Tellier2006-Adam17 Lipid Rafts

You might also like

Download as pdf or txt
Download as pdf or txt
You are on page 1of 12

E XP E RI ME N TA L CE L L RE S E A RCH 3 1 2 ( 2 00 6 ) 3 9 6 9 –39 8 0

a v a i l a b l e a t w w w. s c i e n c e d i r e c t . c o m

w w w. e l s e v i e r. c o m / l o c a t e / y e x c r

Research Article

The shedding activity of ADAM17 is sequestered in lipid rafts

Edwige Tellier, Matthias Canault, Laure Rebsomen, Bernadette Bonardo,


Irène Juhan-Vague, Gilles Nalbone, Franck Peiretti⁎
Inserm, U626, Marseilles, France
Université de la Méditerranée, Faculté de Médecine, 27 Boulevard Jean Moulin, Marseilles 13385 Cedex 5, France

ARTICLE INFORMATION ABS T R AC T

Article Chronology: The tumor necrosis factor-alpha (TNF) converting enzyme (ADAM17) is a metalloprotease-
Received 27 June 2006 disintegrin responsible for the cleavage of several biologically active transmembrane
Revised version received proteins. However, the substrate specificity of ADAM17 and the regulation of its shedding
28 August 2006 activity are still poorly understood. Here, we report that during its transport through the Golgi
Accepted 31 August 2006 apparatus, ADAM17 is included in cholesterol-rich membrane microdomains (lipid rafts)
Available online 5 September 2006 where its prodomain is cleaved by furin. Consequently, ADAM17 shedding activity is
sequestered in lipid rafts, which is confirmed by the fact that metalloproteinase inhibition
Keywords: increases the proportion of ADAM17 substrates (TNF and its receptors TNFR1 and TNFR2) in
ADAM17 lipid rafts. Membrane cholesterol depletion increases the ADAM17-dependent shedding of
TACE these substrates demonstrating the importance of lipid rafts in the control of this process.
TNF-alpha Furthermore, ADAM17 substrates are present in different proportions in lipid rafts, suggesting
TNF receptors that the entry of each of these substrates in these particular membrane microdomains is
Cholesterol specifically regulated. Our data support the idea that one of the mechanisms regulating
Shedding ADAM17 substrate cleavage involves protein partitioning in lipid rafts.
© 2006 Elsevier Inc. All rights reserved.

Introduction [5]), imposing this enzyme as a centerpiece in mammalian cell


biology. The fact that the genetic inactivation of ADAM17 was
ADAMs (proteins containing A disintegrin and metallopro- perinatally lethal for homozygous knockout mice [6] exempli-
tease domain) are a large family of type I transmembrane fied its key role in cell biology and organism development.
glycoproteins. ADAMs have been implicated in numerous No consensus sequence elements were found after align-
cellular processes [1,2], some of which involve shedding of ment of the cleavage sites of ADAM17 substrates, which would
cell-surface molecules. The best-characterized ADAM protein- indicate that the sequence specificity is not primordial for the
ase is ADAM17 (also known as tumor necrosis factor-alpha cleavage of ADAM17 substrates. A model was proposed in
(TNF) converting enzyme (TACE)), originally discovered as the which ADAM17 would cleave an ectodomain as long as the
processing protease of precursor TNF [3,4]. Subsequently, distance of the segment between the membrane plane and the
ADAM17 was shown to be involved in the cleavage of a wide globular portion of the substrate ectodomain allows for
diversity of ligands and receptors (reviewed by Smalley et al. accessibility to the cleavage site [7]. Such a model could

⁎ Corresponding author. Inserm U626, Faculté de Médecine, 27 Boulevard Jean Moulin, Marseilles 13385 Cedex 5, France. Fax: +33 4 91 25 43
36.
E-mail address: Franck.Peiretti@medecine.univ-mrs.fr (F. Peiretti).

0014-4827/$ – see front matter © 2006 Elsevier Inc. All rights reserved.
doi:10.1016/j.yexcr.2006.08.027
3970 E XP E RI ME N TA L CE LL RE S E A RCH 3 1 2 ( 2 00 6 ) 3 9 6 9 –39 8 0

explain why the transfer of the cleavage site from one protein Isolation of lipid rafts
to another may either result in spontaneous or largely
diminished shedding [8,9]. In these conditions, one way to Lipid rafts were isolated by sucrose density gradient centrifu-
consider a specific regulation of ADAM17 substrate release is to gation of cells treated with non-ionic detergents. Approxi-
envisage a precise regulation of the spatial and temporal mately, 9 × 106 cells were washed with ice-cold PBS and then
expression of ADAM17 and its substrates. A substrate will be harvested by scraping in PBS. The cells were collected by
released only if, at a given time point, it is colocalized with centrifugation, suspended in 900 μl of “raft buffer” [20 mM
activated ADAM17. Thus, the sequestration of the activated HEPES, pH 7.4, 150 mM NaCl, 1 mM EGTA, with protease,
enzyme together with the substrate in particular cell compart- phosphatase and metalloproteinase inhibitors], snapped fro-
ments could be considered as an efficient way to regulate the zen in liquid nitrogen and thawed on ice. The suspension was
shedding process. In favor of this notion, we previously passed 10 times through a 25-gauge needle. Lysis with Triton
demonstrated that an increased transport of ADAM17 sub- X-100 or Tween 20 was performed by adding these detergents
strates from their Golgi apparatus pool towards the cell surface at a final concentration of 1% and 2% (v/v), respectively. The
plays a major role in the stimulated shedding of ADAM17 cell suspension was then incubated on ice for 20 min under a
substrates [10]. Recently, it was shown that the shedding of gentle agitation. Lysis with Brij 98 was performed by equili-
some ADAM17 substrates like the amyloid precursor protein brating the cell suspension at 37°C, followed by addition of Brij
[11], CD30 [12], interleukin-6 receptor [13], L-selectin [14] and 98 to a final concentration of 1% (w/v). The incubation was
collagen XVII [15] could be increased by cholesterol-lowering carried out at 37°C for 7 min, then the sample was kept on ice.
drugs, which suggests that membrane microdomains rich in One milliliter of the lysate was combined with an equal
cholesterol may play a crucial regulatory role in the ectodo- volume of 90% (w/v) sucrose, transferred to 12 ml ultracentri-
main shedding of ADAM17 substrates. fuge tubes and overlaid with a discontinuous sucrose gradient
The cholesterol-rich membrane microdomains, also called comprised of 4 ml of 35% sucrose, followed by 2 ml of 5%
“lipid rafts”, are assemblies of sphingolipids and cholesterol sucrose. Separation of the low-density lipid rafts was achieved
within the membrane. They are fluid but tightly packed and by centrifugation at 280,000×g in a swinging bucket rotor for
well ordered, representing the liquid ordered phase that floats 18–22 h at 4°C. Following centrifugation, 0.5 ml fractions were
around in the liquid disordered matrix of the entire membrane harvested from the top of the gradient. Aliquots of selected
[16]. Proteins exhibit different association capacities to these fractions were analyzed by immunoblot or by ELISA. Separa-
cholesterol-rich microdomains, and their incorporation in tion of lipid rafts was also performed from organs collected
lipid rafts is a dynamic process [17]. The distribution of specific from mice (C57/BL6j). Investigations were approved by the
membrane proteins between the lipid raft and non-raft phases local ethic committee of the Faculté de Médecine of Marseille.
and their concentrations in lipid rafts regulate their interac- Mice were killed by excessive anesthesia using isoflurane and
tions and influence their functions [18]. liver, heart and spleen were collected. They were frozen in
In this work, we show that the furin-dependent maturation liquid nitrogen, pulverized using a steel beads shaker,
of ADAM17 occurs in the cholesterol-rich membrane micro- incubated 1 h at 4°C with the “raft buffer”. The homogenate
domains. As a consequence, the mature form of ADAM17 is was treated as described above for lipid raft preparation using
concentrated in lipid rafts, where its substrates (TNF and its Brij 98. In some cases, instead of performing lipid raft
receptors TNFR1 and TNFR2) are cleaved. The importance of separation on sucrose density gradient, the Brij 98 cell lysate
lipid rafts in the regulation of ADAM17 substrate shedding is was centrifuged (1,000×g, 10 min, 4°C) to remove nuclei and
revealed by the fact that lipid rafts disruption increases the cellular debris. Then, the resulting supernatant was submitted
shedding of TNF, TNFR1 and TNFR2. to high-speed centrifugation (100,000×g, 1 h, 4°C ) to pellet the
Brij 98 insoluble material corresponding to the total lipid raft
fraction. The pellet was washed with the “raft buffer”, then
Materials and methods solubilized in the SDS–PAGE sample buffer for immunoblot
analyses or in the “raft buffer” containing 60 mM of octyl beta-
Materials D-glucopyranoside for ELISA analyses.

