Mutational Analysis of the Fractalkine Chemokine Domain
BASIC AMINO ACID RESIDUES DIFFERENTIALLY CONTRIBUTE TO CX3CR1
BINDING, SIGNALING, AND CELL ADHESION*
Jeffrey K.
Harrison
§,
Alan M.
Fong¶,
Peter A. W.
Swain
,
Shuzhen
Chen
,
Yen-Rei A.
Yu
,
Mina N.
Salafranca
,
William B.
Greenleaf
,
Toshio
Imai**, and
Dhavalkumar
D.
Patel¶

From the
Department of Pharmacology and Therapeutics,
University of Florida College of Medicine,
Gainesville, Florida 32610-0267, the Departments of ¶ Medicine
and
Immunology, Duke University Medical Center,
Durham, North Carolina 27710, and ** KAN Research Institute,
Kyoto 600-8815, Japan
Received for publication, November 10, 2000, and in revised form, March 5, 2001
 |
ABSTRACT |
Fractalkine (FKN/CX3CL1) is a unique member of
the chemokine gene family and contains a chemokine domain (CD), a
mucin-like stalk, a single transmembrane region, and a short
intracellular C terminus. This structural distinction affords FKN the
property of mediating capture and firm adhesion of FKN receptor
(CX3CR1)-expressing cells under physiological flow conditions. Shed
forms of FKN also exist, and these promote chemotaxis of
CX3CR1-expressing leukocytes. The goal of the present study was to
identify specific residues within the FKN-CD critical for FKN-CX3CR1
interactions. Two residues were identified in the FKN-CD, namely
Lys-7 and Arg-47, that are important determinants in mediating
an FKN-CX3CR1 interaction. FKN-K7A and FKN-R47A mutants exhibited
30-60-fold decreases in affinity for CX3CR1 and failed to arrest
efficiently CX3CR1-expressing cells under physiological flow
conditions. However, these mutants had differential effects on
chemotaxis of CX3CR1-expressing cells. The FKN-K7A mutant acted as an
equipotent partial agonist, whereas the FKN-R47A mutant had marked
decreased potency and efficacy in measures of chemotactic activity.
These data identify specific structural features of the FKN-CD that are
important in interactions with CX3CR1 including steady state binding,
signaling, and firm adhesion of CX3CR1-expressing cells.
 |
INTRODUCTION |
Leukocyte trafficking is a complex physiological process that is
dependent upon a number of cellular events that include mechanisms of
cell chemotaxis and adhesion (1, 2). In this regard, chemokines are a
growing family of small molecular mass proteins whose role(s) are
primarily involved in leukocyte migration (3). Fractalkine
(FKN)1 is a newly described
member of the chemokine gene family and the only known member of the
CX3C subfamily (4, 5). The extended structure of FKN is distinct among
the chemokine gene family in that it affords the molecule unique
membrane localization where it can mediate FKN receptor, CX3CR1,
-dependent cell adhesion (6-8). FKN is expressed primarily
by neurons (9-11) and endothelial cells (6, 12). Neuronally expressed
FKN is presumably involved in the central nervous system-based
communication with CX3CR1-expressing microglia, whereas FKN expressed
on endothelial cells likely mediates extravasation of CX3CR1-expressing
cells, which include monocytes, T cells, and NK cells, out of the lumen
of the blood vessel into the tissue parenchyma. FKN is also expressed
by dendritic cells and intestinal epithelial cells, and CX3CR1 has
recently been demonstrated on mast cells (13-16).
The dissection of molecular processes involving chemokines and
chemokine receptors is an active area of investigation as development of therapeutic agents targeting these systems are likely to yield novel
beneficial approaches in the treatment of inflammation, cancer, as well
as inhibition of viral (human immunodeficiency virus)
pathogenesis. Given the localization of FKN to endothelial, dendritic,
and neuronal cells, this molecule also constitutes a potential
therapeutic target for modulating several physiological processes
including leukocyte trafficking and/or mechanisms involved neuroinflammation. The goal of the present study was aimed at identifying amino acid residues in the FKN molecule that are important determinants for binding to, and activating its receptor, CX3CR1. We
employed deletional and site-directed mutagenesis in order to evaluate
the effect of different lengths of the mucin stalk and determine the
role of specific amino acid residues present in the FKN chemokine
domain, in mediating FKN binding, signaling, and
CX3CR1-dependent cell adhesion.
 |
EXPERIMENTAL PROCEDURES |
Mutagenesis--
DNA sequences encoding human fractalkine were
cloned to the EcoRI/XhoI site of pcDNA3
(huFKN/pcDNA3). Five truncation mutants of FKN were prepared by
site-directed mutagenesis using huFKN/pcDNA3 as the template and
five paired sets of complementary oligonucleotides encoding a
different region of the mucin stalk (see Fig. 1). The oligonucleotides
used are as follows A, 5'-CGA GTC TGT GGT CCT CGA GCC CGA AGC CAC
AGG-3' and 5'-CCT GTG GCT TCG GGC TCG AGG ACC ACA GAC TCG-3'; B, 5'-CGC
CAA AGG CTC AGG CTC GAG GGC CTG TGG GCA CGG-3' and 5'-CCG TGC CCA CAG
GCC CTC GAG CCT GAG CCT TTG GCG-3'; C, 5'-GGA GAA TGC TCC GTC TCG AGG
CCA GCG TGT GTG G-3' and 5'-CCA CAC ACG CTG GCC TCG AGA CGG AGC ATT CTC
C-3'; D, 5'-CCA TGC CAC CAT GGA CCT CGA GAG GCT GGG CGT CC-3' and
5'-GGA CGC CCA GCC TCT CGA GGT CCA TGG TGG CAT GG-3'; and E, 5'-CGC CCA GGC TGC CAC CTC GAG GCA GGC GGT GGG GC-3' and 5'-GCC CCA CCG CCT GCC
TCG AGG TGG CAG CCT GGG CG-3'. Each mutation introduced an additional
XhoI restriction endonuclease recognition sequence into the
plasmid. Mutant DNA was generated using the PCR; the original template
was excised by subsequent DpnI treatment. Plasmid DNA was
isolated from random ampicillin-resistant bacterial colonies and
subjected to restriction endonuclease digestion analysis using XhoI. The entire protein coding sequence of each mutant
plasmid was subject to DNA sequence analysis in order to confirm the
mutation and the fidelity of the DNA polymerase (Pfu) used
in the PCR. An EcoRI/XhoI fragment of each
truncation mutant was then subcloned to the same site(s) of
pcDNA3.1/myc/His6 (Invitrogen); the specific version
used depended on the frame in which the XhoI site was engineered. DNA sequence analysis of the final plasmid constructs was
also performed in order to verify that the nucleotides encoding the
myc/His6 were in frame with the nucleotides encoding the
FKN.
