Download A consensus sequence in the endothelin

Survey
yes no Was this document useful for you?
   Thank you for your participation!

* Your assessment is very important for improving the workof artificial intelligence, which forms the content of this project

Document related concepts

Ultrasensitivity wikipedia , lookup

Mitogen-activated protein kinase wikipedia , lookup

Polyclonal B cell response wikipedia , lookup

Lipid signaling wikipedia , lookup

Ligand binding assay wikipedia , lookup

Biochemical cascade wikipedia , lookup

Molecular neuroscience wikipedia , lookup

Specialized pro-resolving mediators wikipedia , lookup

NMDA receptor wikipedia , lookup

Endocannabinoid system wikipedia , lookup

Paracrine signalling wikipedia , lookup

Clinical neurochemistry wikipedia , lookup

G protein–coupled receptor wikipedia , lookup

Signal transduction wikipedia , lookup

Transcript
Am J Physiol Renal Physiol 288: F732–F739, 2005.
First published December 14, 2004; doi:10.1152/ajprenal.00300.2004.
A consensus sequence in the endothelin-B receptor second intracellular loop
is required for NHE3 activation by endothelin-1
Kamel Laghmani,1 Aiji Sakamoto,2 Masashi Yanagisawa,3 Patricia A. Preisig,1,* and Robert J. Alpern1,*
1
Department of Internal Medicine and the 3Howard Hughes Medical Institute, University
of Texas Southwestern Medical Center, Dallas, Texas; and 2Division of Biotechnology
and Department of Bioscience, National Cardiovascular Center Research Institute, Osaka, Japan
Submitted 11 August 2004; accepted in final form 2 December 2004
opossum kidney cells; endothelin-A/endothelin-B chimeras; sodium/
hydrogen antiporter activity
THE ENDOTHELINS (ET) are a family of three 21-amino acid
peptides, ET-1, ET-2, and ET-3. They are involved in a variety
of physiological responses, such as vascular smooth muscle
cell contraction and dilation, development, and regulation of
renal function (17, 35). The biological effects of the ETs are
mediated by two receptor subtypes, ETA and ETB (3, 13, 32,
34). ET-1 and ET-2 bind the ETA receptor with high affinities,
whereas ET-3 binds with low affinity. The three isopeptides
bind the ETB receptor with similar high affinities. Both receptors have seven hydrophobic transmembrane domains, characteristic of G protein-coupled receptors. Human ETA and ETB
receptors exhibit significant amino acid sequence identity (55%
overall, 74% within the putative transmembrane helices) (33,
36). Of the receptor subdomains, the extracellular NH2 terminus and the intracellular COOH-terminal tail display the least
sequence similarities, whereas the sequences in intracellular
loops 2 and 3 are relatively similar. Despite their similarities,
studies have demonstrated distinct roles for these intracellular
* P. A. Preisig and R. J. Alpern contributed equally to these studies.
Address for reprint requests and other correspondence: P. Preisig, Univ. of
Texas Southwestern Medical Center, Rm. H5.122, 5323 Harry Hines Blvd.,
Dallas, TX 75390-8856 (E-mail: [email protected]).
F732
loops in the human receptors. Human ETA and ETB selectively
couple to G␣s and G␣i, respectively, with the second and third
intracellular loops of human ETA involved in G␣s activation
and the third intracellular loop of human ETB involved in G␣i
activation (22).
ET-1 increases the activity of the proximal tubule apical
membrane Na⫹/H⫹ antiporter, which mediates the majority of
proximal tubular NaCl and NaHCO3 absorption (10, 12, 29,
30). This proximal tubule apical membrane Na⫹/H⫹ activity is
mediated by NHE3 (1, 4). OKP cells, an opossum kidney cell
line with many proximal tubule characteristics, express NHE3
(2, 23, 24). Using stable transfection, we established OKP cell
lines expressing ETA and ETB receptors and showed that in
cells expressing the ETB, but not the ETA receptor, ET-1
increases NHE3 activity (7). In addition, NHE3 activation by
ET-1 was inhibited by ETB receptor blockers, but not by ETA
receptor blockers. Finally, in proximal tubules harvested from
ETB receptor knockout mice, ET-1 fails to increase NHE3
activity, whereas stimulation is seen in tubules from wild-type
mice (20). These findings agree with immunohistochemistry
and binding studies showing that the ETB receptor is the
predominant ET receptor on the renal proximal tubule (9, 42).
However, the mechanism responsible for the receptor specificity of NHE3 regulation remains unclear. We previously
showed that ET-1 causes similar patterns of protein tyrosine
phosphorylation, adenylyl cyclase inhibition, and increases in
cell [Ca2⫹] in ETA- and ETB-expressing OKP cells, implying
that an additional signaling pathway must mediate the receptor
specificity of NHE3 regulation (7, 8, 28).
To address this, in the current study human ETA/ETB receptor chimeras and site-directed mutagenesis were used to identify the ET receptor domain(s) involved in ET-1 regulation of
NHE3 activity in OKP cells. We found that ETA receptor
activation inhibits NHE3 activity, an effect for which the
COOH-terminal tail is necessary and sufficient. Activation of
NHE3 by ET-1 requires the COOH-terminal domain and the
second intracellular loop of the ETB receptor. Site-directed
mutagenesis within the ETB second intracellular loop identified
residues critical for stimulation of NHE3 activity by ET-1.
These residues define a consensus sequence, KXXXPKXXXV,
required for ET-1 stimulation of NHE3.
METHODS
Materials. All chemicals were obtained from Sigma (St. Louis,
MO) unless otherwise noted as follows. Penicillin and streptomycin
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.
0363-6127/05 $8.00 Copyright © 2005 the American Physiological Society
http://www.ajprenal.org
Downloaded from http://ajprenal.physiology.org/ by 10.220.33.3 on August 11, 2017
