Regulation of a Transient Receptor Potential (TRP)
Channel by Tyrosine Phosphorylation
SRC FAMILY KINASE-DEPENDENT TYROSINE PHOSPHORYLATION OF
TRPV4 ON TYR-253 MEDIATES ITS RESPONSE TO HYPOTONIC STRESS*
Hongshi
Xu
,
Hongyu
Zhao
,
Wei
Tian
,
Kiyotsugu
Yoshida§,
Jean-Baptiste
Roullet
, and
David M.
Cohen
¶
From the
Division of Nephrology, Department of
Medicine, and ¶ Department of Cell and Developmental Biology,
Oregon Health & Science University and the Portland Veterans Affairs
Medical Center, Portland, Oregon 97201 and the § Dana
Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts
02115
Received for publication, October 29, 2002, and in revised form, January 17, 2003
 |
ABSTRACT |
The recently identified transient receptor
potential (TRP) channel family member, TRPV4 (formerly known as OTRPC4,
VR-OAC, TRP12, and VRL-2) is activated by hypotonicity. It is highly
expressed in the kidney as well as blood-brain barrier-deficient
hypothalamic nuclei responsible for systemic osmosensing. Apart from
its gating by hypotonicity, little is known about TRPV4 regulation. We
observed that hypotonic stress resulted in rapid tyrosine
phosphorylation of TRPV4 in a heterologous expression model and in
native murine distal convoluted tubule cells in culture. This tyrosine
phosphorylation was sensitive to the inhibitor of Src family tyrosine
kinases, PP1, in a dose-dependent fashion. TRPV4 associated
with Src family kinases by co-immunoprecipitation studies and confocal
immunofluorescence microscopy, and this interaction required an intact
Src family kinase SH2 domain. One of these kinases, Lyn, was activated
by hypotonic stress and phosphorylated TRPV4 in an immune complex kinase assay and an in vitro kinase assay using recombinant
Lyn and TRPV4. Transfection of wild-type Lyn dramatically potentiated hypotonicity-dependent TRPV4 tyrosine phosphorylation
whereas dominant negative-acting Lyn modestly inhibited it. Through
mutagenesis studies, the site of tonicity-dependent
tyrosine phosphorylation was mapped to Tyr-253, which is conserved
across all species from which TRPV4 has been cloned. Importantly, point
mutation of Tyr-253 abolished hypotonicity-dependent
channel activity. In aggregate, these data indicate that hypotonic
stress results in Src family tyrosine kinase-dependent
tyrosine phosphorylation of the tonicity sensor TRPV4 at residue
Tyr-253 and that this residue is essential for channel function in this
context. This is the first example of direct regulation of TRP
channel function through tyrosine phosphorylation.
 |
INTRODUCTION |
Signaling by hypotonic stress and cell swelling has been well
studied in diverse models (reviewed in Ref. 1). Nonetheless, the sensor
for hypotonic stress, both at the single cell and organismal levels,
remains elusive. A member of the transient receptor potential (TRP)1 family of cation
channels has recently been described that fulfills multiple criteria
for an osmosensing protein. Based upon homology with OSM-9, an
osmosensing TRP-like channel expressed in Caenorhabditis elegans (2), the mammalian candidate osmosensing TRP channel family member, was cloned independently as OTRPC4 (3) and VR-OAC (4).
This protein, also identified as TRP12 and vanilloid receptor-related protein-2 in other contexts, was recently renamed TRPV4 (5).
TRPV4, as a member of the TRPV subfamily of TRP channels, shares close
homology with the following channels: 1) the vanilloid (capsaicin)
receptor and sensor of noxious heat, TRPV1; 2) the heat-sensitive
vanilloid receptor-related channel-1, TRPV2; 3) the calcium channels,
TRPV5 (formerly known as ECaC1 and CaT2) and TRPV6 (formerly known as
CaT1, ECaC2, and CaT-L) (all reviewed in Ref. 5 and references
therein); and 4) the very recently described temperature-sensitive
receptor, TRPV3 (6-8). TRPV4 forms nonselective cation channels that
are activated by changes in extracellular osmolarity; hypertonicity, in
contrast, inactivates the channel (3, 4).
Although TRPV4 is expressed at the mRNA level in multiple tissues
including liver, heart, lung, spleen, and adipose tissue, it is by far
most abundant in the kidney (3, 4). By in situ hybridization, renal expression was localized principally to the distal
convoluted tubule (3). Interestingly, TRPV4 expression was also
detected in the specific blood-brain barrier-deficient hypothalamic
nuclei responsible for systemic osmosensing and in the mechanosensing
hair cells of the inner ear (4). Functional and expression data
were both consistent with a role for TRPV4 in osmosensing at both the
single cell and organismal levels.
Apart from its gating by tonicity, little is known about TRPV4
regulation. We and others have shown that signaling events in response
to hypotonic stress are reminiscent of activation of a
receptor-tyrosine kinase (9-12). We observed that hypotonic stress
resulted in rapid tyrosine phosphorylation of TRPV4 in a heterologous
expression model and in native murine distal convoluted tubule cells in
culture. This tyrosine phosphorylation was sensitive to the inhibitor
of Src family cytoplasmic tyrosine kinases, PP1, in a
dose-dependent fashion. TRPV4 associated with Src family tyrosine kinases by co-immunoprecipitation studies and confocal immunofluorescence microscopy, and this interaction required an intact
SH2 domain. Lyn, a Src family kinase, was activated by hypotonic stress
and phosphorylated TRPV4 in an immune complex kinase assay and kinase
assay with recombinant proteins. Transfection of wild-type Lyn
dramatically potentiated hypotonicity-dependent TRPV4
tyrosine phosphorylation whereas dominant negative-acting Lyn modestly
inhibited it. Phosphorylation occurred principally at residue Tyr-253,
and point mutation of this residue abolished tonicity-dependent calcium entry. In aggregate, these data
indicate that Src family kinase-dependent tyrosine
phosphorylation of Tyr-253 regulates TRPV4 activation in response to hypotonicity.
 |
MATERIALS AND METHODS |
Cells and Cell Culture--
mIMCD3 cells were maintained and
passaged as previously described (13). The murine distal convoluted
tubule mDCT cell line (14), kindly provided by Peter Friedman, was
maintained in Dulbecco's modified Eagle's medium/F12 supplemented
with 10% fetal bovine serum.
Murine TRPV4 Cloning, Transient and Stable Transfection, and
Mutagenesis--
TRPV4 was cloned from murine kidney
poly(A)+ RNA via RT-PCR using the SuperScript
Preamplification System (Invitrogen) for first strand synthesis,
followed by amplification with TRPV4-specific primers, 5'-AGT ATG GCA
GAT CCT GGT GA-3' and 5'-GTA CAG TGG GGC ATC GTC C-3'. Amplified
fragment was TA-cloned into pcDNA3.1/V5-His-TOPO (Invitrogen) to
create an expression plasmid encoding a carboxyl-terminal V5-epitope-tagged murine TRPV4; identity was confirmed by sequencing (Core Facility, Vollum Institute for Advanced Biomedical Research). Following the demonstration of tyrosine phosphorylation of TRPV4, high
probability tyrosine phosphorylation sites were sought using ProSite
(us.expasy.org/prosite, Ref. 15) and NetPhos
(www.cbs.dtu.dk/services/NetPhos, Ref. 16; see "Results"). The
deduced amino acid sequence of the TRPV4 clone in the present study was
identical to that of the canonical sequence, NP_071300, with the
following exceptions: Leu instead of Arg at position 90, Thr instead of
Ile at position 389, and His instead of Tyr at position 860. Leu-90 was
previously reported in the context of murine TRP12 and murine VR-OAC.
