endo-
PLD activity with FIPI [43] also reduces such effects, cytic trafficking of the EGFR might still occur,
demonstrating their dependence on PLD-generated undetectable by our analysis at steady state conditions
PA. All these results indicate that pharmacological but enough to cause an imbalance in EGFR signaling
perturbations or EGFR endocytic trafficking derived towards deleterious effects.
from exaggerated PA signaling have the potential to PAP activity involves two different enzyme families,
selectively counteract the oncogenic influence of PAP1 and PAP2, each comprising three isoforms [34].
EGFR in cancerous cells. These include soluble lipins-1–3 (PAP1), which associ-
PAP inhibition reduces PA-generated DAG [40,41], ate with membranes of the endoplasmic reticulum, and
which depending on its magnitude might decrease transmembrane lipid phosphate phosphatases LPP-1–3
DAG-mediated signaling and lipid homeostasis [63]. (PAP2), which are distributed at the plasma membrane
Propranolol at twice the concentration used here has and endosomal membranes and have a broader range
been shown to induce apoptosis in numerous cell lines of substrates beyond PA. Lipin-1 knockdown cells
associated with impairments in lipid homeostasis [63]. contain increased levels of PA and decreased levels of
Such general deleterious alteration should indiscrimi- cAMP and PKA activity, suggesting that soluble PAP
nately affect all kinds of cells, contrasting with the activity regulates the levels of PA that modulate the
selective cellular sensitivity shown here, which relates PDE4/PKA pathway [64]. There are also examples of
to EGFR oncogenic function. Alterations in lipid cellular effects of propranolol acting mainly upon solu-
homeostasis might contribute together with EGFR ble PAP activity [37,40]. Lipins are thus likely candi-
endocytic trafficking perturbations to determine cell- dates to mediate the effects of PAP inhibitors on
context-dependent deleterious effects. EGFR endocytic traffic perturbation, remaining to be
Our results also show interesting results using com- experimentally proved.
binations of D-propranolol and desipramine at concen- Because PA is involved in a variety of cellular pro-
trations closely related to those reported in the blood cesses, acting through a large number of PA binding
of patients treated with these drugs [44,46]. HeLa- effectors [29], more than a single mechanism might be
EGFR and MKN-EGFR cells, which have increased involved in the anti-proliferation/viability effects of
proliferation compared with their parental counter- PAP inhibition. Spatiotemporal alterations in EGFR
parts, display higher sensitivity to continuous treat- signaling might combine with other PA actions to be
ment (72 h) with 10–30 lM D-propranolol and 1 lM disclosed. These might include defects in DAG produc-
desipramine, while epithelial MDCK cells remain unaf- tion leading to unbalanced lipid homeostasis [63] or
fected with this treatment. H1975 lung cancer cells that alterations in macroautophagy recently involved in