Laghmani, Kamel, Aiji Sakamoto, Masashi Yanagisawa, Patricia A. Preisig, and Robert J. Alpern. A consensus sequence in the
endothelin-B receptor second intracellular loop is required for NHE3
activation by endothelin-1. Am J Physiol Renal Physiol 288: F732–
F739, 2005. First published December 14, 2004; doi:10.1152/ajprenal.
00300.2004.—Endothelin-1 (ET-1) increases the activity of Na⫹/H⫹
exchanger 3 (NHE3), the major proximal tubule apical membrane
Na⫹/H⫹ antiporter. This effect is seen in opossum kidney (OKP) cells
expressing the endothelin-B (ETB) and not in cells expressing the
endothelin-A (ETA) receptor. However, ET-1 causes similar patterns
of protein tyrosine phosphorylation, adenylyl cyclase inhibition, and
increases in cell [Ca2⫹] in ETA- and ETB-expressing OKP cells,
implying that an additional mechanism is required for NHE3 stimulation by the ETB receptor. The present studies used ETA and ETB
receptor chimeras and site-directed mutagenesis to identify the ET
receptor domains that mediate ET-1 regulation of NHE3 activity. We
found that binding of ET-1 to the ETA receptor inhibits NHE3
activity, an effect for which the COOH-terminal tail is necessary and
sufficient. ET-1 stimulation of NHE3 activity requires the COOHterminal tail and the second intracellular loop of the ETB receptor.
Within the second intracellular loop, a consensus sequence was
identified, KXXXVPKXXXV, that is required for ET-1 stimulation of
NHE3 activity. This sequence suggests binding of a homodimeric
protein that mediates NHE3 stimulation.
F733
NHE3 ACTIVATION BY ENDOTHELIN-1
tation-containing DNA using high-fidelity PfuTurbo DNA polymerase. Dpn1, an endonuclease (target sequence: 5⬘-Gm6ATC-3⬘) specific for methylated and hemimethylated DNA, was used to digest the
methylated, nonmutated parental DNA template. The circular mutated
DNA was transformed into XL1-Blue supercompetent cells for nick
repair and the DNA isolated by miniprep.
Binding experiments. To measure ET-1 binding, cells were grown
to confluence in 12-well plates, rendered quiescent, and incubated in
medium containing 0.3% BSA and 20 pM 125I-ET-1, ⫾10⫺6 M cold
ET-1 for 1 h at 37°C. After the binding solution was removed, the
cells were washed twice with cold PBS, harvested in 0.5 ml of 1 N
NaOH, placed in a scintillation cocktail, and bound ET-1 was counted
in a Beckman LS3801 model scintillation counter (Fullerton, CA), as
described (7). Nonspecific binding was defined as binding that occurred in the presence of excess (10⫺6 M) cold ET-1 and was
subtracted from total binding (assayed in the absence of cold ET-1) to
obtain specific binding. Results are expressed as a percentage of
specific binding in cells transfected with the wild-type ETB receptor.
Statistics. Data are reported as means ⫾ SE. Statistical significance
was determined using a paired Student’s t-test (NHE3 activity studies)
or ANOVA (binding studies) and set at P ⬍ 0.05.
RESULTS
We previously showed in stably transfected cells that the
ET-1-induced increase in NHE3 activity is mediated by ETB
and not ETA receptors. The first step of this study was to
compare the effect of ET-1 on NHE3 activity in wild-type OKP
cells and OKP cells transiently transfected with the ETB or
ETA receptor cDNA. In wild-type untransfected cells, 10⫺8 M
ET-1 applied for 30 min caused a consistent small, but statistically insignificant, increase (⫹15%) in NHE3 activity (Fig.
1). When the ETB receptor was expressed, 10⫺8 M ET-1
significantly increased NHE3 activity by 38%, in agreement
with our previous study (7). In contrast, in OKP cells expressing the ETA receptor, 10⫺8 M ET-1 significantly inhibited
NHE3 activity (⫺15%).
Table 1. Sequences of primers
Mutation Name
Direction
Mutant 1
I836V
Mutant 2
K837Q
Mutant 3
V841I
Mutant 4
K843L
Mutant 5 (Also X4)
W844V
Mutant 6
V847I
X1
G838V
X2
I839V
X3
G840A
X5
T845A
X6
A846V
Delete
G838
Sense
Reverse
Sense
Reverse
Sense
Reverse
Sense
Reverse
Sense
Reverse
Sense
Reverse
Sense
Reverse
Sense
Reverse
Sense
Reverse
Sense
Reverse
Sense
Reverse
Sense
Reverse
Sense
Reverse
P842A
Sequence
gtt
cat
gtt
cat
gga
ctg
gga
caa
gga
caa
ggg
ccc
gga
gct
gga
gct
gta
ctg
ggg
cca
ggg
cca
gct
gct
gga
caa
gct
ttt
gct
ttt
gta
ctg
att
aac
att
aac
gtt
aaa
gta
gtc
gta
gtc
gaa
ctg
gtt
att
gtt
aat
tct
gtc
att
ttt
AJP-Renal Physiol • VOL
tct
gga
tct
gga
gaa
tcc
ggg
aat
ggg
aat
cca
tca
gaa
cat
gaa
cat
tta
tcc
cca
caa
cca
caa
tgg
cat
ggg
cta
tgg
acc
tgg
acc
tta
att
gtt
ttc
gtt
ttc
aaa
aaa
tta
ttt
tta
ttt
aag
att
aaa
aac
aaa
aac
agt
ttt
gtt
ctg
agt aga gtt aaa gga att ggg gtt cca aaa tg
cca att cct tta act cta ctc caa gaa gca ac
agt aga att caa gga att ggg gtt cca aaa tg
cca att cct tga att cta ctc caa gaa gca ac
aag ga att g gga ttc caa aat gga cag cag
ttg gaa tcc caa ttc ctt taa ttc tac tcc
cca aaa gtg aca gca gta gaa att gtt ttg
tac tgc tgt cac ttt tgg aac ccc aat tcc
cca aaa gtg aca gca gta gaa att gtt ttg
tac tgc tgt cac ttt tgg aac ccc aat tcc
tgg aca gca ata gaa att gtt ttg att tgg g
caa ttt cta ttg ctg tcc att ttg gaa ccc c
aa gta a ttg ggg ttc caa aat gga cag c
gga acc cca att act tta att cta ctc c
aag ga gtt g ggg ttc caa aat gga cag c
gga acc cca act cct tta att cta ctc c
gaa tt gcg g ttc caa aat gga cag cag
ttg caa ccg caa ttc ctt taa ttc tac
tgg gca gca gta gaa att gtt ttg att tgg
aat tcc tac tgc tgc cca ttt tgg aac ccc
tgg aca gta gta gaa att gtt ttg att tgg
aat ttc tac tac tgt cca ttt tgg aac ccc
aga att aaa ⌬ att ggg gtt cca aaa tgg aca gc
gga acc cca att tta att cta ctc caa gaa gc
gca aaa tgg aca gca gta gaa att g
ctg tcc att ttg caa ccc caa ttc c
288 • APRIL 2005 •
www.ajprenal.org
Downloaded from http://ajprenal.physiology.org/ by 10.220.33.3 on August 11, 2017
were from Whittaker MA Bioproducts (Walkersville, MD);