Of note, the extreme carboxyl-terminal Tyr-860, present in NM_022017 and absent from our clone, was predicted to encode an extremely low-probability tyrosine phosphorylation site (probability 0.075, NetPhos). HEK293 cells were transfected with LipofectAMINE Plus in
accordance with the manufacturer's directions (Invitrogen). Stable
cell lines were derived from pooled clones propagated under selection
pressure with Gly-418. Lyn-WT and Lyn-DN were originally described as
pSR
-Lyn wt and pSR
-Lyn(K-R) (17). Individual point mutants of
TRPV4 were generated using the QuickChange mutagenesis system
(Stratagene), in accordance with the manufacturer's instructions, using PAGE-purified primers obtained from IDT. Multiple point mutants
(e.g. 5×-mutant) were generated through iterated
application of this approach. All mutations were confirmed by direct
sequencing, as above.
Antibodies, Immunoblotting, Immunoprecipitation, and Kinase
Assays--
Anti-peptide polyclonal antibody was raised in two rabbits
against a synthetic peptide representing the carboxyl-terminal sequence
of TRPV4 (CDG HQQ GYA PKW RTD DAP L) and then subjected to affinity
purification with the peptide antigen (Alpha Diagnostic International,
Inc., San Antonio, TX). Sequence analysis (BLAST search with peptide)
indicated that this sequence was not conserved across other TRP family
members including TRPV1 (VR-1), TRPV2 (VRL-1), TRPV5 (ECaC, CAT2),
TRPV6 (ECaC-2, Cat1). It was conserved across murine VRL-2, TRP12,
OTRPC4, and TRPV4, all of which are nearly identical at the amino acid
level and have been collectively designated TRPV4 (5). Other antibodies
were commercially available including anti-PY99 (Upstate
Biotechnology), anti-V5, anti-Lyn, anti-Src, anti-Fyn, anti-Lck,
anti-Hck, anti-Yes (Santa Cruz Biotechnologies). Immunoblot analysis
was performed as previously described (18). Cell monolayers were lysed
in Lysis Buffer (125 mM NaCl, 50 mM Tris (pH
7.5), 0.1% SDS, 0.5% sodium deoxycholate, 1% Nonidet P-40, 1 mM sodium orthovanadate, 1 mM
4-(2-aminoetryl)benzenesulfonyl fluoride, 1 µg/ml leupeptin, 1 µg/ml aprotinin, 1 µg/ml pepstatin A, 25 mM
-glycerophosphate, 2 mM sodium pyrophosphate) for 30 min
at 4 °C and immunoprecipitated with appropriate antibody and protein
A/G bound to Sepharose beads. Lyn immune complex kinase assay was
performed essentially as described by Yoshida et al. (17).
Briefly, monolayers were lysed in Lysis Buffer, immunoprecipitated with
anti-Lyn, washed, and subjected to kinase assay in Complete Kinase
Buffer (50 mM HEPES, pH 7.4, 2 mM
dithiothreitol, 10 mM MgCl2, 10 mM
MnCl2, 0.1 mM sodium orthovanadate, 20 µM ATP, 1% (v/v) [
-32P]ATP [250
µCi/ml], 75 ng/µl acid-activated enolase) for 30 min at 30 °C
prior to SDS/PAGE. Adult mouse kidney tissue blot was obtained from
Alpha Diagnostic International. For Lyn co-immunoprecipitation kinase
assay, monolayers of control or hypotonically stressed HEK293 cells
were lysed in Lysis Buffer, immunoprecipitated with anti-Lyn, washed
extensively, and then subjected to in vitro kinase assay in
Complete Kinase buffer for 30 min at 30 °C in the absence of
acid-activated enolase. In vitro TRPV4-directed LynA kinase assay was performed essentially as described by Chin et al.
(19). Briefly, in vitro transcribed and translated TRPV4 or
TRPV4 (see below), synthesized using the TNT kit
(Promega) in accordance with the manufacturer's instructions, was
immunoprecipitated with anti-V5 antibody. After washing, bead-bound
TRPV4 or
TRPV4 was incubated in In Vitro Kinase buffer (10 mM HEPES, pH 7.5, 50 mM NaCl, 5 mM
MgCl2, 5 mM MnCl2, 0.1 mM sodium orthovanadate, 20 µM ATP, 5% v/v
[
-32P]ATP [250 µCi/ml], and 0.36 µg/µl LynA
[Calbiochem]) for 30 min at 30 °C.
TRPV4 (murine TRPV4 absent
membrane-spanning domains 2 through 6) was prepared through
Bsu36I restriction digestion of TRPV4-pcDNA3.1/V5-His-TOPO
and self-ligation following gel purification of the larger fragment. In
some experiments (e.g. anti-V5 immunoprecipitation followed
by anti-Src family protein-tyrosine kinase immunoblotting), anti-V5 was
covalently cross-linked to protein A/G-Sepharose (Amersham Biosciences)
using dimethyl pimelimidate (20) to eliminate the secondary antibody
detection of immunoglobulin heavy chain, which co-migrates with many
Src family protein-tyrosine kinases.
Intracellular Calcium Measurement--
Intracellular
[Ca2+]i measurements were
conducted on cell suspensions obtained by trypsinization of confluent
cell monolayers of wild-type and TRPV4 stably transfected HEK293 cells. The suspended cells were loaded with Fura-2-AM, washed twice with HEPES-buffered saline solution (HBSS: 130 mM NaCl, 5 mM KCl, 1 mM MgSO4, 1 mM NaHPO4, 2 mM CaCl2,
10 mM HEPES, pH 7.4), and assayed for intracellular calcium
concentration ([Ca2+]i) in a
cuvette under constant, gentle stirring (2-ml final volume).
Fluorescent emission was monitored at 510 nm with alternate excitation
at 340 and 380 nm using a Hitachi F2000 fluorescence spectrophotometer
(Hitachi Instruments, Inc., Naperville, IL). Calibration of the Fura-2
signal was performed as previously described (21) using
Fura-2·Ca2+ dissociation constant of 224 nM
(22).
Immunofluorescence Microscopy--
Monolayers were grown on
LabTek Chamber slides (Nalgene Nunc) and labeled with primary antibody
(anti-Lyn (Santa Cruz Biotechnology sc-15), anti-Src (Santa Cruz
Biotechnology sc-19), or anti-V5 (Invitrogen 46-0705)) and secondary
antibody (AlexaFluor 594 goat anti-rabbit IgG (Molecular Probes
A-11037) or AlexaFluor 488 goat anti-mouse IgG (Molecular Probes
A-11001) prior to confocal epifluorescence imaging. Studies were
performed on wild-type HEK293 cells and on HEK293 cells stably
transfected with TRPV4. The affinity-purified anti-TRPV4 antibody was
insufficiently sensitive in the immunofluorescence assay to permit
examination of the native (mDCT) system. For confocal imaging,
consecutive sections 500 nm apart, bracketing the center of the cell,
were scanned alternating between 488 and 568 nm using a Leica TCS SP
confocal laser scanning microscope. System settings were held constant
for all imaging. Images were digitally captured and then deconvolved
using Power HazeBusterTM imaging software (VayTek, Inc.,
Fairfield, VA). All imaging analysis determining colocalization was
carried out using software by IPLabTM (Scanalytics Inc.,
Fairfield, VA).
 |
RESULTS |
Because tyrosine phosphorylation events may accompany hypotonic
stress, we sought to determine whether hypotonicity resulted in
tyrosine phosphorylation of the tonicity-responsive TRPV4. V5-epitope-tagged TRPV4 was stably overexpressed in HEK293 cells, which
were reported to lack this channel (3). By anti-phosphotyrosine immunoblotting of anti-V5 immunoprecipitates, hypotonic stress (150 mosmol/kg H2O) resulted in pronounced and transient
up-regulation of TRPV4 tyrosine phosphorylation (Fig.
