ArticlePDF Available

The Relative Influence of Metal Ion Binding Sites in the I-like Domain and the Interface with the Hybrid Domain on Rolling and Firm Adhesion by Integrin 4 7

Authors:
  • Shanghai Institute of Biochemistry and Cell Biology

Abstract and Figures

We examined the effect of conformational change at the beta(7) I-like/hybrid domain interface on regulating the transition between rolling and firm adhesion by integrin alpha(4)beta(7). An N-glycosylation site was introduced into the I-like/hybrid domain interface to act as a wedge and to stabilize the open conformation of this interface and hence the open conformation of the alpha(4) beta(7) headpiece. Wild-type alpha(4)beta(7) mediates rolling adhesion in Ca(2+) and Ca(2+)/Mg(2+) but firm adhesion in Mg(2+) and Mn(2+). Stabilizing the open headpiece resulted in firm adhesion in all divalent cations. The interaction between metal binding sites in the I-like domain and the interface with the hybrid domain was examined in double mutants. Changes at these two sites can either counterbalance one another or be additive, emphasizing mutuality and the importance of multiple interfaces in integrin regulation. A double mutant with counterbalancing deactivating ligand-induced metal ion binding site (LIMBS) and activating wedge mutations could still be activated by Mn(2+), confirming the importance of the adjacent to metal ion-dependent adhesion site (ADMIDAS) in integrin activation by Mn(2+). Overall, the results demonstrate the importance of headpiece allostery in the conversion of rolling to firm adhesion.
Content may be subject to copyright.
The Relative Influence of Metal Ion Binding Sites in the
I-like Domain and the Interface with the Hybrid Domain on
Rolling and Firm Adhesion by Integrin
4
7
*
Received for publication, July 9, 2004, and in revised form, September 9, 2004
Published, JBC Papers in Press, September 24, 2004, DOI 10.1074/jbc.M407773200
JianFeng Chen‡, Junichi Takagi§, Can Xie‡, Tsan Xiao‡, Bing-Hao Luo‡,
and Timothy A. Springer‡
From The CBR Institute for Biomedical Research and Department of Pathology, Harvard Medical School,
Boston, Massachusetts 02115 and §Institute for Protein Research, Laboratory of Protein Synthesis and
Expression, Osaka University, 3-2 Yamadaoka, Suita, Osaka 565-0871, Japan
We examined the effect of conformational change at
the
7
I-like/hybrid domain interface on regulating the
transition between rolling and firm adhesion by inte-
grin
4
7
.AnN-glycosylation site was introduced into
the I-like/hybrid domain interface to act as a wedge
and to stabilize the open conformation of this interface
and hence the open conformation of the
4
7
head-
piece. Wild-type
4
7
mediates rolling adhesion in Ca
2
and Ca
2
/Mg
2
but firm adhesion in Mg
2
and Mn
2
.
Stabilizing the open headpiece resulted in firm adhe-
sion in all divalent cations. The interaction between
metal binding sites in the I-like domain and the inter-
face with the hybrid domain was examined in double
mutants. Changes at these two sites can either coun-
terbalance one another or be additive, emphasizing
mutuality and the importance of multiple interfaces in
integrin regulation. A double mutant with counterbal-
ancing deactivating ligand-induced metal ion binding
site (LIMBS) and activating wedge mutations could
still be activated by Mn
2
, confirming the importance
of the adjacent to metal ion-dependent adhesion site
(ADMIDAS) in integrin activation by Mn
2
. Overall,
the results demonstrate the importance of headpiece
allostery in the conversion of rolling to firm adhesion.
Integrins are a family of heterodimeric adhesion molecules
with noncovalently associated
and
subunits that mediate
cell-cell, cell-matrix, and cell-pathogen interactions and that
signal bidirectionally across the plasma membrane (1, 2). The
affinity of integrin extracellular domains is dynamically regu-
lated by “inside-out” signals from the cytoplasm. Furthermore,
ligand binding can induce “outside-in” signaling and activate
many intracellular signaling pathways (3–6). Integrin extra-
cellular domains exist in at least three distinct global confor-
mational states that differ in affinity for ligand (5, 7); the
equilibrium among these different states is regulated by the
binding of integrin cytoplasmic domains to cytoskeletal compo-
nents and signaling molecules (4, 6).
Integrin affinity regulation is accompanied by a series of
conformational rearrangements. Electron micrographic studies
of integrins
V
3
and
5
1
demonstrate that ligand binding, in
the absence of restraining crystal lattice contacts, induces a
switchblade-like extension of the extracellular domain and a
change in angle between the I-like and hybrid domains (5, 7).
Recent crystal structures of integrin
IIb
3
in the open, high
affinity conformation demonstrate that the C-terminal
7-helix
of the
I-like domain moves axially toward the hybrid domain,
causing the
hybrid domain to swing outward by 60°, away
from the
subunit (8). This conversion from the closed to the
open conformation of the ligand-binding domains in the inte-
grin headpiece also destabilizes the bent conformation and
induces integrin extension in which the headpiece extends and
breaks free from an interface with the leg domains that connect
it to the plasma membrane. To stabilize the outward swing of
the hybrid domain and the high affinity open headpiece con-
formation, glycan wedges have been introduced into the inter-
face between the hybrid and I-like domains of
3
and
1
inte-
grins (9). The relation between hybrid domain swing-out and
high integrin affinity has also been strongly supported by the
study of an allosteric inhibitory
1
integrin antibody SG19,
which binds to the outer side of the I-like hybrid domain inter-
face and prevents hybrid domain swing-out as shown by elec-
tron micrographic image averages (10). Allosteric inhibition by
this mAb
1
was confirmed because it did not inhibit ligand
binding to the low affinity state but rather inhibited conversion
to the high affinity state. Binding of SG19 mAb to the
1 wedge
mutant was dramatically decreased compared with wild-type,
further supporting induction of hybrid domain swing-out by the
wedge mutant. Conversely, allosteric activating mAbs have
been shown to map to the face of the
hybrid domain that is
closely opposed to the
subunit in the closed conformation and
therefore appear to induce the high affinity state by favoring
hybrid domain swing-out (11). Disulfide cross-links in the
6–
7 loop (12) and shortening of the
7-helix in the I-like
domain (13) also support the conclusion that downward dis-
placement of the
7-helix induces high affinity for ligand. A
homologous
7-helix displacement in integrin
subunit I do-
mains similarly induces high affinity for ligand (14).
It has long been known that integrin affinity for ligand is
strongly influenced by metal ions, and recently the basis for
this regulation has been deduced for the integrin
4
7
(15). The
integrin
4
7
binds the cell surface ligand mucosal cell adhe-
sion molecule-1 (MAdCAM-1) and mediates rolling adhesion by
* This work was supported by National Institutes of Health Grant
HL48675. The costs of publication of this article were defrayed in part
by the payment of page charges. This article must therefore be hereby
marked advertisement in accordance with 18 U.S.C. Section 1734
solely to indicate this fact.
To whom correspondence should be addressed: Harvard Medical
School, 200 Longwood Ave., Boston MA, 02115. Tel.: 617-278-3225; Fax:
617-278-3232; E-mail: springeroffice@cbr.med.harvard.edu.
1
The abbreviations used are: mAb, monoclonal antibody; MIDAS,
metal ion-dependent adhesion site; LIMBS, ligand-induced metal bind-
ing site; ADMIDAS, adjacent to MIDAS; LMA, LIMBS, MIDAS, and
ADMIDAS; MAdCAM-1, mucosal cell adhesion molecule-1.