2184 FEBS Journal 281 (2014) 2172–2189 ª 2014 FEBS


R. Shaughnessy et al. Targeting endocytosis blocks oncogenic EGFR

EGFR oncogenic function [65]. However, of all these selected with 1 mgmL 1 geneticin sulfate (G418) and main-
possibilities, the inhibitory effect of rolipram down- tained in 0.8 mgmL 1 G418. Cultures of HeLa cells and
stream of PLD-generated PA puts forward a higher H1975 (ATCC, from K. Polity, Yale University, USA) were
role of lowering cAMP levels and PKA activity to in DMEM and MKN45 cells in RPMI, supplemented with
induce EGFR endocytic perturbations in mediating 10% fetal bovine serum and antibiotics (200 UmL 1 penicil-
anti-tumoral effects of PAP inhibition. Desipramine lin and 0.1 mgmL 1 streptomycin; Sigma-Aldrich), kept at
used at high concentrations, enough to trigger the PA/ 37 °C in a humidified atmosphere (95% air, 5% CO2). Cell
PDE4/PKA pathway, has been shown to induce apop- growth was analyzed by counting trypan blue stained cells
(Invitrogen) at a ratio of 1 : 1 using the Countess Auto-
tosis in several carcinoma cells, so far attributed to the
matic Cell Counter (Invitrogen). Cells were seeded at 10 000
activation of JNK [66], endoplasmic reticulum stress
cellswell 1 and drug-containing medium was replaced every
[67] and autophagy [68], although without analyzing
24 h. Data were graphed and analyzed using the Student’s
EGFR internalization. Whatever the complexity of the
t test using GRAPHPAD PRISM software (San Diego, CA, USA).
mechanisms involved, PAP inhibition arises as an
alternative to selectively induce deleterious effects
upon cancerous cells whose malignancy depends on PA determination
oncogenic EGFR dysfunctions.
Cellular PA levels were measured using an enzyme-coupled
fluorometric assay, as previously described [70]. Fluores-
Materials and methods cence emission of resorufin reporter was measured at
580 nm after excitation at 530 nm in a Synergy fluorometer
Antibodies and reagents (Biotek, Winooski, VT, USA) using GEN5 2.0 ALL-IN-ONE MI-
CROPLATE READER software (Biotek). The PA concentration
Anti-phosphotyrosine monoclonal IgG and anti-pEG- was standardized to total protein concentration.
FRY1068 (Millipore, Billerica, MA, USA), anti-EGFR-968
[42], monoclonal anti-EGFR HB8506 (Hybridoma) (ATCC,
Manassas, VA, USA), polyclonal anti-poly ADP ribose Ligand-binding assays
polymerase (PARP) (Cell Signaling Technology, Danvers, Human 125I-EGF was prepared by the chloramine T
MA, USA), rabbit polyclonal anti-GFP (produced in our method obtaining specific activities of 50 000–70 000
laboratory), anti-EEA1 and anti-TfR (BD Transduction cpmng 1 and binding assays were done as described previ-
Laboratory, Franklin Lakes, NJ, USA), anti-Lamp1 (Santa ously [23,24] in Hanks’ solution with 20 mM HEPES and
Cruz, Palo Alto, CA, USA) and anti-b-actin (Abcam, 0.2% BSA during 1 h at 4 °C. After washing the unbound
Cambridge, UK) were used. The ECL system (Thermo ligand, the cells were lysed with 1 M NaOH. Total counts
Fisher Scientific, Waltham, MA, USA) was used for per minute was measured in a Wizard2 Automatic Gamma
immunoblot detection. Secondary antibodies were Alexa 488 Counter (PerkinElmer, Waltham, MA, USA). The samples
and Alexa 555 (Molecular Probes, Carlsbad, CA, USA) and were normalized by total amount of proteins measured
HRP-conjugated goat anti-rabbit and anti-mouse IgG spectrophotometrically at 280 nm.
(Rockland Immunochemicals, Boyertown, PA, USA). D-pro-
pranolol, L-propranolol, desipramine, agarose type II and
bovine serum albumin (BSA) from Sigma-Aldrich, St Louis, Immunoprecipitation and immunoblot assays
MO, USA. Plasmid pEGFP-N1-EGFR for EGFR-GFP
Extracts of 250 lg of total protein was immunoprecipitated
expression was provided by Alexander Sorkin (University of
with the monoclonal EGFR-HB8506 antibody, resolved by
Pittsburgh, PA, USA) [69]. Cell culture reagents were pur-
SDS/PAGE and immunoblotted with anti-phosphotyrosine
chased from Invitrogen (Carlsbad, CA, USA) and Sigma
monoclonal IgG (Millipore) as previously described [24].
Aldrich. EGFR and PLD inhibitors, AG1478 and FIPI, were
To detect EGFR, PARP (Cell Signaling) and b-actin (Ab-
used (Calbiochem, Darmstadt, Germany). Tissue culture
cam) cell extracts were subjected to 7.5% SDS/PAGE and
plastics were purchased from Nalgene Nunc (Thermo Fisher
the proteins were electrotransferred onto nitrocellulose
Scientific).
membranes and immunoblotted with the corresponding
antibodies. Anti-EGFR-968 was used for EGFR [42].
Cell culture, transfections and proliferation Bands were revealed by ECL (Thermo Fisher Scientific)
assays and visualized with the Chemidoc IT system using the VI-
SIONWORKS LS software. Digital pictures were analyzed with
HeLa cells (ATCC) and MKN45 cells (JCRB0254, from IMAGEJ (NIH, Bethesda, MD, USA) and Adobe PHOTOSHOP
Andrew Quest, Universidad de Chile, Santiago, Chile) were CS (Adobe Systems, Mountain View, CA, USA). For
permanently transfected with pEGFP-N1-EGFR (HeLa-EGFR assessing apoptosis we used anti-PARP (Cell Signaling),
or MKN45-EGFR) using the Lipofectamine 2000 method,