1A). This phenomenon was specific to hypotonic stress as it was not observed in the presence of
treatment with the peptide growth factor, epidermal growth factor, or
the activator of classical protein kinase C,
12-O-tetradecanoylphorbol-13-acetate (TPA) (Fig.
1B). There was no effect upon total TRPV4 abundance in these
experiments (data not shown). Two other solute stressors, hypertonic
stress and urea stress, both failed to increase anti-phosphotyrosine immunoreactivity in this model (data not shown). Of note, additional less intense bands migrating more slowly than the principal band corresponding to newly tyrosine-phosphorylated TRPV4 were frequently observed in this heterologous expression model. Treatment of lysates with endoglycosidase-F resulted in the complete disappearance of the
higher molecular mass bands and enhancement of the principal lower
molecular mass band (Fig. 1C). These data were consistent with the presence of forms of TRPV4 exhibiting varying degrees of
glycosylation.

View larger version (34K):
[in this window]
[in a new window]
|
Fig. 1.
TRPV4 is tyrosine-phosphorylated in response
to hypotonic stress. Anti-phosphotyrosine immunoblots of anti-V5
immunoprecipitates prepared from TRPV4 stable transfectants of HEK293
and subjected to the indicated duration of hypotonic stress (150 mosmol/kgH2O (A) or exposure to other cell
activators (EGF, 100 ng/ml × 30 min or the phorbol
ester, TPA, 100 nM × 30 min (B).
C, effect of incubation with the indicated concentration of
endoglycosidase-F upon electrophoretic mobility of TRPV4.
NS, non-specific.
|
|
We next sought to confirm that: 1) stable heterologous transfection of
epitope-tagged TRPV4 resulted in expression of functional TRPV4 and
thereby mimicked endogenous expression, and 2) addition of the epitope
tag did not interfere with channel function. Calcium transients in
response to hypotonicity were examined in untransfected and
TRPV4-transfected HEK293 cells. In both untransfected and TRPV4-transfected cells, there was negligible calcium entry following sham (isotonic) treatment (Fig.
2A). In response to hypotonic stress (150 mosmol/kg H2O), there was a robust response in
the TRPV4-transfected cells but virtually no response in the wild-type cell line. Next, to confirm that the calcium transients required calcium entry, similar studies were performed with only the
TRPV4-transfected cell line in the presence and absence of
extracellular calcium (Fig. 2B). The response to hypotonic
stress was noted again, in the presence of extracellular calcium
([Ca2+]o = 1.25 mM)
but was completely absent when calcium was omitted from the assay
buffer ([Ca2+]o ~0
mM). However, the hypotonicity-dependent,
intracellular calcium signal was restored upon addition of calcium
(CaCl2, 1.25 mM) to the cuvette. In aggregate,
these data indicated that the heterologous expression model
functionally mimicked the native system.

View larger version (15K):
[in this window]
[in a new window]
|
Fig. 2.
Heterologously expressed TRPV4 is a
functional calcium channel. A, effect of control or
hypotonic treatment upon intracellular calcium concentration in
untransfected, wild-type HEK293 cells (HEKwt) or
TRPV4-transfected HEK293 cells (TRPV4-HEK). Cells were loaded with
Fura-2/AM as described under "Materials and Methods," washed, and
concentrated by centrifugation (~2 × 107 cells/ml).
An aliquot (100 µl) of this cell suspension was injected in the assay
cuvette containing 1,900 µl of either isotonic (-Iso) or
hypotonic (-Hypo) HBSS (final osmolality = 150 mOsm/kg), and fluorescence was immediately recorded.
Hypotonicity-induced calcium signals were very modest in wild-type
HEK293 but robust in TRPV4 stable transfectants. B, role of
extracellular calcium in hypotonicity-induced calcium transients in
TRPV4-transfected HEK293 cells. Cells, prepared as in A,
were subjected to hypotonic stress in the presence or absence of
extracellular calcium (1.25 mM). In Ca2+-free
([Ca2+]o ~0 mM),
hypotonic HBSS, no intracellular calcium signal is observed.
Nonetheless, the channel is activated as indicated by the rapid rise in
[Ca2+]i upon addition of calcium
to the cuvette ([Ca2+]o = 1.25 mM).
|
|
To demonstrate that tyrosine phosphorylation of TRPV4 in response to
hypotonic stress occurred in a native system, in a fashion similar to
the HEK293-based heterologous expression system, a rabbit polyclonal,
affinity-purified anti-peptide antibody was generated against the
carboxyl terminus of murine TRPV4 (see "Materials and Methods").
The specific peptide was chosen to avoid cross-reactivity with closely
related members of the TRP channel family (Fig.
3A); VRL-2, OTRPC4, VR-OAC,
and TRP12 are names independently assigned to the protein now
designated TRPV4 (5). Murine renal tissue was screened for anti-TRPV4
immunoreactivity. Expression was detected in whole kidney lysate (Fig.
3B), most of which appeared to be conferred by expression in
the outer cortex. The distal convoluted tubule, where TRPV4 mRNA
had previously been detected (3), is confined to the outer cortex.
Several cell lines were also screened for the presence of TRPV4 by
anti-TRPV4 immunoblotting (Fig. 3C). Consistent with the
reports of others, anti-TRPV4 immunoreactivity was absent in 3T3 and
HEK293 cells. It was, however, present, in the renal distal convoluted
tubule cell line (14), mDCT, and in the renal inner medullary cell
line, mIMCD3. As expected, abundant TRPV4 expression was detected in a
HEK293 cell line stably transfected with TRPV4. To assess utility of
the anti-TRPV4 antiserum in immunohistochemistry, and to confirm that
the TRPV4 overexpression system exhibited membrane-associated
immunoreactivity, anti-TRPV4 immunofluorescence microscopy was
performed with wild-type HEK293 cells and with the TRPV4/HEK stable
cell line studied in Fig. 2. Anti-TRPV4 confocal immunofluorescence
microscopy demonstrated modest nonspecific staining in wild-type HEK293
cells (Fig. 3D) and robust, primarily cell
membrane-associated, expression in TRPV4/HEK stable transfectants (Fig.
3D). The effect of hypotonicity upon TRPV4 tyrosine
phosphorylation was next examined in the native model of the mDCT cell
line. Hypotonic stress (~ 150 mosmol/kg H2O) resulted in
robust time-dependent tyrosine phosphorylation of TRPV4 in
this model (Fig. 3E), confirming that neither heterologous expression nor, potentially, overexpression, was required for this
phenomenon.

View larger version (48K):
[in this window]
[in a new window]
|
Fig. 3.