THE JOURNAL OF BIOLOGICAL CHEMISTRY Vol. 279, No. 53, Issue of December 31, pp. 55556–55561, 2004
© 2004 by The American Society for Biochemistry and Molecular Biology, Inc. Printed in U.S.A.
This paper is available on line at http://www.jbc.org55556
at National Institutes of Health Library on March 18, 2008 www.jbc.orgDownloaded from
lymphocytes in postcapillary venules in mucosal tissues and
the subsequent firm adhesion in endothelium and trans-endo-
thelial migration. These key steps in lymphocyte trafficking in
vivo can be mimicked in vitro by introducing
4
7
transfected
cells into parallel wall flow chambers with MAdCAM-1 coated
on the lower wall. In Ca
2
and Ca
2
/Mg
2
,
4
7
mediates
rolling adhesion, whereas in Mg
2
,Mn
2
, or when
4
7
is
activated from within the cell,
4
7
mediates firm adhesion
(15–17). Unliganded-closed and liganded-closed structures of
V
3
have revealed a linear array of three divalent cation
binding sites in the I-like domain (18, 19). The metal coordi-
nating residues in
3
are 100% identical to those in
7
. Muta-
tion of these residues in
7
, and studies of synergy between
Ca
2
and Mg
2
and competition between Ca
2
and Mn
2
,
revealed the following (15). 1) The middle of the three linearly
arrayed sites, the metal ion-dependent adhesion site (MIDAS),
is absolutely required for rolling and firm adhesion, and it can
bind to MAdCAM-1 (and presumably coordinate) either
through Ca
2
,Mg
2
,orMn
2
. 2) The adjacent to MIDAS
(ADMIDAS) metal ion binding site functions as a negative
regulatory site that stabilizes rolling adhesion. Its mutation
results in firm adhesion in Ca
2
,Mg
2
,orMn
2
. Furthermore,
Ca
2
exerts negative regulation at high concentrations at this
site by favoring the closed I-like domain conformation, and
Mn
2
activates integrins by competing with Ca
2
at this site
and favoring an alternative coordination geometry seen in the
open I-like domain conformation. 3) The ligand-induced metal
binding site (LIMBS) functions as a positive regulatory site
that favors firm adhesion. Its mutation results in rolling adhe-
sion in Ca
2
,Mg
2
,orMn
2
. Furthermore, synergism between
low concentrations of Ca
2
and Mg
2
results from their binding
to the LIMBS and MIDAS, respectively.
Despite these advances in understanding the mechanism by
which metal ions stabilize alternative conformations of integrin
I-like domains, several issues remain unresolved. How do the
closed and open conformations of the
4
7
headpiece affect
rolling and firm adhesion? Does metal ion occupancy at the
LIMBS and ADMIDAS or outward swing of the hybrid domain
have the strongest effect on I-like domain conformation? If
changes occur at both metal binding sites and the I-like/hybrid
domain interface, does one dominate the other, or can they be
counterbalancing or additive? Here we address these questions
and the importance of allostery at the I-like/hybrid domain
interface by introducing a glycan wedge mutation into the
7
subunit to stabilize the open conformation of this interface.
MATERIALS AND METHODS
Monoclonal Antibodies—The human integrin
4
7
-specific mono-
clonal antibody Act-1 was described previously (20, 21).
cDNA Construction, Transient Transfection, and Immunoprecipita-
tion—The
7
site-directed mutations were generated by using
QuikChange (Stratagene). Wild-type human
7
cDNA (22) in vector
pcDNA3.1/Hygro() (Invitrogen) was used as the template. All muta-
tions were confirmed by DNA sequencing. Transient transfection of
293T cells using calcium phosphate precipitation was as described (23).
Transfected 293T cells were metabolically labeled with [
35
S]cysteine
and -methionine, and labeled cell lysates were immunoprecipitated
with 1
l of Act-1 mAb ascites and 20
l of protein G agarose, eluted
with 0.5% SDS, and subjected to non-reducing 7% SDS-PAGE and
fluorography (24). The selected protein bands were quantified using a
Storm PhosphorImager after3hofexposure to storage phosphor
screens (Amersham Biosciences).
Immunofluorescence Flow Cytometry—Immunofluorescence flow cy-
tometry was as described (23) using 10
g/ml purified antibody.
Flow Chamber Assay—A polystyrene Petri dish was coated with a
5-mm diameter, 20-
l spot of 5
g/ml purified h-MAdCAM-1/Fc in
coating buffer (phosphate-buffered saline, 10 mMNaHCO
3
, pH 9.0) for
1 h at 37 °C followed by 2% human serum albumin in coating buffer for
1 h at 37 °C to block nonspecific binding sites (16). The dish was
assembled as the lower wall of a parallel plate flow chamber and
mounted on the stage of an inverted phase-contrast microscope (25).
293T cell transfectants were washed twice with Ca
2
- and Mg
2
-free
Hanks’ balanced salt solution, 10 mMHepes, pH 7.4, 5 mMEDTA, 0.5%
bovine serum albumin and resuspended at 5 10
6
/ml in buffer A (Ca
2
-
FIG.1. Design of the N-glycosylation site (wedge) mutation
and confirmation by immunoprecipitation. A, sequence alignment
of the relevant portion of the human integrin
3
and
7
I-like domains.
The N-glycosylation sites in
3
and
7
wedge mutants are underlined,
and the residues mutated to Thr are shown in red. Helices and
-strands are labeled and overlined.Band C, the
3
I-like/hybrid
domain interface. The interfaces are shown in the closed (B) (19) and
open (8) (C) conformations, with the I-like and hybrid domains shown as
cyan and yellow ribbons, respectively. The structures are shown in the
same orientation after superposition using allosterically invariant por-
tions of the I-like domain (8). The side chain of the Asn that is N-
glycosylated in
3
and
7
wedge mutants is shown. D, lysates from
35
S-labeled 293T cell transfectants were immunoprecipitated with
Act-1 mAb. Precipitated wild-type (WT) and glycan wedge mutant
(Q324T) materials were subjected to non-reducing 7.5% SDS-PAGE and
fluorography. Positions of molecular mass markers are shown on the
left, and the integrin bands are indicated on the right.
TABLE I
Expression of
4
7
mutants
Integrin
4
7
cell surface expression in 293T transient transfectants
was determined with Act-1 mAb and immunofluorescence flow cytom-
etry. The data are mean specific fluorescence intensity as percent of
wild type (WT) difference from the mean for two independent exper-
iments.
Name Mutation Expression
%WT
Wild type 100 12
Glycan wedge Q324T 47 3
LIMBS D237A 99 20
Wedge/LIMBS Q324T/D237A 38 10
ADMIDAS D147A 70 9
Wedge/ADMIDAS Q324T/D147A 35 8
Conversion of Rolling to Firm Adhesion 55557
at National Institutes of Health Library on March 18, 2008 www.jbc.orgDownloaded from
and Mg
2
-free Hanks’ balanced salt solution, 10 mMHepes, and 0.5%
bovine serum albumin) and kept at room temperature. Cells were
diluted to 1 10
6
/ml in buffer A containing different divalent cations
immediately before infusion in the flow chamber using a syringe pump.
Cells were allowed to accumulate for 30 s at 0.3 dyne cm
2
. Then,
shear stress was increased every 10 s from 1 up to 32 dynes cm
2
in
2-fold increments. The number of cells remaining bound at the end of
each 10-s interval was determined. Rolling velocity at each shear stress
was calculated from the average distance traveled by rolling cells in 3 s.
To avoid confusing rolling with small amounts of movement due to
tether stretching or measurement error, a velocity of 2
m/s, which
corresponds to a movement of 1/2 cell diameter during the 3-s meas-
urement interval, was the minimum velocity required to define a cell as
rolling instead of firmly adherent (26). Microscopic images were re-
corded on Hi8 videotape for later analysis.