Metalloproteinase inhibitor RU 36156 active on ADAM17 [19] Isolation of the cytoskeleton fraction
was donated by Dr. S. Roman-Roman (Aventis Pharma,
Romainville, France). Phosphatase inhibitor cocktail 2, α- Cytoskeleton-rich fraction was isolated as previously de-
cyclodextrin, methyl-β-cyclodextrin (CD), cholesterol-loaded scribed [20]. This fraction was solubilized in the SDS–PAGE
cyclodextrin, filipine III, cholesterol oxidase, sphingomyeli- sample buffer for immunoblot analyses or incubated with 1 ml
nase, PMA, calcium ionophore A23187 and LPS from Escherichia of “raft buffer” containing 0.6 M KI (30 min, 4°C) in order to
coli serotype 026:B6 (LPS) were from Sigma. Protease inhibitor depolymerize actin filaments [21], mixed to 1 ml of 90%
cocktail Complete EDTA-free was from Roche. The proprotein sucrose solution and submitted to lipid raft isolation.
convertase inhibitor decanoyl-RVKR-chloromethylketone was
from Bachem. ADAM17 polyclonal antibody, TIMP3, recombi- Expression vectors
nant human ADAM17 and the fluorogenic peptide III were from
R&D Systems. Caveolin-1 and Flotilin-1 polyclonal antibodies Expression vectors for human TNF, TNFR1 and TNFR2 were
were from Santa Cruz Biotechnology. FLAGm2 polyclonal described elsewhere [10,22]. Expression vector for human
antibody was from AffinityBio Reagents. furin-flag (pcDNA3) was given by Dr. G. Thomas (Portland, OR).
E XP E RI ME N TA L CE L L RE S E A RCH 3 1 2 ( 2 00 6 ) 3 9 6 9 –39 8 0 3971

Cell culture, microvesicle preparation and transfection using three different detergents. The very selective Triton X-
100, the less selective Tween 20 and the intermediate Brij 98
The monkey kidney fibroblast cell line COS-7, the rat [24]. When Triton X-100 was used, the majority of the proform
cardiomyoblast cell line H9C2, the human colonic epithelial and mature form of ADAM17 as well as the widely used lipid
cell lines HT29 and LoVo cells and the human monocytic cell raft marker caveolin-1 were detected in the gradient fractions
line THP-1 were cultured as suggested by ATCC (http://www. of the highest density (Fig. 1A). Minute amounts of the mature
lgcpromochem-atcc.com/). Microvesicles (MVs) were genera- form of ADAM17 and caveolin-1 were detected in the light
ted by stimulating THP-1 (5 × 106 cells/ml) with 5 μM calcium density gradient fractions corresponding to the region of lipid
ionophore A23187 for 15 min and isolated as previously raft flotation (Fig. 1A). The large amount of caveolin-1 in the
described [23]. Transient transfections were performed with high-density gradient fractions suggests that Triton X-100
Polyfect reagent (Qiagen) essentially as described by the solubilized a main part of the proteins contained in lipid rafts.
manufacturer. The stable ectopic expression of TNF in the Using Tween 20 or Brij 98, we found that most of the mature
human endothelial cell line ECV-304 (ECVTNF) and their culture form of ADAM17 and caveolin-1 were detected in the light
conditions were described previously [10]. density gradient fractions and minor amounts of these
proteins were detected in the highest density gradient
Fluorogenic assays of metalloprotease activity fractions (Fig. 1A). The large amount of caveolin-1 in the
light density gradient fractions suggests that these two
THP-1 MVs were added to the activity buffer [25 mM Tris/HCl pH detergents maintain the lipid raft cohesion and are thus
8.0, 2.5 μM ZnCl2] containing 10 μM of pro-TNF mimetic fluo- adapted for lipid raft separation. The proform of ADAM17 was
rogenic peptide III that was designed as an ADAM17 substrate. exclusively detected in gradient fractions of the highest
Its fluorescence-related enzymatic cleavage was monitored at density. These results strongly suggest that a part of the
320 nm excitation and 405 nm emission wavelength using a mature form of ADAM17 is contained in lipid rafts. In the
microplate fluorescence reader (Chameleon™, Hidex). Recom-
binant human ADAM17 was used as the positive control. Blank
(buffer and MVs and substrate, separately) was subtracted from
sample measurements for calculations.

Protein assays

Total amounts (free and bound forms) of transmembrane and


soluble forms of human or mouse TNF, TNFR1 and TNFR2 and
transmembrane human ICAM-1 were assayed according to
the specifications of their respective enzyme-linked immu-
nosorbent assays (ELISA) kits (R&D Systems).

Statistical analyses

Isolations of lipid rafts by sucrose density gradient centrifu-


gation were repeated at least three times and a representative
experiment is presented. All other experiments were per-
formed in triplicate and repeated at least three times. All data
are expressed as the mean ± SD. Treatments were compared
with their respective controls and statistical calculation of the
mean differences was performed with the two-tailed t-test. A
value of p < 0.05 was considered statistically significant.

Fig. 1 – The mature form of ADAM17 is sequestered in lipid


Results rafts. (A) ECVTNF cell lysates were prepared as indicated in the
Materials and methods section using Triton X-100, Tween 20
The mature form of ADAM17 is contained in lipid rafts or Brij 98. Where indicated, cells were treated for 30 min with
1% methyl-β-cyclodextrin (CD) prior the harvesting. After
On a physicochemical point of view, lipid rafts could be fractionation by sucrose density gradient centrifugation,
defined as membrane domains that are resistant to extraction ADAM17 and caveolin-1 (Cav1) were detected by immunoblot
in mild neutral detergents and that float in the upper half of a in the indicated gradient fractions. (B) Cells were left
5–30% sucrose density gradient. However, it is well admitted untreated (Cont) or treated for 3 h with 80 nM of PMA, 10 μM
that lipid rafts obtained with different detergents differ of RU 36156 (RU) or both (PMA+RU), prior the harvesting.
considerably in their protein and lipid contents [24]. To avoid Indicated fractions were pooled and ADAM17 and caveolin-1
misinterpretation linked to the ability of the detergent to (Cav1) were detected by immunoblot. Pro and mature forms
selectively solubilize membrane proteins, we separated lipid of ADAM17 are indicated on the right hand side of the blots by
rafts from ECVTNF cells (endothelial cells overexpressing TNF) p and m, respectively.
3972 E XP E RI ME N TA L CE LL RE S E A RCH 3 1 2 ( 2 00 6 ) 3 9 6 9 –39 8 0