Site-directed mutagenesis of the chemokine domain of FKN (FKN-CD) was
also carried out using oligonucleotide-directed PCR and subsequent
DpnI treatment. Mutants were generated in the
huFKN/pcDNA3 (full length) and the truncation mutant, form E
(described above). The specific oligonucleotide pairs used for the
mutagenesis are as follows (see Fig. 1 in reference to the specific
residues mutated): K7A, 5'-GCA CCA CGG TGT GAC GGC ATG CAA CAT CAC GTG
C-3' and 5'-GCA CGT GAT GTT GCA TGC CGT CAC ACC GTG GTG C-3'; K14A,
5'-GCA ACA TCA CGT GCA GCG CGA TGA CAT CAA AGA TAC C-3' and 5'-GGT ATC
TTT GAT GTC ATC GCG CTG CAC GTG ATG TTG C-3'; M15K, 5'-GCA ACA TCA CGT
GCA AGA AGA CAT CAA-3' and 5'-GGT ATC TTT GAT GTC TTC TTG CTG CAC GTG
ATG TTG C-3'; K36A, 5'-CCA GGC ATC ATG CGG CGC ACG CGC AAT CAT CTT
GG-3' and 5'-CCA AGA TGA TTG CGC GTG CGC CGC ATG ATG CCT GG-3'; R37A,
5'-CCA GGC ATC ATG CGG CAA AGC CGC AAT CAT CTT GGA G-3' and 5'-CTC CAA
GAT GAT TGC GGC TTT GCC GCA TGA TGC CTG G-3'; R47A, 5'-GGA GAC GAG ACA
GCA CGC GCT GTT CTG TGC CGA C-3' and 5'-GTC GGC ACA GAA CAG CGC GTG CTG
TCT CGT CTC C-3'; K54A, 5'-GTT CTG TGC CGA CCC GGC GGA GCA ATG GGT CAA
GG-3' and 5'-CCT TGA CCC ATT GCT CCG CCG GGT CGG CAC AGA AC-3'; R74A,
5'-CTG CTG CCC TAA CTG CAA ATG GCG GCA CCT TCG-3' and 5'-CGA AGG TGC CGC CAT TTG CAG TTA GGG CAG CAG-3'; and NIT to EIM, 5'-CAC CAC GGT GTG
ACG AAA TGC GAG ATC ATG TGC AGC AAG ATG ACA TC-3' and 5'-GAT GTC ATC
TTG CTG CAC ATG ATC TCG CAT TTC GTC ACA CCG TGG TG-3.
Expression of FKNs in HEK293T Cells--
Approximately 10 million HEK293T cells (seeded in two 100-mm dishes) were transfected
using 10 µg of plasmid DNA/plate and LipofectAMINE according to the
manufacturer's instructions. Cells were incubated in DMEM containing
10% FBS for the first 20 h post-transfection and subsequently
incubated with DMEM containing 0.5% FBS for an additional 24 h.
Media were harvested and subjected to Ni-NTA chromatography according
to the manufacturer's (Qiagen QIAexpressionist 03/99, Valencia, CA)
recommended procedures. Briefly, conditioned media were mixed with an
equal volume of 2× buffer (100 mM
NaH2PO4, pH 8.0, 600 mM NaCl, 20 mM imidazole, pH 8.0) and 1 ml of the Ni-NTA slurry. The
mixture was rocked for 1 h at 4 °C. After rocking, the mixture
was loaded into a filtration column, and the packed resin bed was
subsequently washed with 8 ml of wash buffer (50 mM
NaH2PO4, pH 8.0, 300 mM NaCl, 20 mM imidazole, pH 8.0). The protein was eluted with elution
buffer (4 × 0.5 ml aliquots, 50 mM
NaH2PO4, pH 8.0, 300 mM NaCl, 250 mM imidazole, pH 8.0), flash-frozen in liquid
N2 in 100-µl aliquots, and stored at
80 °C until
further use.
Western Blot Analysis--
Aliquots of conditioned media, Ni-NTA
column flow-through, column washes, and the elutions were subjected to
SDS-PAGE and Western blot analysis analysis as described previously
(9). Aliquots were electrophoresed through 10% polyacrylamide gels. The proteins were transferred onto a polyvinylidene difluoride 0.45-µm membrane (Millipore, Bedford, MA) and blocked on a rotating table for 0.5 h in TBS-T (Tris-buffered saline/Tween), 5% milk (20 mM Tris-HCl, 150 mM NaCl, 0.1% Tween 20, pH 7.4). After drying, the membranes were incubated for 2 h with a
rabbit anti-fractalkine antibody (1:10,000 dilution) in TBS-T, 5%
nonfat instant milk. The membranes were washed 3 times (10 min per
wash) in TBS-T and subsequently incubated in a goat anti-rabbit
IgG/horseradish peroxidase (1:10,000) in TBS-T, 5% milk for 1 h.
The membranes were washed 3 times (10 min per wash) in TBS-T and then
developed for 5 min with Pierce Supersignal chemiluminescent substrate
(Pierce). Membranes were finally exposed onto Amersham ECL film
(Amersham Pharmacia Biotech).
Whole Cell Radioligand Binding Analysis--
Approximately
50,000 CHO cells stably expressing human or rat CX3CR1, or 300,000 rat
microglia were seeded into the wells of a 12-well plate. Two days
later, whole cell radioligand binding analysis was performed according
to procedures published previously (9). Briefly, cells were rinsed (1 time) with PBS and subsequently incubated for 1 h in 0.5 ml of
HBSS containing 10 mM Hepes-Na, pH 7.4, and 0.1% BSA
(binding buffer) with 0.5-1 nM 125I-huFKN-CD
(specific activity, 200-300 Ci/mmol) in the absence and presence of
various concentrations of unlabeled inhibitors (FKN-CD, truncation
mutants, or FKN-CD mutants). At the end of the incubation cells were
rinsed 3 times (1 ml per wash) with ice-cold binding buffer.
Radioactivity retained in the wells was extracted with 0.2 N NaOH and subject to gamma spectroscopy. Nonspecific binding was determined in the presence of 100 nM huFKN-CD.
Data are expressed as percent of total specific binding. Graphical and
statistical analysis was carried out using Prism 2.0 (GraphPad Software, Inc., San Diego, CA).
Measurement of Intracellular Calcium Levels--
Intracellular
calcium levels in fura-2 AM-loaded CHO-huCX3CR1 cells were assayed as
described previously (9) using a spectrofluorimeter (Photon Technology
International, Monmouth Junction, NJ). Cells were illuminated
alternately with ultraviolet light (1/s) of 340 and 380 nm wavelength
and monitored for fluorescence at 510 nm. Due to the uncertainties
involved in the calculation of [Ca2+]i, the
signals are reported as changes in F340/F380, which gives a relative
measure of [Ca2+]i.
FKN-ELISA--
The concentrations of purified soluble FKN
proteins were determined by ELISA using anti-FKN antibodies. One
hundred µl/well of 2 µg/ml capture anti-FKN (R & D Systems,
Minneapolis, MN) was incubated overnight at room temperature in a
96-well plate. After washing three times in wash buffer (0.05% Tween
20 in PBS), the wells were blocked with 1% BSA, 5% sucrose, 0.05%
NaN3 for 1 h. Following three more washes, 100 µl of
each diluted protein sample was added and incubated for 1 h at
37 °C. After 3 washes, 100 µl of 100 ng/ml biotinylated anti-FKN
(R & D Systems) diluted in PBS containing 0.05% Tween 20, 0.05%
NaN3, 0.2% BSA was added and incubated for 1 h at
37 °C. The plates were then washed 3 times, and 100 µl of 1:4000
dilution streptavidin horseradish peroxidase (Zymed
Laboratories Inc., South San Francisco, CA) was added and
incubated for 20 min at room temperature. After 3 washes, horseradish
peroxidase activity was detected colorimetrically using the TMB
microwell peroxidase substrate system (Kirkegaard & Perry,
Gaithersburg, MD) and the results read on a Dynex microplate reader.