FEBS Journal 281 (2014) 2172–2189 ª 2014 FEBS 2185


Targeting endocytosis blocks oncogenic EGFR R. Shaughnessy et al.

which detects both full-length PARP and its 89 kDa 2 Ciardiello F & Tortora G (2008) EGFR antagonists in
cleaved fragment produced by activated caspase 3 [48]. cancer treatment. N Engl J Med 358, 1160–1174.
Western blot quantification was performed using IMAGEJ 3 Mellinghoff I (2007) Why do cancer cells become
software and graphed using GRAPHPAD PRISM software as a ‘addicted’ to oncogenic epidermal growth factor
percentage of cleaved PARP/total PARP. receptor? PLoS Med 4, 1620–1622.
4 Pao W & Chmielecki J (2010) Rational, biologically
based treatment of EGFR-mutant non-small-cell lung
Immunofluorescence analysis cancer. Nat Rev Cancer 10, 760–774.
Cells grown on glass coverslips were prepared for immuno- 5 Goh LK & Sorkin A (2013) Endocytosis of receptor
fluorescence as described previously [71]. The secondary tyrosine kinases. Cold Spring Harb Perspect Biol 5,
antibodies were anti-mouse AF555 and anti-rabbit AF488 a017459.
[71]. Cells were then mounted on microscope slides with 6 Di Fiore PP & De Camilli P (2001) Endocytosis and
Fluoromount G and digital fluorescence images were signaling: an inseparable partnership. Cell 106, 1–4.
acquired on a Leica DMI600b inverted microscope with a 7 Brankatschk B, Wichert SP, Johnson SD, Schaad O,
639 glycerol immersion lens and an Andor iXon3 EMCCD Rossner MJ & Gruenberg J (2012) Regulation of the
camera. Z stacks of 200 nm steps were acquired and then EGF transcriptional response by endocytic sorting.
3D deconvoluted using SVI HUYGENS ESSENTIAL SOFTWARE Sci Signal 5, ra21.
(SVI, Hilversum, The Netherlands). Colocalization analysis 8 Ceresa BP & Schmid SL (2000) Regulation of signal
was performed using IMAGEJ and JACOP plugin. HeLa and transduction by endocytosis. Curr Opin Cell Biol 12,
MKN45 cells stably expressing EGFR-GFP were fixed as 204–210.
above and mounted on microscope slides with Fluoromount 9 Miaczynska M, Pelkmans L & Zerial M (2004) Not just
G and digital direct fluorescence images were acquired on a a sink: endosomes in control of signal transduction.
Zeiss Axiophot microscope with a Plan-APOCHROMAT Curr Opin Cell Biol 16, 400–406.
100/1.4 oil immersion objective and the 14-bit Zeiss Axio- 10 Goh LK, Huang F, Kim W, Gygi S & Sorkin A (2010)
cam camera, transferred to a computer workstation running Multiple mechanisms collectively regulate clathrin-
AXIOVISION imaging software (Zeiss, Thornwood, NY, mediated endocytosis of the epidermal growth factor
USA), processed and analyzed with IMAGEJ software. receptor. J Cell Biol 189, 871–883.
11 Sigismund S, Algisi V, Nappo G, Conte A, Pascolutti
R, Cuomo A, Bonaldi T, Argenzio E, Verhoef LG,
Soft agar colony formation assay Maspero E et al. (2013) Threshold-controlled
Assays were performed in 12-well plates with ubiquitination of the EGFR directs receptor fate.
300 cellswell 1, resuspended as a single cell suspension in EMBO J 32, 2087–2185.
0.3% agar and layered on top of 0.5% agar, as described 12 Stang E, Blystad FD, Kazazic M, Bertelsen V, Brodahl
previously [72]. The plates were incubated for 14 days and T, Raiborg C, Stenmark H & Madshus IH (2004) Cbl-
formed colonies were counted manually under 109 objec- dependent ubiquitination is required for progression of
tive lenses, using at least three wells per condition in each EGF receptors into clathrin-coated pits. Mol Biol Cell
experiment. 15, 3591–3604.
13 Levkowitz G, Waterman H, Zamir E, Kam Z, Oved S,
Langdon WY, Beguinot L, Geiger B & Yarden Y
Acknowledgements (1998) c-Cbl/Sli-1 regulates endocytic sorting and
ubiquitination of the epidermal growth factor receptor.
This work received financial support from grants
Genes Dev 12, 3663–3674.
CONICYT # PFB12/2007, Fondecyt #1100747, FON-
14 Marmor MD & Yarden Y (2004) Role of protein
DEF # D09I1104, Program MIFAB-FCHBC-MF105
ubiquitylation in regulating endocytosis of receptor
and CONICYT fellowship to RS. We are grateful to tyrosine kinases. Oncogene 23, 2057–2070.
Alexander Sorkin for his generous gift of EGFR-GFP 15 Sigismund S, Argenzio E, Tosoni D, Cavallaro E, Polo
expression plasmid and to Andrew Quest and Katerina S & Di Fiore PP (2008) Clathrin-mediated
Polity for providing cell lines. The authors have no internalization is essential for sustained EGFR
conflicting financial interests. signaling but dispensable for degradation. Dev Cell 15,
209–219.
16 Wegner CS, Rodahl LM & Stenmark H (2011) ESCRT
References
proteins and cell signalling. Traffic 12, 1291–1297.
1 Yarden Y & Pines G (2012) The ERBB network: at 17 Di Fiore PP (2009) Endocytosis, signaling and cancer,
last, cancer therapy meets systems biology. Nat Rev much more than meets the eye. Preface. Mol Oncol 3,
Cancer 12, 553–563. 273–279.