course of the work, separations of lipid rafts were made using


Brij 98 because it solubilizes disordered membrane at 37°C,
which suggests that isolated lipid rafts possess the biochem-
ical characteristics of those present in the cell membrane at
the physiological temperature [25].
To prove that the mature form of ADAM17 detected in the
light density gradient fractions was indeed integrated in
cholesterol-rich membrane microdomains, we disrupted lipid
rafts and analyzed the repartition of the mature form of
ADAM17 in a sucrose gradient. For this purpose, cells were
incubated for 30 min with the cholesterol lowering/raft
disrupter agent methyl-β-cyclodextrin (CD), which resulted
in the loss of about 60% of the cellular cholesterol (data not
shown) without altering the viability of the cells within this
time period. In this situation, the mature form of ADAM17 as
well as caveolin-1 were detected in gradient fractions of a
higher density than that originally containing lipid rafts (Fig.
1A), which supports the concept that these two proteins are Fig. 2 – THP-1 microvesicles are enriched in the active form of
integrated in lipid rafts. To further identify the protein detected ADAM17. (A) THP-1 cell lysate was prepared as described in
by the anti-ADAM17 antibody, we treated cells with phorbol- the Materials and methods section using Brij 98. After
12,13-myristate acetate (PMA), which reduces the amount of fractionation by sucrose density gradient centrifugation,
the mature form of ADAM17 [26], a phenomenon inhibited by ADAM17 and flotilin-1 (Flot1) were detected by immunoblot
metalloproteinase inhibition. The signal detected with the in the indicated gradient fractions and (B) in lysates of THP-1
anti-ADAM17 antibody in lipid rafts prepared from PMA- and THP-1 microvesicles (MVs) with similar protein content.
treated cells was reduced compared with that detected in (C) Fluorogenic assay of the metalloprotease activity of
lipid rafts prepared from untreated cells and this effect was human recombinant ADAM17 (rADAM17) and of MVs was
reversed by the metalloproteinase inhibitor RU 36156 (Fig. 1B), performed as described in the Materials and methods section
strongly suggesting that the protein detected in lipid rafts by in presence or absence of TIMP3 (100 nM). Pro and mature
the anti-ADAM17 antibody is actually the mature form of forms of ADAM17 are indicated on the right hand side of the
ADAM17. RU 36156 alone was without effect on the distribution blots by p and m, respectively. *Significant versus the
of ADAM17 and caveolin-1 (Fig. 1B). situation without TIMP3 (p < 0.001).
The mature form of ADAM17 was detected in lipid rafts
prepared from several cell types (see throughout the text),
suggesting that its integration in lipid rafts is a general Furthermore, it was previously shown that the attachment of
phenomenon. lipid rafts to the cytoskeleton increased their density [28]. We
To further prove that the integration of the mature form of thus hypothesized the existence of very high-density lipid
ADAM17 in lipid rafts was not the consequence of our method rafts attached to the cytoskeleton and containing the mature
of preparation, we took advantage of the ability of THP-1 cells form of ADAM17. This hypothesis is sustained by the
to generate microvesicles (MVs) rich in lipid rafts [27] and we following observations: (i) when more material was used for
analyzed the ADAM17 content of these MVs. As expected, after the detection of caveolin-1, we noticed its presence in the
the separation of lipid rafts from THP-1 cells, the mature form high density gradient fractions (Fig. 3A); and (ii) a small
of ADAM17 and the raft marker flotilin-1 were detected in the amount of the mature form of ADAM17 was detected in the
light density gradient fractions whereas the proform of gradient fraction of the highest density (Fig. 1A). After low-
ADAM17 was excluded from these fractions (Fig. 2A). MVs speed centrifugation of the cell lysate, the mature form of
originated from THP-1 cells were largely enriched in the ADAM17 was mostly recovered in the fraction containing the
mature form of ADAM17 and flotilin-1 compared with the cytoskeleton which also contained most of the actin and
original cells (Fig. 2B). The densitometric analysis of four caveolin-1 (Fig. 3B). The proform of ADAM17 was detected in
independent immunoblots showed that the ratio mature the cytoskeleton-free fraction that contains cytosolic proteins
form/proform of ADAM17 was significantly higher (p = 0.002) and membrane proteins solubilized by Brij 98. To verify that
in MVs (5.5 ± 09) than in THP-1 cells (0.67 ± 0.3). Furthermore, a the mature form of ADAM17 that cosedimented with the
metalloproteinase activity that can be significantly inhibited cytoskeleton was indeed sequestered in lipid rafts, we
at 50% by the ADAM17 inhibitor TIMP3 was measured at the pelleted the cytoskeleton by low-speed centrifugation and
surface of these MVs suggesting that the mature form of performed lipid raft separation on the resulting supernatant
ADAM17 is active (Fig. 2C). (cytoskeleton-free) and pellet (cytoskeleton). In the cytoske-
leton-free preparation, the mature form of ADAM17 and
A subset of lipid rafts containing the mature form of ADAM17 caveolin-1 was mostly detected in the lipid raft fractions (Fig.
is associated with the cytoskeleton 3C). Actin was detected in the lipid raft region, but most of the
actin was found in the gradient fraction of the highest density
We recently described that the mature form of ADAM17 was (Fig. 3C). This result shows that polymerized actin is still
more prone to bind the cytoskeleton than its proform [20]. present in the sample after cytoskeleton elimination. Before
E XP E RI ME N TA L CE L L RE S E A RCH 3 1 2 ( 2 00 6 ) 3 9 6 9 –39 8 0 3973

Thus, to study the membrane localization of furin, one of the


proprotein convertases that catalyzes the cleavage of ADAM17
prodomain [22], we ectopically expressed a furin-flag in COS-7
cells, separated the total lipid raft fraction from the lipid raft-
free fraction by direct high-speed centrifugation (100,000×g,
1 h) of the 1% Brij 98 cell lysates and performed immunoblots
on each fraction. Furin was enriched in the total lipid raft
fraction (Fig. 4A). This fraction was also enriched in the mature
form of ADAM17 compared with its proform and caveolin-1
(Fig. 4A). This result suggests that furin, like the mature form of
ADAM17, is contained in lipid rafts.
Incubation of ECVTNF cells with the proprotein convertase
inhibitor decanoyl-RVKR-chloromethylketone resulted in less
TNF released from the cells (data not shown) and in the
appearance of the proform of ADAM17 in lipid rafts (Fig. 4B).
Furthermore, the proform of ADAM17 was detected in lipid
rafts isolated from the furin-deficient human colon epithelial
cell line LoVo whereas only the mature form of ADAM17 was
detected in lipid rafts from another human colon epithelial
cell line HT29 (Fig. 4C). Altogether, these results indicate that
furin cleaves the prodomain of ADAM17 in lipid rafts, although
this maturation process is not necessary for its integration in
lipid rafts.

Fig. 3 – Lipid rafts containing ADAM17 are linked to the


cytoskeleton. (A) Caveolin-1 (Cav1) was detected by
immunoblot as described in Fig. 1, except that 7-fold more
material was loaded onto the gel. (B) ADAM17, actin and
caveolin-1 (Cav1) were detected by immunoblot in the total
ECVTNF cell lysate (lane 1), the cytoskeleton fraction (lane 2)
and the cytoskeleton-free fraction (lane 3). (C) Rafts were
isolated from the cytoskeleton-free fraction and the
cytoskeleton fraction treated with KI (0.6 M) to depolymerize
actin filaments. ADAM17, actin and caveolin-1 (Cav1) were
detected by immunoblot. Pro and mature forms of ADAM17
are indicated by p and m, respectively.

performing lipid raft separation on the cytoskeleton prepara-


tion, the actin-based cytoskeleton was depolymerized by KI
treatment. Actin was only detected in the light fractions of the Fig. 4 – Furin activates ADAM17 in lipid rafts. (A) Furin-flag
gradient together with the mature form of ADAM17 and with was ectopically expressed in COS-7 cells. 48 h
most of the caveolin-1 (Fig. 3C). Altogether, these results show post-transfection, cells were harvested and lysed. ADAM17,
that the mature form of ADAM17 is contained in lipid rafts furin-flag (Furin) and caveolin-1 (Cav1) were detected by
and that a part of these lipid rafts is associated with the actin- immunoblot in the total cell lysate (lane 1), the lipid raft-free
based cytoskeleton. The proform of ADAM17 is not integrated fraction (lane 2) and the total lipid raft fraction (lane 3). (B)
in lipid rafts and is not associated with the cytoskeleton. ECVTNF cells were left untreated (Cont) or incubated for 24 h
with 50 μM of decanoyl-RVKR-chloromethylketone (CMK),
Furin cleaves the prodomain of ADAM17 in lipid rafts then treated as indicated in Fig. 1 and ADAM17 was detected
by immunoblot in indicated gradient fractions. (C) HT-29 and
Analysis of lipid rafts by sucrose density gradient centrifuga- LoVo cells were treated as described in Fig. 1, indicated
tion necessitates a large number of expressing cells. Due to the fractions were pooled and ADAM17 was detected by
low efficiency of transfection, experiments using transient immunoblot. Pro and mature forms of ADAM17 are indicated
ectopic expression are not compatible with such a technique. on the right hand side of the blots by p and m, respectively.
3974 E XP E RI ME N TA L CE LL RE S E A RCH 3 1 2 ( 2 00 6 ) 3 9 6 9 –39 8 0

