Download as pdf or txt
Download as pdf or txt
You are on page 1of 10

Am J Physiol Renal Physiol 299: F559–F567, 2010.

First published June 24, 2010; doi:10.1152/ajprenal.00617.2009.

Vasopressin increases phosphorylation of Ser84 and Ser486 in Slc14a2


collecting duct urea transporters
Shelly Hwang,1 Ruwan Gunaratne,1 Markus M. Rinschen,1 Ming-Jiun Yu,1 Trairak Pisitkun,1
Jason D. Hoffert,1 Robert A. Fenton,2 Mark A. Knepper,1 and Chung-Lin Chou1
1
Laboratory of Kidney and Electrolyte Metabolism, National Heart, Lung, and Blood Institute, National Institutes of Health,
Bethesda, Maryland; and 2The Water and Salt Research Center, Department of Anatomy, Aarhus University, Aarhus, Denmark
Submitted 26 October 2009; accepted in final form 21 June 2010

Hwang S, Gunaratne R, Rinschen MM, Yu M-J, Pisitkun T, strongly regulates the urea permeability of the IMCD (34).
Hoffert JD, Fenton RA, Knepper MA, Chou C-L. Vasopressin in- However, the mechanisms involved in this regulation remain
creases phosphorylation of Ser84 and Ser486 in Slc14a2 collecting duct unclear. [32P]-labeling experiments revealed that vasopressin
urea transporters. Am J Physiol Renal Physiol 299: F559 –F567, 2010. signaling increases phosphorylation of UT-A1 (5, 44). Candi-
First published June 24, 2010; doi:10.1152/ajprenal.00617.2009.— date phosphorylation sites (Ser486 and Ser499) in UT-A1 have

