Role of gangliosides in the association of ErbB2 with lipid rafts in mammary epithelial HC11 cells

10
Role of gangliosides in the association of ErbB2 with lipid rafts in mammary epithelial HC11 cells Elena Sottocornola 1 , Roberta Misasi 2 , Vincenzo Mattei 2,3 , Laura Ciarlo 2 , Roberto Gradini 2,4 , Tina Garofalo 2,3 , Bruno Berra 1 , Irma Colombo 1 and Maurizio Sorice 2,3 1 Institute of General Physiology and Biological Chemistry, University of Milan, Italy 2 Department of Experimental Medicine and Pathology, University of Rome ‘La Sapienza’, Italy 3 Laboratory of Experimental Medicine and Environmental Pathology, Rieti, Italy 4 INM Neuromed, Pozzilli, Italy Gangliosides, ubiquitous components of eukaryotic membranes, are not uniformly distributed within the outer leaflet of the plasma membrane, but segregate, together with cholesterol, glycosylphosphatidylinositol- anchored proteins and signaling-transduction mole- cules, into unique, more or less stable clusters or microdomains called as ‘glycosphingolipid-enriched microdomains’ (GEM), which contribute to membrane structure, organization and, more importantly, func- tion. Indeed, GEM are viewed as a dynamic and pref- erential association of sphingolipids and cholesterol into moving platforms, termed lipid rafts, which can selectively incorporate or exclude proteins [1] and con- tribute to lipid-mediated protein trafficking and signal transduction [2]. The growth factor receptor tyrosine kinase ErbB2 is a 185 kDa transmembrane glycoprotein intensively investigated because of its important role in normal mammary gland development and in the deregulation of growth displayed by cancer cells, including breast and ovarian tumor cells [3,4]. A ligand which binds directly and specifically to ErbB2 has not been identi- fied to date, but it can be activated in trans by ligands binding to epidermal growth factor receptor (EGFR), such as epidermal growth factor (EGF) and transform- ing growth factor a1 [3]. Indeed, in cells coexpressing both ErbB2 and EGFR, EGF preferentially stimulates the formation of ErbB2 EGFR heterodimers in which cross-phosphorylation occurs [5,6]. Data concerning the structural and mechanistic aspects required for Keywords epidermal growth factor receptor; ErbB2; GM3; HC11 cells; lipid rafts Correspondence M. Sorice, Department of Experimental Medicine and Pathology, University of Rome ‘La Sapienza’, viale Regina Elena 324, Rome 00161, Italy Fax: +39 6 445 4820 Tel: +39 6 499 72675 E-mail: [email protected] (Received 24 December 2005, revised 15 February 2006, accepted 27 February 2006) doi:10.1111/j.1742-4658.2006.05203.x We analyzed the role of gangliosides in the association of the ErbB2 recep- tor tyrosine-kinase (RTK) with lipid rafts in mammary epithelial HC11 cells. Scanning confocal microscopy experiments revealed a strict ErbB2– GM3 colocalization in wild-type cells. In addition, analysis of membrane fractions obtained using a linear sucrose gradient showed that ErbB2, epi- dermal growth factor receptor (EGFR) and Shc-p66 (proteins correlated with the ErbB2 signal transduction pathway) were preferentially enriched in lipid rafts together with gangliosides. Blocking of endogenous ganglioside synthesis by (+ –)-threo-1-phenyl-2-decanoylamino-3-morpho- lino-1-propanol hydrochloride ([D]-PDMP) induced a drastic cell-surface redistribution of ErbB2, EGFR and Shc-p66, within the Triton-soluble fractions, as revealed by linear sucrose-gradient analysis. This redistribution was partially reverted when exogenous GM3 was added to ganglioside- depleted HC11 cells. The results point out the key role of ganglioside GM3 in retaining ErbB2 and signal-transduction-correlated proteins in lipid rafts. Abbreviations [D]-PDMP, (+ –)-threo-1-phenyl-2-decanoylamino-3-morpholino-1-propanol hydrochloride; EGF, epidermal growth factor; EGFR, epidermal growth factor receptor; FITC, fluorescein isothiocyanate; GEM, glycosphingolipid-enriched microdomains; HRP, horseradish peroxidase; Rf, retardation factor; RTK, receptor tyrosine-kinase; TX-100, Triton X-100. FEBS Journal 273 (2006) 1821–1830 ª 2006 The Authors Journal compilation ª 2006 FEBS 1821

Transcript of Role of gangliosides in the association of ErbB2 with lipid rafts in mammary epithelial HC11 cells

Role of gangliosides in the association of ErbB2 with lipidrafts in mammary epithelial HC11 cellsElena Sottocornola1, Roberta Misasi2, Vincenzo Mattei2,3, Laura Ciarlo2, Roberto Gradini2,4,Tina Garofalo2,3, Bruno Berra1, Irma Colombo1 and Maurizio Sorice2,3

1 Institute of General Physiology and Biological Chemistry, University of Milan, Italy

2 Department of Experimental Medicine and Pathology, University of Rome ‘La Sapienza’, Italy

3 Laboratory of Experimental Medicine and Environmental Pathology, Rieti, Italy

4 INM Neuromed, Pozzilli, Italy

Gangliosides, ubiquitous components of eukaryotic

membranes, are not uniformly distributed within the

outer leaflet of the plasma membrane, but segregate,

together with cholesterol, glycosylphosphatidylinositol-

anchored proteins and signaling-transduction mole-

cules, into unique, more or less stable clusters or

microdomains called as ‘glycosphingolipid-enriched

microdomains’ (GEM), which contribute to membrane

structure, organization and, more importantly, func-

tion. Indeed, GEM are viewed as a dynamic and pref-

erential association of sphingolipids and cholesterol

into moving platforms, termed lipid rafts, which can

selectively incorporate or exclude proteins [1] and con-

tribute to lipid-mediated protein trafficking and signal

transduction [2].