BCECF-AM was from Molecular Probes (Eugene, OR); ET-1 was
from Peptides International (Louisville, KY); and 125I-ET-1 was from
Amersham (Arlington Heights, IL).
Cell culture and transfections. OKP cells were grown in DMEM
supplemented with 10% fetal bovine serum, 100 U/ml penicillin, and
100 ␮g/ml streptomycin at 37°C in a CO2 incubator. To overexpress
human ETA and ETB receptors, cells were transiently transfected with
wild-type, chimera, or mutant receptors using the pME18S expression
vector driven by a constitutively active SR␣ promoter (18). OKP cells
were plated on coverslips in 35-mm plastic dishes, grown to 80%
confluence, transfected with 1.1 ␮g DNA using Lipofectamine Plus
according to the manufacturer’s instructions, and grown to 100%
confluence. Confluent cells were rendered quiescent by the removal of
serum for 24 h before study. Cells on control and experimental
coverslips were derived from the same flask and passage and were
studied on the same day. ETA/ETB receptor chimeric cDNAs were
generated as described (33).
Na⫹/H⫹ antiporter activity. Cell pH was measured in a temperature-controlled spectrofluorometer (SLM 8000C, Rochester, NY) as
the ratio of BCECF fluorescence with 500- and 450-nm excitation
(emission wavelength 530 nm), as previously described (7, 8, 27, 28).
To measure Na⫹/H⫹ antiporter activity, cells were acidified with
nigericin and the initial rate of Na-dependent intracellular alkalinization was measured, as previously described (7, 8, 28). In all studies,
control and experimental cells were from the same passage and were
assayed on the same day. Exposure to 10⫺8 M ET-1 was begun at the
time of BCECF loading (30 min before study). Control cells were
exposed to the vehicle (0.1% acetic acid).
Site-directed mutagenesis. Point mutations were generated using a
Stratagene QuikChange Site-Directed Mutagenesis Kit (La Jolla, CA),
as per kit protocol. Two primers complementary to each other were
designed to contain the desired mutation (Table 1). The plasmid
containing the wild-type receptor was grown up in a dam⫹ Escherichia coli strain, resulting in the plasmid DNA methylation. Thermal
cycling was performed as per kit instructions (12 cycles of 95°C ⫻
30 s, 55°C ⫻ 1 min, and 68°C ⫻ 1 min/kb plasmid length) to denature
the template, anneal the mutagenic primers, and synthesize the mu-
F734
NHE3 ACTIVATION BY ENDOTHELIN-1
To identify the domain(s) of the ETB and ETA receptors
involved in ET-1 regulation of NHE3 activity, we transfected
cells with chimeric ETA and ETB receptor cDNAs. Figure 2
shows the restriction sites used to generate the chimeric
cDNAs. Nomenclature will indicate the specific domains derived from each receptor. For example, chimera A(N-III)B(IV-C)
designates a chimeric receptor consisting of the ETA sequence
from the NH2-terminal extracellular tail to the third transmembrane domain followed by the ETB sequence from the second
intracellular loop through the COOH-terminal tail. To generate
this chimera wild-type ETA and ETB receptor, cDNA was cut
with BssHI (Fig. 2) (33). OKP cells were transiently transfected with each chimeric cDNA and NHE3 activity was
measured following exposure to 10⫺8 M ET-1 or vehicle.
Role of the COOH-terminal domain. Experiments shown in
Fig. 3 examined the role of the COOH-terminal domain.
Again, activation of the ETA receptor led to inhibition of
Fig. 2. Schematic diagram showing the location of the restriction sites used to
generate the chimeric cDNAs. The number of circles is based on the human
ETA sequence. Dotted circles are sequence gaps placed in the sequence to align
the ETA and ETB sequences. F Represent amino acid residues that are identical
between the aligned human ETA and ETB sequences; E represent nonidentical
residues. Positions of the restriction sites used to construct the chimeric
receptors are shown by arrows along with the name of the restriction enzymes.
TMD, transmembrane domain.
AJP-Renal Physiol • VOL
Fig. 3. Role of the COOH-terminal tail. Cells were grown, treated, and
assayed as in Fig. 1. A(N-VII)B(C) and B(N-VII)A(C) were generated and the
COOH-terminal tail was removed in construct B(N-VII) using the restriction
enzyme EcoRI (Fig. 2). Blue, ETB receptor sequence. Red, ETA receptor
sequence. ETA: n ⫽ 3; A(N-VII)B(C): n ⫽ 7; B(N-VII)A(C): n ⫽ 7; B(N-VII):
n ⫽ 4. *P ⬍ 0.01. **P ⫽ 0.057.
NHE3 activity (Fig. 3). Substitution of the ETA COOH-terminal tail with the ETB COOH-terminal tail [A(N-VII)B(C)]
prevented this effect, whereas substitution of the ETB COOHterminal tail with the ETA COOH-terminal tail [B(N-VII)A(C)]
mediated inhibition (⫺26%, P ⬍ 0.01). Thus the presence of
the COOH-terminal domain of the ETA receptor is necessary
and sufficient to mediate inhibition of NHE3 activity by ET-1.
By contrast, the above results using the chimera A(NVII)B(C) suggest that the ETB COOH-terminal domain is not
sufficient to mediate stimulation of NHE3 activity by ET-1.
The failure of B(N-VII)A(C) to mediate ET-1-induced stimulation of NHE3 could indicate a requirement for the COOH
terminus or could be due to ETA-induced inhibition mediated
by the ETA COOH terminus. To determine whether the ETB
COOH-terminal tail is required for ET-1 stimulation of NHE3,
we examined a truncated ETB receptor in which the COOHterminal tail was removed (18). As shown in Fig. 3, with
removal of the COOH-terminal tail ET-1 was without effect on
NHE3 activity. Taken together, these results suggest that the
ETB COOH-terminal tail is necessary but not sufficient to
mediate stimulation of NHE3 activity by ET-1.
Role of the ETB receptor intracellular loops. To identify
other key domains of the ETB receptor required for ET-1
stimulation of NHE3 activity, we used a series of chimeras in