2186 FEBS Journal 281 (2014) 2172–2189 ª 2014 FEBS


R. Shaughnessy et al. Targeting endocytosis blocks oncogenic EGFR

18 Mosesson Y, Mills GB & Yarden Y (2008) Derailed 33 Selvy PE, Lavieri RR, Lindsley CW & Brown HA
endocytosis: an emerging feature of cancer. Nat Rev (2011) Phospholipase D: enzymology, functionality, and
Cancer 8, 835–850. chemical modulation. Chem Rev 111, 6064–6119.
19 Bache KG, Slagsvold T & Stenmark H (2004) Defective 34 Kok BP, Venkatraman G, Capatos D & Brindley DN
downregulation of receptor tyrosine kinases in cancer. (2012) Unlike two peas in a pod: lipid phosphate
EMBO J 23, 2707–2712. phosphatases and phosphatidate phosphatases. Chem
20 Rush JS, Quinalty LM, Engelman L, Sherry DM & Rev 112, 5121–5146.
Ceresa BP (2012) Endosomal accumulation of the 35 Houslay MD (2001) PDE4 cAMP-specific
activated epidermal growth factor receptor (EGFR) phosphodiesterases. Prog Nucleic Acid Res Mol Biol 69,
induces apoptosis. J Biol Chem 287, 712–722. 249–315.
21 Sorkin A & von Zastrow M (2009) Endocytosis and 36 Blackman BE, Horner K, Heidmann J, Wang D,
signalling: intertwining molecular networks. Nat Rev Richter W, Rich TC & Conti M (2011) PDE4D and
Mol Cell Biol 10, 609–622. PDE4B function in distinct subcellular compartments in
22 Pelkmans L, Fava E, Grabner H, Hannus M, mouse embryonic fibroblasts. J Biol Chem 286, 12590–
Habermann B, Krausz E & Zerial M (2005) Genome- 12601.
wide analysis of human kinases in clathrin- and 37 Meier KE, Gause KC, Wisehart-Johnson AE, Gore
caveolae/raft-mediated endocytosis. Nature 436, 78–86. AC, Finley EL, Jones LG, Bradshaw CD, McNair AF
23 Norambuena A, Metz C, Jung JE, Silva A, Otero C, & Ella KM (1998) Effects of propranolol on
Cancino J, Retamal C, Valenzuela JC, Soza A & phosphatidate phosphohydrolase and mitogen-activated
Gonzalez A (2010) Phosphatidic acid induces ligand- protein kinase activities in A7r5 vascular smooth
independent epidermal growth factor receptor endocytic muscle cells. Cell Signal 10, 415–426.
traffic through PDE4 activation. Mol Biol Cell 21, 38 Alexander RW, Williams LT & Lefkowitz RJ (1975)
2916–2929. Identification of cardiac beta-adrenergic receptors by
24 Salazar G & Gonzalez A (2002) Novel mechanism for (minus) [3H]alprenolol binding. Proc Natl Acad Sci
regulation of epidermal growth factor receptor USA 72, 1564–1568.
endocytosis revealed by protein kinase A inhibition. 39 Howe R & Shanks RG (1966) Optical isomers of
Mol Biol Cell 13, 1677–1693. propranolol. Nature 210, 1336–1338.