The growth factor receptor tyrosine kinase ErbB2

is a 185 kDa transmembrane glycoprotein intensively

investigated because of its important role in normal

mammary gland development and in the deregulation

of growth displayed by cancer cells, including breast

and ovarian tumor cells [3,4]. A ligand which binds

directly and specifically to ErbB2 has not been identi-

fied to date, but it can be activated in trans by ligands

binding to epidermal growth factor receptor (EGFR),

such as epidermal growth factor (EGF) and transform-

ing growth factor a1 [3]. Indeed, in cells coexpressing

both ErbB2 and EGFR, EGF preferentially stimulates

the formation of ErbB2 ⁄EGFR heterodimers in which

cross-phosphorylation occurs [5,6]. Data concerning

the structural and mechanistic aspects required for

Keywords

epidermal growth factor receptor; ErbB2;

GM3; HC11 cells; lipid rafts

Correspondence

M. Sorice, Department of Experimental

Medicine and Pathology, University of Rome

‘La Sapienza’, viale Regina Elena 324,

Rome 00161, Italy

Fax: +39 6 445 4820

Tel: +39 6 499 72675

E-mail: [email protected]

(Received 24 December 2005, revised 15

February 2006, accepted 27 February 2006)

doi:10.1111/j.1742-4658.2006.05203.x

We analyzed the role of gangliosides in the association of the ErbB2 recep-

tor tyrosine-kinase (RTK) with lipid rafts in mammary epithelial HC11

cells. Scanning confocal microscopy experiments revealed a strict ErbB2–

GM3 colocalization in wild-type cells. In addition, analysis of membrane

fractions obtained using a linear sucrose gradient showed that ErbB2, epi-

dermal growth factor receptor (EGFR) and Shc-p66 (proteins correlated

with the ErbB2 signal transduction pathway) were preferentially enriched

in lipid rafts together with gangliosides. Blocking of endogenous

ganglioside synthesis by (+ ⁄ –)-threo-1-phenyl-2-decanoylamino-3-morpho-

lino-1-propanol hydrochloride ([D]-PDMP) induced a drastic cell-surface

redistribution of ErbB2, EGFR and Shc-p66, within the Triton-soluble

fractions, as revealed by linear sucrose-gradient analysis. This redistribution

was partially reverted when exogenous GM3 was added to ganglioside-

depleted HC11 cells. The results point out the key role of ganglioside GM3

in retaining ErbB2 and signal-transduction-correlated proteins in lipid

rafts.

Abbreviations

[D]-PDMP, (+ ⁄ –)-threo-1-phenyl-2-decanoylamino-3-morpholino-1-propanol hydrochloride; EGF, epidermal growth factor; EGFR, epidermal

growth factor receptor; FITC, fluorescein isothiocyanate; GEM, glycosphingolipid-enriched microdomains; HRP, horseradish peroxidase;

Rf, retardation factor; RTK, receptor tyrosine-kinase; TX-100, Triton X-100.

FEBS Journal 273 (2006) 1821–1830 ª 2006 The Authors Journal compilation ª 2006 FEBS 1821

EGF-dependent ‘trans activation’ of EGFR and ErbB2

in ErbB2 ⁄EGFR heterodimers, as well as data con-

cerning the overall effects induced by changes in the

relative expression levels of EGFR and ErbB2, are

growing rapidly and several lines of evidence have

shown that ErbB2 is associated with lipid microdo-

mains [7–10]. By contrast, the role of cell-membrane

components, such as gangliosides, for determining the

plasma membrane distribution and relative densities of

receptors has not yet been investigated thoroughly.

Recently, we reported the first evidence that ganglio-

side depletion associates with increased levels of the

activated ErbB2 and EGFR, whereas increased gan-

glioside GM3 content correlates with the downregula-

tion of both receptors [11].

In this study, we provide evidence of ErbB2–GM3

association on the plasma membrane of mouse mam-

mary epithelial HC11 cells and demonstrate that gan-

gliosides, and particularly GM3, play a key role in

retaining ErbB2 and proteins correlated with its signal-

transduction pathway in lipid rafts.

Results

ErbB2–GM3 colocalization in mammary epithelial

HC11 cells

In order to evaluate ErbB2 distribution and its pos-

sible association with GM3, we performed immunoflu-

orescence labeling, followed by scanning confocal

microscopy analysis. Cells were labeled with anti-

ErbB2 polyclonal serum and then with anti-GM3

monoclonal serum.

Analysis of GM3 expression and distribution in

untreated HC11 cells (Fig. 1A) revealed that GM3

staining appeared uneven over the cell surface, similar

to that seen on ErbB2 molecule fluorescence. A

merged image of the two stainings clearly revealed

orange areas, resulting from the overlap of green and

red fluorescence, which corresponded to colocalization

areas.

To analyze the effect of ganglioside depletion on

the association of ErbB2 with ganglioside GM3, we

preliminary treated the cells with (+ ⁄ –)-threo-1-phe-nyl-2-decanoylamino-3-morpholino-1-propanol hydro-

chloride ([D]-PDMP), which blocks endogenous

ganglioside biosynthesis, resulting in the almost com-

plete disappearance of all ganglioside species [11]. Vir-

tually no staining was observed in cells labeled with

anti-GM3 serum (Fig. 1B). The lack of immunolabe-

ling demonstrates the effect of [D]-PDMP on the

depletion of gangliosides and the specificity of the anti-

GM3 serum. The distribution of ErbB2 appeared more

diffuse compared with control untreated cells. No

colocalization areas between GM3 and ErbB2 were

detected.

By contrast, overlain areas were reverted when exo-

genous GM3 was added to ganglioside-depleted HC11

cells (Fig. 1C).

Scatter-plot diagrams showed how the dual labels

are colocalized. Figure 1D shows a colocalization area

that is evident in untreated HC11 cells. In cells treated

A

B

C

D

Fig. 1. Scanning confocal microscopy analysis of GM3–ErbB2

association on HC11 cells. Cells were fixed with 4% paraformalde-

hyde, permeabilized with 0.5% TX-100 and then incubated with

anti-ErbB2 polyclonal serum, followed by the addition of FITC-conju-

gated goat anti-(rabbit IgG) serum. Cells were then labeled with

anti-GM3 monoclonal serum (GMR6), followed by the addition of

Texas red-conjugated anti-(mouse IgM) serum. Merge: dual immuno-

labeling of anti-GM3 (red) and anti-ErbB2 (green). Colocalization

areas are stained in orange. (A) Untreated HC11 cells. (B) HC11

cells treated with 30 lM [D]-PDMP for 5 days. (C) HC11 cells treat-

ed with 30 lM [D]-PDMP for 5 days and then with 125 lM GM3

for 5 min. (D) Two-dimensional scatter plot analysis of the dual-

labeled fluorochromes (pseudocolor) GM3–ErbB2. Diagrams show

the pixel intensity distribution of a dual-channel section. The x-axis

represents intensity from the red channel; the y-axis represents

intensity from the green channel; a colocalization area is evident in

untreated HC11 cells. In cells treated with [D]-PDMP and then with

GM3 a major colocalization index is evident because the blue area

is larger and more directed towards the diagonal line. Figure shows

analysis of about 40 cells.