Tonicity-dependent tyrosine
phosphorylation of TRPV4 is evident in a native system.
A, alignment of carboxyl termini of TRPV family members
(c, C. elegans; h, human;
m, murine; r, rat) with TRPV4 (formerly known as
VRL-2, OTRPC4, VR-OAC, or TRP12) depicting location of synthetic
peptide to which anti-TRPV4 polyclonal antibody was raised.
B, immunodetection of TRPV4 in murine kidney tissue lysates
prepared from whole kidney (K), outer cortex
(OC), inner cortex (IC), medulla (M),
and papilla (P). C, anti-TRPV4 immunoblot of
whole cell lysates prepared from the indicated untransfected
(wild-type) cell types, as well from TRPV4-HEK293 stable transfectants.
D, confocal microscopy of subconfluent monolayers of
wild-type HEK293 cells (left) and TRPV4/HEK293 stable
transfectants (right) labeled with anti-TRPV4 and
AlexaFluor-594-conjugated anti-rabbit secondary antibody (see
"Materials and Methods"). E,
anti-phosphotyrosine immunoblot of anti-TRPV4 immunoprecipitates
prepared from a murine distal convoluted tubule cell line (mDCT)
treated for the indicated interval with hypotonic stress.
|
|
To ascertain in preliminary fashion the nature of this tyrosine
phosphorylation event, a panel of tyrosine kinase inhibitors was
applied prior to hypotonic stress. The general tyrosine kinase inhibitor, genistein, only modestly inhibited the effect of
hypotonicity whereas the Src family kinase-specific inhibitor, PP1, was
more effective. In addition, this effect was dose-dependent
(Fig. 4). Of note, piceatannol, which is
reportedly specific for Syk (23), a related cytoplasmic
protein-tyrosine kinase not belonging to the Src family (reviewed in
Ref. 24), failed to influence tonicity-dependent tyrosine
phosphorylation. These data suggested in a preliminary fashion that
tonicity-dependent activation of a Src family kinase may be
responsible for TRPV4 tyrosine phosphorylation.

View larger version (25K):
[in this window]
[in a new window]
|
Fig. 4.
Effect of hypotonicity is inhibited by
PP1. Anti-phosphotyrosine immunoblot of anti-V5 immunoprecipitates
prepared from TRPV4-HEK293 stable transfectants subjected to control
treatment or hypotonic stress for 30 min, in the presence or absence of
the tyrosine kinase inhibitors, genistein (100 µM), PP1
(100 nM/1 µM), or piceatannol (30 µM).
|
|
To confirm our inference from the inhibitor data, we first sought a
physical association between TRPV4 and members of the Src family of
tyrosine kinases. For these studies, the HEK293 heterologous expression
model was used primarily because of the clean background afforded by
the commercially available monoclonal anti-V5 antibody. (In most
experiments, this antibody was covalently linked to Sepharose beads for
immunoprecipitation because of the similarity in molecular mass between
Src family protein-tyrosine kinases and immunoglobulin heavy chain.)
Multiple Src family members were immunodetectable in the anti-V5
(anti-TRPV4) immunoprecipitates (Fig.
5A); the association between
TRPV4 and both Lyn and Yes increased with duration of hypotonic stress.
Of note, on immunoblots with anti-Src, a faint band co-migrating with
bona fide Src was evident following anti-V5
immunoprecipitation even in untransfected HEK cells. The three
antibodies in this panel are reported to be free from cross-reactivity
with other Src family kinases in murine cells. Reciprocal experiments
were performed (Fig. 5B) wherein lysates prepared from
untransfected and V5-TRPV4-transfected HEK293 cells were
immunoprecipitated with anti-Src family kinase antibodies and then
immunoblotted with anti-V5. TRPV4 interaction with Lyn, Src, Fyn, Hck,
Lck, and Yes was detected in this fashion. Only Lyn exhibited
time-dependent interaction with TRPV4 such that the anti-V5
(i.e. anti-TRPV4) immunoreactivity in anti-Lyn immunoprecipitates was much stronger at 30 min than at time zero. These
data suggested a physical interaction between TRPV4 and the Src family
tyrosine kinases.

View larger version (52K):
[in this window]
[in a new window]
|
Fig. 5.
TRPV4 co-immunoprecipitates with Src family
tyrosine kinases. A, anti-V5 immunoprecipitates
prepared from wild-type HEK293 cells and TRPV4-HEK293 stable
transfectants subjected to hypotonic stress for the indicated interval,
and then immunoblotted for the indicated Src family tyrosine kinase.
B, anti-V5 immunoblot of immunoprecipitates prepared from
wild-type HEK293 cells and TRPV4-HEK293 stable transfectants; cells
were subjected to hypotonic stress for the indicated duration and then
immunoprecipitated with antibodies specific for the indicated Src
family protein-tyrosine kinase.
|
|
To further support a physical interaction between these proteins,
confocal immunofluorescence microscopy was performed. In HEK293 cells
stably transfected with TRPV4, near total co-localization of TRPV4 with
Lyn was observed (Fig. 6). This
co-localization, however, was unaffected by up to 30 min of exposure to
hypotonicity. In contrast, only a small fraction of immunodetectable
TRPV4 exhibited co-localization with Src (data not shown).

View larger version (84K):
[in this window]
[in a new window]
|
Fig. 6.
TRPV4 and Lyn co-localize by confocal
microscopy. TRPV4-transfected HEK293 cells were subjected to
confocal imaging analysis after immunofluorescence staining with
anti-Lyn and anti-V5. Panel A depicts Lyn
immunolocalization, panel B is TRPV4 localization, and
panel C is a merging of the Lyn and V5-TRPV4 images showing
co-localization. A, B, and C are
corresponding individual layers (1 µm); D represents the
sum of all merged layers.
|
|
We next sought to confirm the interaction in the native mDCT system and
to determine in preliminary fashion the Src family tyrosine kinase
motif conferring interaction with TRPV4. Fyn was selected for these
studies because of its constitutive interaction with TRPV4 by
immunoprecipitation and because of the availability of agarose
bead-bound deletion mutants of this kinase. Murine Fyn is a 534 (or 537 for FynT) amino acid Src family protein-tyrosine kinase containing
three well described structural motifs: an SH3 domain (amino acids
~87-141), an SH2 domain (amino acids ~149-231), and a
carboxyl-terminal kinase domain (amino acids ~268-517). Agarose-conjugated SH3-SH2 from Fyn (absent the kinase domain) effectively affinity-precipitated TRPV4 from both the heterologous expression (V5-TRPV4-HEK293, Fig.
7A) and the native (mDCT, Fig. 7B) models. In both models, agarose-conjugated Fyn SH2 alone
(Fyn-(145-247)) but not agarose-conjugated Fyn SH3 alone
(Fyn-(85-139)) reproduced this effect (Fig. 7C). These data
suggested that the TRPV4-Src family tyrosine kinase interaction was
mediated via the SH2 domain of the latter, presumably in conjunction
with one or more cognate phosphotyrosine motifs on TRPV4.

View larger version (46K):
[in this window]
[in a new window]
|
Fig. 7.
TRPV4 associates with Src family kinases via
SH2 domain. A, lysates prepared from control-treated
TRPV4-HEK293 stable transfectants, affinity-precipitated with agarose
bead-bound Fyn-(85-247) or control protein A/G-bound beads, and then
immunoblotted with anti-V5 to detect the presence of V5-TRPV4.