Protein concentrations were determined by averaging the results of
three independent ELISA assays.
Chemotaxis Assays--
Chemotaxis toward soluble FKN and FKN-CD
mutants was addressed by migration through transwells. Various
dilutions of Ni-NTA-eluted fractions containing wild type and mutant
proteins were made in 600 µl of medium (RPMI 1640 containing 10%
FBS) and placed in the bottom of a transwell (6.5 mm diameter, 3-µm
pore size, Costar, Corning, NY). One million L1.2 cells expressing
CX3CR1-GFP were resuspended in 100 µl of RPMI medium and placed into
the top chamber of a transwell. After 4 h of incubation at
37 °C, the bottom well was harvested. The number of migrating cells
was determined by counting a 20-µl aliquot using flow cytometry
(Coulter Epics XL-MCL). Ni-NTA column elution buffer had no inhibitory
or enhancing effects on L1.2 cell chemotaxis. For each experiment,
values were normalized to wild type FKN with maximal chemotaxis to wild
type FKN set at 100%. Concentration-response curves were generated by
curve fit interpolation analysis using Cricket Graph III (Computer
Associates International, Islandia, NY). Statistical analysis was
carried out using Statistica 3.0b (Statsoft Inc.)
Expression of FKNs in EA.hy 926 Cells--
EA.hy 926 cells were
grown in DMEM containing 10% FBS. Cells (grown to 80% confluence)
were transfected with 10 µg of DNA using a calcium phosphate
precipitation-based method. Forty eight hours post-transfection, cells
were seeded into media containing 0.5 mg/ml G418. G418-resistant cells
were analyzed for cell surface levels of fractalkine by staining with a
monoclonal antibody specific for the mucin domain of fractalkine (mAb
1D6). Results were visualized on a Coulter Epics-XL flow cytometer.
Capture and Firm Adhesion under Physiological Flow--
Firm
adhesion of CX3CR1-expressing cells was assessed by a parallel plate
flow chamber assay as described previously (7). Briefly, EA.hy 926 cells were plated onto glass coverslips and allowed to grow overnight
at 37 °C. The coverslips were then placed in a parallel plate flow
chamber. K562 or K562-CX3CR1 cells were resuspended in flow buffer (PBS
containing 0.75 mM CaCl2, 0.75 mM
MgCl2, 0.5% BSA) at a concentration of 1 × 106 cells/ml. The cells were perfused at a shear stress of
0.25 dynes/cm2 for 5 min. Following subsequent 1 min washes
of 0.5, 1, and 1.85 dynes/cm2 for 1 min each, the cells
were subjected to a high shear stress of 5 dynes/cm2 in
flow buffer. The results of each run were recorded onto videotape. The
number of K562 cells remaining bound to the monolayer was then counted.
The number of captured cells on EA.hy 926 cell transfectants was
determined by counting the number of cells that were in contact with
the monolayer for at least 5 s during the initial loading phase in
the flow chamber (shear stress of 0.25 dynes/cm2). For all
monolayers, greater than 90% of the captured cells remained firmly
adherent (n = 3).
 |
RESULTS |
Although FKN is expressed as a membrane-attached protein,
extracellular forms of this protein are readily detected. The specific cleavage site(s) that generate these shed forms of FKN are not known.
FKN released from the cell surface has an observed molecular mass
greater than predicted and thus is likely to contain all or a portion
of the glycosylated mucin-like stalk. Our first goal was directed
toward determining the effect of various lengths of this mucin-like
domain on the affinity and efficacy of the FKN-CD for binding and
activating CX3CR1. Fig. 1 depicts the
entire protein sequence of human FKN, in alignment with the chemokine domains of the two known rodent orthologs (rat and murine FKN-CD). In
order to evaluate the effect of various lengths of the mucin stalk on
FKN-CD function, five truncation mutants of human FKN (designated A-E)
were prepared in which DNA sequences encoding the signal peptide, the
chemokine domain, and differential lengths of the mucin stalk were
cloned in frame with C-terminal six histidine residues
(His6). The C-terminal addition of the
His6 residues facilitated purification of the various
secreted FKN truncation mutants from the conditioned media of the
HEK293T cell transfectants (using Ni-NTA chromatography). In addition,
the figure highlights conserved lysine and arginine residues, present
in the chemokine domain of the various fractalkine orthologs, that were
targeted for site-directed mutagenesis (to alanine). Since chemokine
receptors typically contain a number of aspartate and glutamate
residues within their extracellular regions, these negatively charged
side chains present in the receptor are likely candidate amino acids for interaction with positively charged amino acids that are prevalent not only in FKN but also in other chemokine sequences. Thus, we hypothesized that specific basic residues present within the FKN molecule are critical for interactions with its receptor CX3CR1. In
addition, a single methionine residue (at position 15) and a unique
N-linked glycosylation sequence (NIT) present in human and
rat FKN are highlighted. Met-15 was previously suggested to interact
with a peptide containing the sequences of the N terminus of human
CX3CR1 (17); this residue was mutated to a lysine. From structural
studies, the single N-linked glycosylation in the human FKN
sequence, which is also present in the cognate region of rat FKN,
appears to be a readily available substrate for this post-translational
modification. Thus, in order to assess the role of this putative
glycosylation site in the interaction of FKN with CX3CR1, we mutated
the N-linked glycosylation sequence to "EIM"; this
tri-amino acid motif is EIM in the murine ortholog.

View larger version (36K):
[in this window]
[in a new window]
|
Fig. 1.
Alignment of human fractalkine (huFKN) with
rat and murine fractalkine chemokine domains (ratFKN-CD and
murFKN-CD). Basic residues conserved within the three FKN
orthologs (Lys-7, Lys-14, Lys-36, Arg-37, Arg-47, Lys-54, and Arg-74)
were changed to alanine and are indicated in boldface.
Boldface methionine (Met-15) was mutated to a lysine. The unique
N-linked glycosylation consensus sequence (NIT) present in
human and rat FKN-CDs is underlined. The starred
residues denote the four conserved cysteine residues; the
boldface and underlined sequences correspond to
the transmembrane spanning domain.
|
|
The effect of the length of the mucin stalk on the binding of the
FKN-CD to CX3CR1 and the stimulation of intracellular calcium mobilization in CX3CR1-expressing cells were assessed. Western blot
analysis (Fig. 2A) of the five
293T cell-expressed truncation mutants of FKN, purified by Ni-NTA
chromatography, indicated that all five forms were expressed and
secreted into the HEK293T cell-conditioned media. All of the secreted
proteins migrated as broad, diffuse bands thru SDS-PAGE at molecular
masses greater than their calculated molecular masses based upon the
predicted protein backbones (Table I).