Surface Calculations—Accessible surfaces were calculated with
probes of the indicated radii using the buried surface routine of Crys-
tallography and NMR System software (27).
RESULTS
Activation of
4
7
with a Glycan Wedge Mutation—The mu-
tation Gln-324 3Thr in
7
introduced an N-glycosylation site
at Asn-322 in the
4–
5 loop of the I-like domain (Fig. 1A), the
same position as used previously for the wedge mutant in the
highly homologous
3
subunit (9). Crystal structures have been
defined for the
3
I-like/hybrid domain interface in both closed
low affinity and open high affinity conformations (5, 8, 19). In
the
3
subunit, the identical Asn residue has 5-fold more sol-
vent-accessible surface area as determined with a 1.4-Å probe
radius to simulate a water molecule in the open conformation
(Fig. 1C) than in the closed conformation (Fig. 1B)ofthe
hybrid/I-like interface. To approximate the size of the first four
carbohydrate residues of an N-linked glycan, we used a 10-Å
probe radius (see “Materials and Methods”), and we found that
the Asn side chain was accessible in the open but not the closed
conformation, as would be expected from visual inspection of
the interface (Fig. 1, Band C). Similarly to other wedge mu-
tants (9), the
4
7
wedge mutant was expressed somewhat less
well than wild type in 293T transfectants (Table I). Immuno-
precipitation and SDS-PAGE of [
35
S]cysteine- and -methio-
nine-labeled
4
7
showed a 3,000 M
r
increase for the Q324T
mutant
7
subunit compared with wild type, confirming N-
glycosylation of the introduced site (Fig. 1D).
During maturation and processing of
4
1
and
4
7
, a por-
tion of the intact
4
subunit, which migrates at 150,000 and
180,000 M
r
, is cleaved to fragments of 80,000 and 70,000 M
r
(28–31). Cleavage occurs after a dibasic Lys-Arg sequence in
the
4
thigh domain (29, 31). Activation of T lymphocytes
increases cleavage of the
4
subunit (29, 32, 33). Interestingly,
addition of the glycan wedge markedly increased
4
subunit
cleavage from 51% (wild type) to 90% (Q324T mutant) (Fig. 1C
and Table II). This finding directly demonstrates that
4
inte-
grin activation (see below) enhances proteolytic processing of
the
4
subunit. This could result either from greater exposure
of the cleavage site in open, extended
4
7
or longer residence
in the post-endoplasmic reticulum compartments where proc-
essing occurs (34).
The adhesive behavior in shear flow of 293T
4
7
cell trans-
fectants was characterized by allowing them to adhere to
MAdCAM-1 in a parallel wall flow chamber, incrementally in-
creasing the wall shear stress, and determining the velocity of
the adherent cells. In 1 mMCa
2
, 293T transient transfectants
expressing wild-type
4
7
rolled with increasing velocity as shear
stress was increased (Fig. 2). By contrast, wild-type transfectants
were firmly adherent in 1 mMMg
2
(Fig. 2). In Mn
2
, adhesive-
FIG.2.Adhesion in shear flow of wild-type and glycan wedge mutant
4
7
cell transfectants on MAdCAM-1 substrates. Cells were
infused into the flow chamber in buffer containing 1 mMCa
2
,1mMCa
2
1mMMg
2
,1mMMg
2
, or 0.5 mMMn
2
. Cells transfected with
4
cDNA alone (Mock)or
4
7
transfectants treated with 5 mMEDTA did not accumulate on MAdCAM-1 substrates. Rolling velocities of individual
cells were measured at a series of increasing wall shear stresses, and cells within a given velocity range were enumerated to give the population
distribution. dyn, dynes.
TABLE II
Effect of the glycan wedge in
4
7
on cleavage of
4
subunit
293T cells were transiently transfected with wild-type (WT) or mu-
tant integrin
4
7
using calcium phosphate precipitation and metabol-
ically labeled with [
35
S]cysteine and -methionine as in Fig. 1D. Labeled
cell lysates were immunoprecipitated with Act-1 antibody and sub-
jected to non-reducing 7% SDS-PAGE and fluorography. The selected
protein bands were quantified using a Storm PhosphorImager after 3 h
of exposure to storage phosphor screens. The percent of radioactivity in
each
4
subunit band was calculated, and cleavage was calculated as
percent of (
4/80
4/70
)/(
4/180
4/150
4/80
4/70
).
Radioactivity in each
4
subunit band Cleavage
4/180
4/150
4/80
4/70
%
WT 10 39 25 26 51
Q324T 2 8 41 49 90
Conversion of Rolling to Firm Adhesion55558
at National Institutes of Health Library on March 18, 2008 www.jbc.orgDownloaded from
ness was more activated than in Mg
2
because more cells accu-
mulated and fewer cells detached at the highest wall shear stress
of 32 dynes/cm
2
. By contrast with wild type, the
4
7
Q324T
glycan wedge mutant mediated firm adhesion regardless of the
divalent cation present (Fig. 2). Furthermore, the accumulation
efficiency and shear resistance of the wedge mutant was identical
in Ca
2
,Ca
2
/Mg
2
,Mg
2
, and Mn
2
and similar to that of the
wild-type
4
7
293T transfectants in Mn
2
. Thus, integrin
4
7
was constitutively activated by the glycan wedge introduced into
the hybrid/I-like domain interface.
Mutation of the
4
cleavage site residue Arg-558 abolishes
4
subunit cleavage and has no effect on
4
1
adhesion on fibronec-
tin or VCAM-1 (29, 35). We tested the effect of the same mutation
in
4
7
transfectants, and we found it to have no effect on adhe-
sion in shear flow to MAdCAM-1 (data not shown).
As described previously (15), mutation of LIMBS residues
stabilizes integrin
4
7
in the low affinity state. For example,
the LIMBS mutant D237A mediates rolling adhesion regard-
less of the divalent cations that are present (Fig. 3A). The
wedge/LIMBS double mutant (Q324T/D237A) was expressed
as well as the wedge mutant in 293T transfectants (Table I).
Compared with the LIMBS mutation, the wedge/LIMBS double
mutation reproducibly increased the number of firmly adher-
ent cells at low shear (1 and 2 dynes cm
2
)inCa
2
/Mg
2
and
Mg
2
(Fig. 3). In Mn
2
, the wedge/LIMBS mutant mediated
firm adhesion, whereas the LIMBS mutant mediated rolling
adhesion (Fig. 3). These data show that the LIMBS is required
for full activation by the wedge mutation in Ca
2
,Ca
2
/Mg
2
,
FIG.3. Interaction of glycan wedge and LIMBS mutations. A, adhesive modality and resistance to detachment in shear flow of LIMBS
(D237A) and wedge/LIMBS double mutant (Q324T/D237A)
4
7
293T transfectants on MAdCAM-1 substrates in the presence of 1 mMCa
2
,1mM
Ca
2
1mMMg
2
,1mMMg
2
, or 0.5 mMMn
2
.B, the number of rolling and firmly adherent
4
7
293T transient transfectants was measured
in the same divalent cations as in Aat a wall shear stress of 1 and 2 dynes cm
2
. Data are S.D. (n3). dyn, dynes.
Conversion of Rolling to Firm Adhesion 55559
at National Institutes of Health Library on March 18, 2008 www.jbc.orgDownloaded from
and Mg
2
(Q324T/D237A mutant in Fig. 3Acompared with
Q324T mutant in Fig. 2). Furthermore, activation by Mn
2
of
the double wedge/LIMBS Q324T/D237A mutant definitively
establishes that the LIMBS is not required for activation by
Mn
2
.