ErbB2–raft association in HC11 cells E. Sottocornola et al.

1822 FEBS Journal 273 (2006) 1821–1830 ª 2006 The Authors Journal compilation ª 2006 FEBS

with [D]-PDMP and then with GM3 a major colocali-

zation index is evident, because the blue area is larger

and more directed towards the diagonal line.

Effect of ganglioside depletion on cholesterol and

caveolin-1 distribution in HC11 cells

In order to verify that [D]-PDMP does not interfere

with the membrane distribution of known lipid-raft

markers, we investigated the cholesterol and caveolin-1

content of membrane fractions obtained by a 5–30%

linear sucrose gradient from HC11 cells in the absence

or presence of treatment with [D]-PDMP.

As expected, cholesterol was present mainly in frac-

tions 4–6 (Fig. 2A), which, under our experimental

conditions, correspond to lipid-raft fractions. About

90% of the cholesterol content of the total cell extract

was recovered in fractions 4–6. Virtually the same cho-

lesterol distribution was observed in cells treated with

[D]-PDMP.

Similar findings were found in western blot analysis

of caveolin-1 distribution in the sucrose gradient frac-

tions. The analysis was performed loading fraction

samples by volume. Because the protein content of

Triton X-100 (TX-100)-soluble fractions 10 and 11

was higher than that of TX-100-insoluble fractions 4–6

(not shown) [12], we can observe that caveolin-1

was consistently enriched in TX-100-insoluble fractions

in untreated, as well as in [D]-PDMP-treated, cells

(Fig. 2B).

ErbB2 preferential association with lipid-raft

fractions in HC11 cells

To evaluate the distribution of ErbB2 in raft fractions

of HC11 cells, treated or not with [D]-PDMP, EGF or

[D]-PDMP and EGF, all fractions obtained by sucrose

gradient were analyzed by western blot (Fig. 3). The

results revealed that in non-EGF-stimulated cells

ErbB2 was present mainly in fractions 5 and 6, but

also in fractions 7–11 (Fig. 3A), indicating that ErbB2

is preferentially present in raft fractions. EGF stimulat-

ion did not seem to appreciably modify this distribut-

ion (Fig. 3B).

Interestingly, [D]-PDMP treatment induced a drastic

cell-surface redistribution of ErbB2 (Fig. 3C). Indeed,

the receptor became completely Triton soluble and was

present exclusively in fractions 10 and 11. Identical

profile redistribution of ErbB2 was also evident in

A

B

Fig. 2. (A) Densitometric analysis of cholesterol content in HC11

sucrose-gradient membrane fractions. HC11 cells, either untreated

or treated with 30 lM [D]-PDMP for 5 days, were lyzed in lysis buf-

fer and the supernatant (postnuclear fraction) was subjected to

sucrose density gradient separation. After centrifugation, the gradi-

ent was fractionated and free cholesterol of each fraction was anal-

yzed by TLC and quantified by densitometric scanning analysis. (B)

western blot analysis of caveolin-1 distribution in HC11 sucrose

gradient membrane fractions. Lysates from HC11 cells, either

untreated or treated with 30 lM [D]-PDMP for 5 days, were anal-

yzed by western blot with anti-(caveolin-1) polyclonal serum, fol-

lowed by incubation with an HRP-conjugated anti-(rabbit IgG)

serum, as a secondary antibody.

A

B

C

D

E

Fig. 3. ErbB2 distribution in HC11 sucrose gradient membrane fract-

ions. HC11 cells were lyzed in lysis buffer and the supernatant

(postnuclear fraction) was subjected to sucrose density gradient

separation. After centrifugation, the gradient was fractionated and

each fraction was analyzed by western blotting with anti-ErbB2

polyclonal serum, followed by incubation with a HRP-conjugated

anti-(rabbit IgG) serum, as a secondary antibody. (A) Untreated

HC11 cells. (B) HC11 cells treated with 10 nM EGF for 15 min. (C)

HC11 cells treated with 30 lM [D]-PDMP for 5 days. (D) HC11 cells

treated with 30 lM [D]-PDMP for 5 days and then with 10 nM EGF

for 15 min. (E) HC11 cells treated with 30 lM [D]-PDMP for 5 days

and then with 125 lM GM3 for 5 min.

E. Sottocornola et al. ErbB2–raft association in HC11 cells

FEBS Journal 273 (2006) 1821–1830 ª 2006 The Authors Journal compilation ª 2006 FEBS 1823

HC11 cells treated with [D]-PDMP and EGF

(Fig. 3D), indicating that EGF is not determinant in

defining the retention of ErbB2 into the lipid rafts.

The effect of [D]-PDMP treatment was partially

abolished by addition of exogenous GM3 to ganglio-

side-depleted HC11 cells. In fact, after [D]-PDMP

incubation, followed by GM3 treatment, a significant

proportion of ErbB2 returned to fractions 4 and 5

(Fig. 3E).

In order to better clarify the functional role of the

association between ErbB2 and lipid rafts, we analyzed

the distribution of phospho-ErbB2 in sucrose-gradient

fractions obtained from HC11 cells in the absence or

presence of treatment with EGF, [D]-PDMP and

[D]-PDMP ⁄GM3. Although, as expected, virtually no

phosphorylated ErbB2 was detected in all the fractions

obtained from control cells (Fig. 4A), after triggering

with EGF (Fig. 4B), phosphorylated ErbB2 was found

in both the TX-100-insoluble fractions and the

TX-100-soluble fractions. Interestingly, in cells treated

with [D]-PDMP and EGF (Fig. 4D) a band corres-

ponding to phosphorylated ErbB2 was detected in

fractions 10 and 11, whereas in ganglioside-depleted

cells and cells treated with [D]-PDMP and GM3 no

ErbB2 phosphorylation was observed (Fig. 4C,E

respectively). These findings support the view that

GM3 is mainly involved in retaining ErbB2 in lipid-

raft domains, but that it is not involved in ErbB2

phosphorylation.