This is likely due to the glycosylated characteristics of the mucin
stalk. Nevertheless, the relative sizes of the various truncation
mutants were consistent with the protein containing the CD and
progressively increasing lengths of the mucin stalk. Competition
binding analysis (125I-FKN-CD binding to huCX3CR1-CHO
cells) of each of these truncation mutants indicated that each form
displayed a comparable affinity for human CX3CR1 (Fig. 2B).
Furthermore, each form of the protein stimulated similar increases in
intracellular calcium levels in the fura-2-loaded huCX3CR1-CHO cells
(Fig. 2C). Collectively the data indicate that the mucin
stalk does not contribute to FKN binding affinity and efficacy for
stimulating calcium mobilization in CX3CR1-expressing cells.

View larger version (28K):
[in this window]
[in a new window]
|
Fig. 2.
Binding and activation of CX3CR1 by huFKN-CD
is not modulated by the length of the mucin stalk. Truncated forms
of human FKN were prepared as indicated under "Experimental
Procedures." A, Western blot analysis of huFKN-CD with
various lengths of the mucin stalk. 293T cells were transfected with
plasmid DNA encoding five different forms of huFKN (A-E, as
designated in Fig. 1). Myc/His6-tagged proteins present in
the conditioned media 2 days post-transfection were purified using
Ni-NTA chromatography, and an aliquot (1 µl) of one of the elution
fractions was subject to SDS-PAGE and Western blot analysis using the
anti-hufractalkine antibody according to procedures indicated under
"Experimental Procedures." The 1st lane contains 2.5 pmol of human FKN chemokine domain (CD) purchased from R & D
Systems. Numbers to the left denote relative
molecular mass of protein standards. B, competition binding
analysis of huFKN truncation mutants. Radioligand binding analysis of
125I-huFKN-CD binding to huCX3CR1-CHO cells. Whole cell
binding analysis of 125I-huFKN-CD in the absence and
presence of increasing amounts of the NI-NTA-eluted protein was
performed according to procedures outlined under "Experimental
Procedures." Data are plotted as % Bound as a function of
volume of the eluate added. Results are expressed as mean ± S.E.
from three independently conducted experiments performed in triplicate.
Inset depicts competition by huFKN-CD. C,
stimulation of intracellular calcium mobilization by huFKN truncation
mutants. Representative traces from a single experiment are depicted.
Results are representative of three independently conducted
experiments.
|
|
View this table:
[in this window]
[in a new window]
|
Table I
Predicted and observed molecular masses of FKN truncation mutants
Shown are the calculated molecular masses (in kilodaltons) of the
predicted protein backbones of the various FKN truncation mutants as
well as the molecular mass ranges of the expressed proteins as observed
by SDS-PAGE and Western blot analysis (depicted in Fig. 2). The
predicted molecular masses exclude the mass contributed by the putative
signal peptide sequence.
|
|
The effect of mutating the species conserved basic residues, the single
N-linked glycosylation consensus sequence, and Met-15, in
the FKN-CD, on the binding of FKN to CX3CR1 was evaluated. Site-directed mutants of these residues were prepared in the context of
the longest secreted truncation mutant (CD with all of the mucin stalk,
i.e. form E) and purified by Ni-NTA chromatography. Western
blot analysis (Fig. 3A)
indicated that all of the Ni-NTA-purified mutants were present at
concentrations equivalent to the levels of the purified "wild type"
form. Ni-NTA-purified proteins were then evaluated for their relative
potency for inhibiting the binding of 125I-FKN-CD to the
huCX3CR1-CHO cells. The K7A and R47A mutants displayed significantly
lower binding potencies than the wild type and all other mutant forms
(Fig. 3B). Estimates of the relative shift in potencies of
these two forms were ~30-60-fold. FKN-CD mutants showing comparable
binding affinities for CX3CR1 were evaluated for their ability to
stimulate increases in intracellular calcium in fura-2-loaded
huCX3CR1-CHO cells. At concentrations that achieve near full receptor
occupancy (~90%), each of these FKN mutants stimulated increases in
intracellular calcium that were comparable to the "wild type" FKN
(Fig. 3C). At concentrations equivalent to ~75% receptor
occupancy by the wild type FKN, FKN-K7A, and -R47A mutants stimulated
lower increases in intracellular calcium that correlated with the
decreased binding affinity of these mutants.

View larger version (24K):
[in this window]
[in a new window]
|
Fig. 3.
Lysine 7 and arginine 47 are critical amino
acids in the binding of FKN to human CX3CR1. Western blot and
binding analysis of huFKN-CD mutants. Site-directed mutagenesis of
conserved basic residues within the huFKN-CD was performed according to
procedures indicated under "Experimental Procedures." Plasmid DNA
encoding the longest truncation mutant (CD with all of the mucin stalk)
served as the template in the mutagenesis reactions. Proteins were
expressed in 293T cells and purified by Ni-NTA chromatography.
A, Western blot analysis of wild type (WT) and CD
mutants (K7A, K14A, M15K, K36A, R37A, R47A, K54A, R74A, NIT to EIM) was
performed as described above. The equivalent of 0.25 µl of each
mutant were electrophoresed. The numbers below each lane
indicate the site of the specific mutation. B, competition
binding analysis of huFKN-CD mutants. Radioligand binding analysis of
125I-huFKN-CD binding to huCX3CR1-CHO cells is shown. Whole
cell binding analysis of 125I-huFKN-CD in the absence and
presence of increasing amounts of the Ni-NTA-eluted protein was
performed according to procedures indicated above. Data are plotted as
% Bound as a function of volume of the eluate of a single
fraction added. Results are expressed as mean ± S.E. from three
independently conducted experiments performed in triplicate. WT
( ), K7A ( ), K14A ( ), M15K ( ), K36A ( ), R37A ( ), R47A
( ), K54A ( ), R74A ( ), NIT to EIM (×). C,
stimulation of intracellular calcium mobilization by huFKN-CD mutants.
Representative traces from a single experiment are depicted. Results
are representative of three independently conducted experiments.
|
|
The concentration-dependent effect of FKN to promote
chemotaxis of CX3CR1-expressing cells was evaluated for each of the
mutants (Fig. 4). FKN-K7A and -R47A
mutants displayed reduced chemotactic activity relative to that
observed with wild type FKN. The FKN-R47A mutant was nearly ineffective
at stimulating a chemotactic response, with maximal activity less than
10% of the wild type activity, whereas the FKN-K7A mutant
retained ~60% of the wild type maximal activity. The FKN-K36A mutant
had slightly enhanced maximal chemotactic activity. Mutation of Met-15
(to Lys) yielded an FKN that was ~10-fold less potent at promoting
chemotaxis; these data are consistent with the slight reduction in
binding affinity of FKN-M15K (see Fig. 3B). Table
II summarizes the effect of the mutations
on the relative concentration dependence and maximal chemotactic
activities of the mutant FKNs relative to the respective wild type FKN
parameters.

View larger version (43K):
[in this window]
[in a new window]
|
Fig. 4.