Increased Firm Adhesion by Double ADMIDAS/Wedge Mu-
tant—Mutation of the negative regulatory ADMIDAS activates
firm adhesion even in Ca
2
(15) (D147A mutant in Fig. 4
compared with wild type in Fig. 2). The double wedge/
ADMIDAS Q324T/D147A mutant was somewhat less well ex-
pressed than the ADMIDAS D147A mutant (Table I). Nonethe-
less, the double Q324T/D147A mutant showed more firmly
adherent cells in Ca
2
and Mn
2
than did the single D147A
mutant (Fig. 4) or the single Q324T mutant (Fig. 2).
DISCUSSION
Allosteric transition to the high affinity integrin headpiece
conformation is proposed to involve rearrangement of the
I-like LIMBS, MIDAS, and ADMIDAS (LMA) sites, downward
displacement of the
I-like
7-helix that connects to the hybrid
domain, and outward swing of the
hybrid domain (5, 7–9, 11,
12, 36). Outward swing of the hybrid domain has been demon-
strated by electron micrographic studies of liganded
V
3
and
5
1
integrins and crystal studies of liganded
IIb
3
but not in
an
V
3
crystal structure in which crystal lattice and head-
piece-leg interactions presumably prevented swing-out when a
ligand was soaked into crystals (8, 18). Conversely, introduc-
tion of a glycan wedge into the
1
and
3
subunits has been
demonstrated to induce high affinity for ligand by
5
1
and
IIb
3
integrins (9). Both of these integrins recognize ligands
with RGD sequences. We have extended these results here to
the
4
7
integrin, which does not recognize RGD in its ligands
and which mediates both rolling and firm adhesion. In
4
7
,
the glycan wedge converted rolling adhesion in Ca
2
and Ca
2
/
Mg
2
to firm adhesion, demonstrating that stabilizing the open
conformation at the I-like/hybrid domain interface is sufficient
to stabilize high affinity firm adhesion.
Furthermore, we examined here for the first time the inter-
play between the LMA metal binding sites at the ligand-bind-
ing interface on the “top” of the
I-like domain and the inter-
face with the hybrid domain on the opposite, “bottom” face of
the I-like domain. We asked whether one of these two inter-
faces would dominate regulation of rolling or firm adhesion, or
whether there would be mutuality in which mutations in each
of these interfaces influenced the equilibrium between rolling
and firm adhesion. The results demonstrate the latter. That is,
stabilization of rolling adhesion by LIMBS mutation was par-
tially counteracted by the wedge mutation in Ca
2
/Mg
2
and
Mg
2
and fully counteracted in Mn
2
, where firm adhesion
occurred. Conversely, stabilization of firm adhesion by the
wedge mutation was fully counteracted by the LIMBS muta-
tion in Ca
2
, where rolling occurred, and largely counteracted
in Ca
2
/Mg
2
and Mg
2
. Therefore, the equilibrium at the
LMA sites strongly influences that at the
I-like/hybrid do-
main interface and vice versa, and changes in equilibrium at
one site can counterbalance those at the other. The combined
effects of the ADMIDAS and wedge mutations also demon-
strated additive effects at the LMA sites and I-like/hybrid
interface because changes at both of these sites stabilized firm
adhesion more strongly than changes at either alone.
Another notable finding of these studies is that Mn
2
can
still activate firm adhesion when the LIMBS is mutated. Pre-
viously, the LIMBS and ADMIDAS were found to be positive
and negative regulatory sites, respectively, and positive regu-
lation by low Ca
2
concentrations was found to be intact when
the ADMIDAS was mutated (15). This, together with struc-
tural considerations, suggested that negative regulation by
high Ca
2
concentrations was effected at the ADMIDAS. Scat-
chard plots showed competitive rather than noncompetitive
inhibition by Ca
2
of stimulation by Mn
2
, suggesting that the
ADMIDAS was also the stimulatory site for Mn
2
. However, it
was not possible to confirm the role of the ADMIDAS in stim-
ulation of firm adhesion by Mn
2
because rolling adhesion
occurred in LIMBS mutants even in Mn
2
. By contrast, in the
double LIMBS/wedge mutant, the equilibrium between rolling
and firm adhesion is not far from that in wild type, and it is
regulated by divalent cations. Mn
2
was found to fully activate
FIG.4.Interaction of glycan wedge and ADMIDAS mutations. Adhesive modality and resistance to detachment in shear flow of ADMIDAS
(D147A) and double wedge/ADMIDAS (Q324T/D147A) mutant
4
7
transfectants on the MAdCAM-1 substrates in the presence of the indicated
divalent cations. The divalent cation concentrations are the same as in Fig. 2. dyn, dynes.
Conversion of Rolling to Firm Adhesion55560
at National Institutes of Health Library on March 18, 2008 www.jbc.orgDownloaded from
firm adhesion by the LIMBS/wedge mutant, showing that the
LIMBS is not required for regulation by Mn
2
and providing
strong support for the previous conclusion that the ADMIDAS
is the site for activation by Mn
2
.
Although much progress has been made recently in defining
different integrin conformational states, questions remain
about how signals are transduced from the cytoplasm to the
ligand binding site and whether intermediate conformational
states have intermediate affinity for ligand. It appears that to
mediate rolling adhesion, integrins must be in one of the ex-
tended conformations rather than in the bent conformation (15,
37). The extended conformation with the closed headpiece is an
intermediate in the conformational pathway between the bent
conformation, which contains a closed headpiece, and the ex-
tended conformation with the open headpiece (5). The current
study demonstrates that stabilization of the open headpiece by
a glycan wedge at the
I-like/hybrid interface is sufficient to
convert low affinity rolling adhesion to high affinity firm adhe-
sion. It appears that the glycan wedge converts the extended
conformation with the closed headpiece to the extended confor-
mation with the open headpiece. Therefore, this study strongly
suggests that within the extended integrin conformation, con-
version of the closed to the open headpiece is sufficient to
convert rolling adhesion to firm adhesion. In an intact integrin,
marked separation in the plane of the membrane of the trans-
membrane domains of the integrin
and
subunits would also
stabilize the open headpiece and therefore may be the mecha-
nism for converting rolling adhesion to firm adhesion.
Acknowledgment—We thank Dr. Michael J. Briskin for providing the
human MAdCAM-1/Fc.