25 Taylor SS, Kim C, Cheng CY, Brown SH, Wu J & 40 Asp L, Kartberg F, Fernandez-Rodriguez J, Smedh M,
Kannan N (2008) Signaling through cAMP and cAMP- Elsner M, Laporte F, Barcena M, Jansen KA, Valentijn
dependent protein kinase: diverse strategies for drug JA, Koster AJ et al. (2009) Early stages of Golgi vesicle
design. Biochim Biophys Acta 1784, 16–26. and tubule formation require diacylglycerol. Mol Biol
26 Wettschureck N & Offermanns S (2005) Mammalian G Cell 20, 780–790.
proteins and their cell type specific functions. Physiol 41 Baron CL & Malhotra V (2002) Role of diacylglycerol
Rev 85, 1159–1204. in PKD recruitment to the TGN and protein transport
27 Grange M, Sette C, Cuomo M, Conti M, Lagarde M, to the plasma membrane. Science 295, 325–328.
Prigent AF & Nemoz G (2000) The cAMP-specific 42 Buvinic S, Bravo-Zehnder M, Boyer JL, Huidobro-
phosphodiesterase PDE4D3 is regulated by Toro JP & Gonzalez A (2007) Nucleotide P2Y1
phosphatidic acid binding. Consequences for cAMP receptor regulates EGF receptor mitogenic signaling
signaling pathway and characterization of a and expression in epithelial cells. J Cell Sci 120, 4289–
phosphatidic acid binding site. J Biol Chem 275, 33379– 4301.
33387. 43 Su W, Yeku O, Olepu S, Genna A, Park JS, Ren H,
28 Grange M, Picq M, Prigent AF, Lagarde M & Nemoz Du G, Gelb MH, Morris AJ & Frohman MA (2009) 5-
G (1998) Regulation of PDE-4 cAMP Fluoro-2-indolyl des-chlorohalopemide (FIPI), a
phosphodiesterases by phosphatidic acid. Cell Biochem phospholipase D pharmacological inhibitor that alters
Biophys 29, 1–17. cell spreading and inhibits chemotaxis. Mol Pharmacol
29 Liu Y, Su Y & Wang X (2013) Phosphatidic acid- 75, 437–446.
mediated signaling. Adv Exp Med Biol 991, 159–176. 44 Murray KT, Reilly C, Koshakji RP, Roden DM,
30 Roth MG (2008) Molecular mechanisms of PLD Lineberry MD, Wood AJ, Siddoway LA, Barbey JT &
function in membrane traffic. Traffic 9, 1233–1239. Woosley RL (1990) Suppression of ventricular
31 Antonescu CN, Danuser G & Schmid SL (2010) arrhythmias in man by D-propranolol independent of
Phosphatidic acid plays a regulatory role in clathrin- beta-adrenergic receptor blockade. J Clin Invest 85,
mediated endocytosis. Mol Biol Cell 21, 2944–2952. 836–842.
32 Shen Y, Xu L & Foster DA (2001) Role for 45 Frazer A (2001) Serotonergic and noradrenergic
phospholipase D in receptor-mediated endocytosis. Mol reuptake inhibitors: prediction of clinical effects from in
Cell Biol 21, 595–602. vitro potencies. J Clin Psychiatry 62 (Suppl 12), 16–23.