Chemotaxis of CX3CR1-GFP-expressing L1.2
cells to FKN-CD mutants. Chemotaxis toward different
concentrations of soluble FKN mutant proteins was tested as described
under "Experimental Procedures." Mutant FKN protein concentrations
were determined by ELISA. Shown are the concentration-response curves
for mutant FKN proteins (filled circles with solid
lines) compared with wild type FKN (open circles with
dashed lines). Shown are the means ± S.E. from three
independent experiments. The numbers of migrating cells were normalized
to wild type FKN with 100% representing the peak chemotaxis to wild
type FKN in each experiment.
|
|
View this table:
[in this window]
[in a new window]
|
Table II
Relative EC50 values and maximal chemotaxis of FKN-CD mutants
Shown are EC50 values and maximal chemotactic activity for
FKN-CD mutants, relative to EC50 and maximal response of wild
type (WT) FKN at stimulating chemotaxis of CX3CR1-L1.2 cells. Results
are expressed as means ± S.D. from three independent experiments.
ND, not determined.
|
|
As stated, the specific basic residues targeted for mutagenesis are
conserved in human, rat, and mouse FKNs. Analysis of the relative
potencies of the mutants in binding to rat CX3CR1 revealed similar
decreased affinities of the K7A and R47A mutants. This was evident
regardless if the source of rat CX3CR1 was recombinant (Fig.
5A, rat CX3CR1-CHO cells) or
native cell expressed (Fig. 5B, rat microglia). These data
indicate similar interactions of the FKN with CX3CR1 in humans and
rodents and point to conserved amino acids in these receptor orthologs
as the counter amino acid residues interacting with amino acids Lys-7
and Arg-47 in FKN.

View larger version (16K):
[in this window]
[in a new window]
|
Fig. 5.
Analysis of huFKN-CD mutant binding to rat
CX3CR1. A, radioligand binding analysis of
125I-huFKN-CD binding to rat CX3CR1-CHO cells in the
presence of huFKN-CD mutants. Whole cell binding analysis of
125I-huFKN-CD in the absence and presence of increasing
amounts of the Ni-NTA-eluted protein was performed according to
procedures indicated above. Data are plotted as % Bound as
a function of volume of the eluate of a single fraction added. Results
are expressed as mean ± S.E. from one experiment performed in
triplicate. The results are representative of two experiments.
WT ( ), K7A ( ), K14A ( ), M15K ( ), K36A ( ), R37A
( ), R47A ( ), K54A ( ), R74A ( ), NIT to EIM (×).
B, radioligand binding analysis of 125I-huFKN-CD
binding to rat microglial cells in the presence of huFKN-CD mutants.
Whole cell binding analysis of 125I-huFKN-CD in the absence
and presence of increasing amounts of the Ni-NTA-eluted protein was
performed. Data are plotted as % Bound as a function of
volume of the eluate of a single fraction added. Results are expressed
as mean ± S.E. from one experiment performed in triplicate. The
results are representative of two experiments. WT ( ), K7A ( ),
R47A ( ).
|
|
Parallel mutations were generated in the full-length FKN, in order to
assess the role of these residues on FKN-dependent cell adhesion. The endothelial cell line, EA.hy 926, was transfected with
each of these mutant constructs, and G418-resistant clonal cells were
sorted. EA.hy 926 cells are derived from human umbilical vein
endothelial cells and display a number of endothelial cell-like properties. Cell surface expression levels of these mutant FKNs in the
stably transfected cells were roughly equivalent in all cell lines
generated, as evidenced by flow cytometry (Fig.
6). The ability of CX3CR1-expressing
cells to bind to the EA.hy 926 cells expressing the mutant FKNs was
determined by a parallel plate flow chamber assay. Fig.
7 shows that K562-CX3CR1 cells efficiently bind to all of the mutant FKN EA.hy 926 cells to the same
extent as the wild type FKN-expressing EA.hy 926 cells with the
exception of the K7A and R47A mutant FKN-expressing cells. A 60%
reduction in both the capture and firm adhesion of K562-CX3CR1 cells to
these two mutant FKN-expressing EA.hy 926 cells, as compared with wild
type FKN-EA.hy 926 cells, was evident.

View larger version (41K):
[in this window]
[in a new window]
|
Fig. 6.
Expression analysis of "full-length"
huFKN-CD mutants in EA.hy 926 cells. EA.hy 926 cells stably
transfected with various full-length FKN-CD mutants were tested for
surface expression by flow cytometry. Shown is the expression of FKN as
determined using mAb 1D6 (dark histograms) that recognizes
the mucin domain. Background staining with the control mAb P3 is
indicated by the clear histograms.
|
|

View larger version (26K):
[in this window]
[in a new window]
|
Fig. 7.
CX3CR1-K562 cell capture by and firm adhesion
to FKN mutants expressed on EA.hy 926 cells. Left
panel, cell capture. Shown are the relative amounts of K562-CX3CR1
cells captured at low shear stress by EA.hy 926 cells expressing the
various FKN mutants compared with wild type FKN at a shear stress of
0.25 dynes/cm2. K562-CX3CR1 cells were allowed to interact
with confluent EA.hy 926 monolayers under physiological flow conditions
using the parallel plate flow chamber. Capture was defined as cell
contact with the monolayer for at least 5 s. Right
panel, firm adhesion. Shown are the relative amounts of
K562-CX3CR1 cells remaining firmly bound to EA.hy 926 monolayers
expressing various FKN mutants following high shear stress. After the
initial 5-min loading time at 0.25 dynes/cm2, the cells
were subjected to increasing shear stresses as outlined under
"Experimental Procedures." After 2 min at 10 dynes/cm2,
the number of K562-CX3CR1 cells remaining bound to the monolayer was
counted. The values for capture and firm adhesion have been normalized
with 100% capture and firm adhesion being the number of cells
interacting with or firmly adherent to EA.hy 926 monolayers expressing
wild type FKN. Shown are the means and standard deviations from three
independent experiments. The FKN mutants with a statistically
significant (p < 0.05) difference from the wild type
protein are indicated with an asterisk.
|
|
A chief concern in the analysis of functional properties of
site-directed mutants is whether or not the specific mutation alters
global protein conformation, as opposed to disrupting a specific
protein-protein interaction. Thus, the effect of the various
site-specific mutations on global FKN-CD conformation was assessed
using a panel of monoclonal antibodies directed against the CD of human
FKN. Cell surface reactivity of each of the mAbs was assessed using the
EA.hy 926-expressed proteins. Reactivity of each of the mAbs toward the
wild type FKN and the K7A, K14A, K54A, R74A, and NIT-EIM FKN mutants
were equivalent (Table III). In contrast,
the reactivities of mAbs 2A5 and 8C5 were reduced in the M15K, K36A,
and R37A mutants. The monoclonal antibody, mAb 1B7, was less reactive
toward the M15K and R37A mutants, whereas mAb 4F2 did not react with
the R47A mutant. The spatial proximity of Met-15, Lys-36, and Arg-37
(Fig. 8) is consistent with similar reductions in reactivity by mAbs 2A5, 8C5, and 4F2.
View this table:
[in this window]
[in a new window]
|
Table III
Epitope mapping of FKN-CD mutants
Shown are the cell surface reactivities of monoclonal antibodies
directed to the mucin (1D6) and chemokine domains of FKN. Cell surface
reactivity with FKN mutants was determined by indirect
immunofluorescence and flow cytometry. Reactivity was considered
negative ( ) if the mean fluorescence was less than 20% of maximal.
|
|

View larger version (43K):
[in this window]
[in a new window]
|
Fig. 8.