REFERENCES
1. Hynes, R. O. (2002) Cell 110, 673–687
2. Takagi, J., and Springer, T. A. (2002) Immunol. Rev. 186, 141–163
3. Kim, M., Carman, C. V., and Springer, T. A. (2003) Science 301, 1720–1725
4. Giancotti, F. G., and Ruoslahti, E. (1999) Science 285, 1028–1032
5. Takagi, J., Petre, B. M., Walz, T., and Springer, T. A. (2002) Cell 110, 599–611
6. Vinogradova, O., Velyvis, A., Velyviene, A., Hu, B., Haas, T. A., Plow, E. F.,
and Qin, J. (2002) Cell 110, 587–597
7. Takagi, J., Strokovich, K., Springer, T. A., and Walz, T. (2003) EMBO J. 22,
4607–4615
8. Xiao, T., Takagi, J., Wang, J.-h., Coller, B. S., and Springer, T. A. (2004) 432,
59–67
9. Luo, B.-H., Springer, T. A., and Takagi, J. (2003) Proc. Natl. Acad. Sci. U. S. A.
100, 2403–2408
10. Luo, B.-H., Strokovich, K., Walz, T., Springer, T. A., and Takagi, J. (2004)
J. Biol. Chem. 279, 27466–27471
11. Mould, A. P., Barton, S. J., Askari, J. A., McEwan, P. A., Buckley, P. A., Craig,
S. E., and Humphries, M. J. (2003) J. Biol. Chem. 278, 17028–17035
12. Luo, B.-H., Takagi, J., and Springer, T. A. (2004) J. Biol. Chem. 279,
10215–10221
13. Yang, W., Shimaoka, M., Chen, J. F., and Springer, T. A. (2004) Proc. Natl.
Acad. Sci. U. S. A. 101, 2333–2338
14. Springer, T. A., and Wang, J.-h. (2004) in Cell Surface Receptors (Garcia, K. C.,
ed) pp. 29–63, Elsevier, San Diego, CA
15. Chen, J. F., Salas, A., and Springer, T. A. (2003) Nat. Struct. Biol. 10,
995–1001
16. de Chateau, M., Chen, S., Salas, A., and Springer, T. A. (2001) Biochemistry
40, 13972–13979
17. Berlin, C., Bargatze, R. F., von Andrian, U. H., Szabo, M. C., Hasslen, S. R.,
Nelson, R. D., Berg, E. L., Erlandsen, S. L., and Butcher, E. C. (1995) Cell
80, 413–422
18. Xiong, J. P., Stehle, T., Zhang, R., Joachimiak, A., Frech, M., Goodman, S. L.,
and Arnaout, M. A. (2002) Science 296, 151–155
19. Xiong, J.-P., Stehle, T., Diefenbach, B., Zhang, R., Dunker, R., Scott, D. L.,
Joachimiak, A., Goodman, S. L., and Arnaout, M. A. (2001) Science 294,
339–345
20. Lazarovits, A. I., Moscicki, R. A., Kurnick, J. T., Camerini, D., Bhan, A. K.,
Baird, L. G., Erikson, M., and Colvin, R. B. (1984) J. Immunol. 133,
1857–1862
21. Schweighoffer, T., Tanaka, Y., Tidswell, M., Erle, D. J., Horgan, K. J., Luce,
G. E., Lazarovits, A. I., Buck, D., and Shaw, S. (1993) J. Immunol. 151,
717–729
22. Tidswell, M., Pachynski, R., Wu, S. W., Qiu, S.-Q., Dunham, E., Cochran, N.,
Briskin, M. J., Kilshaw, P. J., Lazarovits, A. I., Andrew, D. P., Butcher,
E. C., Yednock, T. A., and Erle, D. J. (1997) J. Immunol. 159, 1497–1505
23. Lu, C., Oxvig, C., and Springer, T. A. (1998) J. Biol. Chem. 273, 15138–15147
24. Luo, B.-H., Springer, T. A., and Takagi, J. (2004) PLoS Biol. 2, 776–786
25. Lawrence, M. B., and Springer, T. A. (1991) Cell 65, 859–873
26. Salas, A., Shimaoka, M., Chen, S., Carman, C. V., and Springer, T. A. (2002)
J. Biol. Chem. 277, 50255–50262
27. Brunger, A. T., Adams, P. D., Clore, G. M., DeLano, W. L., Gros, P., Grosse-
Kunstleve, R. W., Jiang, J.-S., Kuszewski, J., Nilges, M., Pannu, N. S.,
Read, R. J., Rice, L. M., Simonson, T., and Warren, G. L. (1998) Acta
Crystallogr. Sect. D 54, 905–921
28. Holzmann, B., and Weissman, I. L. (1989) EMBO J. 8, 1735–1741
29. Teixido, J., Parker, C. M., Kassner, P. D., and Hemler, M. E. (1992) J. Biol.
Chem. 267, 1786–1791
30. Parker, C. M., Pujades, C., Brenner, M. B., and Hemler, M. E. (1993) J. Biol.
Chem. 268, 7028–7035
31. Hemler, M. E., Elices, M. J., Parker, C., and Takada, Y. (1990) Immunol. Rev.
114, 45–65
32. Sanchez-Madrid, F., De Landazuri, M. O., Morago, G., Cebrian, M., Acevedo,
A., and Bernabeu, C. (1986) Eur. J. Immunol. 16, 1343–1349
33. McIntyre, B. W., Evans, E. L., and Bednarczyk, J. L. (1989) J. Biol. Chem. 264,
13745–13750
34. Bednarczyk, J. L., Szabo, M. C., and McIntyre, B. W. (1992) J. Biol. Chem. 267,
25274–25281
35. Bergeron, E., Basak, A., Decroly, E., and Seidah, N. G. (2003) Biochem. J. 373,
475–484
36. Mould, A. P., Symonds, E. J., Buckley, P. A., Grossmann, J. G., McEwan, P. A.,
Barton, S. J., Askari, J. A., Craig, S. E., Bella, J., and Humphries, M. J.
(2003) J. Biol. Chem. 278, 39993–39999
37. Salas, A., Shimaoka, M., Kogan, A. N., Harwood, C., von Andrian, U. H., and
Springer, T. A. (2004) Immunity 20, 393–406
Conversion of Rolling to Firm Adhesion 55561
at National Institutes of Health Library on March 18, 2008 www.jbc.orgDownloaded from
... In each of these natural ligands the aspartic acid coordinates with a Mg ++ ion that sits in the metal ion dependent adhesion site (MIDAS) of β 7 . Mg ++ ion coordination is a strict requirement for ligand binding [23,38]. Cardozo and colleagues identified nearby amino acids, glutamine-arginine-valine (QRV) (170-172) that also influence V2-α 4 β 7 interactions, demonstrating that the binding site is not limited to the LDV/I tripeptide [12]. ...
... Rolling adhesion is associated with lower affinity binding to MAdCAM, while firm adhesion is associated with a higher affinity interaction. Manipulation of Ca ++ , Mg ++ , and Mn ++ concentrations provides a way to manipulate the affinity of α 4 β 7 -MAdCAM interactions in vitro [38]. Strength of adhesion is highest in Mn ++ , followed by Mg ++ > Mg ++ /Ca ++ , > Ca ++ . ...
Article
Full-text available
The GI tract is preferentially targeted during acute/early HIV-1 infection. Consequent damage to the gut plays a central role in HIV pathogenesis. The basis for preferential targeting of gut tissues is not well defined. Recombinant proteins and synthetic peptides derived from HIV and SIV gp120 bind directly to integrin α4β7, a gut-homing receptor. Using both cell-surface expressed α4β7 and a soluble α4β7 heterodimer we demonstrate that its specific affinity for gp120 is similar to its affinity for MAdCAM (its natural ligand). The gp120 V2 domain preferentially engages extended forms of α4β7 in a cation -sensitive manner and is inhibited by soluble MAdCAM. Thus, V2 mimics MAdCAM in the way that it binds to α4β7, providing HIV a potential mechanism to discriminate between functionally distinct subsets of lymphocytes, including those with gut-homing potential. Furthermore, α4β7 antagonists developed for the treatment of inflammatory bowel diseases, block V2 binding to α4β7. A 15-amino acid V2 -derived peptide is sufficient to mediate binding to α4β7. It includes the canonical LDV/I α4β7 binding site, a cryptic epitope that lies 7–9 amino acids amino terminal to the LDV/I, and residues K169 and I181. These two residues were identified in a sieve analysis of the RV144 vaccine trial as sites of vaccine -mediated immune pressure. HIV and SIV V2 mAbs elicited by both vaccination and infection that recognize this peptide block V2-α4β7 interactions. These mAbs recognize conformations absent from the β- barrel presented in a stabilized HIV SOSIP gp120/41 trimer. The mimicry of MAdCAM-α4β7 interactions by V2 may influence early events in HIV infection, particularly the rapid seeding of gut tissues, and supports the view that HIV replication in gut tissue is a central feature of HIV pathogenesis.