TNF, TNFR1 and TNFR2 are normally processed by ADAM17 shown). Metalloproteinase inhibition also significantly in-
in lipid rafts creased the percentage of TNF associated with the total lipid
raft fraction isolated by direct high-speed centrifugation of the
Because the mature form of ADAM17 is sequestered in lipid 1% Brij 98 cell lysates (Fig. 5B, inset). This result corroborates
rafts, we hypothesized that the cleavage of ADAM17 sub- that obtained by separation on sucrose gradient and validates
strates occurs in this place. TNF, TNFR1 and TNFR2 were the use of this faster although less discriminative method to
previously described as being ADAM17 substrates [3,4,6,29]. study the membrane repartition and cleavage of ectopically
We first analyzed the repartition of these substrates between expressed TNFR1 and TNFR2. We showed in two different cell
the lipid raft and non-raft regions of the membrane and lines (COS-7 and H9C2) that metalloproteinase inhibition
defined the localization of their cleavage under basal condi- significantly increased the proportion of ectopically expressed
tions. A small proportion of TNF from ECVTNF cells was TNFR1 and TNFR2 in the total lipid raft fraction (Fig. 5C),
detected in the light density fractions of the gradient (11% of suggesting that TNFR1 and TNFR2 are cleaved by ADAM17 in
the total TNF was contained in fractions 3 to 7). Cholesterol lipid rafts. By comparison, ICAM-1, that does not behave like
depletion by CD treatment reduced this proportion (4% of the an ADAM17 substrate [10,30], was detected in lipid rafts, but its
total TNF was contained in fractions 3 to 7) and increased that amount was not modified by the treatment with the metallo-
in the fractions of the gradient of higher density (Fig. 5A). This proteinase inhibitor (Fig. 5D). We previously described that
result suggests that a small proportion of TNF is contained in PMA stimulation of ECVTNF cells increased the synthesis and
lipid rafts. Inhibition of ADAM17 by RU 36156 increased the the ADAM17-dependent shedding of ectopically expressed
proportion of TNF detected in lipid rafts and conversely TNF [10]. However, TNF distributed similarly in the sucrose
reduced that in non-raft fractions (Fig. 5B) without affecting gradient whether these cells were treated or not with PMA (Fig.
the sedimentation profile of ADAM17 and caveolin-1 (data not 6A). Inhibition of metalloproteinases increased the proportion

Fig. 5 – The cleavage of TNF, TNFR1 and TNFR2 occurs in lipid rafts. (A) ECVTNF cell were left untreated or treated for 30 min with
1% CD, then lysates were prepared as described in Fig. 1. After fractionation by sucrose density gradient centrifugation,
TNF was measured by ELISA in each fraction. (B) Same as panel A except that ECVTNF cells were treated or not for 3 h with 10 μM
of RU 36156 (RU). (B, inset) Total lipid raft fractions from ECVTNF cell lysates were prepared by high-speed centrifugation.
(C) COS-7 and H9C2 cells transiently expressing ectopic TNFR1 and TNFR2 were treated as in (B, inset). TNF, TNFR1 and TNFR2
were measured by ELISA in the raft and non-raft fractions. The % of ADAM17 substrate present in the total lipid raft fraction is
given by the formula: [substrate]raft × 100 / ([substrate]raft + [substrate]non-raft). (D) Same as panel B except that ICAM-1 was
measured (AU: arbitrary unit).
E XP E RI ME N TA L CE L L RE S E A RCH 3 1 2 ( 2 00 6 ) 3 9 6 9 –39 8 0 3975

rafts whereas a portion of the mature form of ADAM17 and


caveolin-1 were detected in lipid rafts (Fig. 7A). Comparable
results were obtained on the heart and the spleen (data not
shown). The relative high proportion of the mature form of
ADAM17 detected in the heavy fractions of the gradient could
result from the association of lipid rafts with the cytoskeleton
as shown above. To make TNF levels confidently measurable
in liver, mice were injected with LPS 5 h before organ
collection. This treatment did not alter the presence of the
mature form of ADAM17 in lipid rafts (data not shown). A large
proportion of TNF (40% of the total) was detected in lipid rafts
(Fig. 7B). TNFR1 and TNFR2 were measurable upon unstimu-
lated (data not shown) and LPS-stimulated conditions and
were present in lipid rafts (18% and 40% of the total,
respectively) (Fig. 7C). This result demonstrates that the
localization of the mature form of ADAM17, TNF, TNFR1 and
TNFR2 in cholesterol-rich membrane microdomains is a
physiological phenomenon that does not result from the cell
culture conditions or the ectopic expression.

Alteration of lipid raft integrity increases ADAM17 substrate


release

Because cholesterol depletion reduced the amount of the


mature form of ADAM17 present in lipid rafts (Fig. 1A), we

Fig. 6 – The PMA-stimulated cleavage of TNF occurs in lipid


rafts. (A) ECVTNF cells were left untreated (Control) or
treated for 3 h with 80 nM of PMA in absence or presence of
10 μM of RU 36156 (PMA + RU). After fractionation by sucrose
density gradient centrifugation, TNF was measured by
ELISA in each fraction. (B) Same as panel A to simplify the
reading only the PMA and PMA + RU situations are shown.
TNF in each fraction is expressed as the percentage of the
total amount of TNF present in the gradient.

of TNF in the lipid rafts, reflecting ADAM17 inhibition, and also


in the non-raft regions (Fig. 6A), which reveals the PMA-
stimulated TNF synthesis. However, when the amount of TNF
present in each gradient fraction was expressed as the
percentage of the total amount of TNF contained in the entire
gradient (Fig. 6B), it is clear that under PMA stimulation,
metalloproteinase inhibition increased the proportion of TNF
only in the lipid raft region. This result demonstrates that the
PMA-stimulated TNF shedding by ADAM17 occurs in lipid
rafts.

In mouse organs, the mature form of ADAM17 and a fraction


of TNF and TNFRs are sequestered in lipid rafts
Fig. 7 – TNF, TNFR1 and TNFR2 are contained in lipid rafts
Freshly collected mouse organs were used to verify that the isolated from mouse organs. Mouse livers were prepared as
above described distribution of ADAM17, TNF, TNFR1 and described under in the Materials and methods section and
TNFR2 in the lipid raft and non-raft regions of the membrane lipid rafts were isolated by sucrose density gradient
was not restricted to cultured cells or due to the ectopic centrifugation. (A) ADAM17 and caveolin-1 (Cav1) were
expression. Mouse livers were collected and lipid rafts were detected by immunoblot in the indicated fractions. (B) TNF
isolated by sucrose density gradient centrifugation. As and (C) TNFR1 and TNFR2 were measured by ELISA in each
expected, the proform of ADAM17 was excluded from lipid fraction.
3976 E XP E RI ME N TA L CE LL RE S E A RCH 3 1 2 ( 2 00 6 ) 3 9 6 9 –39 8 0

Table 1 – Lipid raft disruption increases the release of TNF, TNFR1 and TNFR2
ECVTNF COS-7 H9C2

TNF TNF TNFR1 TNFR2 TNF TNFR1 T NFR2

Control 193.9 ± 96 4320.5 ± 888 98.9 ± 8.5 592.5 ± 42.6 579.2 ± 96 17.3 ± 0.4 83 ± 6.2
CD 385.4 ± 25 6171.1 ± 421 154.6 ± 14 1243 ± 110 1144.2 ± 237 21.9 ± 1.7 152.4 ± 37.4
t-test, p 0.0001 0.0005 0.0014 0.0001 0.0011 0.0056 0.0057

ECVTNF cells, COS-7 and H9C2 cells ectopically expressing TNF, TNFR1 and TNFR2 were left untreated (Control) or treated for 30 min with 1% CD.
TNF, TNFR1 and TNFR2 accumulated in the media over this period of time were measured by ELISA and expressed as pg/ml. The p values
(Control vs. CD) are indicated in the table.