Model of the FKN-CD depicting the
location of the Lys-7 and Arg-47 amino acid residues. The
ribbon diagram was generated using the program MOLSCRIPT
(25), and the atomic coordinates and structure factors
(1f2l) are deposited in the Protein Data Bank,
Research Collaboratory for Structural Bioinformatics, Rutgers
University, New Brunswick, NJ (21). The side chains of Lys-7 and Arg-47
are depicted in red. The side chains of Met-15 and Arg-37
are in blue, and the side chain of Lys-36 is shown in
purple. The conserved Cys residues are green. The
N and C termini of this structure are marked with N and
C, respectively.
|
|
 |
DISCUSSION |
The major goal of the current study was aimed at identifying
specific basic residues in the chemokine domain of FKN that are critical for receptor interactions. Two amino acid residues, namely Lys-7 and Arg-47, were identified as important amino acid determinants in the binding of FKN to CX3CR1. The significance of these residues in
binding interactions was confirmed in two independent methods used to
assess binding of FKN to CX3CR1, namely in steady state binding
analysis and in in vitro flow chamber assays in which the
FKN-CX3CR1 interaction is measured under conditions mimicking physiological blood flow. Whereas the K7A and R47A mutants behaved similarly in assays quantifying binding activities, these mutants had
differential effects in measures of signaling via CX3CR1. Although
FKN-R47A was essentially ineffective at stimulating intracellular calcium mobilization and chemotaxis of CX3CR1-expressing cells, the
FKN-K7A mutant retained signaling activity. Three additional findings
were noted in the present study. First, the single N-linked glycosylation site present in human and rat FKN is not glycosylated to
any appreciable extent in heterologous expression systems and also does
not appear to have any functional significance in the binding or
activation of CX3CR1 by FKN. Second, a methionine residue (Met-15),
previously suggested to interact with the N terminus of CX3CR1, does
not appear to be as critical at interacting with CX3CR1, as expressed
in mammalian cells. Third, the affinity and efficacy of the FKN-CD are
independent of the length of the mucin stalk.
FKN is a dual function chemokine capable of promoting CX3CR1-expressing
cell chemotaxis and cell adhesion. Shed or secreted forms of FKN
stimulate leukocyte chemotaxis, whereas the membrane-attached form of
FKN mediates cell adhesion (4, 6, 7). A limited level of
structure-function analysis of FKN and CX3CR1 has been reported.
Although the chemokine domain of FKN is critical for interaction of FKN
with CX3CR1, less information is available on the role of the mucin
stalk. Under conditions mimicking physiological blood flow, cells
expressing "full-length" FKN with the entire mucin domain are more
efficient at capturing leukocytes as compared with FKN-CD proteins,
i.e. without the mucin stalk (18). Although the arrest of
CX3CR1-expressing cells under flow conditions is dependent on the
chemokine domain of FKN, the function of the mucin stalk in this
setting appears to be as a molecular extender of the FKN-CD to
circulating leukocytes. The ability of FKN to arrest cells appears to
be a unique property of the FKN chemokine domain as chimeric molecules
in which the mucin domain was replaced with a stalk from E-selectin was
fully functional in leukocyte arrest (18). Furthermore, chimeric
molecules in which other chemokines were "fused" to the FKN mucin
stalk failed to arrest cells expressing the cognate chemokine receptor
(19).
Given the importance of the chemokine domain of FKN in binding and
functional responses of CX3CR1-expressing cells, our studies primarily
addressed the roles of specific amino acids in the FKN-CD in
interacting with the receptor. A single N-linked
glycosylation site is present in the human and rat orthologs of FKN
and, according to structural data, is spatially available for
post-translational modification (17). Although the human and rat FKN
molecules have the potential N-glycosylation site
(CNITC), mouse FKN does not contain an
N-glycosylation site in the CX3C bulge (CEIMC), indicating
that glycosylation may not be important. However, murine FKN does not
support arrest of CX3CR1-expressing cells under flow (20). Furthermore,
evidence that N-glycosylation may play a role under certain
conditions comes from the fact that synthetic, non-glycosylated FKN has
diminished activity for monocytes (4). One possibility is that
N-glycosylation contributes to monocyte activation and
adhesion. In our heterologous expression system, we did not find
evidence for glycosylation of this site in the human protein. The
relative migration of the NIT to EIM FKN mutant in SDS-PAGE was not
significantly different than the mobility of the wild type FKN.
Furthermore, these amino acid changes did not affect binding and
functional properties, including affinity for the receptor, stimulation
of intracellular calcium mobilization, potency, and efficacy in
promoting chemotaxis and cell adhesion.
There are six basic residues present within the FKN-CD that are
conserved in human, rat, and murine FKN. The similar binding affinities
of human and rodent FKN for rat CX3CR1 (9) suggests that conserved
residues in FKN are the likely amino acid residues that interact
directly with motifs present in the receptor. Mutation of each of these
residues to an alanine appeared to have little effect on the global
conformation of the FKN-CD. This conclusion is based on two
observations from the mAb epitope mapping studies. First, in general
the mAbs used in this study reacted as efficiently with the mutants as
they did with the wild type FKN. Second, those mutants that showed
reduced mAb reactivity (e.g. M15K, K36A, and R37A) did not
display significant differences in binding affinities, stimulation of
calcium mobilization, or cell adhesive properties; a slight reduction
in the potency of the M15K mutant at stimulating chemotaxis was seen.
Our approach identified two key residues, namely Lys-7 and Arg-47, as
important amino acids for interaction with CX3CR1. These mutant FKNs
displayed reduced affinity for CX3CR1 in steady state whole cell
binding assays and did not support either efficient capture or firm
adhesion of CX3CR1-expressing cells under physiological flow. However,
despite similar effects of these mutations on FKN-CX3CR1-binding
interactions, differential effects of these mutations on CX3CR1
signaling were observed. Mutation of Arg-47 produced a protein largely
ineffective at stimulating either intracellular calcium mobilization or
chemotaxis, at least at the concentrations tested. However, mutation of
Lys-7 yielded a molecule that retained most of the agonist activity,
although maximal chemotaxis of the L1.2 cells expressing CX3CR1 toward the K7A mutant was slightly reduced. More notable, the potency of this
mutant in this assay was similar to the wild type FKN, which was not
expected based on measures of affinity in the whole cell binding
assays. This difference may reflect receptor host cell differences
(L1.2 versus CHO). Alternatively, K7A may be stimulating
chemotaxis via a state of CX3CR1 that is not sensitive to mutation at
this amino acid position in FKN. Measurement of the relative binding
affinities of the mutant panel in the whole cell binding assay is based
upon inhibition of the binding of subsaturating levels of
125I-FKN-CD and thus represents binding to a high affinity
state of the receptor. This state of the receptor may be all or
partially distinct from the population of receptors that mediate the
chemotaxis response. Nonetheless, the results identify regions of the
FKN-CD that differentiate binding and signaling properties of the FKN molecule.