... However, ADMIDAS is a negative regulatory site that mainly mediates rolling adhesion. Notably, a high concentration of Ca 2+ exerts a negative regulatory effect at this site by blocking the I-like domain, causing Mn 2+ to compete with Ca 2+ for ADMIDAS and thus activating integrins [17,42]. Conversely, SyMBS acts as a positive regulatory site that favors stable adhesion. ...
Article
Full-text available
The interaction between integrin α4β7 and mucosal vascular addressin cell-adhesion molecule-1 (MAdCAM-1) facilitates the adhesion of circulating lymphocytes to the surface of high endothelial venules in inflammatory bowel diseases (IBDs). Lymphocyte adhesion is a multistep cascade involving the tethering, rolling, stable adhesion, crawling, and migration of cells, with integrin α4β7 being involved in rolling and stable adhesions. Targeting the integrin α4β7–MAdCAM-1 interaction may help decrease inflammation in IBDs. This interaction is regulated by force; however, the underlying mechanism remains unknown. Here, we investigate this mechanism using a parallel plate flow chamber and atomic force microscopy. The results reveal an initial increase in the lifetime of the integrin α4β7–MAdCAM-1 interaction followed by a decrease with an increasing force. This was manifested in a two-state curve regulated via a catch-bond–slip-bond conversion regardless of Ca2+ and/or Mg2+ availability. In contrast, the mean rolling velocity of cells initially decreased and then increased with the increasing force, indicating the flow-enhanced adhesion. Longer tether lifetimes of single bonds and lower rolling velocities mediated by multiple bonds were observed in the presence of Mg2+ rather than Ca2+. Similar results were obtained when examining the adhesion to substrates co-coated with chemokine CC motif ligand 25 and MAdCAM-1, as opposed to substrates coated with MAdCAM-1 alone. In conclusion, the integrin α4β7–MAdCAM-1 interaction occurs via ion- and cytokine-dependent flow-enhanced adhesion processes and is regulated via a catch-bond mechanism.
... Integrin α 4 β 7 mediates rolling and firm adhesion of lymphocytes when inactive and activated, respectively [22,26], controlling two critical steps in tissue-specific homing of lymphocytes [27,28]. Integrin activation can be dynamically regulated via a cluster of three metal ionbinding sites in the β I domain [26,29]. A cation-π interaction in the human β 7 I domain between F185 in the SDL and the synergistic metal ion-binding site (SyMBS) cation has been shown to be essential for the activation of α 4 β 7 [30]. ...
Article
Full-text available
Background: β7 integrins are responsible for the efficient recruitment of lymphocytes from the blood and their retention in gut-associated lymphoid tissues. Integrin α4β7 binds MAdCAM-1, mediating rolling adhesion of lymphocytes on blood vessel walls when inactive and firm adhesion when activated, thereby controlling two critical steps of lymphocyte homing to the gut. By contrast, integrin αEβ7 mediates the adhesion of lymphocytes to gut epithelial cells by interacting with E-cadherin. Integrin β7 blocking antibodies have shown efficacy in clinical management of inflammatory bowel disease (IBD); however, fully blocking β7 function leads to the depletion of colonic regulatory T (Treg) cells and exacerbates dextran sulfate sodium (DSS)-induced colitis by evoking aberrant innate immunity, implying its potential adverse effect for IBD management. Thus, a better therapeutic strategy targeting integrin β7 is required to avoid this adverse effect. Results: Herein, we inhibited integrin α4β7 activation in vivo by creating mice that carry in their integrin β7 gene a mutation (F185A) which from structural studies is known to lock α4β7 in its resting state. Lymphocytes from β7-F185A knock-in (KI) mice expressed α4β7 integrins that could not be activated by chemokines and showed significantly impaired homing to the gut. The β7-F185A mutation did not inhibit αEβ7 activation, but led to the depletion of αEβ7+ lymphocytes in the spleen and a significantly reduced population of αEβ7+ lymphocytes in the gut of KI mice. β7-F185A KI mice were resistant to T cell transfer-induced chronic colitis, but did not show an increased susceptibility to DSS-induced innate colitis, the adverse effect of fully blocking β7 function. Conclusions: Our findings demonstrate that specific inhibition of integrin α4β7 activation is a potentially better strategy than fully blocking α4β7 function for IBD treatment.
... The importance of divalent metal cations in integrin conformational, as well as affinity, regulation was demonstrated by several studies involving mutation of residues involved in cation coordination [36][37][38]. When these divalent metal cations are absent, as seen in the "metal-free/deficient" experimental conditions, this negative regulation of integrin conformation and affinity is removed, since no calcium cations are available for the coordination of the ADMIDAS site and subsequent capture of the α7 helix. ...
Article
CD23 is involved in a myriad of immune reactions. It is not only a receptor for IgE, but also functions in the regulation of IgE synthesis, isotype switching in B cells, and induction of the inflammatory response. These effector functions of CD23 arise through its interaction with another leukocyte-specific cell surface receptor – the β 2 integrin subfamily. It has been shown that CD23 is also capable of interacting with the β 3 and β 5 integrin β-subunit of integrins via a basic RKC motif in a metal cation-independent fashion. In this study the interaction was probed for whether or not the RKC motif governs the interaction between CD23 and the α X β 2 integrin as well. This was done by performing bioinformatic docking predictions between CD23 and α X β 2 integrin αI domain and SPR spectroscopy analysis of the interaction. This revealed that in the absence of cations, the RKC motif is involved in interaction with the integrin αI domain. However, in the presence of divalent metal cations the interaction showed the involvement of a novel acidic motif within the CD23 protein. This same pattern of interaction was seen in docking predictions between CD23 and the β 3 I-like domain. This study thus presents an alternative site as a possible contributor to the CD23-integrin interaction exhibiting cation-dependence.
... Previously, the rolling and firm phases of  4  7 adhesion were proposed to, respectively, correlate with the low-affinity, closed-headpiece, and high-affinity, open-headpiece states seen with other integrins (Chen et al., 2003(Chen et al., , 2004 ...
Article
Abbreviations used in this paper: ADMIDAS, adjacent to MIDAS; CDR, comple-mentarity-determining region; IgSF, immunoglobulin superfamily; MIDAS, metal ion-dependent adhesion site; SAD, single-wavelength anomalous dispersion; SDL, specificity-determining loop; SyMBS, synergistic metal ion binding site.
... Previously, the rolling and firm phases of  4  7 adhesion were proposed to, respectively, correlate with the low-affinity, closed-headpiece, and high-affinity, open-headpiece states seen with other integrins (Chen et al., 2003(Chen et al., , 2004 ...