(Fyn-(85-247) encodes the SH3-SH2 domains of Fyn.) B,
lysates prepared from wild-type mDCT cells treated for the indicated
interval with hypotonic stress, affinity-precipitated with agarose
bead-bound Fyn-(85-247) or control protein A/G-bound beads, and then
immunoblotted with anti-TRPV4. C, lysates prepared from
wild-type mDCT cells (upper panel) and TRPV4-HEK293 stable
transfectants (lower panel) treated for the indicated
interval with hypotonic stress, affinity-precipitated with agarose
bead-bound Fyn-(85-139) or Fyn-(145-247), and then immunoblotted with
anti-TRPV4. Fyn-(85-139) encodes the SH3 domain and Fyn-(145-247)
encodes the SH2 domain of Fyn.
|
|
The ability of Src family tyrosine kinases to phosphorylate TRPV4 was
next examined. HEK293 cells stably expressing V5-epitope-tagged TRPV4
were transiently transfected with wild-type Lyn, dominant negative-acting Lyn, or with vector alone. TRPV4 was immunoprecipitated with anti-V5 and phosphorylation assessed by anti-phosphotyrosine immunoblotting. Overexpression of Lyn resulted in a marked increase in
TRPV4 tyrosine phosphorylation under control conditions, which was even
more evident in the presence of hypotonic stress (Fig. 8). These data suggested that Lyn can
phosphorylate TRPV4 in intact cells. Dominant negative-acting Lyn
modestly but reproducibly suppressed tonicity-dependent
phosphorylation of TRPV4 but did not eliminate it entirely, suggesting
that Lyn is perhaps a principal kinase involved in this process. These
data could not, however, demonstrate whether this effect was direct or
indirect.

View larger version (24K):
[in this window]
[in a new window]
|
Fig. 8.
Transfection of Lyn enhances and dominant
negative-acting Lyn inhibits tonicity-dependent TRPV4
tyrosine phosphorylation. TRPV4-HEK293 stable transfectants were
transiently transfected with the indicated plasmid and subjected to
control treatment or hypotonic stress (150 mosmol/kgH2O × 30 min) prior to anti-V5
immunoprecipitation and anti-phosphotyrosine immunoblotting. The
discontinuity between lanes 2 and 3 reflects the
removal of several intervening lanes; all depicted lanes are taken from
the same exposure of the same autoradiograph.
|
|
We performed in vitro kinase assays to determine whether Lyn
can directly phosphorylate TRPV4. We first confirmed that Lyn was
activated in our model by hypotonic stress using an anti-Lyn immune
complex kinase assay with acid-treated enolase as a substrate (Fig.
9A). In a different type of
immunoprecipitation kinase assay, Lyn was immunoprecipitated (in
conjunction with its associated proteins) from control and
hypotonically stressed cells and then subjected to an in
vitro kinase assay in the presence of [
-32P]ATP,
cold ATP, Mg2+, and Mn2+. Up-regulated
phosphorylation of a band of the appropriate size for TRPV4 was evident
in the anti-Lyn immunoprecipitates from hypotonically stressed cells at
30 min of treatment (Fig. 9B). To exclude the possibility
that a kinase co-immunoprecipitating with Lyn or TRPV4 was responsible
for the TRPV4-directed kinase activity, similar experiments were
performed using in vitro transcribed and translated TRPV4
and recombinant LynA (Calbiochem). To facilitate expression in this
model and to produce a more soluble protein product, 5 of 6 transmembrane-spanning domains were deleted from murine TRPV4 cDNA
to generate
TRPV4 (see "Materials and Methods"). No candidate
tyrosine phosphorylation sites were present in this domain of TRPV4.
Recombinant Lyn phosphorylated the in vitro transcribed and
translated TRPV4 and
TRPV4 producing bands precisely co-migrating with immunodetectable TRPV4 and
TRPV4 (Fig. 9C).
Importantly, no substrate phosphorylation was evident in the absence of
exogenous Lyn (data not shown). These data suggested that in addition
to associating with Lyn, TRPV4 serves as a bona fide Lyn
substrate.

View larger version (47K):
[in this window]
[in a new window]
|
Fig. 9.
Lyn is activated by hypotonic stress and
phosphorylates TRPV4. A, autoradiogram of immune
complex kinase assay of anti-Lyn immunoprecipitates prepared from
control and hypotonically treated (150 mosmol/kgH2O × 15 min) HEK293; arrowhead denotes in vitro
phosphorylated Lyn substrate (enolase). B,
co-immunoprecipitation kinase assay of anti-Lyn immunoprecipitates
prepared from TRPV4-HEK293 stable transfectants; immunoprecipitates
were subjected to kinase assay in absence of exogenous substrate.
Phosphorylation of band co-migrating with TRPV4 is detected following
hypotonic treatment. C, in vitro kinase assay
with recombinant LynA and in vitro transcribed/translated
TRPV4 or TRPV4 (a TRPV4 deletion mutant lacking five of six
membrane-spanning domains to facilitate expression). A specifically
phosphorylated band, co-migrating with either immunodetectable TRPV4 or
TRPV4 was evident. No phosphorylation, specific or nonspecific, was
noted in the absence of LynA (data not shown).
|
|
We next sought to identify the site of the tyrosine phosphorylation in
TRPV4 through site-directed mutagenesis studies. Preliminary sequence
analysis of mTRPV4 using a standard protein motif search (ProSite,
www.expasy.ch/prosite/) detected a single canonical ((RK)X2(DE)X3Y or
(RK)X3(DE)X2Y, Refs.
25-27) tyrosine phosphorylation site at amino acid Tyr-411. Sequence
alignment indicated conservation of the site across all 4 species in
which TRPV4 had been cloned. Therefore, of the 36 tyrosines present in
VR-OAC, we performed site-directed mutagenesis to replace this residue
with phenylalanine (Y411F). When cells were transiently
transfected with this mutant (Y411F-TRPV4), however, tyrosine
phosphorylation of TRPV4 was as robust as in cells transfected with the
wild-type TRPV4 (data not shown).
Subsequent analyses were performed to reconcile the inability of the
Y411F mutant to abrogate tyrosine phosphorylation of TRPV4. Guided by a
recently described (16) neural net-based contextual amino acid
phosphorylation site identification shareware (NetPhos 2.0, www.cbs.dtu.dk/services/NetPhos/), we evaluated the likelihood of
encountering regulated tyrosine phosphorylation at each of the 36 tyrosines in TRPV4. Interestingly, the single "classical" tyrosine
phosphorylation motif in this protein (upon which we based our initial
mutagenesis approach, Tyr-411) was actually a suboptimal candidate for
phosphorylation in vivo because of an adjacent bulky
tryptophan residue rendering the tyrosine sterically inaccessible.
Eight other sites were identified with a high likelihood of undergoing
regulated tyrosine phosphorylation. Based upon conservation across
species in which TRPV4 has been cloned and upon predicted intracellular
localization, we selected five sites for mutagenesis. The 5×-mutant
TRPV4 construct included the following mutations (numbered according to
accession number NP_071300, the murine consensus TRPV4): Y253F, Y281F,
Y415F, Y508F, and Y805F. Hypotonicity-inducible tyrosine
phosphorylation was evident in anti-V5 immunoprecipitates of mDCT cells
transiently transfected with wild-type TRPV4 but not in cells
transfected with the 5×-mutant (Fig.