Slight reductions in steady state binding affinity and potency in the
chemotaxis assay were observed for the M15K mutant. Met-15 was
previously suggested to be a key determinant in the interaction of the
FKN-CD with CX3CR1. From solution NMR measurements of bacterially
expressed protein, Mizoue et al. (17) determined that this
residue displayed a significant chemical shift upon titration with a
chemically synthesized peptide corresponding to the N terminus of human
CX3CR1. Our approach toward identifying amino acids in FKN important
for CX3CR1 binding and signaling was based on investigation of these
interactions using longer forms of the FKN molecule, expressed by
mammalian cells as either a membrane-attached or -secreted form, as
well as full-length, mammalian cell-expressed receptor. Thus, the
identification of Lys-7 and Arg-47 as critical amino acids suggests
that regions other than the N terminus of CX3CR1 contribute to the
binding energy. Furthermore, post-translational modifications of the
receptor may also confer FKN binding determinants.
Shed forms of FKN are evident in both transfected and native cell
systems (3, 9). Transfection of 293 cells with the full-length form of
FKN leads to the detection of both cell surface protein as well as
protein in the conditioned media (3). Brief exposure of primary rat
neurons with glutamate leads to increased levels of shed FKN and
corresponding decreases in membrane-attached FKN. This
neurotoxicity-induced proteolytic cleavage of FKN may be mediated by
specific proteases as the matrix metalloproteinase inhibitor batimastat
prevents this cleavage (20). Alterations in the levels of low molecular
weight forms of FKN are also evident in the facial motor nucleus after
peripheral nerve injury and may play a role in neuronal regeneration
(9). The presence of various forms of shed FKN could suggest distinct
physiological roles for the cleaved forms of the protein. Previously
published data (4, 6, 18) indicated that FKN-CD and FKN-CD with the
entire mucin stalk were equally effective at binding and stimulating CX3CR1; these earlier studies did not address the functionality of the
FKN-CD with intermediate lengths of the mucin stalk. In order to
examine possible biphasic effects of the mucin stalk region
(i.e. determine if shorter stalks reduced or enhanced a FKN-CX3CR1 interaction), we generated five soluble recombinant FKN
constructs in which the FKN-CD contains various lengths of the mucin
stalk. Western blot analysis of each of these forms demonstrated that
each form migrated in SDS-PAGE at molecular masses greater than the
masses predicted by their respective protein backbones. This indicates
that glycosylation of the mucin stalk occurs along the entire length of
this domain. From a functional standpoint, our data indicate that the
length of the stalk does not influence either the binding affinity of
the FKN-CD for CX3CR1 or the efficacy in stimulating a calcium
mobilization response in receptor-expressing cells. Although there may
be multiple sites of cleavage in the FKN mucin domain, the
physiological significance of potentially distinct forms of shed FKN
remains to be determined. Furthermore, the context within which the
receptor is expressed, i.e. the specific cell type, may
determine the differential sensitivity toward various shed forms of
FKN.
In summary, these data exemplify the importance of amino acid
residues within the FKN-CD as the critical determinants in an FKN-CX3CR1 interaction. The structure of the FKN-CD has been reported using both NMR and x-ray crystallography (17, 21). Considering this
structural information, the side chains of Lys-7 and Arg-47, while
remotely distant from one another, appear to extend out from the same
face of the FKN-CD (Fig. 8). Thus, it is conceivable that both of these
residues are simultaneously interacting with specific, different amino
acid residues found in the extracellular domain of CX3CR1. Mutation of
both residues interfered to a similar extent, with the ability of FKN
to bind to CX3CR1, as measured in steady state binding and adhesion
assays. However, the K7A mutant retained the potency and most of the
efficacy of the FKN for stimulating chemotaxis of CX3CR1-expressing
cells. The results indicate that residues within the FKN-CD
differentially regulate binding to and activation of CX3CR1.
Site-directed mutagenesis of other chemokines has revealed similar
interactions between these chemokines and their cognate receptors. For
instance, mutation of the Arg residue in the ELR motif of
interleukin-8 (equivalent to Lys-7 in FKN) yields a molecule
that displays reduced affinity for its neutrophil receptor (22).
Similarly, mutation of Lys-49 in MCP-1, an amino acid found in the
analogous position as Arg-47 of the FKN-CD, yields a mutant with
reduced affinity for CCR2 (23). Therefore, common mechanisms of
chemokine-chemokine receptor protein interactions seem likely. The
identification of the specific counter amino acids residing on the
receptor, which interact specifically with the FKN-CD, will require
additional experimentation and will have to include further mutational
analysis and/or structural determination of the receptor in complex
with the ligand. Toward the latter goal, the recent demonstration of
the x-ray crystal structure of rhodopsin (24) suggests that more
precise structures of other members of the G-protein-coupled receptor
superfamily may be forthcoming. Identification of specific FKN-CX3CR1
interactions will aid future development of therapeutic agents that
target this receptor system in disease.
 |
ACKNOWLEDGEMENTS |
We thank Aron Boney for performing the FKN
ELISA assays and Drs. Art Edison for help with constructing the FKN-CD
model, Wolfgang Streit for assistance in culturing rat microglia, and
Stephen Baker for critical discussions of the data.
 |
FOOTNOTES |
*
This work was supported in part by National Institutes of
Health Grants NS34901 (to J. K. H.) and AR39162 (to D. D. P) and an
American Heart Association-Florida Affiliate grant-in-aid (to J. K. H.).The costs of publication of this
article were defrayed in part by the
payment of page charges. The article
must therefore be hereby marked
"advertisement" in
accordance with 18 U.S.C. Section
1734 solely to indicate this fact.
§
To whom correspondence may be addressed: Dept. of Pharmacology and
Therapeutics, College of Medicine, Box 100267, University of Florida,
Gainesville, FL 32610-0267. Tel.: 352-392-3227; Fax: 352-392-9696;
E-mail: harrison@pharmacology.ufl.edu.

To whom correspondence may be addressed: Depts. of Medicine and
Immunology, Duke University Medical Center, Box 2632, 223 MSRB, Durham,
NC 27710. Tel.: 919-684-4234; Fax: 919-681-9399; E-mail:
patel003@mc.duke.edu.
Published, JBC Papers in Press, March 8, 2001, DOI, 10.1074/jbc.M010261200
 |
ABBREVIATIONS |
The abbreviations used are:
FKN, fractalkine;
CD, chemokine domain;
CX3CR1, receptor for fractalkine;
DMEM, Dulbecco's modified Eagle's medium;
FBS, fetal bovine serum;
PBS, phosphate-buffered saline;
CHO, Chinese hamster ovary;
HEK, human
embryonic kidney;
Ni-NTA, nickel-nitrilotriacetic acid;
BSA, bovine
serum albumin;
PAGE, polyacrylamide gel electrophoresis;
PCR, polymerase chain reaction;
WT, wild type;
ELISA, enzyme-linked
immunosorbent assay;
mAb, monoclonal antibody.
 |
REFERENCES |
1.
|
Springer, T. A.
(1994)
Cell
76,
310-314
|
2.
|
Butcher, E. C.,
and Picker, L. J.
(1996)
Science
272,
60-66[Abstract]
|
3.
|
Murphy, P. M.,
Baggiolini, M.,
Charo, I. F.,
Hebert, C. A.,
Horuk, R.,
Matsushima, K.,
Miller, L. H.,
Oppenheim, J. J.,
and Power, C. A.
(2000)
Pharmacol. Rev.