Chapter
α4 integrins (including α4β1 and α4β7) are primarily expressed on leukocytes. By interacting with their ligands expressed on high endothelial venules (HEVs), α4 integrins mediate the recruitment of leukocytes from blood circulation to lymphoid organs and inflamed tissues, thereby playing essential roles in immune surveillance and host defense. The function of integrins is dynamically regulated by their activation and signaling. Integrin activation is associated with global conformational rearmament from a bent to extend conformation, mainly triggered by intracellular activation signals, termed “inside-out” signaling. The ligand binding to the integrin can also activate multiple intracellular pathways, which is termed “outside-in” signaling. In this chapter, we summarize the major findings regarding α4 integrins over the last decades, including their structures, extracellular ligands, intracellular adaptor proteins, and functions in homeostasis and diseases, including cancer, multiple sclerosis (MS), inflammatory bowel disease (IBD), and other autoimmune diseases.KeywordIntegrinα4β1α4β7StructureExtracellular ligandsIntracellular adaptor proteinsActivationHomeostasisDisease
Article
Integrins are main cell adhesion receptors serving as linker attaching cells to extracellular matrix (ECM) and bidirectional hubs transmitting biochemical and mechanical signals between cells and their environment. Integrin αvβ3 is a critical family member of integrins and interacts with ECM proteins containing RGD tripeptide sequence. Accumulating evidence indicated that the abnormal expression of integrin αvβ3 was associated with various tumor progressions, including tumor initiation, sustained tumor growth, distant metastasis, drug resistance development, maintenance of stemness in cancer cells. Therefore, αvβ3 has been explored as a therapeutic target in various types of cancers, but there is no αvβ3 antagonist approved for human therapy. Targeting-integrin αvβ3 therapeutics has been a challenge, but lessons from the past are valuable to the development of innovative targeting approaches. This review systematically summarized the structure, signal transduction, regulatory role in cancer, and drug development history of integrin αvβ3, and also provided new insights into αvβ3-based therapeutics in cancer from bench to clinical trials, which would contribute to developing effective targeting αvβ3 agents for cancer treatment.
Article
In vivo imaging is creating great opportunities for disease diagnosis as a research tool. Probes are usually used to observe physiological structures in vivo clearly. Recent progresses of nanoprobes are important for the generation of high resolution and high contrast images required by accurate and precision disease diagnosis. In vivo self-assembled peptide(SAP) nanoprobes are playing major roles in in vivo imaging by modularity of design, high imaging contrast, response to the location of the lesion, and long-time retention in the lesion. And the response to lesion and long-term retention in there can enhance imaging sensitivity and specificity of in vivo SAP nanoprobes. Therefore, in vivo SAP nanoprobes are simple ancillary contrast entities to optimize the imaging effect. In this review, the recent progress of in vivo SAP nanoprobes for in vivo imaging, from molecular design of peptides to biomedical and clinical applications including disease diagnosis and disease-related molecular imaging is systematically summarized. We evaluate their ability, including sensitivity and specificity to provide relevant information under preoperative and during surgery circumstances and critically their likelihood to be clinically translated. Finally, a brief outlook on remaining challenges and potential directions for future research in this area is presented.
Chapter
The understanding, at the molecular level, of the interactions between innate and adaptive arms of the immune system is currently a hot topic, particularly to those interested in immunology - especially susceptibility to infectious diseases. This book provides a survey of topics, in the area of innate and adaptive immunity, which have been researched within the MRC Immunochemistry Unit, at Oxford University, over a period of forty years. The topics include: " antibody structure - for which the first Director of the Immunochemistry Unit, Professor RR Porter, was awarded a Nobel prize in 1972 " the characterization of membrane proteins on lymphoid cells - leading to the concept of these molecules belonging to an immunoglobulin super family " the proteins of the human serum complement system - one of the body's major defences against microbial infection " the human cell -surface integrins and the hyaluronan- binding proteins, which are involved in regulation of inflammation at cell surfaces and within the extracellular matrix " the family of collectin molecules - containing distinct globular carbohydrate -binding domains linked to collagen-like regions - which play important roles in innate immunity in the lungs and bloodstream by immediate recognition and clearance of microbial pathogens Each chapter in the book gives a brief historical background to a topic and then provides a survey of recent advances in the field and are written by internationally recognised renowned experts. The theme running through the chapters is that of protein structure-function relationships - including, amongst others, descriptions of quaternary structures of large oligomeric proteins, of Factor H and C1q binding to specific ligands, and of the chemistry of the mechanism of catalysis of covalent binding of activated C3 and C4 proteins to nucleophilic groups on microbial surfaces. In several chapters excellent descriptions are given with respect to how the immune system can be recruited to combat microbial infection - via proteins of both the innate and adaptive immune systems. The book also includes notable chapters which are excellent examples of the importance of how the isolation, characterisation, protein engineering and crystallisation has resulted in a full understanding of complex protein-protein interactions involved in the recognition and triggering events of important sections of the immune system: -Structure and Function of the C1 Complex - GÚrard J. Arlaud -Chemical Engineering of Therapeutic Antibodies - George T Stevenson -Leukocyte surface proteins - purification and characterisation - A. Neil Barclay -Cell Surface Integrins - Suet-Mien Tan and S.K. Alex Law This book is aimed primarily at established senior research scientists, postdoctoral research scientists and PhD students who have an interest in proteins of the immune system. However, the wide range of immunity system topics, while staying broadly within innate/adaptive immunity will also appeal to a wider audience.
Article
Full-text available
The leukocyte integrin alpha 4 beta 1 (VLA-4, CD49d/CD29) is a receptor for the extracellular matrix protein fibronectin and the endothelial adhesion protein VCAM-1. We have analyzed the biosynthesis and post-translational modifications of the two subunits of this receptor complex. The alpha 4 subunit was initially synthesized as a single-chain polypeptide that underwent the formation of complex endoglycosidase H-resistant oligosaccharide side chains and which could be proteolytically cleaved into two noncovalently associated fragments. The level and rate of alpha 4 subunit cleavage was dependent on the cell studied. The T cell tumor line HPB-ALL expressed both intact and fragmented alpha 4 on the cell surface. The interleukin-2-dependent natural killer line NK 3.3 and long term interleukin-2-dependent activated T lymphocytes cleaved the alpha 4 polypeptide earlier and more efficiently than did HPB-ALL cells and did not have detectable levels of intact alpha 4 on the cell surface. The proteolysis of alpha 4 was blocked by treating cells with either the lysosomotrophic amine NH4Cl or the carboxylic ionophore monensin. The presence of complex N-linked oligosaccharides did not seem to be necessary for alpha 4 cleavage or for binding of the alpha 4 beta 1 complex to a synthetic peptide corresponding to the binding site for this receptor on fibronectin.
Article
Full-text available
The cell surface heterodimer VLA-4 (alpha 4 beta 1), a member of the integrin family of adhesion receptors, is involved in both cell-extracellular matrix and cell-cell adhesion. Unlike any other integrin alpha subunit, the intact (150 kDa) alpha 4 subunit of VLA-4 can sometimes be cleaved into two noncovalently associated fragments (80 and 70 kDa). Using biosynthetic and mixing experiments, we found that human alpha 4 cleavage is a regulated, compartmentalized event, occurring soon after maturation of the beta 1-associated alpha 4 subunit. Cleavage of alpha 4, which is increased following T cell activation, has been suggested to correlate with altered VLA-4 functions. To address directly the functional importance of alpha 4 cleavage, we have studied VLA-4-mediated adhesion functions in cells expressing intact alpha 4 in comparison with cells expressing cleaved alpha 4. For this purpose, we first sequenced the N terminus of the endogenously produced 70-kDa alpha 4 fragment and identified the alpha 4 cleavage site between Lys557-Arg558 and Ser559. To abolish cleavage, we converted Arg558 to Leu or Lys557 to Gln by site-directed mutagenesis of the alpha 4 cDNA and then transfected both mutant and wild type alpha 4 cDNAs into VLA-4-negative K562 cells. Whereas transfection with wild type alpha 4 cDNA yielded predominantly cleaved alpha 4 subunit, the Leu558-alpha 4 yielded only intact alpha 4 subunit, and Gln557-alpha 4 yielded mostly intact alpha 4 subunit. Transfectants with the intact or the cleaved alpha 4 were equally capable of engaging in VLA-4-dependent adhesion to vascular cell adhesion molecule-1 and to the Hep II fragment of fibronectin (40 kDa) and aggregated equally well in response to anti-alpha 4 antibodies. Thus, cleavage of the alpha 4 subunit in these transfectants did not alter any of the known VLA-4-mediated adhesion functions.