10A, upper
panel). Background nonspecific immunoreactivity co-migrating with
TRPV4 (Fig. 10A) precluded detailed analysis of the tyrosine
residues undergoing phosphorylation so an additional model system was
sought. The COS7 cell line, in which essentially no co-migrating
background was detectable, was selected for further study. Consistent
with the mDCT model, mutagenesis of the 5 key tyrosine residues of
TRPV4 prevented hypotonicity-dependent phosphorylation in
the COS7 cells (Fig. 10A, lower panel).

View larger version (54K):
[in this window]
[in a new window]
|
Fig. 10.
Hypotonicity results in phosphorylation of
Tyr-253 of TRPV4. A, anti-PY99 immunoreactivity
(tyrosine phosphorylation) of anti-V5 immunoprecipitates prepared from
untransfected mDCT cells (None), or mDCT cells transiently
transfected with either wild-type (+TRPV4) or 5× tyrosine
mutant (5×-mut) TRPV4 (upper panel); the lower
panel depicts the identical experiment performed using stable
transfectants in COS7 cells. B, anti-PY99 immunoreactivity
(tyrosine phosphorylation; upper panel) and anti-V5
immunoreactivity (lower panel) of anti-V5 immunoprecipitates
prepared from COS7 cells stably transfected with an expression vector
encoding wild-type TRPV4 (WT) or the indicated
tyrosine-directed Tyr Phe point mutant of TRPV4, and then subjected
to the presence (+) or absence ( ) of hypotonicity. The smear in the
left-most lane of the upper panel indicates
molecular mass markers. C, alignment of murine
(m), rat (r), and human (h) TRPV4 with
TRPV1-TRPV6 (murine clones, where available), and C. elegans
OSM-9 showing restriction of Tyr-253 to TRPV4.
|
|
A series of stable COS7 cell lines expressing single Tyr
Phe
mutants of TRPV4 was generated and compared with cells expressing wild-type TRPV4 (Fig. 10B). Although there was some
variability in the level of expression of each of the mutants, it was
clear that Tyr-253 accounted for the vast majority of regulated (and constitutive) tyrosine phosphorylation of TRPV4. Interestingly, Tyr-253
is conserved across all species from which TRPV4 has been cloned
(mouse, rat, and human), yet is not shared by any of the other TRPV
family members or the prototypical C. elegans channel, OSM-9
(Fig. 10C). In addition, it is not shared by any members of
the TRPC or TRPM families (data not shown).
Further studies were carried out to determine the importance of the
putative tyrosine phosphorylation sites in TRPV4 function in response
to hypotonicity. Wild-type HEK293 cells were compared with cells stably
transfected with TRPV4 or with TRPV4 harboring mutations (Tyr
Phe)
in the five key tyrosine residues (Fig. 11A). Again, a robust
increase in intracellular calcium was observed in TRPV4-transfected
cells but not in untransfected HEK cells. Importantly, the 5×-mutant
transfectants essentially lacked hypotonicity responsiveness, with a
response indistinguishable from that of untransfected HEK. This effect
was not a consequence of lack of expression of the mutant protein or
aberrant targeting of the mutant protein because immunodetectable TRPV4
was evident in both whole cell lysates and membrane preparations from
the 5×-mutant cell line (Fig. 11A). Because Tyr-253 was the
principal target of tyrosine phosphorylation in response to hypotonic
stress based upon our mutagenesis data (Fig. 10), we next assessed the
effect of the single Tyr-253 point mutant upon TRPV4 channel function (Fig. 11B). The Y253F mutant, similar to the 5×-mutant,
exhibited negligible hypotonicity-dependent calcium
response. Again, TRPV4 expression and membrane targeting were
comparable to the overexpressed wild-type TRPV4 as demonstrated through
immunoblotting and preparation of membrane and cytosolic proteins (Fig.
11B). In addition, the subcellular distribution of the
heterologously expressed Tyr-253 was indistinguishable from that of the
heterologously expressed wild-type TRPV4 (data not shown). These data
indicate that Tyr-253 is essential not only for tyrosine
phosphorylation of TRPV4 but also for function of TRPV4 in response to
hypotonic stress; they are also consistent with the observation of
Liedtke et al. (4) and Watanabe et al. (28) who
noted a decrease in the hypotonicity responsiveness of mutant TRPV4
proteins lacking much of the amino terminus. Of note, mutation of a
different single tyrosine among the five principal candidates
(Tyr-281), upon stable transfection of the resultant plasmid, failed to
influence hypotonicity-dependent calcium transients (data
not shown), further underscoring the specificity of the Tyr-253
effect.

View larger version (23K):
[in this window]
[in a new window]
|
Fig. 11.
Mutation of Tyr-253 abolishes
hypotonicity-dependent calcium transients.
A, HEK293 cells (HEK) or HEK cells stably transfected with
TRPV4, or TRPV4 in which the five principal putative tyrosine
phosphorylation sites have been mutated (5×-mut-TRPV4), were subjected
to isotonicity or hypotonic stress and the effect upon intracellular
calcium concentration was determined ratiometrically. The filled
arrowhead indicates the time of addition of the cells to the
cuvette; the transient upward and downward deflections noted at this
time are artifacts of chamber opening. Expression of TRPV4 in
whole-cell extracts and in cell membrane preparations was assessed via
anti-TRPV4 immunoblotting. B, studies similar to those
performed in A were performed with HEK cells stably
transfected with either TRPV4 or the point mutant Y253F-TRPV4. The
effect of hypotonicity was virtually abolished (i.e.
indistinguishable from untransfected HEK cells) by the point mutation.
Whole-cell and membrane-associated expression were again confirmed
through immunoblotting.
|
|
 |
DISCUSSION |
To the best of our knowledge, these are the first data
demonstrating regulated tyrosine phosphorylation of a TRP channel or its association with a Src family protein-tyrosine kinase. Moreover, they are the first to describe regulation of function of a TRP channel
through direct tyrosine phosphorylation. We observed the physical and
functional interaction between TRPV4 and Src family kinases in both the
heterologous overexpression model and in the native expression model
afforded by the murine distal convoluted tubule cell line. We show that
TRPV4 becomes newly tyrosine-phosphorylated in response to hypotonic
stress, and that point mutation of the tyrosine residue conferring
essentially all of the de novo phosphorylation virtually
abolishes tonicity-dependent calcium mobilization.
In contrast to the present model of direct regulation, several TRP
family members are indirectly regulated by upstream activation of a
receptor-tyrosine kinase, via intermediary action of a second messenger
system. For example, TRPM7 is indirectly inactivated upon
receptor-tyrosine kinase activation, an effect mediated through phospholipase C activation and PIP2 hydrolysis (29). TRPC4 and TRPC5,
in contrast, are activated in response to receptor-tyrosine kinase
activation (30). TRPC3, upon heterologous expression in chicken
B-cells, is activated in a Syk-dependent fashion, although the actual effector appears to be phospholipase C
(31).
Although kinase activation and the role of kinase-dependent
signaling in anisotonicity has been extensively studied in diverse models (reviewed in Ref. 1), comparatively fewer data describe the role
of tyrosine kinase activity in this process. In cell culture models,
tyrosine kinase activity is up-regulated by hypotonic stress (9, 10)
and is required for immediate-early gene transcription and expression
(10, 32). Pharmacological inhibition of tyrosine kinase activity
blocks, and potentiation of tyrosine kinase activity inhibits,
regulatory volume decrease-associated efflux of cations (9), chloride
(33-39), and the amino acid osmolyte, taurine (39, 40).