EGFR and Shc-p66 preferential association with

lipid-raft fractions in HC11 cells

Because in cells coexpressing both ErbB2 and EGFR,

as is the case of HC11 cells, the two proteins strictly

interact and EGF preferentially stimulates the forma-

tion of ErbB2 ⁄EGFR heterodimers [5,6], in the same

raft fractions of HC11 cells analyzed previously, we

also examined the distribution of EGFR. EGFR was

present in fractions 5 and 6, but also in fractions 7–11

(Fig. 5A). In EGF-stimulated cells, movement of the

receptor to TX-100-soluble fractions was observed (Fig.

5B). After [D]-PDMP treatment, the receptor became

completely Triton soluble and was present exclusively

in fractions 10 and 11 (Fig. 5C). After [D]-PDMP incu-

bation, followed by GM3 treatment, a proportion of

EGFR returned to fractions 4–6 (Fig. 5D).

Because the Shc proteins are translocated into the

lipid rafts of the plasma membrane after phosphoryla-

tion by ErbB2 and EGFR receptors [13], we also ana-

lyzed the distribution of Shc-p66 in the same fractions

(Fig. 6). The results revealed that in control cells only

a small amount of Shc-p66 was detectable in fractions

5 and 6, corresponding to lipid rafts; a higher amount

of Shc-p66 was detected in TX-100-soluble fractions

(mainly 10 and 11) (Fig. 6A). In cells treated with

EGF the higher amount of Shc-p66 was detected in

the Triton-insoluble fractions (Fig. 6B), indicating that

A

B

C

D

E

Fig. 4. Analysis of the distribution of phosphorylated ErbB2 in

HC11 sucrose gradient membrane fractions. HC11 cells were lyzed

in lysis buffer and the supernatant (postnuclear fraction) was sub-

jected to sucrose density gradient separation. After centrifugation,

the gradient was fractionated and each fraction was analyzed by

western blotting with anti-(phospho-ErbB2) polyclonal serum, fol-

lowed by incubation with an HRP-conjugated anti-(rabbit IgG)

serum, as a secondary antibody. (A) Untreated HC11 cells. (B)

HC11 cells treated with 10 nM EGF for 15 min. (C) HC11 cells treat-

ed with 30 lM [D]-PDMP for 5 days. (D) HC11 cells treated with

30 lM [D]-PDMP for 5 days and then with 10 nM EGF for 15 min.

(E) HC11 cells treated with 30 lM [D]-PDMP for 5 days and then

with 125 lM GM3 for 5 min.

A

B

C

D

Fig. 5. EGFR distribution in HC11 sucrose gradient membrane fract-

ions. HC11 cells were lyzed in lysis buffer and the supernatant

(postnuclear fraction) was subjected to sucrose density gradient

separation. After centrifugation, the gradient was fractionated and

each gradient fraction analyzed by western blotting with anti-EGFR

polyclonal serum, followed by incubation with an HRP-conjugated

anti-(rabbit IgG), as a secondary antibody. (A) Untreated HC11 cells.

(B) HC11 cells treated with 10 nM EGF for 15 min. (C) HC11 cells

treated with 30 lM [D]-PDMP for 5 days. (D) HC11 cells treated

with 30 lM [D]-PDMP for 5 days and then with 125 lM GM3 for

5 min.

ErbB2–raft association in HC11 cells E. Sottocornola et al.

1824 FEBS Journal 273 (2006) 1821–1830 ª 2006 The Authors Journal compilation ª 2006 FEBS

EGF induced Shc-p66 recruitment to lipid rafts.

Importantly, [D]-PDMP treatment caused a significant

modification of its membrane distribution, inducing,

as for ErbB2 and EGFR, an almost complete shift

of Shc-p66 to Triton-soluble fractions 10 and 11

(Fig. 6C), which was partially reverted in ganglioside-

depleted HC11 cells by the addition of GM3 and EGF

(Fig. 6D).

Association of ErbB2 with Shc-p66

To verify whether Shc-p66 may interact with activated

ErbB2, lysates from TX-100-insoluble fractions (frac-

tions 4–6 pooled) and TX-100-soluble fractions (frac-

tions 10 and 11 pooled), obtained from EGF-treated

and untreated cells, were immunoprecipitated with the

anti-Shc-p66 Ab, followed by protein G–acrylic beads.

The results in Fig. 7A show that, in control unsti-

mulated cells, ErbB2 was slightly associated with Shc-

p66 mainly in TX-100 soluble fractions. By contrast,

after triggering with EGF, a significant proportion of

Shc-p66 also became associated with ErbB2 in the

TX-100-insoluble fractions, suggesting that, after EGF

stimulation, Shc-p66 may associate with activated

ErbB2 within lipid rafts.

No bands were detected after EGF stimulation in

control immunoprecipitation experiments with a rabbit

IgG having irrelevant specificity. Immunoprecipitation

was verified by western blot (Fig. 7B).

Profile distribution of raft markers in plasma

membrane fractions of HC11 cells

Because gangliosides are well-known markers of lipid-

raft domains, we examined the ganglioside profile of

sucrose-gradient fractions from HC11 cells. Ganglio-

sides were extracted in chloroform ⁄methanol ⁄waterand separated by HPTLC. Resorcinol-positive bands

were identified on the basis of their HPTLC mobility,

compared with standard reference molecules. Three

main resorcinol positive bands, having a retardation

factor (Rf) analogous to GM3, GM2 (the most prom-

inent) and GD1a, respectively, were detected

(Fig. 8A). The observation that the main band comi-

grates with GM2 is not surprising, because this mole-

cule is the main ganglioside constituent in these cells,

as reported previously [11]. All the ganglioside bands

were exclusively detectable in fractions 4–6, which,

under our experimental conditions, correspond to

lipid rafts.

A

B

C

D

Fig. 6. Shc-p66 distribution in HC11 sucrose gradient membrane

fractions. HC11 cells were lyzed in lysis buffer and the supernatant

(postnuclear fraction) was subjected to sucrose density gradient

separation. After centrifugation, the gradient was fractionated and

each gradient fraction was analyzed by western blotting with anti-

Shc polyclonal serum, followed by incubation with an HRP-conju-

gated anti-(rabbit IgG), as a secondary antibody. (A) Untreated

HC11 cells. (B) HC11 cells treated with 10 nM EGF for 15 min. (C)

HC11 cells treated with 30 lM [D]-PDMP for 5 days. (D) HC11 cells

treated with 30 lM [D]-PDMP for 5 days and then with 125 lM

GM3 for 5 min plus 10 nM EGF for 15 min.