which domains of the ETB receptor were replaced with the
corresponding region of the ETA receptor. As shown in Fig. 4,
when the ETB sequence extended from the COOH terminus to
the third intracellular loop, [A(N-V)B(VI-C) and A(N-IV)B(VC)], ET-1 had no effect on NHE3 activity. By contrast, when
the second intracellular loop of the ETB receptor was included
[A(N-III)B(IV-C)], ET-1 stimulation of NHE3 activity was
restored (⫹35%). These results suggest that in addition to the
COOH-terminal tail, a specific region including transmem-
288 • APRIL 2005 •
www.ajprenal.org
Downloaded from http://ajprenal.physiology.org/ by 10.220.33.3 on August 11, 2017
Fig. 1. Endothelin (ET)A and ETB receptor regulation of Na⫹/H⫹ exchanger 3
(NHE3) activity. Opossum kidney (OKP) cells were grown, transfected, and
exposed to 10⫺8 M ET-1 or vehicle, and NHE3 activity was assayed as
described in METHODS. Left: untransfected cells, n ⫽ 8. Middle: ETB receptor
sequence, n ⫽ 18. Right: ETA receptor sequence, n ⫽ 10. *P ⬍ 0.01.
NHE3 ACTIVATION BY ENDOTHELIN-1
brane helix III and/or the second intracellular loop of the ETB
receptor is required for ET-1 stimulation of NHE3 activity.
To confirm this, an additional series of chimeras were used
(Fig. 5). When the sequence extending from transmembrane
helices III to VI of the ETB receptor was replaced with the
corresponding ETA sequence [B(N-III)A(IV-VI)B(VII-C)],
ET-1 was without effect on NHE3 activity. When only the
region between transmembrane helices V and VI of the ETB
receptor was replaced with the corresponding ETA sequence
[B(N-IV)A(V-VI)B(VII-C)], ET-1 stimulation of NHE3 activity was restored (⫹36%). Together, these data confirm that the
domain containing the second intracellular loop and transmembrane helix IV of the ETB receptor is critical in mediating
ET-1-induced NHE3 activation.
To further confirm this conclusion, we used a chimera in
which both the second intracellular loop and COOH-terminal
tail were derived from the ETB receptor and all other intracellular sequences were from the ETA receptor [A(N-III)B(IVV)A(VI-VII)B(C)]. As shown in Fig. 5, in cells expressing this
chimera ET-1 increased NHE3 activity by 35%, confirming
that the second intracellular loop and COOH-terminal tail of
ETB are both required and sufficient for ET-1 stimulation of
NHE3 activity.
Amino acid residues in the ETB receptor second intracellular loop required for ET-1 stimulation of NHE3 activity. The
above studies all confirmed a key role for the domain including
the second intracellular loop and transmembrane helix IV in
NHE3 stimulation. We reasoned that the key domain was most
likely intracellular and further pursued the second intracellular
loop. The second intracellular loop is highly conserved with
only six amino acids that differ between the ETA and ETB
receptors (Fig. 6). The specific role of each of these amino
acids was analyzed by site-directed mutagenesis converting
each of the six ETB amino acids individually to their corresponding ETA receptor amino acid (Fig. 6 and Table 1). As
AJP-Renal Physiol • VOL
Fig. 5. Critical TMDs of the ETB receptor. Cells were grown, treated, and
assayed as in Fig. 1. B(N-III)A(IV-VI)B(VII-C), B(N-IV)A(V-VI)B(VII-C),
and A(N-III)B(IV-V)A(VI-VII)B(C) were generated by using restriction enzymes BssHI/ClaI, NcoI/ClaI, and BssHI/BglII/EcoRI, respectively (Fig. 2).
Blue, ETB receptor sequence. Red, ETA receptor sequence. ETB: n ⫽ 10;
B(N-III)A(IV-VI)B(VII-C): n ⫽ 7; B(N-IV)A(V-VI)B(VII-C): n ⫽ 6; A(NIII)B(IV-V)A(VI-VII)B(C): n ⫽ 5. *P ⬍ 0.01.
shown in Fig. 7, in cells expressing wild-type ETB receptor,
ET-1 increased NHE3 activity by 46% (P ⬍ 0.001). Individual
mutation of ETB residues I836 and W844 to the corresponding
ETA residues V835 and V843, respectively, did not prevent this
effect (ET-1 increased NHE3 activity 44 and 52%, respectively, both P ⬍ 0.001). By contrast, when ETB receptor
residues K837, V841, K843, and V847 were individually replaced
with the corresponding ETA receptor residues (Q836, I840, L842,
and I846, respectively), the ETB receptor lost its ability to
mediate ET-1 stimulation of NHE3 activity (Fig. 7). Thus ETB
Fig. 6. Schematic diagram of the ETA and ETB receptor second intracellular
loop sequences. The diagram of the receptor is as shown in Fig. 2. The dashed
lines below the TMD indicate the region shown in the gray box. Amino acid
residues that are the same in the ETA and ETB receptor are in black. Those that
are different between the receptors are noted in italic.
288 • APRIL 2005 •
www.ajprenal.org
Downloaded from http://ajprenal.physiology.org/ by 10.220.33.3 on August 11, 2017
Fig. 4. Role of ETB receptor intracellular loops. Cells were grown, treated, and
assayed as in Fig. 1. A(N-V)B(VI-C), A(N-IV)B(V-C), and A(N-III)B(IV-C)
were generated using restriction enzymes BglII, NcoI, and BssHI, respectively
(Fig. 2). Blue, ETB receptor sequence. Red, ETA receptor sequence. ETB: n ⫽
16; A(N-V)B(VI-C): n ⫽ 4; A(N-IV)B(V-C): n ⫽ 4; A(N-III)B(IV-C): n ⫽ 8.
*P ⬍ 0.01.
F735
F736
NHE3 ACTIVATION BY ENDOTHELIN-1
ETB receptor and those expressing key truncated, chimeric, or
mutated receptors.
DISCUSSION
receptor residues K837, V841, K843, and V847 play a critical role
in ET-1 stimulation of NHE3 activity, whereas I836 and W844
are not required.
The two pairs of K and V residues (K837 and V841; K843 and
847
V ) that were found to be required for ET-1 stimulation of
NHE3 activity are each separated by three amino acids. We
next tested the role of the six amino acid residues (G838, I839,
G840, W844, T845, and A846) lying between the two “K-V” pairs
(Fig. 6 and Table 1). W844 is one of the 6 amino acid residues
that differs between the ETB and ETA receptors. Mutation of
this residue (W844V) had already been shown to have no effect