Several cytoplasmic protein-tyrosine kinases have previously been
implicated in the hypotonic stress response. In a marine elasmobranch
model of hypotonic stress, Syk- and Lyn-phosphorylated band 3, an
efflux pathway for the osmolyte, taurine (41). In lymphocytes, lck may
mediate activation of swelling-induced chloride efflux, based in part
upon experiments using p56lck
/
mice (35). Lastly, in
cells derived from mammalian kidney, chloride channel activity is
activated by fyn, although the dependence of this phenomenon upon
anisotonicity, per se, remains unexplored (42). Our data
extend the range of substrates phosphorylated by Src family kinases in
response to hypotonicity.
It is of interest that of the 36 tyrosine residues in murine TRPV4, a
single residue (Tyr-411) was identified through a standard approach
(ProSite. www.expasy.ch/prosite/) as constituting a candidate phosphorylation site. Mutation of this site, however, failed to influence hypotonicity-dependent tyrosine phosphorylation
of TRPV4. A neural net-derived, context-based approach (NetPhos 2.0, www.cbs.dtu.dk/services/NetPhos, Ref. 16) permitted us to identify the
eight most likely candidate tyrosine phosphorylation sites, from which
five were selected for further study based upon sequence conservation
and intracellular location. Interestingly, Tyr-411 was not among these
sites. Systematic mutagenesis identified Tyr-253 as the principal site
of regulated phosphorylation (Fig. 10B). Consistent with a
role in TRPV4 signal transduction in response to hypotonicity, Tyr-253
is conserved across all species from which TRPV4 has been cloned and is
not shared by any other members of the TRPV family, or by the
prototypical C. elegans osmosensing channel, OSM-9 (Fig.
10C). The apparent requirement for an intact SH2 domain for
the Src family protein-tyrosine kinase
TRPV4 interaction
suggests
perhaps a constitutive level of tyrosine phosphorylation of this
protein, which is supported by our anti-phosphotyrosine immunoblot
analyses. In addition, the up-regulation in Lyn co-immunoprecipitation
with TRPV4 with increasing duration of hypotonic stress suggests a more
regulated interaction for this kinase and the possible presence of a
positive feedback loop.
The structural role of Tyr-253 in TRPV4 channel gating remains unclear.
Tyr-253 resides within the first of three putative ankyrin repeat
domains in the TRPV4 amino terminus. Liedtke et al. (4)
showed that elimination of the amino-terminal amino acids 1-402 of
TRPV4 (including all three ankyrin repeat domains) decreased
tonicity-responsiveness of TRPV4. In a similar fashion, Watanabe
et al. (28) showed that deletion of residues 235-398 dramatically decreased TRPV4 calcium channel activity in response to
heat. These data are in excellent agreement with our assignment of a
pivotal role to Tyr-253.
TRPV4 is expressed predominantly in the kidney and much of this
expression is confined to the distal convoluted tubule (3). We noted
expression in a cell line derived from the renal medulla, as well. In
mammals, the distal nephron is intimately involved with both local and
systemic osmoregulation. It is tempting to speculate that TRPV4 is
instrumental in this process by responding to changes in ambient
tonicity in this tightly regulated epithelium. Whether TRPV4 serves to
"sense" osmotic changes, as has been proposed by others (3, 4), or
whether it also serves as an effector of anisotonicity,
through post-translational modification by Src family receptor-tyrosine
kinases, remains unknown.
 |
ACKNOWLEDGEMENT |
We thank Peter Friedman for the generous gift
of the mDCT cell line.
 |
FOOTNOTES |
*
These studies were supported by the National Institutes of
Health (Grant DK52494 to D. M. C.), by the American Heart
Association, and by the Department of Veterans Affairs.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 should be addressed: Mailcode PP262,
Oregon Health & Science University, 3314 S. W. U. S. Veterans Hospital Rd., Portland, OR 97201. Tel.: 503-220-8262 (ext. 56654); Fax:
503-721-7810; E-mail: cohend@ohsu.edu.
Published, JBC Papers in Press, January 21, 2003, DOI 10.1074/jbc.M211061200
 |
ABBREVIATIONS |
The abbreviations used are:
TRP, transient
receptor potential;
SH, Src homology domain;
HEK, human embryonic
kidney cell.
 |
REFERENCES |
1.
|
Lang, F.,
Busch, G. L.,
Ritter, M.,
Volkl, H.,
Waldegger, S.,
Gulbins, E.,
and Haussinger, D.
(1998)
Physiol. Rev.
78,
247-306[Abstract/Free Full Text]
|
2.
|
Colbert, H. A.,
Smith, T. L.,
and Bargmann, C. I.
(1997)
J. Neurosci.
17,
8259-8269[Abstract/Free Full Text]
|
3.
|
Strotmann, R.,
Harteneck, C.,
Nunnenmacher, K.,
Schultz, G.,
and Plant, T. D.
(2000)
Nat. Cell Biol.
2,
695-702[CrossRef][Medline]
[Order article via Infotrieve]
|
4.
|
Liedtke, W.,
Choe, Y.,
Marti-Renom, M. A.,
Bell, A. M.,
Denis, C. S.,
Sali, A.,
Hudspeth, A. J.,
Friedman, J. M.,
and Heller, S.
(2000)
Cell
103,
525-535[Medline]
[Order article via Infotrieve]
|
5.
|
Montell, C.,
Birnbaumer, L.,
Flockerzi, V.,
Bindels, R. J.,
Bruford, E. A.,
Caterina, M. J.,
Clapham, D. E.,
Harteneck, C.,
Heller, S.,
Julius, D.,
Kojima, I.,
Mori, Y.,
Penner, R.,
Prawitt, D.,
Scharenberg, A. M.,
Schultz, G.,
Shimizu, N.,
and Zhu, M. X.
(2002)
Mol. Cell
9,
229-231[Medline]
[Order article via Infotrieve]
|
6.
|
Peier, A. M.,
Reeve, A. J.,
Andersson, D. A.,
Moqrich, A.,
Earley, T. J.,
Hergarden, A. C.,
Story, G. M.,
Colley, S.,
Hogenesch, J. B.,
McIntyre, P.,
Bevan, S.,
and Patapoutian, A.
(2002)
Science
296,
2046-2049[Abstract/Free Full Text]
|
7.
|
Smith, G. D.,
Gunthorpe, M. J.,
Kelsell, R. E.,
Hayes, P. D.,
Reilly, P.,
Facer, P.,
Wright, J. E.,
Jerman, J. C.,
Walhin, J. P.,
Ooi, L.,
Egerton, J.,
Charles, K. J.,
Smart, D.,
Randall, A. D.,
Anand, P.,
and Davis, J. B.
(2002)
Nature
418,
186-190[CrossRef][Medline]
[Order article via Infotrieve]
|
8.
|
Xu, H.,
Ramsey, I. S.,
Kotecha, S. A.,
Moran, M. M.,
Chong, J. A.,
Lawson, D.,
Ge, P.,
Lilly, J.,
Silos-Santiago, I.,
Xie, Y.,
DiStefano, P. S.,
Curtis, R.,
and Clapham, D. E.