Downloaded from http://ajprenal.physiology.org/ at EBSCO on April 7, 2013


Vasopressin-regulated urea transport in the renal inner medullary
collecting duct (IMCD) is mediated by two urea channel proteins,
been mutated and shown to be involved in urea transport
UT-A1 and UT-A3, derived from the same gene (Slc14a2) by alter- regulation, although sites in UT-A3 have not been investigated
native splicing. The NH2-terminal 459 amino acids are the same in in this manner (6). More recently, mass spectrometry-based
both proteins. To study UT-A1/3 phosphorylation, we made phospho- phosphoproteomic studies identified several sites in addition to
specific antibodies to UT-A sequences targeting phospho-serines at Ser486 and Ser499 (2, 16), namely Ser10, Ser62, Ser63, and
positions 84 and 486, sites identified previously by protein mass Ser84 (2). Among these sites, three have been demonstrated by
spectrometry. Both antibodies proved specific, recognizing only the mass spectrometry to be regulated by vasopressin: Ser84,
phosphorylated forms of UT-A1 and -A3. Immunoblotting of rat Ser486, and Ser499 (2, 6). Of these, Ser84 is present in both
IMCD suspensions or whole inner medullas showed that the V2R- UT-A1 and UT-A3, whereas the other two sites are only
selective vasopressin analog 1-deamino-8-D-arginine vasopressin present in UT-A1 (Fig. 1).
(dDAVP) increases phosphorylation at Ser84 (in UT-A1 and UT-A3) To investigate regulated phosphorylation at Ser84, we pro-
and Ser486 (in UT-A1) by about eightfold. Time course studies in rat
duced a phospho-specific antibody to this site and used it in
IMCD suspensions showed maximum phosphorylation within 1 min
of dDAVP exposure, consistent with the time course of vasopressin-
immunoblotting and immunohistochemical experiments in
stimulated phosphorylation of the vasopressin-sensitive water channel rats. The results show that vasopressin stimulates phosphory-
aquaporin-2 at Ser256. Confocal immunofluorescence in Brattleboro lation of both UT-A1 and UT-A3 at Ser84 with a time course
rat medullary tissue showed labeling limited to the IMCD, which similar to that of vasopressin-induced phosphorylation of aqua-
increased markedly in response to dDAVP. Immuno-electron micros- porin-2 at Ser256. In addition, to confirm the role of the Ser486
copy studies showed that both phosphorylated forms were present site in UT-A1 and investigate its regulation by vasopressin, we
mainly in intracellular compartments in the presence of vasopressin. produced a phospho-specific antibody to this site and con-
These studies demonstrate regulated phosphorylation of both UT-A1 firmed that phosphorylation of this site is also increased by
and UT-A3 in response to vasopressin in a manner consistent with vasopressin.
coordinate regulation of UT-A and aquaporin-2 in the renal IMCD.
The findings add to prior evidence for vasopressin-induced phosphor-
METHODS
ylation of UT-A1, providing evidence that UT-A3 may be regulated
by phosphorylation as well. Experimental animals. Pathogen-free male Sprague-Dawley rats
phospho-specific antibody; confocal microscopy; immunofluores- (Taconic Farms, Germantown, NY) or homozygous Brattleboro rats
cence immunocytochemistry; immuno-electron microscopy (Harland Sprague-Dawley, Indianapolis, IN) weighing 150 –200 g
were used in these studies. All experiments were conducted in accord
with animal protocol H-0110R1 approved by the Animal Care and
THE RENAL INNER MEDULLARY collecting duct (IMCD) is the sole Use Committee of the National Heart, Lung, and Blood Institute or the
site of expression of two urea transport proteins, UT-A1 and boards of the Institute of Anatomy and Institute of Clinical Medicine,
UT-A3 (Fig. 1), which are derived from a single gene Aarhus University, according to the licenses for use of experimental
animals issued by the Danish Ministry of Justice.
(Slc14a2) by alternative splicing. Based on recent X-ray crys-
Phospho-specific UT-A antibodies. Rabbit polyclonal phospho-
tallography studies, these proteins probably function as urea specific UT-A1/3 antibodies were generated against 12-amino acid
channels (20), rather than carriers as previously believed. synthetic phosphopeptides surrounding rat Ser84 or Ser486 (see
These urea channels mediate transepithelial urea transport, NP_062220.2 for sequence). For each antibody, two rabbits were
which allows rapid equilibration of urea between the lumen of immunized and affinity-purified (PhosphoSolutions, Aurora, CA),
the IMCD and the inner medullary interstitium. Thus, urea, P7281 and P7282 for pSer84-UT-A1/3; P7283 and P7284 for
which is generated as the chief nitrogen waste product from pSer486-UT-A1. The specificities of these antibodies were tested by
protein metabolism, can be excreted at high rates without dot blotting. We also used a previously characterized rabbit antibody
impairing urine-concentrating ability (4, 12). Vasopressin L446 targeted for the NH2-terminal tail of both UT-A1 and UT-A3
(7). IgG concentration of each antibody was quantified by a NanoDrop
spectrophotometer (model ND-1000, NanoDrop Technologies, Wil-
Address for reprint requests and other correspondence: M. A. Knepper, 10 mington, DE; P7281, 50 ␮g/ml; P7282, 60 ␮g/ml; P7283, 90 ␮g/ml;
Center Dr., MSC 1603, Bldg. 10, Rm. 6N260, National Institutes of Health, P7284, 70 ␮g/ml; L446, 140 ␮g/ml). Other antibodies used in this
Bethesda, MD 20892-1603 (e-mail: knep@helix.nih.gov). paper were rabbit anti-aquaporin-2 (AQP2; K5007) (14), rabbit anti-
http://www.ajprenal.org F559
F560 UREA CHANNEL PHOSPHORYLATION AT Ser84 AND Ser486