A

B

Fig. 7. HC11 cells, treated or not with EGF (10 nM for 5 min at 37 �C), were lyzed in lysis buffer and the supernatant (postnuclear fraction)

was subjected to sucrose density gradient separation. After centrifugation, the gradient was fractionated and coimmunoprecipitation of Shc-

p66 with ErbB2 was performed in TX-100-insoluble fractions (4–6 pooled together) or in TX-100-soluble fractions (10 and 11 pooled together)

with an anti-Shc-p66 specific serum. The immunoprecipitates were analyzed by western blot with anti-ErbB2 (A) and anti-Shc-p66 (B) sera.

E. Sottocornola et al. ErbB2–raft association in HC11 cells

FEBS Journal 273 (2006) 1821–1830 ª 2006 The Authors Journal compilation ª 2006 FEBS 1825

To further confirm the correct sucrose density gradi-

ent separation of TX-100-insoluble and TX-100-soluble

fractions, we also analyzed the distribution pattern of

the known raft protein flotillin-2.

The analysis was performed loading fraction samples

by volume. Because the protein content of TX-100-

soluble fractions 10 and 11 was much higher than that

of TX-100-insoluble fractions 4–6 (not shown) [12], we

can observe that flotillin-2 was consistently enriched in

rafts (TX-100-insoluble fractions) (Fig. 8B).

Discussion

In this study we analyzed primarily the localization of

ErbB2 in lipid rafts of mouse mammary epithelial

HC11 cells. Laser scanning confocal microscopy obser-

vations revealed colocalization areas between GM3, a

well-known marker of lipid rafts [14], and ErbB2. This

finding is in agreement with and extends previous

observations about the surface distribution of ErbB2,

which is mostly excluded from clathrin-coated pits on

the cell plasma membrane [15], and it gives further

support to the conclusions of Nagy et al. [9], who

hypothesized the association of ErbB proteins (ErbB2

and ErbB3) with these microdomains by quantitative

fluorescence microscopy in SKBR-3 breast cancer cells.

In addition, in CHO-K1 cells, expression of GD3

affected, to some extent, the plasma membrane distri-

bution of endogenous ErbB2 [16]. The preferential

distribution of ErbB2 in lipid rafts was clearly demon-

strated by our membrane fractionation experiments,

which also revealed that EGF is not able to modify

the receptor localization. However, analysis revealed

that ErbB2 is not exclusively associated with the raft

fractions. This finding is consistent with the observa-

tions of Hommelgaard et al. [10], prompting us to

hypothesize that ErbB2 is in dynamic equilibrium with

lipid rafts in the membrane protrusions so that, at a

single time point, only a fraction of ErbB2 is directly

interacting with the raft gangliosides. This transient

ErbB2–gangliosides interaction could potentially regu-

late the function of ErbB2 (heterodimerization, signa-

ling and metabolic fate) [10].

We therefore analyzed the role of gangliosides in the

association of ErbB2 with lipid rafts in mammary epi-

thelial HC11 cells. The key role played by gangliosides

in defining the distribution of ErbB2 into signaling

specialized plasma membrane domains was shown by

treatment of HC11 cells with [D]-PDMP. Ganglioside

depletion, due to the inhibition of endogenous ganglio-

side synthesis, was shown to have striking effects upon

the plasma membrane localization of ErbB2. Indeed,

ErbB2 underwent complete redistribution within the

high-density TX-100-soluble fractions of the plasma

membrane, indicating, by a novel approach, that gan-

gliosides play a key role in the retention of this protein

in lipid rafts. These findings are strongly supported by

the observation that [D]-PDMP does not destroy the

organization of lipid rafts, because cholesterol as well

as caveolin-1 could still be detected in TX-100-insol-

uble fractions after treatment with [D]-PDMP. In addi-

tion, treatment of ganglioside-depleted HC11 cells with

exogenous ganglioside GM3 induced the return of a

significant proportion of ErbB2 in raft fractions. How-

ever, both ErbB2 localized in TX-100-soluble and

TX-100-insoluble fractions were phosphorylated. These

data, together with previous results [11], strengthen the

view that GM3 plays an important role in ErbB2

membrane localization but not in its phosphorylation,

suggesting that gangliosides might influence the signa-

ling-transduction pathways after EGF stimulation by

compartmentalizing the receptor in different membrane

domains.

Because in cells coexpressing ErbB2 and EGFR, like

HC11 cells, ligand stimulation largely favors the for-

mation of ErbB2 ⁄EGFR heterodimers [5,6], EGFR

distribution in the same plasma membrane fractions

was also analyzed. Conflicting results have been repor-

ted in the literature on the presence of EGFR within

lipid rafts [7,17–21]. It may depend on the cell type,

the use of different detergents [17,18] and, mainly, the

ganglioside composition of the cells. The latter may

also be influenced by cell cycle and ⁄or cell density [19].

Zurita et al. [16] found EGFR mainly in TX-100-

soluble fractions in CHO-K1 cells, although they

observed that EGFR and GD3 colocalized on the cell

surface. In the same vein, Wang et al. demonstrated

1 2 3 4 5 6 7 8 9 10 11 St

GM3GM2GM1

GD1aGD1bGT1b

1

Flotillin

Fraction 2 3 4 5 6 7 8 9 10 11

45 kDa

A

B

Fig. 8. Ganglioside distribution in HC11 sucrose gradient membrane

fractions. HC11 cells were lyzed in lysis buffer and the supernatant

(postnuclear fraction) was subjected to sucrose density gradient

separation. After centrifugation, 11 gradient fractions were recov-

ered. (A) Gangliosides were extracted in chloroform ⁄methanol ⁄water from each fraction and analyzed by HPTLC. St: standard gan-

gliosides GM3, GM2, GM1, GD1a, GD1b, GT1b. (B) Western blot-

ting with anti-(flotillin-2) polyclonal serum in the same fractions.

ErbB2–raft association in HC11 cells E. Sottocornola et al.