suspected that it should also alter the release of its substrates. The responsibility of cholesterol depletion in the CD-
Our results showed that an incubation time of 30 min with CD stimulated TNF release was confirmed by showing that α-
significantly increased the release of TNF, TNFR1 and TNFR2 cyclodextrin and cholesterol-loaded cyclodextrin, that are
ectopically expressed in several cell lines (Table 1). It is unlikely less efficient than CD to remove cholesterol, were unable to
that CD increase the CMV-driven synthesis of ectopically significantly increase TNF release (Fig. 8A). Other molecules
expressed TNF, TNFR1 and TNFR2 over such a short period of susceptible of disrupting lipid rafts also affected TNF
time. Nevertheless, to rule out the possibility of an increased release. Treatment of ECVTNF cells with the sterol binding
TNF synthesis, ECVTNF cells were preincubated for 30 min with polyene antibiotic filipin III, with sphingomyelinase that
cycloheximide before the treatment with CD. In this condition, degrades sphingomyelin and liberates cholesterol from lipid
the CD-stimulated TNF release was not modified (data not rafts or with cholesterol oxidase that oxidizes cellular
shown) indicating that it only involves post-translational cholesterol leading to lipid raft degradation, all significantly
events. The post-translational origin of this regulation was increased the release of TNF in the cell culture media (Fig.
further proven by showing that the spontaneous release of TNF 8A) without significantly altering its cell-associated amount
from membranes isolated from ECVTNF was significantly (data not shown) ruling out an effect on its production. The
increased by a treatment with CD (control = 1002 ± 31 pg/ml; effect of cholesterol withdrawal on the release of endoge-
CD = 1850 ± 200 pg/ml; p = 0.0019). nous ADAM17 substrates was tested using human umbilical
vein endothelial cells. The release of endothelial TNFR1 and
TNFR2 was increased by a factor 1.4 (control = 74.97 ±
10.86 pg/ml; CD = 108.1 ± 6.1 pg/ml; p < 0.0001) and 1.7 (con-
trol = 23.81 ± 8.5 pg/ml; CD = 41.28 ± 7.8 pg/ml; p = 0.0007),
respectively.
Finally, the role of ADAM17 in the CD-stimulated TNF
release was confirmed by the inhibitory effect of TIMP3
(Fig. 8B).

Discussion

In this work we show conclusively that the shedding activity


of ADAM17 is sequestered in the cholesterol-rich membrane
microdomains called lipid rafts.
The mature form of ADAM17 was detected in lipid rafts
prepared from cell lines of different origins and from freshly
collected mouse organs. In our study, the detergent Brij 98 was
chosen for the preparation of lipid rafts because it respects
their integrity that exists at physiological temperature [25].
However, with the more selective detergent Triton X-100, the
mature form of ADAM17 was also detected in the detergent
resistant membrane fraction. The fact that the proform of
Fig. 8 – Modulation of cholesterol homeostasis alters the ADAM17 did not follow the same profile of flotation as the
release of ADAM17 substrates. (A) ECVTNF cells were left mature form of ADAM17 is an argument in favor of the
untreated or treated for 30 min with 1% CD, α-cyclodextrin discriminative capacity of our lipid raft separation method.
(αCD), cyclodextrin loaded with cholesterol (Chol) or for 1 h However, there is no standard method to prepare and to study
with 500 ng/ml of filipin III (Fil), 0.5 U/ml of sphingomyelinase lipid rafts and it could be objected that the observed
(SM) or with 5 U/ml of cholesterol oxidase (CO). (B) ECVTNF localization of the mature form of ADAM17 in the lipid rafts
cells were treated for 30 min with 1% CD, 400 nM of TIMP3 or results from the selected method of separation. Thus, instead
TIMP3 + CD. TNF released in the cell culture media was of preparing lipid rafts by a conventional biochemical
measured by ELISA. *Significant versus control p < 0.0002. approach, we took advantage of the ability of THP-1 cells to
E XP E RI ME N TA L CE L L RE S E A RCH 3 1 2 ( 2 00 6 ) 3 9 6 9 –39 8 0 3977

generate MVs that are rich in lipid raft regions of the The lower proportion of ADAM17 substrates in lipid rafts
membrane [27]. Expectedly, we show that these MVs are compared with that in non-raft region should not preclude its
enriched in the mature and active form of ADAM17. role because this pool should be considered in a dynamic way:
The maturation of ADAM17 is not a prerequisite for its at every moment, molecules are cleaved in lipid rafts and
entry in lipid rafts, as demonstrated by experiments showing replaced by molecules coming from the non-raft region. The
that either the inhibition or the absence of furin, led to the effect of PMA argues in favor of this interpretation: it increases
appearance of the proform of ADAM17 in lipid rafts. Further- the synthesis and the shedding of TNF without altering its
more, furin was localized in lipid rafts, which is in accordance membrane repartition, suggesting a faster transit of TNF from
with data showing its interaction with caveolin-1 [31] and non-raft to the raft regions. A survey of the literature shows
suggests that the maturation of ADAM17 takes place in lipid that more than half of the identified ADAM17 substrates are
rafts. Because it was previously demonstrated that the localized in lipid rafts (Table 2). However, to our knowledge,
cleavage of ADAM17 prodomain by furin occurs in the medial our work originally describes the processing of ADAM17
Golgi [22,32], our data suggest that the proform of ADAM17 is substrates in lipid rafts.
integrated in lipid rafts present in the medial Golgi and is then The sequestration of the mature form of ADAM17 in
processed by furin. cholesterol-rich membrane microdomains suggests a poten-
Other secretases are integrated in lipid rafts, as it is the tial regulation of ADAM17 substrate shedding through the
case for the mature form of the beta secretase BACE [33] and modulation of cholesterol homeostasis. It was already
for all four components of the gamma secretase complex,
namely presenilin 1-derived fragments, mature nicastrin,
APH-1 and PEN-2 [34]. However, to our knowledge, this is the
Table 2 – ADAM17 substrates present in lipid rafts
first demonstration of the maturation of a secretase, herein
ADAM17, in lipid rafts. ADAM17 substrate Presence in
We previously demonstrated that the mature form of lipid rafts
ADAM17 is associated with the cytoskeleton [20]. Herein, we Amphiregulin
confirm this result because we find that a fraction of lipid rafts Amyloid-beta precursor protein [50]
containing the mature form of ADAM17 is associated with the AXL Receptor Tyrosine Kinase
cytoskeleton. The association of lipid rafts with the cytoske- CD30 [12]
CD40 [51]
leton increases their density [28], which could explain why a
CD117 (cKIT) [52]
previous report localized the mature form of ADAM17 in the Cellular prion protein PrP [53]
heaviest fractions of the sucrose gradient corresponding to the Collagen XVII [15]
supposed non-raft region of the cell membrane [12]. Two other Epiregulin
reports suggest that ADAM17 is not integrated in lipid rafts Fractalkine (CX3CL1) [54]
[15,35]. However, these works did not distinguish the mature Glycoprotein GP of Ebola virus [55]
Glycoprotein IB α [56]
form of ADAM17 from its proform.
Glycoprotein V [57]
The cleavage of ADAM17 prodomain by furin is a prere-
Growth hormone receptor [58]
quisite to its activation, although it is not the only event Heparin binding epidermal growth factor
involved because dissociation of the prodomain [36], phos- Human epidermal growth factor receptor 4 [59]
phorylation events [37,38] and association with other proteins Human Meprin
[20,39,40] were described to be likely important. However, the IL-6 receptor α [13]
presence of the mature form of ADAM17 in lipid rafts suggests Interleukin-1 receptor 2
Interleukin-15 receptor α
that its cleavage activity occurs in these membrane micro-
Low-density lipoprotein receptor [60]
domains. We studied this aspect by analyzing the lipid raft
L-selectin [61]
repartition and cleavage of three substrates of ADAM17: TNF, Macrophage colony stimulating factor receptor
TNFR1 and TNFR2. The presence of these proteins was Mucin 1 [62]
established in lipid rafts from different cell lines and mouse Nectin 4
organs. Moreover, the proportion of these proteins in lipid Neuregulins [63]
rafts was increased upon metalloproteinase inhibition, sug- Neurogenic locus notch homolog protein
P75 neurotrophin receptor [64]
gesting that the cleavage of TNF, TNFR1 and TNFR2 occurs in
Sortilin-related receptor
lipid rafts. For some substrates, ADAM10 was found to be Transforming growth factor-α
involved in the constitutive shedding whereas ADAM17 was TNF-related activation-induced cytokine
involved in the stimulated shedding [41,42]. However, this Transforming tyrosine kinase protein [65]
situation cannot be generalized to all ADAM17 substrates. Tumor necrosis factor-α
ADAM17 was implicated in both the constitutive and stimu- Tumor necrosis factor receptor I [66]
Tumor necrosis factor receptor II [67]
lated shedding of several substrates [43] and ADAM10 was not
Vascular cell adhesion molecule 1
shown to be responsible for the constitutive or stimulated
shedding of several others [44,45], in particular that of TNF The list of ADAM17 substrates is from [5] and actualized for
[46]. Our result showing that the PMA-stimulated shedding of collagen XVII [47] and nectin-4 [48]. References indicate the
TNF also occurs in lipid rafts further arguments in favor of the presence of the ADAM17 substrate in lipid rafts. The absence of
reference indicates that no data is available except for interleukin-
sequestration of ADAM17 activity in these membrane
15 receptor α that was described to be a non-raft protein [49].
microdomains.
3978 E XP E RI ME N TA L CE LL RE S E A RCH 3 1 2 ( 2 00 6 ) 3 9 6 9 –39 8 0