Fig. 1. Phospho-Ser84 UT-A1/3 and phospho-Ser486 UT-A1 antibody recognition sites. The schematic drawing of UT-A1 and UT-A3 shows that the
NH2-terminal 1– 459 amino acids of UT-A1 are identical to UT-A3. The filled boxes represent hydrophobic regions that contain transmembrane domains of the
protein. The residue numbers indicated here and elsewhere in this paper correspond to the rat sequence (RefSeq no. NP_062220.2). The antibodies recognizing
phosphorylated Ser84 of both UT-A1 and UT-A3 (P7181 and P7282) were raised against a synthetic phosphopeptide targeting to Ser84. The antibody recognizing

Downloaded from http://ajprenal.physiology.org/ at EBSCO on April 7, 2013


phosphorylated Ser486 of UT-A1 (P7284) was raised against a synthetic phosphopeptide targeting to Ser486. A previously characterized antibody recognizing total
UT-A1 and UT-A3 (L446) was raised against amino acids 56 –78 (7, 38).

phospho-Ser256-AQP2 (14), and chicken anti-AQP2 (LL265) (3). All abdominal aorta. The kidneys were first perfused with PBS for 10 s to
of these antibodies have been previously characterized. wash out the blood, followed by ice-cold 4% paraformaldehyde for 5
IMCD suspensions. IMCD suspensions were prepared as previ- min. The fixed kidneys were trimmed to expose all three major
ously described with slight modification (9). After rats were eutha- regions (cortex, outer medulla, and inner medulla), embedded in
nized, kidney inner medullas were dissected, minced, and digested paraffin, and sectioned (4 ␮m) for immunofluorescence studies.
into suspensions by incubation at 37°C for 70 –90 min in digestion Immunofluorescence confocal microscopy. Immunostaining was
solution (250 mM sucrose, 10 mM triethanolamine, pH 7.6) contain- performed as previously described (43). In brief, paraffin-embedded
ing collagenase B (3 mg/ml; Roche, Indianapolis, IN) and hyaluron- whole kidney sections were dewaxed using xylene and rehydrated
idase (1,400 U/ml; Worthington, Lakewood, NJ). The resulting inner sequentially in 100, 95, 90, and 70% ethanol. Antigen retrieval was
medullary suspension (whole IM) was subjected to three low-speed performed with microwave treatment for 15 min in TEG buffer (10
centrifugations (at 70 g, 30 s) to separate the IMCD-enriched fraction mM Tris and 0.5 mM EGTA, pH 9.0) followed by neutralization in 50
in the pellet from the non-IMCD fraction in the supernatant. When all mM NH4Cl (in PBS). Blocking was performed using 1% BSA, 0.2%
three fractions were used, they were harvested by centrifugation at gelatin, and 0.05% saponin in PBS. Incubation with the primary
1,000 g for 5 min and washed once. The pellets were resuspended in antibody (diluted in 0.1% BSA and 0.3% Triton X-100 in PBS) was
1⫻ Laemmli buffer (1.5% SDS, 10 mM Tris, pH 6.8) containing performed overnight (4°C). After being washed with 0.1% BSA, 0.2%
protease and phosphatase inhibitors and passed through a DNA gelatin, and 0.05% saponin in PBS, tissue sections were incubated for
shredder (Qiagen, Valencia, CA). After protein concentration deter- 1 h with secondary antibody (conjugated with either Alexa 488 or
mination by the BCA method (Thermo Scientific, Rockford, IL), Alexa 568; Invitrogen) diluted in 0.1% BSA and 0.3% Triton X-100
samples were stored in the presence of 6% glycerol and 40 mM DTT in PBS. After subsequent washes with PBS, nuclei were counter-
after heating at 60°C for 15 min. When IMCD suspensions were used stained with DAPI (4 ␮l of 0.2 mg/ml DAPI stock solution diluted in
in physiological experiments, the IMCD-enriched pellet was gently 10 ml PBS). The sections were then preserved in fluorescence mount-
washed and resuspended in bicarbonate-buffered solution containing ing medium (S3023, Dako North America). For peptide blocking
(in mM) 118 NaCl, 4 Na2HPO4, 5 KCl, 25 NaHCO3, 2 CaCl2, 1.2 controls, antibody was preincubated with appropriate peptides at a
MgSO4, and 5.5 glucose (290 mosmol/kgH2O). IMCD suspensions 1:25 molar ratio for 2 h at 4°C before use. Sections were also
were allowed to equilibrate with gentle stirring with an aid of a incubated without primary antibody as a negative control. Confocal
micro-stirring bar at 37°C with 95% air-5% CO2 supply for 10 min fluorescence images were acquired using a Zeiss LSM 510 META
before use. Time course experiments were carried out by exposing microscope and software (Carl Zeiss MicroImaging, Thornwood, NY).
IMCD suspensions to 1-deamino-8-D-arginine vasopressin (dDAVP) Immunogold-electron microscopy. We carried out immunogold
or vehicle for various lengths of time (0, 1, 5, 15, 30 min). Hormone labeling of rat renal inner medulla tissues following the procedure
incubation was terminated by spinning the suspensions at 14,000 rpm described by Moeller et al. (27), with rats treated with dDAVP for 60
for 1 min to harvest the pellet containing IMCD segments. Samples min and control rats. Anti-pS84 (no. 7281, dilution 1:50), anti-pS486
were resuspended in 1⫻ Laemmli buffer and treated as described (no. 7284, dilution 1:50), and anti-UT-A1/3 (L446, dilution, 1:300)
above. were used.
Semiquantitative immunoblotting. Proteins were resolved by SDS- Statistical analysis. Data are presented as means ⫾ SE. All statis-
PAGE on polyacrylamide gels (Criterion, Bio-Rad, Hercules, CA) and tical comparisons were made by t-tests. P ⬍ 0.05 was considered
transferred electrophoretically onto nitrocellulose membranes. Mem- significant.
branes were blocked for 30 min with Odyssey blocking buffer (Li-
Cor, Lincoln, NE), rinsed, and probed with the respective affinity-
RESULTS
purified antibodies at proper dilution (in Odyssey blocking buffer
containing 0.1% Tween 20) overnight at 4°C. After 1-h incubation
Specificities of phospho-specific UT-A1/3 antibodies. Figure
with secondary antibody (Alexa Fluor 680 goat anti-rabbit immuno-
globulin G; Invitrogen, Carlsbad, CA) at 1:5,000 dilution, sites of 1 shows the locations of phosphoserines targeted by the phos-
antibody-antigen reaction were detected using an Odyssey infrared pho-specific antibodies. Figure 2 shows the results of dot
imager (Li-Cor). blotting testing the specificities of the antibodies. As shown,
Perfusion fixation of rat kidneys. Rats under anesthesia were phospho-Ser84 UT-A1/3 antibody P7282 recognized only the
surgically prepared for retrograde perfusion of the kidneys via the phosphopeptide targeted to Ser84 of UT-A1/3 but not the