FEBS Journal 281 (2014) 2172–2189 ª 2014 FEBS 2187


Targeting endocytosis blocks oncogenic EGFR R. Shaughnessy et al.

46 Hursting MJ, Clark GD, Raisys VA, Miller SJ & of the Ras/MEK/Erk signaling cascade. FEBS Lett 531,
Opheim KE (1992) Measurement of free desipramine in 65–68.
serum by ultrafiltration with immunoassay. Clin Chem 59 Lee CS, Kim KL, Jang JH, Choi YS, Suh PG &
38, 2468–2471. Ryu SH (2009) The roles of phospholipase D in
47 Bailey DN & Jatlow PI (1976) Gas-chromatographic EGFR signaling. Biochim Biophys Acta 1791, 862–
analysis for therapeutic concentration of imipramine 868.
and disipramine in plasma, with use of a nitrogen 60 Zhang Y & Du G (2009) Phosphatidic acid signaling
detector. Clin Chem 22, 1697–1701. regulation of Ras superfamily of small guanosine
48 Nicholson DW, Ali A, Thornberry NA, Vaillancourt triphosphatases. Biochim Biophys Acta 1791, 850–855.
JP, Ding CK, Gallant M, Gareau Y, Griffin PR, 61 Santarpia L, Lippman SM & El-Naggar AK (2012)
Labelle M, Lazebnik YA et al. (1995) Identification Targeting the MAPK-RAS-RAF signaling pathway in
and inhibition of the ICE/CED-3 protease necessary for cancer therapy. Expert Opin Ther Targets 16, 103–
mammalian apoptosis. Nature 376, 37–43. 119.
49 Hanahan D & Weinberg RA (2011) Hallmarks of 62 Griner EM & Kazanietz MG (2007) Protein kinase C
cancer: the next generation. Cell 144, 646–674. and other diacylglycerol effectors in cancer. Nat Rev
50 Hotz B, Keilholz U, Fusi A, Buhr HJ & Hotz HG Cancer 7, 281–294.
(2012) In vitro and in vivo antitumor activity of 63 Fuentes L, Perez R, Nieto ML, Balsinde J & Balboa
cetuximab in human gastric cancer cell lines in relation MA (2003) Bromoenol lactone promotes cell death by a
to epidermal growth factor receptor (EGFR) expression mechanism involving phosphatidate phosphohydrolase-
and mutational phenotype. Gastric Cancer 15, 252–264. 1 rather than calcium-independent phospholipase A2. J
51 Pao W, Miller VA, Politi KA, Riely GJ, Somwar R, Biol Chem 278, 44683–44690.
Zakowski MF, Kris MG & Varmus H (2005) Acquired 64 Mitra MS, Chen Z, Ren H, Harris TE, Chambers KT,
resistance of lung adenocarcinomas to gefitinib or Hall AM, Nadra K, Klein S, Chrast R, Su X et al.
erlotinib is associated with a second mutation in the (2013) Mice with an adipocyte-specific lipin 1
EGFR kinase domain. PLoS Med 2, e73. separation-of-function allele reveal unexpected roles for
52 Ohashi K, Maruvka YE, Michor F & Pao W (2013) phosphatidic acid in metabolic regulation. Proc Natl
Epidermal growth factor receptor tyrosine kinase Acad Sci USA 110, 642–647.
inhibitor-resistant disease. J Clin Oncol 31, 1070–1080. 65 Wei Y, Zou Z, Becker N, Anderson M, Sumpter R,
53 Scott SA, Selvy PE, Buck JR, Cho HP, Criswell TL, Xiao G, Kinch L, Koduru P, Christudass CS, Veltri
Thomas AL, Armstrong MD, Arteaga CL, Lindsley RW et al. (2013) EGFR-mediated Beclin 1