52,
145-176[Abstract/Free Full Text]
|
4.
|
Bazan, J. F.,
Bacon, K. B.,
Hardiman, G.,
Wang, W.,
Soo, K.,
Rossi, D.,
Greaves, D. R.,
Zlotnik, A.,
and Schall, T. J.
(1997)
Nature
385,
640-644[CrossRef][Medline]
[Order article via Infotrieve]
|
5.
|
Pan, Y.,
Lloyd, C.,
Zhou, H.,
Dolich, S.,
Deeds, J.,
Gonzalo, J. A.,
Vath, J.,
Gosselin, M.,
Ma, J.,
Dussault, B.,
Woolf, E.,
Alperin, G.,
Culpepper, J.,
Gutierrez-Ramos, J. C.,
and Gearing, D.
(1997)
Nature
387,
611-617[CrossRef][Medline]
[Order article via Infotrieve]
|
6.
|
Imai, T.,
Hieshima, K.,
Haskell, C.,
Baba, M.,
Nagira, M.,
Kakizaki, M.,
Takagi, S.,
Nomiyama, H.,
Schall, T. J.,
and Yoshie, O.
(1997)
Cell
91,
521-530[Medline]
[Order article via Infotrieve]
|
7.
|
Fong, A. M.,
Robinson, L. A.,
Steeber, D. A.,
Tedder, T. F.,
Yoshie, O.,
Imai, T.,
and Patel, D. D.
(1998)
J. Exp. Med.
188,
1413-1419[Abstract/Free Full Text]
|
8.
|
Haskell, C. A.,
Cleary, M. D.,
and Charo, I. F.
(1999)
J. Biol. Chem.
274,
10053-10058[Abstract/Free Full Text]
|
9.
|
Harrison, J. K.,
Jiang, Y.,
Chen, S.,
Xia, Y.,
Maciejewski, D.,
McNamara, R. K.,
Streit, W. J.,
Salafranca, M. N.,
Adhikari, S.,
Thompson, D. A.,
Botti, P.,
Bacon, K. B.,
and Feng, L.
(1998)
Proc. Natl. Acad. Sci. U. S. A.
95,
10896-10901[Abstract/Free Full Text]
|
10.
|
Nishiyori, A.,
Masbumi, M.,
Ohtani, Y.,
Takami, S.,
Yamamoto, J.,
Kawaguchi, N.,
Kume, T.,
Akaike, A.,
and Satoh, M.
(1998)
FEBS Lett.
429,
167-172[CrossRef][Medline]
[Order article via Infotrieve]
|
11.
|
Schwaeble, W. J.,
Stover, C. M.,
Schall, T. J.,
Dairaghi, D. J.,
Trinder, P. K. E.,
Linington, C.,
Iglesias, A.,
Schubart, A.,
Lynch, N. J.,
Weihe, E.,
and Schafer, M. K. H.
(1998)
FEBS Lett.
439,
203-207[CrossRef][Medline]
[Order article via Infotrieve]
|
12.
|
Harrison, J. K.,
Jiang, Y.,
Wees, E. A.,
Salafranca, M.,
Liang, H.-X.,
Feng, L.,
and Belardinelli, L.
(1999)
J. Leukocyte Biol.
66,
937-944[Abstract]
|
13.
|
Kanazawa, N.,
Nakamura, T.,
Tashiro, K.,
Muramatsu, M.,
Morita, K.,
Yoneda, K.,
Inaba, K.,
Imamura, S.,
and Honjo, T.
(1999)
Eur. J. Immunol.
29,
1925-1932[CrossRef][Medline]
[Order article via Infotrieve]
|
14.
|
Muehlhoefer, A.,
Saubermann, L. J.,
Gu, X.,
Leudtke-Heckenkamp, K.,
Xavier, R.,
Blumberg, R. S.,
Podolsky, D. K.,
MacDermott, R. P.,
and Reinecker, H.-C.
(2000)
J. Immunol.
164,
3368-3376[Abstract/Free Full Text]
|
15.
|
Papadopoulos, E. J.,
Sassetti, C.,
Saeki, H.,
Yamada, N.,
Kawamura, T.,
Fitzhugh, D. J.,
Saraf, M. A.,
Schall, T.,
Blauvelt, A.,
Rosen, S. D.,
and Hwang, S. T.
(1999)
Eur. J. Immunol.
29,
2551-2559[CrossRef][Medline]
[Order article via Infotrieve]
|
16.
|
Papadopoulos, E. J.,
Fitzhugh, D. J.,
Tkaczyk, C.,
Gilfillan, A. M.,
Sassetti, C.,
Metcalfe, D. D.,
and Hwang, S. T.
(2000)
Eur. J. Immunol.
30,
2355-2361[CrossRef][Medline]
[Order article via Infotrieve]
|
17.
|
Mizoue, L. S.,
Bazan, J. F.,
Johnson, E. C.,
and Handel, T. M.
(1999)
Biochemistry
38,
1402-1414[CrossRef][Medline]
[Order article via Infotrieve]
|
18.
|
Fong, A. M.,
Erickson, H. P.,
Zachariah, J. P.,
Poon, S.,
Schamberg, N. J.,
Imai, T.,
and Patel, D. D.
(2000)
J. Biol. Chem.
275,
3781-3786[Abstract/Free Full Text]
|
19.
|
Haskell, C. A.,
Cleary, M. D.,
and Charo, I. F.
(2000)
J. Biol. Chem.
275,
34183-34189[Abstract/Free Full Text]
|
20.
|
Chapman, G. A.,
Moores, K.,
Harrison, D.,
Campbell, C. A.,
Stewart, B. R.,
and Strijbos, P. J. L. M.
(2000)
J. Neurosci.
20,
RC87[Medline]
[Order article via Infotrieve]
|
21.
|
Hoover, D. M.,
Mizoue, L. S.,
Handel, T. M.,
and Lubkowski, J.
(2000)
J. Biol. Chem.
275,
23187-23193[Abstract/Free Full Text]
|
22.
|
Hebert, C. A.,
Vitangcol, R. V.,
and Baker, J. B.
(1991)
J. Biol. Chem.
266,
18989-18994[Abstract/Free Full Text]
|
23.
|
Hemmerich, S.,
Paavola, C.,
Bloom, A.,
Bhakta, S.,
Freedman, R.,
Grunberger, D.,
Krstenansky, J.,
Lee, S.,
McCarley, D.,
Mulkins, M.,
Wong, B.,
Pease, J.,
Mizoue, L.,
Mirzadegan, T.,
Polsky, I.,
Thompson, K.,
Handel, T. M.,
and Jarnagin, K.
(1999)
Biochemistry
38,
13013-13025[CrossRef][Medline]
[Order article via Infotrieve]
|
24.
|
Palczewski, K.,
Kumasaka, T.,
Hori, T.,
Behnke, C. A.,
Motoshima, H.,
Fox, B. A.,
Le Trong, I.,
Teller, D. C.,
Okada, T.,
Stenkamp, R. E.,
Yamamoto, M.,
and Miyano, M.
(2000)
Science
289,
739-745[Abstract/Free Full Text]
|
25.
|
Kraulis, P. J.
(1991)
J. Appl. Crystallogr.
24,
946-950[CrossRef]
|
Copyright © 2001 by The American Society for Biochemistry and Molecular Biology, Inc.