described that lipid raft disruption by different means Carter, W.J. Chen, W.C. Clay, J.R. Didsbury, D. Hassler, C.R.
increased the release of ADAM17 substrates like CD30 Hoffman, T.A. Kost, M.H. Lambert, M.A. Leesnitzer, P.
McCauley, G. McGeehan, J. Mitchell, M. Moyer, G. Pahel, W.
interleukin-6 receptor and collagen XVII [12,13,15]. Herein,
Rocque, L.K. Overton, F. Schoenen, T. Seaton, J.L. Su, J.
we show that lipid raft disruption also increases the release of
Warner, J.D. Becherer, Cloning of a disintegrin
TNF, TNFR1 and TNFR2. The precise mechanism at the basis of metalloproteinase that processes precursor tumour-necrosis
this effect was not studied. However, we showed that factor-alpha, Nature 385 (1997) 733–736.
disruption of lipid rafts displaced the mature form of [4] R.A. Black, C.T. Rauch, C.J. Kozlosky, J.J. Peschon, J.L. Slack,
ADAM17 in the non-raft region of the membrane that contains M.F. Wolfson, B.J. Castner, K.L. Stocking, P. Reddy, S.
the major part of ADAM17 substrates. We propose that the Srinivasan, N. Nelson, N. Boiani, K.A. Schooley, M. Gerhart,
R. Davis, J.N. Fitzner, R.S. Johnson, R.J. Paxton, C.J. March,
cleavage of ADAM17 substrates outside the lipid rafts likely
D.P. Cerretti, A metalloproteinase disintegrin that releases
accounts for a dysregulated and increased release of these tumour-necrosis factor-alpha from cells, Nature 385 (1997)
substrates induced by cholesterol depletion and it could be 729–733.
predicted that a drastic cholesterol depletion would increase [5] D.M. Smalley, K. Ley, L-selectin: mechanisms and
the release of an ADAM17 substrate as long as it is mostly physiological significance of ectodomain cleavage, J. Cell.
present in the non-raft region of the membrane. Therefore, in Mol. Med. 9 (2005) 255–266.
[6] J.J. Peschon, J.L. Slack, P. Reddy, K.L. Stocking, S.W.
normal conditions, the sequestration of the mature form of
Sunnarborg, D.C. Lee, W.E. Russell, B.J. Castner, R.S. Johnson,
ADAM17 in lipid rafts can be considered as the rate-limiting
J.N. Fitzner, R.W. Boyce, N. Nelson, C.J. Kozlosky, M.F.
process of its shedding activity. Indeed, outside of lipid rafts, Wolfson, C.T. Rauch, D.P. Cerretti, R.J. Paxton, C.J. March, R.A.
ADAM17 substrates are not accessible to the enzyme and the Black, An essential role for ectodomain shedding in
fine tuning of their processing could imply a regulation of their mammalian development, Science 282 (1998) 1281–1284.
entry in lipid rafts. In favor of a substrate-specific regulation of [7] M.J. Mohan, T. Seaton, J. Mitchell, A. Howe, K. Blackburn, W.
the entry in lipid rafts we showed that, in presence of a Burkhart, M. Moyer, I. Patel, G.M. Waitt, J.D. Becherer, M.L.
Moss, M.E. Milla, The tumor necrosis factor-alpha converting
metalloproteinase inhibitor that prevents their release, differ-
enzyme (TACE): a unique metalloproteinase with highly
ent proportions of ectopically expressed TNFR1 and TNFR2
defined substrate selectivity, Biochemistry 41 (2002)
were present in lipid rafts (Fig. 5C). Lipid rafts are thus an 9462–9469.
obligatory membrane platform that participates to the control [8] K. Althoff, J. Mullberg, D. Aasland, N. Voltz, K. Kallen, J.
of the proteolytic cleavage of ADAM17 substrates. Grotzinger, S. Rose-John, Recognition sequences and
In conclusion, we found that the maturation of ADAM17 structural elements contribute to shedding susceptibility of
occurs in lipid rafts and that its shedding activity is seques- membrane proteins, Biochem. J. 353 (2001) 663–672.
[9] K. Althoff, P. Reddy, N. Voltz, S. Rose-John, J. Mullberg,
tered in these cholesterol-rich membrane microdomains.
Shedding of interleukin-6 receptor and tumor necrosis factor
Furthermore, TNF, TNFR1 and TNFR2 are present in different alpha. Contribution of the stalk sequence to the cleavage
proportions in lipid rafts, which suggests a specific regulation pattern of transmembrane proteins, Eur. J. Biochem. 267
of their entry into these structures. However, the identification (2000) 2624–2631.
of the posttranslational modifications that could modulate the [10] F. Peiretti, M. Canault, D. Bernot, B. Bonardo, P.
entry of ADAM17 substrates in lipid rafts was out of the scope Deprez-Beauclair, I. Juhan-Vague, G. Nalbone, Proteasome
inhibition activates the transport and the ectodomain
of this present work. These findings point out a key role of lipid
shedding of TNF-{alpha} receptors in human endothelial
rafts in the regulation of ADAM17-dependent shedding events.
cells, J. Cell Sci. 118 (2005) 1061–1070.
They also offer new perspectives about the possibility of [11] E. Kojro, G. Gimpl, S. Lammich, W. Marz, F. Fahrenholz, From
modulating ADAM17 substrate shedding by altering the fatty the cover: low cholesterol stimulates the nonamyloidogenic
acid and/or cholesterol content of the membrane. pathway by its effect on the alpha-secretase ADAM 10, Proc.
Natl. Acad. Sci. U. S. A. 98 (2001) 5815–5820.
[12] B. von Tresckow, K.-J. Kallen, E.P. von Strandmann, P.
Borchmann, H. Lange, A. Engert, H.P. Hansen, Depletion of
Acknowledgments cellular cholesterol and lipid rafts increases shedding of
CD30, J. Immunol. 172 (2004) 4324–4331.
E.T. is a recipient of the Ministère de l'Enseignement Supér- [13] V. Matthews, B. Schuster, S. Schutze, I. Bussmeyer, A. Ludwig,
ieur, de la Recherche et de la Technologie. M.C. is a recipient of C. Hundhausen, T. Sadowski, P. Saftig, D. Hartmann, K.-J.
Nouvelle Société Française d'Athérosclérose (Paris). This work Kallen, S. Rose-John, Cellular cholesterol depletion triggers
shedding of the human interleukin-6 receptor by ADAM10
was supported by funds of Inserm (Paris) and Université de la
and ADAM17 (TACE), J. Biol. Chem. 278 (2003) 38829–38839.
Méditerranée (Marseilles).
[14] I. Walev, D. Tappe, E. Gulbins, S. Bhakdi, Streptolysin
O-permeabilized granulocytes shed L-selectin concomitantly
with ceramide generation via neutral sphingomyelinase,
REFERENCES J. Leukocyte Biol. 68 (2000) 865–872.
[15] E.P. Zimina, L. Bruckner-Tuderman, C.-W. Franzke,
Shedding of collagen XVII ectodomain depends on plasma
[1] P. Primakoff, D.G. Myles, The ADAM gene family: surface membrane microenvironment, J. Biol. Chem. 280 (2005)
proteins with adhesion and protease activity, Trends Genet. 34019–34024.
16 (2000) 83–87. [16] D.A. Brown, E. London, Structure and function of
[2] S. Yamamoto, Y. Higuchi, K. Yoshiyama, E. Shimizu, M. sphingolipid- and cholesterol-rich membrane rafts, J. Biol.
Kataoka, N. Hijiya, K. Matsuura, ADAM family proteins in the Chem. 275 (2000) 17221–17224.
immune system, Immunol. Today 20 (1999) 278–284. [17] L.J. Pike, Lipid rafts: heterogeneity on the high seas, Biochem.
[3] M.L. Moss, S.L. Jin, M.E. Milla, D.M. Bickett, W. Burkhart, H.L. J. 378 (2004) 281–292.
E XP E RI ME N TA L CE L L RE S E A RCH 3 1 2 ( 2 00 6 ) 3 9 6 9 –39 8 0 3979