1826 FEBS Journal 273 (2006) 1821–1830 ª 2006 The Authors Journal compilation ª 2006 FEBS

that endogenous overexpression of GM3 promotes co-

immunoprecipitation of GM3 with EGFR [20]. How-

ever, it has been shown that GM3 can specifically

interact with the purified recombinant extracellular

domain of EGFR [22] and that this tyrosine kinase

receptor contains a structural domain with targeting

information for lipid domains [23]. Our results dis-

played variations in the distribution profiles rather

similar to that of ErbB2. Indeed, in control cells,

EGFR is mainly enriched in TX-100-insoluble

fractions, whereas treatment with [D]-PDMP and

[D]-PDMP ⁄EGF shifts the receptor towards the TX-

100-soluble fractions, confirming the direct correlation

between the two receptors [3,5]. However, after stimu-

lation with EGF, we observed a movement of EGFR

to Triton-soluble fractions, in agreement with previous

studies showing that EGFR is initially concentrated in

caveolae within lipid rafts, but rapidly moves out of

this membrane domain in response to EGF [7]. The

inefficient movement of ErbB2 out of these micro-

domains may be related to its impaired internalization

by clathrin-coated pits [24].

ErbB receptors, and ErbB2 in particular, are able to

activate the ras ⁄MAP kinase signaling pathway via the

Shc proteins [25]. In order to elucidate whether this sig-

nal transduction pathway triggered by ErbB2 may take

place inside rafts in HC11 cells, we investigated the

presence of Shc-p66 in raft fractions. Our findings from

fractionation experiments showed a preferential associ-

ation of Shc-p66 with lipid rafts after EGF stimulation.

These data were also confirmed by coimmunoprecipitat-

ion experiments, in which a consistent proportion of

Shc-p66 coimmunoprecipitates with ErbB2 in the lipid-

raft fractions, suggesting that actually Shc-p66 is

recruited by ErbB2 after triggering via EGF.

In conclusion, we demonstrated a key role for gan-

gliosides in the association of ErbB2 with lipid rafts

in mammary epithelial HC11 cells. This finding was

strongly supported by the observation that addition of

GM3 after [D]-PDMP treatment induced a marked

redistribution of ErbB2 and proteins correlated with

the its signal transduction pathway (i.e. Shc-p66) to

Triton-insoluble fractions.

Experimental procedures

Cell culture and treatments

Mouse mammary epithelial HC11 cells were a gift from

E. Garattini (Institute for Pharmacological Research ‘M.

Negri’, Milan, Italy). Cells were maintained in RPMI-1640

(Gibco-BRL, Life Technologies Italia srl, Italy), supplemen-

ted with 10% heat-inactivated newborn bovine serum,

8 mm glutamine, 50 lgÆmL)1 gentamycin and 5 lgÆmL)1

insulin from bovine pancreas (Sigma, St. Louis, MO), in a

humidified 5% CO2 atmosphere at 37 �C.As described previously [11], total ganglioside depletion

was obtained by treating cells for 5 days at 37 �C with

30 lm [D]-PDMP (Sigma), a competitive inhibitor of gluco-

sylceramide synthetase, resulting in ganglioside biosynthesis

inhibition.

Exogenous ganglioside GM3 treatment of ganglioside-

depleted HC11 cells was performed by incubating the cells at

37 �C for 5 min with 125 lm GM3 (Alexis, S. Diego, CA),

dissolved in routine medium without serum. EGF stimulation

was carried out by incubating the cells, treated or not with

[D]-PDMP, with 10 nm EGF (Sigma) for 15 min at 37 �C.

Analysis of ErbB2–GM3 colocalization by scanning

confocal microscopy

HC11 cells, treated or not with [D]-PDMP and GM3, were

fixed in situ with 4% paraformaldehyde in NaCl ⁄Pi for

30 min at room temperature and then permeabilized with

0.5% TX-100 in NaCl ⁄Pi for 30 min at room temperature.

Cells were labeled with rabbit anti-ErbB2 polyclonal serum

(C18, Santa Cruz Biotechnology, Santa Cruz, CA) for 1 h

at 4 �C, followed by the addition (30 min at 4 �C) of fluo-rescein isothiocyanate (FITC)-conjugated anti-(rabbit IgG)

serum (Calbiochem, La Jolla CA). After three washes in

NaCl ⁄Pi, cells were incubated with GMR6 anti-GM3

monoclonal serum (Seikagaku Corp., Chuo-ku, Tokyo,

Japan) [26] for 1 h at 4 �C, followed by three washes in

NaCl ⁄Pi and the addition (30 min at 4 �C) of Texas

red-conjugated goat anti-(mouse IgM) serum (Sigma). In

parallel experiments, cells were stained with anti-GM3

monoclonal serum before fixing the cells. Alternatively,

control experiments were performed omitting the monoclo-

nal antibody from the immunolabeling procedure. After

washing as above, cells were mounted upside down onto a

glass slide in 5 lL of glycerol ⁄Tris ⁄HCl (6 : 4, v : v), pH 9.2.

As a control, cells were mounted in glycerol ⁄NaCl ⁄Pi (6 : 4,

v : v), pH 7.4 and the results were virtually the same. The

images were acquired using a high-resolution ·63 objective

through a confocal laser scanning microscope Zeiss LSM

510 (Zeiss, Oberkochen, Germany) equipped with argon

and HeNe ion lasers. The green (FITC) and red (Texas

Red) fluorophores were excited simultaneously at 488 and

543 nm. Acquisition of single FITC-stained samples in

dual-fluorescence scanning configuration did not show con-

tribution of green signal in red. Images were collected at

512 · 512 pixels.

Isolation and analysis of lipid-raft fractions

GEM fractions from HC11 cells, treated or not with EGF

(10 nm for 15 min at 37 �C), [D]-PDMP (30 lm for 5 days

E. Sottocornola et al. ErbB2–raft association in HC11 cells

FEBS Journal 273 (2006) 1821–1830 ª 2006 The Authors Journal compilation ª 2006 FEBS 1827

at 37 �C), [D]-PDMP and EGF, [D]-PDMP and GM3

(125 lm for 5 min at 37 �C), or [D]-PDMP and GM3 plus

EGF, were isolated as described previously [27]. Briefly,

2 · 108 cells were suspended in 1 mL of lysis buffer, con-

taining 1% TX-100, 10 mm Tris ⁄HCl pH 7.5, 150 mm

NaCl, 5 mm EDTA, 1 mm Na3VO4, and 75 U aprotinin,

and allowed to stand for 20 min at 4 �C. The cell suspen-

sion was mechanically disrupted by Dounce homogeniza-

tion (10 strokes). The lysate was centrifuged for 5 min at

1300 g to remove nuclei and large cellular debris. The

supernatant fraction (postnuclear fraction) was subjected to

sucrose density gradient centrifugation, i.e. the fraction was

mixed with an equal volume of 85% sucrose (w ⁄ v) in lysis

buffer (10 mm Tris ⁄HCl pH 7.5, 150 mm NaCl, 5 mm

EDTA). The resulting diluent was placed at the bottom of

a linear sucrose gradient (5–30%) in the same buffer and

centrifuged at 200 000 g for 16–18 h at 4 �C in a SW41

rotor (Beckman Institute, Palo Alto, CA). After centrifuga-

tion, the gradient was fractionated, and 11 fractions were

collected starting from the top of the tube. All steps were

performed at 0–4 �C. The amount of protein in each frac-

tion was first quantified by Bio-Rad protein assay (Bio-Rad

Laboratory GmbH, Munchen, Germany).