AJP-Renal Physiol • VOL 299 • SEPTEMBER 2010 • www.ajprenal.org


UREA CHANNEL PHOSPHORYLATION AT Ser84 AND Ser486 F561
receptor-selective vasopressin analog dDAVP (1 nM, 30 min)
strongly increased the abundance of pSer84-UT-A1, pSer84-
UT-A3, and pSer486-UT-A1, but not total UT-A1/3, in IMCD,
confirming that phosphorylation of Ser84 of UT-A1 and
UT-A3 and Ser486 of UT-A1 is regulated by vasopressin.
Figure 3B confirms that the tubule fractionation procedure
successfully enriched the collecting duct-specific marker
AQP2 and deenriched the loop of Henle marker AQP1 in the
IMCD fractions. Preadsorption controls confirm the specifici-
ties of the phospho-specific antibodies (Fig. 3C), showing that
the same bands described above were ablated by preincubation
with the specific phospho-peptide. Note that in the pS486 blot,
there is a pair of bands at ⬃75 and ⬃55 kDa (labeled with
asterisks) that appear to be compatible with similar bands seen
Fig. 2. Specificities of phospho-Ser84 UT-A1/3 antibody (P7281 and P7282)
previously with the same antibody after limited chymotrypsin
and phospho-Ser486 UT-A1 antibody (P7283 and P7284). In each dot blot, cleavage of UT-A1 (7). Thus, it appears possible that a small