[18] T. Golub, S. Wacha, P. Caroni, Spatial and temporal control of {gamma}-secretase with lipid rafts in post-Golgi and
signaling through lipid rafts, Curr. Opin. Neurobiol. 14 (2004) endosome membranes, J. Biol. Chem. 279 (2004)
542–550. 44945–44954.
[19] S. Gallea-Robache, V. Morand, S. Millet, J.M. Bruneau, N. [35] J.G. Kay, R.Z. Murray, J.K. Pagan, J.L. Stow, Cytokine secretion
Bhatnagar, S. Chouaib, S. Roman-Roman, A via cholesterol-rich lipid raft-associated SNAREs at the
metalloproteinase inhibitor blocks the shedding of soluble phagocytic cup, J. Biol. Chem. 281 (2006) 11949–11954.
cytokine receptors and processing of transmembrane [36] Z. Zhang, J.K. Kolls, P. Oliver, D. Good, P.O. Schwarzenberger,
cytokine precursors in human monocytic cells, Cytokine 9 M.S. Joshi, J.L. Ponthier, J.R. Lancaster, Activation of tumor
(1997) 340–346. necrosis factor-alpha-converting enzyme-mediated
[20] M. Canault, E. Tellier, B. Bonardo, E. Mas, M. Aumailley, I. ectodomain shedding by nitric oxide, J. Biol. Chem. 275 (2000)
Juhan-Vague, G. Nalbone, F. Peiretti, FHL2 interacts with both 15839–15844.
ADAM-17 and the cytoskeleton and regulates ADAM-17 [37] S.M. Soond, B. Everson, D.W.H. Riches, G. Murphy,
localization and activity, J. Cell. Physiol. 208 (2006) 363–372. ERK-mediated phosphorylation of Thr735 in
[21] B. Payrastre, P. van Bergen en Henegouwen, M. Breton, J. TNF{alpha}-converting enzyme and its potential role in TACE
den Hartigh, M. Plantavid, A. Verkleij, J. Boonstra, protein trafficking, J. Cell Sci. 118 (2005) 2371–2380.
Phosphoinositide kinase, diacylglycerol kinase, and [38] E. Diaz-Rodriguez, J.C. Montero, A. Esparis-Ogando, L. Yuste,
phospholipase C activities associated to the cytoskeleton: A. Pandiella, Extracellular signal-regulated kinase
effect of epidermal growth factor, J. Cell Biol. 115 (1991) phosphorylates tumor necrosis factor alpha-converting
121–128. enzyme at threonine 735: a potential role in regulated
[22] F. Peiretti, M. Canault, P. Deprez-Beauclair, V. Berthet, B. shedding, Mol. Biol. Cell 13 (2002) 2031–2044.
Bonardo, I. Juhan-Vague, G. Nalbone, Intracellular maturation [39] F. Peiretti, P. Deprez-Beauclair, B. Bonardo, H. Aubert, I.
and transport of tumor necrosis factor alpha converting Juhan-Vague, G. Nalbone, Identification of SAP97 as an
enzyme, Exp. Cell Res. 285 (2003) 278–285. intracellular binding partner of TACE, J. Cell Sci. 116 (2003)
[23] L. Miguet, K. Paucaud, C. Felden, B. Hugel, M.C. Martinez, J.-M. 1949–1957.
Freyssinet, R. Herbrecht, N. Potier, A. van Dorsselaer, L. [40] Y. Zheng, J. Schlondorff, C.P. Blobel, Evidence for regulation of
Mauvieux, Proteomic analysis of malignant lymphocyte the tumor necrosis factor alpha-convertase (TACE) by
membrane microparticles using double ionization coverage protein-tyrosine phosphatase PTPH1, J. Biol. Chem. (2002)
optimization, Proteomics 6 (2006) 153–171. 42463–42470.
[24] S. Schuck, M. Honsho, K. Ekroos, A. Shevchenko, K. Simons, [41] C. Hundhausen, D. Misztela, T.A. Berkhout, N. Broadway, P.
Resistance of cell membranes to different detergents, Proc. Saftig, K. Reiss, D. Hartmann, F. Fahrenholz, R. Postina, V.
Natl. Acad. Sci. U. S. A. 100 (2003) 5795–5800. Matthews, K.-J. Kallen, S. Rose-John, A. Ludwig, The
[25] P. Drevot, C. Langlet, X.-J. Guo, A.-M. Bernard, O. Colard, J.-P. disintegrin-like metalloproteinase ADAM10 is involved in
Chauvin, R. Lasserre, H.-T. He, TCR signal initiation constitutive cleavage of CX3CL1 (fractalkine) and regulates
machinery is pre-assembled and activated in a subset of CX3CL1-mediated cell–cell adhesion, Blood 102 (2003)
membrane rafts, EMBO J. 21 (2002) 1899–1908. 1186–1195.
[26] J.R. Doedens, R.A. Black, Stimulation-induced [42] B. Vincent, E. Paitel, P. Saftig, Y. Frobert, D. Hartmann, B.
down-regulation of tumor necrosis factor-alpha converting De Strooper, et al., The disintegrins ADAM10 and TACE
enzyme, J. Biol. Chem. 275 (2000) 14598–14607. contribute to the constitutive and phorbol ester-regulated
[27] I. del Conde, C.N. Shrimpton, P. Thiagarajan, J.A. Lopez, normal cleavage of the cellular prion protein, J. Biol. Chem.
Tissue-factor-bearing microvesicles arise from lipid rafts and 276 (2001) 37743–37746.
fuse with activated platelets to initiate coagulation, Blood 106 [43] U. Sahin, G. Weskamp, K. Kelly, H.-M. Zhou, S. Higashiyama, J.
(2005) 1604–1611. Peschon, et al., Distinct roles for ADAM10 and ADAM17 in
[28] S. Bodin, C. Soulet, H. Tronchere, P. Sie, C. Gachet, M. ectodomain shedding of six EGFR ligands, J. Cell Biol. 164
Plantavid, B. Payrastre, Integrin-dependent interaction of (2004) 769–779.
lipid rafts with the actin cytoskeleton in activated human [44] V. Budagian, E. Bulanova, Z. Orinska, A. Ludwig, S. Rose-John,
platelets, J. Cell Sci. 118 (2005) 759–769. P. Saftig, et al., Natural soluble interleukin-15R{alpha} is
[29] P. Reddy, J.L. Slack, R. Davis, D.P. Cerretti, C.J. Kozlosky, R.A. generated by cleavage that involves the tumor necrosis
Blanton, D. Shows, J.J. Peschon, R.A. Black, Functional factor-{alpha}-converting enzyme (TACE/ADAM17), J. Biol.
analysis of the domain structure of tumor necrosis Chem. 279 (2004) 40368–40375.
factor-alpha converting enzyme, J. Biol. Chem. 275 (2000) [45] G. Weskamp, J. Schlondorff, L. Lum, J.D. Becherer, T.-W. Kim,
14608–14614. P. Saftig, et al., Evidence for a critical role of the tumor
[30] K.J. Garton, P.J. Gough, J. Philalay, P.T. Wille, C.P. Blobel, R.H. necrosis factor {alpha} convertase (TACE) in ectodomain
Whitehead, P.J. Dempsey, E.W. Raines, Stimulated shedding shedding of the p75 neurotrophin receptor (p75NTR), J. Biol.
of vascular cell adhesion molecule 1 (VCAM-1) is mediated by Chem. 279 (2004) 4241–4249.
tumor necrosis factor-{alpha}-converting enzyme (ADAM 17), [46] Y. Zheng, P. Saftig, D. Hartmann, C. Blobel, Evaluation of the
J. Biol. Chem. 278 (2003) 37459–37464. contribution of different ADAMs to tumor necrosis factor
[31] G. Mayer, G. Boileau, M. Bendayan, The proprotein {alpha} (TNF{alpha}) shedding and of the function of the TNF
convertase furin colocalizes with caveolin-1 in the Golgi {alpha} ectodomain in ensuring selective stimulated
apparatus and endosomes of hepatocytes, Cell Tissue Res. shedding by the TNF{alpha} convertase (TACE/ADAM17),
316 (2004) 55–63. J. Biol. Chem. 279 (2004) 42898–42906.
[32] J. Schlondorff, J.D. Becherer, C.P. Blobel, Intracellular [47] C.-W. Franzke, K. Tasanen, H. Schacke, Z. Zhou, K.
maturation and localization of the tumour necrosis factor Tryggvason, C. Mauch, et al., Transmembrane collagen XVII,
alpha convertase (TACE), Biochem. J. 347 (2000) 131–138. an epithelial adhesion protein, is shed from the cell surface by
[33] D.R. Riddell, G. Christie, I. Hussain, C. Dingwall, ADAMs, EMBO J. 21 (2002) 5026–5035.
Compartmentalization of [beta]-secretase (Asp2) into [48] S. Fabre-Lafay, S. Garrido-Urbani, N. Reymond, A. Goncalves,
low-buoyant density, noncaveolar lipid rafts, Curr. Biol. 11 P. Dubreuil, M. Lopez, Nectin-4, a new serological breast
(2001) 1288–1293. cancer marker, is a substrate for tumor necrosis
[34] K.S. Vetrivel, H. Cheng, W. Lin, T. Sakurai, T. Li, N. Nukina, P.C. factor-{alpha}-converting enzyme (TACE)/ADAM-17, J. Biol.
Wong, H. Xu, G. Thinakaran, Association of Chem. 280 (2005) 19543–19550.
3980 E XP E RI ME N TA L CE LL RE S E A RCH 3 1 2 ( 2 00 6 ) 3 9 6 9 –39 8 0