on the ability of ET-1 to stimulate NHE3 activity (Fig. 7). The
other five potential X residues were replaced with either a
valine or alanine residue. As shown in Fig. 8, individually
mutating these residues had no effect on the ability of ET-1 to
stimulate NHE3 activity (% stimulation varied between 27 and
52%, P ⬍ 0.05 in all 6 experiments). However, removal of one
of the X residues, G838, prevented the ET-1 effect (Fig. 9).
Thus, while the specific amino acids separating the K and V
residues are not critical, the number of amino acids is important.
The two KXXXV sequences in the second intracellular loop
are separated by a proline that is conserved in ETB and ETA
receptors. Mutation of this proline residue (P842) to alanine also
prevented ET-1 from stimulating NHE3 activity (Fig. 9).
Taken together, these data suggest the presence of a consensus
sequence (KXXXVPKXXXV) in the ETB receptor second
intracellular loop that mediates ET-1 stimulation of NHE3
activity.
ET-1 binding to receptors. To address the possibility that the
varying effect of ET-1 on NHE3 activity in the above studies
was a consequence of a difference in receptor expression
and/or failure of ET-1 to bind to a mutated receptor, ligand
binding studies were performed in wild-type OKP cells and
OKP cells expressing intact, and key truncated, chimeric, or
mutated receptors in which ET-1 did not stimulate NHE3
activity (Table 2). As shown, specific ET-1 binding was not
statistically different between cells expressing the wild-type
AJP-Renal Physiol • VOL
Fig. 8. Role of conserved residues in the proposed consensus sequence. Cells
were grown, treated, and assayed as in Fig. 1. Five amino acid residues lying
between the 2 K-V pairs were individually mutated. Mutations were made as
indicated in Table 1. ETB: n ⫽ 8; G838V: n ⫽ 12; I839V: n ⫽ 8; G840A: n ⫽
7; T845A: n ⫽ 8; A846V: n ⫽ 13. *P ⬍ 0.02.
288 • APRIL 2005 •
www.ajprenal.org
Downloaded from http://ajprenal.physiology.org/ by 10.220.33.3 on August 11, 2017
Fig. 7. Role of amino acid residues in the second intracellular loop that differ
between the ETA and ETB receptors. Cells were grown, treated, and assayed as
in Fig. 1. Mutations were made as indicated in Table 1. ETB: n ⫽ 21; I836V:
n ⫽ 7; K837Q: n ⫽ 9; V841I: n ⫽ 9; K843L: n ⫽ 10; W844V: n ⫽ 11; V847I:
n ⫽ 8. *P ⬍ 0.01.
The ETs are a family of three conserved 21 amino acid
peptides that bind to and mediate their effects through two G
protein-coupled receptor subtypes, ETA and ETB (3, 13, 32).
Despite significant amino acid homology, the receptors can be
pharmacologically distinguished by their affinity for the three
ET isopeptides and their distinct physiological roles (22, 26,
36). The ETB receptor mediates release of relaxing factors
causing vasodilation and is involved in inflammatory pain,
whereas the ETA receptor mediates vasoconstriction, stimulates mitogenesis and matrix formation, is antiapoptotic, and is
involved in acute or neuropathic pain (26). Typically, the ETB
receptor activates G␣i, whereas the ETA receptor activates
G␣s, but this G protein coupling is not fixed in all cells (22,
36). In the kidney, while both ETA and ETB are present in renal
vessels and on tubular epithelial cells, ETB expression predominates in tubular epithelial cells (17, 31, 37).
We previously showed that ET-1 stimulates NHE3 activity
in cultured OKP cells, an effect that is mediated through the
ETB and not the ETA receptor (7, 19). ET-1/ETB stimulation of
NHE3 activity involves NHE3 phosphorylation and exocytic
insertion of NHE3 into the apical membrane (27, 28). As
shown in Fig. 1, ET-1/ETA signaling not only fails to stimulate
NHE3 activity but inhibits the transporter. The physiological
significance of NHE3 inhibition is unclear as ET-1 stimulates
the proximal tubule Na⫹/H⫹ antiporter, an effect mediated by
the ETB receptor (19, 20).
Because of these differing effects on NHE3, we initially
sought to determine signaling pathways that were differentially
activated by the ETB and ETA receptors in OKP cells. Using
inhibitors, we found that NHE3 activation by the ETB receptor
was mediated by increases in cell Ca2⫹ and activation of
Ca2⫹-calmodulin kinase and tyrosine kinases. On ET-1 addition, both receptors were found to increase cell Ca2⫹ with
patterns that were indistinguishable. In addition, activation of
NHE3 ACTIVATION BY ENDOTHELIN-1
both receptors resulted in tyrosine phosphorylation of proteins
with a pattern consistent with focal adhesion protein phosphorylation. Finally, although activation of ETA and ETB receptors
can have opposing effects on adenylyl cyclase in many cells, in
OKP cells both receptors inhibited adenylyl cyclase (7, 28).
These findings led us to hypothesize that an additional signaling pathway must be required for ETB receptor activation of
NHE3, perhaps a protein that binds selectively to the ETB
receptor. To address this, the present studies were designed to
define domains unique to the ETB receptor that were required
for NHE3 activation.
The ETA and ETB receptors exhibit 55% overall amino acid
homology (22, 36). Within the receptor domains, the least
homology is seen in the intracellular COOH-terminal domain
and the most homology is associated with the transmembrane
helices. In addition, intracellular loops, a likely site of protein
binding, are highly homologous. In the present studies, OKP
cells were transiently transfected with human ETA and ETB
receptor chimera as well as mutated ETB receptor cDNAs to
determine the domains and sequences responsible for ET-1/
ETB stimulation of NHE3. Our laboratory previously showed
that we achieve ⬃80% transfection efficiency with transient
transfection in OKP cells (14). In addition, we completely