Finally, fractions were subjected to cholesterol analysis,

western blot, immunoprecipitation experiments or ganglio-

side extraction.

Analysis of cholesterol content

All the fractions obtained as reported above from HC11

cells, treated or not with [D]-PDMP, were subjected to cho-

lesterol analysis. The amount of cholesterol was evaluated

as described previously [28]. Free cholesterol was quantified

from TLC plates by densitometric scanning. The density of

the bands used to quantitate cholesterol concentration fell

within the linear range of compound concentration vs.

absorbance.

Immunoblotting analysis of plasma membrane

fractions

All the fractions obtained as reported above were subjected

to 7.5 or 10% SDS ⁄PAGE. Equal volumes of each fraction

were loaded in SDS ⁄PAGE, according to Parolini et al.

[12]. The proteins were electrophoretically transferred to

nitrocellulose membrane (Bio-Rad, Hercules, CA) and then,

after blocking with NaCl ⁄Pi containing 1% albumin,

probed with rabbit anti-(ErbB2 IgG) polyclonal serum, rab-

bit anti-(phospho ErbB2) polyclonal serum (Sigma), rabbit

anti-(EGFR IgG) polyclonal serum, clone 1005 (Santa Cruz

Biotechnology), rabbit anti-(Shc IgG) polyclonal serum

(Transduction Laboratories, Lexington, KY) or, as con-

trols, rabbit anti-(caveolin-1) polyclonal serum (N-20, Santa

Cruz Biotechnology) or goat anti-(flotillin-2) polyclonal

serum (C-20, Santa Cruz Biotechnology). Bound antibodies

were visualized with horseradish peroxidase (HRP)-conju-

gated anti-(rabbit IgG) or anti-(goat IgG) serum (Sigma)

and immunoreactivity was assessed by chemiluminescence

reaction using the ECL western blotting detection system

(Amersham, UK). As a control for nonspecific reactivity,

parallel blots were performed as above, using an anti-

(rabbit IgG) serum (Sigma).

Immunoprecipitation experiments

Briefly, TX-100-insoluble (fractions 4–6) or TX-100-soluble

(fractions 10–11) fractions from HC11 cells, untreated or

treated with 10 nm EGF (Sigma) for 15 min at 37 �C were

lyzed in lysis buffer (10 mm Tris ⁄HCl (pH 8.0), 150 mm

NaCl, 1% Nonidet P-40, 1 mm phenylmethylsulfonyl fluor-

ide, 10 lg of leupeptinÆmL)1). Cell-free lysates were mixed

with protein G–acrylic beads and stirred by a rotary shaker

for 2 h at 4 �C to preclear nonspecific binding. After cen-

trifugation (500 g for 1 min), the supernatant was immuno-

precipitated with the rabbit polyclonal anti-(Shc IgG)

serum (Transduction Laboratories) plus protein G–acrylic

beads. A rabbit IgG isotypic control (Sigma) was

employed.

Immunoprecipitates were subjected to western blot anal-

ysis with the rabbit anti-ErbB2 polyclonal serum (Santa

Cruz Biotechnology). Immunoreactivity was assessed by

chemiluminescence reaction using the ECL western blotting

detection system (Amersham).

Ganglioside extraction and analysis by HPTLC

Ganglioside extraction was performed according to the

method of Svennerholm & Fredman [29] with minor modi-

fications. Briefly, glycosphingolipids were extracted twice in

chloroform ⁄methanol ⁄water (4 : 8 : 3 v ⁄ v ⁄ v) and subjected

to Folch partition by the addition of water resulting in

a final chloroform ⁄methanol ⁄water ratio of 1 : 2 : 1.4

(v ⁄ v ⁄ v). The upper phase, containing polar glycosphingo-

lipids, was purified of salts and low molecular mass

contaminants using Bond Elut-C18 columns, 3 mL

(Superchrom, Harbor City, CA), according to the method

of Williams & McCluer [30]. The eluted glycosphingolipids

were dried and separated by HPTLC, using silica gel 60

HPTLC plates (Merck, Darmstadt, Germany). Chromato-

graphy was performed in chloroform ⁄methanol ⁄ 0.25%aqueous KCl (5 : 4 : 1 v ⁄ v ⁄ v). Plates were then air-dried

and gangliosides visualized with resorcinol.

References

1 Simons K & Ikonen E (1997) Functional rafts in cell

membranes. Nature 387, 569–572.

2 Ikonen E (2001) Roles of lipid rafts in membrane trans-

port. Curr Opin Cell Biol 13, 470–477.

ErbB2–raft association in HC11 cells E. Sottocornola et al.

1828 FEBS Journal 273 (2006) 1821–1830 ª 2006 The Authors Journal compilation ª 2006 FEBS

3 Hynes NE & Stern DF (1994) The biology of erbB-2 ⁄neu ⁄HER-2 and its role in cancer. Biochim Biophys Acta

1198, 165–184.

4 Dougall WC, Qian X, Peterson NC, Miller MJ,

Samanta A & Greene MI (1994) The neu-oncogene:

signal transduction pathways, transformation mechan-

isms and evolving therapies. Oncogene 9, 2109–2123.

5 Qian X, LeVea CM, Freeman JK, Dougall WC &

Greene MI (1994) Heterodimerization of epidermal

growth factor receptor and wild-type or kinase-deficient

Neu: a mechanism of interreceptor kinase activation

and transphosphorylation. Proc Natl Acad Sci USA 91,

1500–1504.

6 Gulliford TJ, Huang GC, Ouyang X & Epstein RJ

(1997) Reduced ability of transforming growth factor-

alpha to induce EGF receptor heterodimerization and

downregulation suggests a mechanism of oncogenic

synergy with ErbB2. Oncogene 15, 2219–2223.