[49] J. Goebel, K. Forrest, L. Morford, T.L. Roszman, Differential localization of the growth hormone receptor and its signaling
localization of IL-2- and -15 receptor chains in membrane elements: impact on growth hormone signaling, J. Biol. Chem.
rafts of human T cells, J. Leukocyte Biol. 72 (2002) 199–206. 279 (2004) 20898–20905.
[50] K.S. Vetrivel, H. Cheng, S.-H. Kim, Y. Chen, N.Y. Barnes, A.T. [59] C. Mineo, G.N. Gill, R.G.W. Anderson, Regulated migration of
Parent, et al., Spatial segregation of {gamma}-secretase and epidermal growth factor receptor from caveolae, J. Biol. Chem.
substrates in distinct membrane domains, J. Biol. Chem. 280 274 (1999) 30636–30643.
(2005) 25892–25900. [60] G.C. Ness, N. Kohlruss, K.R. Gertz, Association of the
[51] P.-O. Vidalain, O. Azocar, C. Servet-Delprat, C. low-density lipoprotein receptor with caveolae in hamster
Rabourdin-Combe, D. Gerlier, S. Manie, CD40 signaling in and rat liver, Biochem. Biophys. Res. Commun. 303 (2003)
human dendritic cells is initiated within membrane rafts, 177–181.
EMBO J. 19 (2000) 3304–3313. [61] R.G. Sitrin, D.R. Johnson, P.M. Pan, D.M. Harsh, J. Huang, H.R.
[52] T. Jahn, P. Seipel, S. Coutinho, S. Urschel, K. Schwarz, C. Petty, et al., Lipid raft compartmentalization of urokinase
Miething, et al., Analysing c-kit internalization using a receptor signaling in human neutrophils, Am. J. Respir. Cell
functional c-kit-EGFP chimera containing the fluorochrome Mol. Biol. 30 (2004) 233–241.
within the extracellular domain, Oncogene 21 (2002) [62] K. Handa, F. Jacobs, B.M. Longenecker, S.-I. Hakomori,
4508–4520. Association of MUC-1 and PSGL-1 with low-density
[53] G.S. Baron, K. Wehrly, D.W. Dorward, B. Chesebro, B. Caughey, microdomain in T-lymphocytes: a preliminary note,
Conversion of raft associated prion protein to the Biochem. Biophys. Res. Commun. 285 (2001) 788–794.
protease-resistant state requires insertion of PrP-res (PrPSc) [63] K.E. Frenzel, D.L. Falls, Neuregulin-1 proteins in rat brain and
into contiguous membranes, EMBO J. 21 (2002) 1031–1040. transfected cells are localized to lipid rafts, J. Neurochem. 77
[54] A. Muehlhoefer, L.J. Saubermann, X. Gu, K. (2001) 1–12.
Luedtke-Heckenkamp, R. Xavier, R.S. Blumberg, et al., [64] T.R. Bilderback, R.J. Grigsby, R.T. Dobrowsky, Association of
Fractalkine is an epithelial and endothelial cell-derived p75NTR with caveolin and localization of
chemoattractant for intraepithelial lymphocytes in the small Neurotrophin-induced sphingomyelin hydrolysis to
intestinal mucosa, J. Immunol. 164 (2000) 3368–3376. caveolae, J. Biol. Chem. 272 (1997) 10922–10927.
[55] S. Bavari, C.M. Bosio, E. Wiegand, G. Ruthel, A.B. Will, T.W. [65] M. Nishio, S. Fukumoto, K. Furukawa, A. Ichimura, H.
Geisbert, et al., Lipid raft microdomains: a gateway for Miyazaki, S. Kusunoki, T. Urano, K. Furukawa, Overexpressed
compartmentalized trafficking of Ebola and Marburg viruses, GM1 suppresses Nerve Growth Factor (NGF) signals by
J. Exp. Med. 195 (2002) 593–602. modulating the intracellular localization of NGF receptors
[56] D.J. Dorahy, L.F. Lincz, C.J. Meldrum, G.F. Burns, Biochemical and membrane fluidity in PC12 cells, J. Biol. Chem. 279 (2004)
isolation of a membrane microdomain from resting platelets 33368–33378.
highly enriched in the plasma membrane glycoprotein CD36, [66] V. Cottin, A. Van Linden, D.W.H. Riches, Phosphorylation of
Biochem. J. 319 (1996) 67–72. tumor necrosis factor receptor CD120a (p55) by p42mapk/erk2
[57] C.N. Shrimpton, G. Borthakur, S. Larrucea, M.A. Cruz, J.-F. induces changes in its subcellular localization, J. Biol. Chem.
Dong, J.A. Lopez, Localization of the adhesion receptor 274 (1999) 32975–32987.
glycoprotein Ib-IX-V complex to lipid rafts is required for [67] X. Feng, M.L. Gaeta, L.A. Madge, J.-H. Yang, J.R. Bradley, J.S.
platelet adhesion and activation, J. Exp. Med. 196 (2002) Pober, Caveolin-1 associates with TRAF2 to form a complex
1057–1066. that is recruited to tumor necrosis factor receptors, J. Biol.
[58] N. Yang, Y. Huang, J. Jiang, S.J. Frank, Caveolar and lipid raft Chem. 276 (2001) 8341–8349.

You might also like