Downloaded from http://ajprenal.physiology.org/ at EBSCO on April 7, 2013


0.01 ␮g of phosphopeptides was loaded to the upper end of the nitrocellulose fraction of total UT-A1 may be subject to physiological pro-
membrane and corresponding nonphosphopeptides were loaded to the lower teolytic cleavage of the middle loop downstream from Ser486.
end. All 4 antibodies were used at a 1:1,000 dilution (see IgG concentrations Immunofluorescence labeling showed that both total and
in METHODS). Antibody P7282 and P7284 recognized phosphopeptides but not
nonphosphopeptides. phosphorylated UT-A1/3 (both sites) are specifically localized
to IMCD cells, with no labeling of other cell types (Fig. 4).
Within the IMCD cell, phospho-UT-A1/3 were found in both
corresponding nonphosphopeptide. Antibody P7281 was sim- the cytoplasm and apical region of the cell, a result consistent
ilar in specificity and produced only a very faint nonphos- with previous immunolocalization studies of UT-A1 (30) or
phopeptide signal that is barely visible in Fig. 2. Antibody UT-A3 (38).
P7282 was consequently used in immunoblotting. Antibody Time course of UT-A1 and UT-A3 phosphorylation. Previ-
P7281, however, yielded better immunostaining in tissue sec- ously, using [32P] radioisotope labeling in IMCD suspensions,
tions than P7282, so it was used in immunofluorescence studies Zhang et al. (44) demonstrated that vasopressin could rapidly
only with appropriate labeling controls. Similarly, one phos- (within 2–5 min) increase phosphorylation of UT-A1. To test
pho-Ser486-UT-A1 antibody (P7284) showed specificity for its whether vasopressin stimulates phosphorylation of Ser84 or
phosphopeptide. Antibody P7284 was used in both immuno- Ser486 of UT-A1 or UT-A3 in a similar fashion, IMCD
blotting and immunofluorescence studies. suspensions were incubated with dDAVP for various lengths of
Phospho-UT-A1 and UT-A3 localization in rat kidney. Figure 3A time. As shown in Fig. 5A, there was a substantial increase in
shows immunoblots testing for the presence of phosphorylated phosphorylation of UT-A1 (117- and 97-kDa bands) at Ser84
UT-A1 and UT-A3 proteins in a whole inner medullary sus- and Ser486 in response to dDAVP, beginning at the shortest
pension and two subfractions, non-IMCD and IMCD-enriched incubation time (1 min) and persisting throughout the entire
(first 3 lanes of each blot). The results show that all three experimental period (30 min). (Note that the ratio of the upper
phosphorylation sites, pSer84-UT-A1, pSer84-UT-A3, and UT-A1 band density to the lower UT-A1 band density appears
pSer486-UT-A1, are enriched in the IMCD fraction, consistent to be greater for Ser486-phosphorylated UT-A1 than for Ser84-
with the known exclusive distribution of UT-A1 and UT-A3 in phosphorylated UT-A1.) Similarly, a rapid increase in phos-
IMCD (30, 38). Previous studies of UT-A1 demonstrated that phorylation can be seen at Ser84 in UT-A3. In contrast, there
it exists in the IMCD chiefly at two distinct molecular weights, was no change in the abundance of total UT-A1 or UT-A3 in
97 and 117 kDa (on 10% polyacrylamide gel), that represent response to dDAVP. Phosphorylation of AQP2 at Ser256 was
two distinct levels of glycosylation (deglycosylated UT-A1 ran also increased within 1 min of dDAVP addition as previously
at 88 kDa.) (7). In Fig. 3A, these two previously identified demonstrated (8, 15). Figure 5B summarizes the time course
bands are indicated by arrows labeled “UT-A1.” Also, previous data graphically, showing the magnitude of UT-A1 and UT-A3
studies of UT-A3 demonstrated that it is present chiefly at two phosphorylation induced by dDAVP as log2(dDAVP:control)
distinct molecular weights, 67 and 44 kDa, that represent two as a function of time (n ⫽ 3).
distinct levels of glycosylation (deglycosylated UT-A3 ran at In vivo UT-A1 and UT-A3 phosphorylation in response to
40 kDa) (38). In Fig. 3A, these previously identified bands are vasopressin. We next tested whether these phosphorylation
indicated by arrows labeled “UT-A3.” Both phospho-specific events can occur in the animal. The experiment was conducted
antibodies P7282 and P7284 recognized pSer84-UT-A1 or on six Sprague-Dawley rats given 200 mM sucrose for drink-
pSer486-UT-A1 as a higher band at 117 kDa and a lower band ing for 16 h before the experiment to lower the baseline level
at 97 kDa, consistent with previous observations showing that of phospho-UT-A. Thereafter, three rats received an intramus-
these bands represent two different glycosylation states (7). cular injection of 2 nmol dDAVP 1 h before euthanasia and the
The phospho-specific antibody P7282 also recognized pSer84- other three rats received saline as control. Rats receiving
UT-A3 at two molecular weights, ⬃67 and ⬃44 kDa, consis- dDAVP treatment had a large increase in urine osmolality
tent with what was reported previously for UT-A3 (38) and (before: 184 ⫾ 13, after: 1,389 ⫾ 65 mosmol/kgH2O, n ⫽ 3,
confirmed here in an immunoblot using an antibody to total P ⬍ 0.0001), whereas the control rats had only a small increase
UT-A1/3 (antibody LL446; Fig. 3A, right). Furthermore, as in urine osmolality over the 1-h waiting period (before: 155 ⫾
shown in the last two lanes of each blot in Fig. 3A, V2 3, after: 297 ⫾ 3 mosmol/kgH2O, n ⫽ 3, P ⬍ 0.05). Immu-