(2002)
Nature
418,
181-186[CrossRef][Medline]
[Order article via Infotrieve]
|
9.
|
Tilly, B. C.,
van den Berghe, N.,
Tertoolen, L. G.,
Edixhoven, M. J.,
and de Jonge, H. R.
(1993)
J. Biol. Chem.
268,
19919-19922[Abstract/Free Full Text]
|
10.
|
Sadoshima, J.,
Qiu, Z. H.,
Morgan, J. P.,
and Izumo, S.
(1996)
EMBO J.
15,
5535-5546[Abstract]
|
11.
|
Zhang, Z.,
and Cohen, D. M.
(1997)
Am. J. Physiol.
273,
F837-F842[Abstract/Free Full Text]
|
12.
|
Zhang, Z.,
Yang, X. Y.,
and Cohen, D. M.
(1998)
Am. J. Physiol.
275,
C1104-C1112[Abstract/Free Full Text]
|
13.
|
Rauchman, M. I.,
Nigam, S.,
Delpire, E.,
and Gullans, S. R.
(1993)
Am. J. Physiol.
265,
F416-F424[Abstract/Free Full Text]
|
14.
|
Gesek, F. A.,
and Friedman, P. A.
(1992)
J. Clin. Invest.
90,
429-438[Medline]
[Order article via Infotrieve]
|
15.
|
Bucher, P.,
and Bairoch, A.
(1994)
Proc. Int. Conf. Intel. Syst. Mol. Biol.
2,
53-61[Medline]
[Order article via Infotrieve]
|
16.
|
Blom, N.,
Gammeltoft, S.,
and Brunak, S.
(1999)
J. Mol. Biol.
294,
1351-1362[CrossRef][Medline]
[Order article via Infotrieve]
|
17.
|
Yoshida, K.,
Kharbanda, S.,
and Kufe, D.
(1999)
J. Biol. Chem.
274,
34663-34668[Abstract/Free Full Text]
|
18.
|
Cohen, D. M.,
Chin, W. W.,
and Gullans, S. R.
(1994)
J. Biol. Chem.
269,
25865-25870[Abstract/Free Full Text]
|
19.
|
Chin, H.,
Arai, A.,
Wakao, H.,
Kamiyama, R.,
Miyasaka, N.,
and Miura, O.
(1998)
Blood
91,
3734-3745[Abstract/Free Full Text]
|
20.
|
Harlow, E.,
and Lane, D.
(1999)
in
Using antibodies: a laboratory manual
(Harlow, E.
, and Lane, D., eds)
, pp. 321-325, Cold Spring Harbor Laboratory Press, Cold Spring Harbor, NY
|
21.
|
Roullet, J. B.,
Luft, U. C.,
Xue, H.,
Chapman, J.,
Bychkov, R.,
Roullet, C. M.,
Luft, F. C.,
Haller, H.,
and McCarron, D. A.
(1997)
J. Biol. Chem.
272,
32240-32246[Abstract/Free Full Text]
|
22.
|
Grynkiewicz, G.,
Poenie, M.,
and Tsien, R. Y.
(1985)
J. Biol. Chem.
260,
3440-3450[Abstract]
|
23.
|
Oliver, J. M.,
Burg, D. L.,
Wilson, B. S.,
McLaughlin, J. L.,
and Geahlen, R. L.
(1994)
J. Biol. Chem.
269,
29697-29703[Abstract/Free Full Text]
|
24.
|
Blume-Jensen, P.,
and Hunter, T.
(2001)
Nature
411,
355-365[CrossRef][Medline]
[Order article via Infotrieve]
|
25.
|
Patschinsky, T.,
Hunter, T.,
Esch, F. S.,
Cooper, J. A.,
and Sefton, B. M.
(1982)
Proc. Natl. Acad. Sci. U. S. A.
79,
973-977[Abstract]
|
26.
|
Hunter, T.
(1982)
J. Biol. Chem.
257,
4843-4848[Abstract/Free Full Text]
|
27.
|
Cooper, J. A.,
Esch, F. S.,
Taylor, S. S.,
and Hunter, T.
(1984)
J. Biol. Chem.
259,
7835-7841[Abstract/Free Full Text]
|
28.
|
Watanabe, H.,
Vriens, J.,
Suh, S. H.,
Benham, C. D.,
Droogmans, G.,
and Nilius, B.
(2002)
J. Biol. Chem.
277,
47044-47051[Abstract/Free Full Text]
|
29.
|
Runnels, L. W.,
Yue, L.,
and Clapham, D. E.
(2002)
Nat. Cell Biol.
4,
329-336[CrossRef][Medline]
[Order article via Infotrieve]
|
30.
|
Schaefer, M.,
Plant, T. D.,
Obukhov, A. G.,
Hofmann, T.,
Gudermann, T.,
and Schultz, G.
(2000)
J. Biol. Chem.
275,
17517-17526[Abstract/Free Full Text]
|
31.
|
Venkatachalam, K.,
Ma, H. T.,
Ford, D. L.,
and Gill, D. L.
(2001)
J. Biol. Chem.
276,
33980-33985[Abstract/Free Full Text]
|
32.
|
Zhang, Z.,
Yang, X. Y.,
and Cohen, D. M.
(1998)
Am. J. Physiol.
275,
C1104-12[Abstract/Free Full Text]
|
33.
|
Sorota, S.
(1995)
Pflugers Arch.
431,
178-185[Medline]
[Order article via Infotrieve]
|
34.
|
Voets, T.,
Manolopoulos, V.,
Eggermont, J.,
Ellory, C.,
Droogmans, G.,
and Nilius, B.
(1998)
J. Physiol.
506,
341-352[Abstract/Free Full Text]
|
35.
|
Lepple-Wienhues, A.,
Szabo, I.,
Laun, T.,
Kaba, N. K.,
Gulbins, E.,
and Lang, F.
(1998)
J. Cell Biol.
141,
281-286[Abstract]
|
36.
|
Nilius, B.,
Prenen, J.,
Voets, T.,
Eggermont, J.,
and Droogmans, G.
(1998)
J. Physiol.
506,
353-361[Abstract/Free Full Text]
|
37.
|
Crepel, V.,
Panenka, W.,
Kelly, M. E.,
and MacVicar, B. A.
(1998)
J. Neurosci.
18,
1196-1206[Abstract/Free Full Text]
|
38.
|
Shuba, Y. M.,
Prevarskaya, N.,
Lemonnier, L.,
Van Coppenolle, F.,
Kostyuk, P. G.,
Mauroy, B.,
and Skryma, R.
(2000)
Am. J. Physiol. Cell Physiol.
279,
C1144-1154[Abstract/Free Full Text]
|
39.
|
Shen, M. R.,
Chou, C. Y.,
Browning, J. A.,
Wilkins, R. J.,
and Ellory, J. C.
(2001)
J. Physiol.
537,
347-362[Abstract/Free Full Text]
|
40.
|
Deleuze, C.,
Duvoid, A.,
Moos, F. C.,
and Hussy, N.
(2000)
J. Physiol.
523,
291-299[Abstract/Free Full Text]
|
41.
|
Musch, M. W.,
Hubert, E. M.,
and Goldstein, L.
(1999)
J. Biol. Chem.
274,
7923-7928[Abstract/Free Full Text]
|
42.
|
Edwards, J. C.,
and Kapadia, S.
(2000)
J. Biol. Chem.
275,
31826-31832[Abstract/Free Full Text]
|
Copyright © 2003 by The American Society for Biochemistry and Molecular Biology, Inc.