7 Mineo C, Gill GN & Anderson RCW (1999) Regulated

migration of epidermal growth factor receptor from

caveolae. J Biol Chem 274, 30636–30643.

8 Zhou W & Carpenter G (2001) Heregulin-dependent

translocation and hyperphosphorylation of ErbB-2.

Oncogene 20, 3918–3920.

9 Nagy P, Vereb G, Sebestyen Z, Horvath G, Lockett SJ,

Damjanovich S, Park JW, Jovin TM & Szollosi J (2002)

Lipid rafts and the local density of ErbB proteins influ-

ence the biological role of homo- and heteroassociation

of ErbB2. J Cell Sci 115, 4251–4262.

10 Hommelgaard AM, Lerdrup M & van Deurs B (2004)

Association with membrane protrusions makes ErbB2

an internalization-resistant receptor. Mol Biol Cell 15,

1557–1567.

11 Sottocornola E, Berra B & Colombo I (2003) GM3 con-

tent modulates the EGF-activated p185c-neu levels, but

not those of the constitutively activated oncoprotein

p185neu. Biochim Biophys Acta 1635, 55–66.

12 Parolini I, Topa S, Sorice M, Pace A, Ceddia P, Mon-

tesoro E, Pavan A, Lisanti MP, Peschle C & Sargia-

como M (1999) Phorbol ester-induced disruption of the

CD4–Lck complex occurs within a detergent-resistant

microdomain of the plasma membrane. Involvement of

the translocation of activated protein kinase C isoforms.

J Biol Chem 274, 14176–14187.

13 Lotti LV, Lanfrancone L, Migliaccio E, Zompetta C,

Pelicci G, Salcini AE, Falini B, Pelicci PG & Torrisi

MR (1996) Shc proteins are localized on endoplasmic

reticulum membranes and are redistributed after tyro-

sine kinase receptor activation. Mol Cell Biol 16, 1946–

1954.

14 Garofalo T, Lenti L, Longo A, Misasi R, Mattei V,

Pontieri GM, Pavan A & Sorice M (2002) Association

of GM3 with Zap-70 induced by T cell activation in

plasma membrane microdomains: GM3 as a marker of

microdomains in human lymphocytes. J Biol Chem 277,

11233–11238.

15 Lotti LV, Di Lazzaro C, Zompetta C, Frati L & Torrisi

MR (1992) Surface distribution and internalization of

erbB-2 proteins. Exp Cell Res 202, 274–280.

16 Zurita AR, Crespo PM, Koritschoner NP & Daniotti

JL (2004) Membrane distribution of epidermal growth

factor receptors in cells expressing different gangliosides.

Eur J Biochem 271, 2428–2437.

17 Roepstorff K, Thomsen P, Sandvig K & van Deurs B

(2002) Sequestration of epidermal growth factor recep-

tors in non-caveolar lipid rafts inhibits ligand binding.

J Biol Chem 277, 18954–18960.

18 Pike L, Han X & Gross RW (2005) Epidermal growth

factor receptors are localized to lipid rafts that contain

a balance of inner and outer leaflet lipids. J Biol Chem

280, 26796–26804.

19 Rosner H, Greis CH & Rodemann HP (1990) Density-

dependent expression of ganglioside GM3 by human

skin fibroblasts in an all-or-none fashion, as a possible

modulator of cell growth in vitro. Exp Cell Res 190,

161–169.

20 Wang XQ, Sun P & Paller AS (2002) Ganglioside

induces caveolin)1 redistribution and interaction with

the epidermal growth factor receptor. J Biol Chem 277,

47028–47034.

21 Puri C, Tosoni D, Comai R, Rabellino A, Segat D,

Caneva F, Luzzi P, Di Fiore PP & Tacchetti C (2005)

Relationships between EGFR signaling-competent and

endocytosis-competent membrane microdomains. Mol

Biol Cell 16, 2704–2718.

22 Miljan EA, Meuillet EJ, Mania-Farnell B, George D,

Yamamoto H, Simon HG & Bremer EG (2002) Interac-

tion of the extracellular domain of the epidermal growth

factor receptor with gangliosides. J Biol Chem 277,

10108–10113.

23 Yamabhai M & Anderson RGW (2002) Second

cysteine-rich region of epidermal growth factor receptor

contains targeting information for caveolae ⁄ rafts. J Biol

Chem 277, 24843–24846.

24 Baulida J, Kraus MH, Alimandi M, Di Fiore PP &

Carpenter G (1996) All ErbB receptors other than the

epidermal growth factor receptor are endocytosis

impaired. J Biol Chem 271, 5251–5257.

25 Deb TB, Wong L, Salomon DS, Zhou G, Dixon JE,

Gutkind JS, Thompson SA & Johnson GR (1998) A

common requirement for the catalytic activity and both

SH2 domains of SHP-2 in mitogen-activated protein

(MAP) kinase activation by the ErbB family of recep-

tors. A specific role for SHP-2 in MAP, but not c-Jun

amino-terminal kinase activation. J Biol Chem 273,

16643–16646.

26 Kotani M, Ozawa H, Kawashima I, Ando S & Tai T

(1992) Generation of one set of monoclonal antibodies

E. Sottocornola et al. ErbB2–raft association in HC11 cells

FEBS Journal 273 (2006) 1821–1830 ª 2006 The Authors Journal compilation ª 2006 FEBS 1829

specific for a-pathway ganglio-series gangliosides. Bio-

chim Biophys Acta 1117, 97–103.

27 Rodgers W & Rose JK (1996) Exclusion of CD45 inhi-

bits activity of p56lck associated with glycolipid-

enriched membrane domains. J Cell Biol 135, 1515–

1523.

28 Huber LA, Xu Q, Jurgens G, Bock G, Buhler E, Gey

KF, Schonitzer D, Trall KN & Wick G (1991) Correla-

tion of lymphocyte lipid composition membrane micro-

viscosity and mitogen response in the aged. Eur J

Immunol 21, 2761–2765.

29 Svennerholm L & Fredman P (1980) A procedure for

the quantitative isolation of brain gangliosides. Biochim

Biophys Acta 617, 97–109.

30 Williams MA & McCluer RH (1980) The use of Sep-

pak C18 cartridges during the isolation of gangliosides.

J Neurochem 35, 266–269.

ErbB2–raft association in HC11 cells E. Sottocornola et al.

1830 FEBS Journal 273 (2006) 1821–1830 ª 2006 The Authors Journal compilation ª 2006 FEBS