inhibited a number of cellular processes in cells transiently
transfected with dominant-negative constructs, again suggesting high transfection efficiency (21, 39, 40).
The studies led to a number of conclusions. First, the
COOH-terminal tail of the ETA receptor is necessary and
sufficient for inhibition of the NHE3 activity by ET-1. Thus a
chimeric receptor that contains only the COOH-terminal tail of
ETA, with the remainder of the receptor derived from the ETB
receptor, inhibits NHE3. Similarly, an ETA receptor in which
the COOH-terminal domain has been replaced with the relevant ETB sequence loses the ability to inhibit NHE3 activity.
Second, the COOH-terminal tail of ETB is required for
stimulation of NHE3. Deletion of this sequence eliminates
regulation, and replacement with the corresponding ETA sequence leads to ET-1-induced inhibition of NHE3 activity.
However, unlike results with the ETA receptor, the COOHterminal tail is not sufficient for stimulation of NHE3. The
AJP-Renal Physiol • VOL
chimera containing the COOH-terminal ETB sequence with the
remainder of the protein derived from the ETA receptor fails to
regulate NHE3 activity. This suggests that in addition to the
COOH-terminal tail, at least one other sequence is required for
NHE3 activation by ET-1/ETB.
The role of the COOH-terminal tail of G protein-coupled
receptors has not been studied extensively. In the parathyroid
Ca2⫹-sensing receptor, individual point mutations of His880
and Phe882 in the COOH-terminal tail to alanine residues were
associated with decreased total inositol phosphate production
and retention of the receptors in the endoplasmic reticulum,
suggesting that the COOH-terminal tail is required for efficient
targeting of the receptor to the cell surface (6). In the present
studies, however, absence of the ETB COOH-terminal tail did
not decrease ET-1 binding, demonstrating that trafficking to
the plasma membrane had not been impaired.
In addition to a requirement for the COOH-terminal tail,
NHE3 activation by ET-1/ETB required a sequence within the
second intracellular loop. Comparing a series of chimeras,
results consistently demonstrated that the presence of sequences in the second intracellular loop and transmembrane
domain IV, between the BssHI and NcoI restriction sites, was
required for NHE3 activation. Mutational studies localized the
key sequences to the second intracellular loop and further
defined the specific sequence required: two essential lysine (K)
and two essential valine (V) resides that are spaced as follows:
KXXXVPKXXXV (Fig. 6). Both the spacing between the
lysine and valine residues and the proline (P) residue were
found to be essential to confer ET-1 stimulation of NHE3. The
presence of the repeat sequence (KXXXV) and the space
requirement for three residues between the K and V pairs
Table 2. ET-1 binding to receptors
Values are means ⫾ SE. Cells were grown and treated as in Fig. 1. Ligand
binding was assayed as described in METHODS. Wild-type opossum kidney
cells: n ⫽ 9; endothelin (ET)B cells: n ⫽ 6; ETA cells: n ⫽ 6; ETB minus C-tail;
n ⫽ 6; A(N-V)B(VI-C): n ⫽ 3; B(N-III)A(IV-VI)B(VII-C): n ⫽ 3; K837Q:
n ⫽ 3; V841I: n ⫽ 6; K843L: n ⫽ 6; V847I: n ⫽ 6; Delete G838: n ⫽ 3.
288 • APRIL 2005 •
www.ajprenal.org
Downloaded from http://ajprenal.physiology.org/ by 10.220.33.3 on August 11, 2017
Fig. 9. G838 deletion and P842A mutation. Cells were grown, treated, and
assayed as in Fig. 1. Mutations were made as indicated in Table 1. G838
deletion: n ⫽ 6; P842A: n ⫽ 9.
F737
F738
NHE3 ACTIVATION BY ENDOTHELIN-1
AJP-Renal Physiol • VOL
this region matches more closely to the NH2 terminus (address
domain) of ET-1 compared with ET-3, and thus ETA binds
ET-1 with higher affinity than ET-3 (33). Subtle differences in
the ETB receptor allow the address recognition domain to
interact with a wider spectrum of ligand address domains,
making the ETB receptor more promiscuous (33). The COOHterminal portion of the ligand interacts with transmembrane
helices I, II, III, and VII of the ETA and ETB receptors to
transmit the ligand message (22). The present studies demonstrate that the amino acid sequence in the second intracellular
loop and first part of transmembrane helix IV transmit the
signal mediating ET-1 stimulation of NHE3 activity. The
combination of a required repeat sequence, required spacing
between essential amino acid residues, and a required proline
residue between the repeat sequences suggests that the protein
that binds the receptor might have symmetrical “arms” that
interact with the receptor. Such a protein could be either a
single protein or a homodimer.
In summary, the present studies identify the COOH-terminal
tail and a sequence within the second intracellular loop of the
ETB receptor that are necessary and sufficient to mediate ET-1
stimulation of NHE3 activity. The presence of a repeat sequence with specific space requirements and a required proline
residue separating the repeat sequences suggests that signal
transmission is mediated by the binding of a homodimeric
protein to the receptor.
ACKNOWLEDGMENTS
Technical assistance was provided by Kavita Mathi and Ebtesam AbdelSalam.
GRANTS
These studies were supported by National Institutes of Health Grant
DK-39298. M. Yanagisawa is an Investigator of the HHMI.
Present address of K. Laghmani: Institut National de la Santé et de la
Recherche Médicale, Institut Biomedicale des Cordeliers, Paris, France.
Present address of R. J. Alpern: Yale School of Medicine, New Haven, CT
06520.
REFERENCES
1. Alpern RJ. Renal acidification mechanisms. In: The Kidney, edited by
Brenner BM. Philadelphia, PA: W. B. Saunders, 2000, p. 455–519.
2. Amemiya M, Yamaji Y, Cano A, Moe OW, and Alpern RJ. Acid
incubation increases NHE-3 mRNA abundance in OKP cells. Am J