CW & Brown HA (2009) Design of isoform-selective phosphorylation in autophagy suppression, tumor
phospholipase D inhibitors that modulate cancer cell progression, and tumor chemoresistance. Cell 154,
invasiveness. Nat Chem Biol 5, 108–117. 1269–1284.
54 Foster DA (2009) Phosphatidic acid signaling to 66 Chang HC, Huang CC, Huang CJ, Cheng JS, Liu SI,
mTOR: signals for the survival of human cancer cells. Tsai JY, Chang HT, Huang JK, Chou CT & Jan CR
Biochim Biophys Acta 1791, 949–955. (2008) Desipramine-induced apoptosis in human PC3
55 Su W, Chen Q & Frohman MA (2009) Targeting prostate cancer cells: activation of JNK kinase and
phospholipase D with small-molecule inhibitors as a caspase-3 pathways and a protective role of [Ca2+]i
potential therapeutic approach for cancer metastasis. elevation. Toxicology 250, 9–14.
Future Oncol 5, 1477–1486. 67 Ma J, Qiu Y, Yang L, Peng L, Xia Z, Hou LN, Fang
56 Zhang F, Wang Z, Lu M, Yonekubo Y, Liang X, C, Qi H & Chen HZ (2011) Desipramine induces
Zhang Y, Wu P, Zhou Y, Grinstein S, Hancock JF apoptosis in rat glioma cells via endoplasmic reticulum
et al. (2014) Temporal production of the signaling lipid stress-dependent CHOP pathway. J Neurooncol 101,
phosphatidic acid by phospholipase d2 determines the 41–48.
output of extracellular signal-regulated kinase signaling 68 Ma J, Hou LN, Rong ZX, Liang P, Fang C, Li HF, Qi
in cancer cells. Mol Cell Biol 34, 84–95. H & Chen HZ (2013) Antidepressant desipramine leads
57 Ghosh S, Strum JC, Sciorra VA, Daniel L & Bell RM to C6 glioma cell autophagy: implication for the
(1996) Raf-1 kinase possesses distinct binding domains adjuvant therapy of cancer. Anticancer Agents Med
for phosphatidylserine and phosphatidic acid. Chem 13, 254–260.
Phosphatidic acid regulates the translocation of Raf-1 69 Carter RE & Sorkin A (1998) Endocytosis of functional
in 12-O-tetradecanoylphorbol-13-acetate-stimulated epidermal growth factor receptor-green fluorescent
Madin Darby canine kidney cells. J Biol Chem 271, protein chimera. J Biol Chem 273, 35000–35007.
8472–8480. 70 Morita S-Y, Ueda K & Kitagawa S (2009) Enzymatic
58 Andresen BT, Rizzo MA, Shome K & Romero G measurement of phosphatidic acid in cultured cells. J
(2002) The role of phosphatidic acid in the regulation Lipid Res 50, 1945–1952.

2188 FEBS Journal 281 (2014) 2172–2189 ª 2014 FEBS


R. Shaughnessy et al. Targeting endocytosis blocks oncogenic EGFR

71 Cancino J, Torrealba C, Soza A, Yuseff MI, Gravotta 72 Lu H, Wang X, Urvalek AM, Li T, Xie H, Yu L &
D, Henklein P, Rodriguez-Boulan E & Gonzalez A Zhao J (2014) Transformation of human ovarian
(2007) Antibody to AP1B adaptor blocks biosynthetic surface epithelial cells by Kruppel-like factor 8.
and recycling routes of basolateral proteins at recycling Oncogene 33, 10–18.
endosomes. Mol Biol Cell 18, 4872–4884.

FEBS Journal 281 (2014) 2172–2189 ª 2014 FEBS 2189

You might also like