Article
The leukocyte integrin alpha4beta1 (VLA-4, CD49d/CD29) is a receptor for the extracellular matrix protein fibronectin and the endothelial adhesion protein VCAM-1. We have analyzed the biosynthesis and post-translational modifications of the two subunits of this receptor complex. The alpha4 subunit was initially synthesized as a single-chain polypeptide that underwent the formation of complex endoglycosidase H-resistant oligosaccharide side chains and which could be proteolytically cleaved into two noncovalently associated fragments. The level and rate of alpha4 subunit cleavage was dependent on the cell studied. The T cell tumor line HPB-ALL expressed both intact and fragmented alpha4 on the cell surface. The interleukin-2-dependent natural killer line NK 3.3 and long term interleukin-2-dependent activated T lymphocytes cleaved the alpha4 polypeptide earlier and more efficiently than did HPB-ALL cells and did not have detectable levels of intact alpha4 on the cell surface. The proteolysis of alpha4 was blocked by treating cells with either the lysosomotrophic amine NH4Cl or the carboxylic ionophore monensin. The presence of complex N-linked oligosaccharides did not seem to be necessary for alpha4 cleavage or for binding of the alpha4beta1 complex to a synthetic peptide corresponding to the binding site for this receptor on fibronectin.
Article
Activation of the ligand binding function of integrin heterodimers requires transmission of an “inside-out” signal from their small intracellular segments to their large extracellular domains. The structure of the cytoplasmic domain of a prototypic integrin αIIbβ3 has been solved by NMR and reveals multiple hydrophobic and electrostatic contacts within the membrane-proximal helices of its α and the β cytoplasmic tails. The interface interactions are disrupted by point mutations or the cytoskeletal protein talin that are known to activate the receptor. These results provide a structural mechanism by which a handshake between the α and the β cytoplasmic tails restrains the integrin in a resting state and unclasping of this interaction triggers the inside-out conformational signal that leads to receptor activation.
Article
Integrins contain two structurally homologous but distantly related domains: an I-like domain that is present in all -subunits and an I domain that is present in some -subunits. Atomic resolution and mutagenesis studies of I domains demonstrate a C-terminal, axial displacement of the 7-helix that allosterically regulates the shape and affinity of the ligand-binding site. Atomic resolution studies of I-like domains have thus far demonstrated no similar 7-helix displacement; however, other studies are consistent with the idea that I and I-like domains undergo structurally analogous rearrangements. To test the hypothesis that C-terminal, axial displacement of the 7-helix, coupled with 6-7 loop reshaping, activates I-like domains, we have mimicked the effect of 7-helix displacement on the 6-7 loop by shortening the 7-helix by two independent, four-residue deletions of about one turn of -helix. In the case of integrin alphaLbeta2, each mutant exhibits constitutively high affinity for the physiological ligand intercellular adhesion molecule 1 and full exposure of a beta I-like domain activation-dependent antibody epitope. In the case of analogous mutants in integrin alpha4beta7, each mutant shows the activated phenotype of firm adhesion, rather than rolling adhesion, in shear flow. The results show that integrins that contain or lack alpha I domains share a common pathway of beta I-like domain activation, and they suggest that beta I-like and alpha I domain activation involves structurally analogous alpha7-helix axial displacements.
Article
We studied interactions in shear flow of cells bearing integrins alpha4 beta1 or alpha4 beta7 with VCAM-1 and MAdCAM-1 substrates in different divalent cations. Interestingly, Ca2+ was essential for tethering in flow and rolling, interactions through both alpha4 integrins. Mg2+ promoted firm adhesion of alpha4 beta7-expressing cells on MAdCAM-l but with much lower tetherina efficiency in shear flow. The k,,ff' of 1.28 s-1 and L resistance of the receptor-ligand bond to force (estimated as a bond interaction distance or Gr) for transient tethers on MAdCAM-1 were similar to values for E- and P-selectins. By contrast to results in Ca2+ or Ca2+ + Mg2+ in Mg2+ the alpha4 beta7-MAdCAM-1 k(off)degrees decreased 20-fold to 0.046 s(-1), and the bond was weaker, providing an explanation for the finding of firm adhesion under these conditions. Shear enhanced tethering to MAdCAM-1, thereby contributing to the stability of rolling. Comparisons to selectins demonstrate that the kinetic and mechanical properties of the alpha4 beta7 integrin are well suited to its intermediate position in adhesion cascades, in which it bridges rapid rolling through selectins to firm adhesion through beta2 integrins.
Article
Rolling of leukocytes on vascular endothelial cells, an early event in inflammation, can be reproduced in vitro on artificial lipid bilayers containing purified CD62, a selectin also named PADGEM and GMP-140 that is inducible on endothelial cells. Neutrophils roll on this selectin under flow conditions similar to those found in postcapillary venules. Adhesion of resting or activated neutrophils through the integrins LFA-1 and Mac-1 to ICAM-1 in a lipid bilayer does not occur at physiologic shear stresses; however, static incubation of activated neutrophils allows development of adhesion that is greater than 100-fold more shear resistant than found on CD62. Addition of a chemoattractant to activate LFA-1 and Mac-1 results in the arrest of neutrophils rolling on bilayers containing both CD62 and ICAM-1. Thus, at physiologic shear stress, rolling on a selectin is a prerequisite for activation-induced adhesion strengthening through integrins.
Article
The family of human T cell activation-associated proteins, named VLA complex, is formed by the molecular association of cell surface glycoproteins of 210, 165, 130 and 80 kDa. In this report, we describe eight different monoclonal antibodies (HP mAb) specific for the 80-kDa polypeptide which preferentially associates with the 165-kDa member. Comparative immunoprecipitation and cell-binding studies demonstrated that the HP mAb recognize an epitope(s) on the 165/80 kDa subset different from those recognized by other anti-VLA mAb previously described. Furthermore, cellular and tissue distribution studies by flow cytometry and peroxidase staining showed that the HP reactivity pattern is different from other VLA specificities. The 165/80-kDa complex defined by HP mAb is present on human thymocytes, peripheral blood lymphocytes as well as on T, B and myelomonocytic cell lines. However, only the 80-kDa subunit was precipitated by HP mAb from activated T lymphocytes. These results suggest that the association between the 165- and 80-kDa subunits diminishes during the activation process, and that the epitopes recognized by the HP mAb are located on the 80-kDa protein. The novel polypeptide association of 165/80 kDa has been termed VLA-3.
Article
Lymphocytes home to various lymphoid organs by adhering to and migrating through specialized high endothelial venules (HEV). The murine cell surface heterodimer LPAM-1 is involved in the homing of lymphocytes to mucosal sites (Peyer's patches). LPAM-1 has an alpha subunit (alpha 4m) analogous to the alpha chain of the human integrin molecule VLA-4. Here we show that the LPAM-1 beta subunit (beta p) is immunochemically and biochemically distinct from previously defined integrin beta subunits, suggesting that beta p represents a novel integrin beta subunit. Depending on the cellular source two alternative beta subunits, beta p and integrin beta 1, can be isolated in association with alpha 4m. Therefore, alpha 4m is the common subunit of the unique integrin LPAM-1 (alpha 4m beta p) and of the heterodimer LPAM-2 (alpha 4m beta 1), which is analogous to VLA-4. Antibody-blocking experiments suggest that, in addition to LPAM-1, LPAM-2 is also involved in the organ-specific adhesion of lymphocytes to Peyer's patch HEV.