Physiol Cell Physiol 269: C126 –C133, 1995.
3. Arai H, Hori S, Aramori I, Ohkubo H, and Nakanishi S. Cloning and
expression of a cDNA encoding an endothelin receptor. Nature 348:
730 –732, 1990.
4. Biemesderfer D, Pizzonia J, Abu-Alfa A, Exner M, Reilly R, Igarashi
P, and Aronson PS. NHE3: a Na/H exchanger isoform of renal brush
border. Am J Physiol Renal Fluid Electrolyte Physiol 265: F736 –F742,
1993.
5. Burstein ES, Spalding TA, and Brann MR. The second intracellular
loop of the m5 muscarinic receptor is the switch which enables G-protein
coupling. J Biol Chem 273: 24322–24327, 1998.
6. Chang W, Pratt S, Chen TH, Bourguignon L, and Shoback D. Amino
acids in the cytoplasmic C terminus of the parathyroid Ca2⫹-sensing
receptor mediate efficient cell-surface expression and phospholipase C
activation. J Biol Chem 276: 44129 – 44136, 2001.
7. Chu TS, Peng Y, Cano A, Yanagisawa M, and Alpern RJ. EndothelinB
receptor activates NHE-3 by a Ca2⫹-dependent pathway in OKP cells.
J Clin Invest 97: 1454 –1462, 1996.
8. Chu TS, Tsuganezawa H, Peng Y, Cano A, Yanagisawa M, and
Alpern RJ. Role of tyrosine kinase pathways in ETB receptor activation
of NHE3. Am J Physiol Cell Physiol 271: C763–C771, 1996.
288 • APRIL 2005 •
www.ajprenal.org
Downloaded from http://ajprenal.physiology.org/ by 10.220.33.3 on August 11, 2017
suggest that this portion of the receptor may be involved in the
binding of a homodimer to the receptor. The presence of the
proline residue, typically associated with flexibility in a protein, between the two repeat sequences would also facilitate
binding of a homodimer.
The second and/or third intracellular loops have been shown
to be critical for signaling in other G protein-coupled receptors.
The second intracellular loop of the thrombin receptor confers
specificity to Gq coupling, is hypothesized to be the relevant
domain involved in G protein coupling and to mediate conformational changes that prevent or promote G protein coupling in
the muscarinic receptor, and is critical for activating the phospholipase C (PLC) pathway through the Gq ␣-subunit in the
PTH/PTH-related protein signaling pathway (5, 15, 25, 41).
The second and third intracellular loops are involved in inhibiting angiotensin-dependent activation of PLC via the angiotensin 1a receptor (38). The second intracellular loop of the
metabotropic glutamate receptor 1 is critical for signal transduction, but unlike most other G protein-coupled receptors,
requires cooperation with another intracellular domain (11). In
Chinese hamster ovary cells expressing ETA/ETB receptor
chimeric proteins, intracellular loops 2 and/or 3 of the ETA
receptor mediate ET-1-induced stimulation of cAMP formation, whereas intracellular loop 3 of the ETB receptor mediates
ET-1-induced inhibition of cAMP formation, demonstrating
that intracellular loops 2 and 3 direct signaling through G␣s
and G␣i, respectively (36). Thus it appears that the intracellular
loops of G protein-coupled receptors mediate signal transduction. In the present studies, we show that intracellular loop 2 of
the ETB receptor mediates ET-1 stimulation of NHE3 activity.
The lack of an effect on ET-1 binding to the chimeric and
mutated receptors is consistent with studies in COS-7 cells
(33). Similarly, the lack of an effect on ET-1 binding with
removal of the ETB COOH-terminal tail is consistent with
studies from Sakamoto et al. (18) showing that the COOHterminal tail of the ETB receptor is involved with signaling but
not ligand binding. These observations suggest that the tertiary
structure of the ETB receptor has been maintained in the
chimeric and mutated receptors and that failure of ET-1 to
stimulate NHE3 activity is not due to failure of the chimeric or
mutated receptors to be expressed on the apical membrane.
In many of the studies in which cells were transfected with
either the ETA receptor or one of the inactive receptors,
baseline NHE3 activity was observed to be increased, compared with cells transfected with the ETB receptor or a functional receptor. One possible explanation for this observation is
that the vacant ETA receptor stimulates NHE3 activity and/or
the vacant ETB receptor inhibits NHE3 activity, both effects
due to binding of a NHE3-regulatory protein. Binding of ET-1
to these receptors could then inhibit binding of the regulatory
protein, relieving NHE3 regulation. Thus binding of ET-1 to
the ETB receptor may prevent NHE3 inhibition and/or binding
of ET-1 to the ETA receptor may prevent NHE3 stimulation by
the vacant receptor.
Receptors have two functions, ligand recognition and binding (“address” function) and signal transmission (“message”
function). Ligand binding studies have proposed that the region
of transmembrane helices IV-VI and the adjacent loops of the
ETA and ETB receptors determine the selection of ligandreceptor interaction and thus are considered to be the address
recognition subdomain (33). In the case of the ETA receptor,
NHE3 ACTIVATION BY ENDOTHELIN-1
AJP-Renal Physiol • VOL
27. Peng Y, Amemiya M, Yang X, Fan L, Moe OW, Yin HL, Preisig PA,
Yanagisawa M, and Alpern RJ. ETB receptor activation causes exocytic
insertion of NHE3 in OKP cells. Am J Physiol Renal Physiol 280:
F34 –F42, 2001.
28. Peng Y, Moe OW, Chu TS, Preisig PA, Yanagisawa M, and Alpern
RJ. ETB receptor activation leads to activation and phosphorylation of
NHE3. Am J Physiol Cell Physiol 276: C938 –C945, 1999.
29. Preisig PA, Ives HE, Cragoe EJ, Alpern RJ, and Rector FC Jr. Role
of the Na/H antiporter in rat proximal tubule bicarbonate absorption.
J Clin Invest 80: 970 –978, 1987.
30. Preisig PA and Rector FC Jr. Role of Na-H antiport in rat proximal
tubule NaCl absorption. Am J Physiol Renal Fluid Electrolyte Physiol 255:
F461–F465, 1988.
31. Rabelink TJ, Kaasjager KAH, Stroes EG, and Koomans HA. Endothelin in renal pathophysiology: from experimental to therapeutic application. Kidney Int 50: 1827–1833, 1996.
32. Sakamoto A, Yanagisawa M, Sakurai T, Takuwa Y, Yanagisawa H,
and Masaki T. Cloning and functional expression of human cDNA for the
ETB endothelin receptor. Biochem Biophys Res Commun 178: 656 – 663,
1991.
33. Sakamoto A, Yanagisawa M, Sawamura T, Enoki T, Ohtani T,
Sakurai T, Nakao K, Toyo-oka T, and Masaki T. Distinct subdomains
of human endothelin receptors determine their selectivity to endothelinAselective antagonist and endothelinB-selective agonists. J Biol Chem 268:
8547– 8553, 1993.
34. Sakurai T, Yanagisawa M, Takuwa Y, Miyazaki H, Kimura S, Goto
K, and Masaki T. Cloning of a cDNA encoding a non-isopeptideselective subtype of the endothelin receptor. Nature 348: 732–735, 1990.
35. Simonson MS. Endothelins: multifunctional renal peptides. Physiol Rev
73: 375– 411, 1993.
36. Takagi Y, Ninomiya H, Sakamoto A, Miwa S, and Masaki T. Structural
basis of G protein specificity of human endothelin receptors: a study with
endothelin A/B chimeras. J Biol Chem 270: 10072–10078, 1995.
37. Terada Y, Tomita K, Nonoguchi H, and Marumo F. Different localization of two types of endothlin receptor mRNA in microdissected rat
nephron segments using reverse transcription and polymerase chain reaction assay. J Clin Invest 90: 107–112, 1992.
38. Thompson JB, Wade SM, Harrison JK, Salafranca MN, and Neubig
RR. Cotransfection of second and third intracellular loop gragments
inhibit angiotensin AT1a receptor activation of phospholipase C in HEK293 cells. J Pharmacol Exp Ther 285: 216 –222, 1998.
39. Tsuganezawa H, Preisig PA, and Alpern RJ. Dominant negative c-Src
inhibits angiotensin II induced activation of NHE3 in OKP cells. Kidney
Int 54: 394 –398, 1998.
40. Tsuganezawa H, Sato S, Yamaji Y, Preisig PA, Moe OW, and Alpern
RJ. Role of c-Src and ERK in acid-induced activation of NHE3. Kidney
Int 62: 41–50, 2002.
41. Verrall S, Ishii M, Chen M, Wang L, Tram T, and Coughlin SR. The
thrombin receptor second cytoplasmic loop confers coupling to Gq-like G
proteins in chimeric receptors. J Biol Chem 272: 6898 – 6902, 1997.
42. Yamamoto T and Uemura H. Distribution of endothelin-B receptor-like
immunoreactivity in rat brain, kidney, and pancreas. J Cardiovasc Pharmacol 31, Suppl 1: S207–S211, 1998.
288 • APRIL 2005 •
www.ajprenal.org
Downloaded from http://ajprenal.physiology.org/ by 10.220.33.3 on August 11, 2017
9. Edwards RM, Trizna W, and Stack EJ. [125I]endothelin-1 binding to
renal brush-border and basolateral membranes. Eur J Pharmacol 345:
229 –232, 1998.
10. Eiam-Ong S, Hilden SA, King AJ, Johns CA, and Madias NE.
Endothelin-1 stimulates the Na⫹/H⫹ and Na⫹/HCO⫺
3 transporters in rabbit
renal cortex. Kidney Int 42: 18 –24, 1992.
11. Gomeza J, Joly C, Kuhn R, Knöpfel T, Bockaert J, and Pin JP. The
second intracellular loop of metabotropic glutamate receptor 1 cooperates
with the other intracellular domains to control coupling to G- proteins.
J Biol Chem 271: 2199 –2205, 1996.
12. Guntupalli J and DuBose TD. Effects of endothelin on rat renal proximal
tubule Na⫹-Pi cotransport and Na⫹/H⫹ exchange. Am J Physiol Renal
Fluid Electrolyte Physiol 266: F658 –F666, 1994.
13. Hosoda K, Nakao K, Tamura N, Arai H, Ogi K, Suga S, Nakanishi S,
and Imura H. Organization, structure, chromosomal assignment, and
expression of the gene encoding the human endothelin-A receptor. J Biol
Chem 267: 18797–18804, 1992.
14. Hu MC, Fan L, Crowder LA, Karim-Jimenez Z, Murer H, and Moe
OW. Dopamine acutely stimulates Na⫹/H⫹ exchanger (NHE3) endocytosis via clathrin-coated vesicles. J Biol Chem 276: 26915, 2001.
15. Iida-Klein A, Guo J, Takemura M, Drake MT, Potts JT Jr, AbouSamra A, Bringhurst FR, and Segre GV. Mutations in the second
cytoplasmic loop of the rat parathyroid hormone (PTH)/PTH-related
protein receptor result in selective loss of PTH-stimulated phospholipase
C activity. J Biol Chem 272: 6882– 6889, 1997.
17. Kedzierski RM and Yanagisawa M. Endothelin system: the doubleedged sword in health and disease. Annu Rev Pharmacol Toxicol 41:
851– 876, 2001.
18. Koshimizu TA, Tsujimoto G, Ono K, Masaki T, and Sakamoto A.
Truncation of the receptor carboxyl terminus impairs membrane signaling
but not ligand binding of human ETB endothelin receptor. Biochem
Biophys Res Commun 217: 354 –362, 1995.
19. Laghmani K, Preisig PA, and Alpern RJ. Role of endothelin in proximal tubule proton secretion and the adaptation to a chronic metabolic
acidosis. J Nephrol 15, Suppl 5: S75–S87, 2002.
20. Laghmani K, Preisig PA, Moe OW, Yanagisawa M, and Alpern RJ.
Endothelin-1/endothelin-B receptor-mediated increases in NHE3 activity
in chronic metabolic acidosis. J Clin Invest 107: 1563–1569, 2001.
21. Li S, Sato S, Yang X, Preisig PA, and Alpern RJ. Pyk2 activation is
integral to acid stimulation of NHE3. J Clin Invest 114: 1782–1789, 2004.
22. Masaki T, Ninomiya H, Sakamoto A, and Okamoto Y. Structural basis
of the function of endothelin receptor. Mol Cell Biochem 190: 153–156,
1999.
23. Moe OW, Miller RT, Horie S, Cano A, Preisig PA, and Alpern RJ.
Differential regulation of Na/H antiporter by acid in renal epithelial cells
and fibroblasts. J Clin Invest 88: 1703–1708, 1991.
24. Montrose MH and Murer H. Polarity and kinetics of Na-H exchange in
cultured opossum kidney cells. Am J Physiol Cell Physiol 259: C121–
C133, 1990.
25. Moro O, Lameh J, Högger P, and Sadée W. Hydrophobic amino acid in
the i2 loop plays a key role in receptor G protein coupling. J Biol Chem
268: 22273–22276, 1993.
26. Nelson J, Bagnato A, Battistini B, and Nisen P. The endothelin axis:
emerging role in cancer. Nat Rev Cancer 3: 110 –116, 2003.
F739