AJP-Renal Physiol • VOL 299 • SEPTEMBER 2010 • www.ajprenal.org


F562 UREA CHANNEL PHOSPHORYLATION AT Ser84 AND Ser486

Fig. 3. Immunoblotting using anti-phospho-Ser84


UT-A1 and UT-A3 and anti-phospho-Ser486
UT-A1 antibodies in rat inner medullary samples.
A: in each blot, whole inner medullary suspension
(whole IM) and 2 subfractions, non-inner medul-
lary collecting duct (non-IMCD) and IMCD,
were loaded (first 3 lanes) to test the presence of
phosphorylated or nonphosphorylated UT-A1 and
UT-A3 in IMCD. In addition, IMCD samples
with and without treatment of 1-deamino-8-D-
arginine vasopressin (dDAVP; 1 nM, 30 min)
were loaded (last 2 lanes) to test the effect of
vasopressin on phosphorylation of UT-A1 and
UT-A3 on targeted sites. Samples were loaded on
a 4 –15% polyacrylamide gel. Loading was 30 ␮g
per lane except for dDAVP-treated IMCD which
was 20 ␮g. Antibodies P7282, P7284, and L446
labeled UT-A1 as 117- and 97-kDa bands (ar-
rows) as previously found (7). Antibodies P7282
and L446 labeled UT-A3 as broad ladder-like 67-

Downloaded from http://ajprenal.physiology.org/ at EBSCO on April 7, 2013


and 44-kDa bands (arrows), as prevously found
(38). All antibodies, P7282, P7284, and L446,
were used at a 1:1,000 dilution. B: an enrichment
test for IMCD suspension preparation using col-
lecting duct aquaporin (AQP2) as a marker for
IMCD and thin descending limb aquaporin
(AQP1) as a marker for non-IMCD. C: pread-
sorption control. Twenty micrograms IMCD sam-
ples treated with dDAVP were loaded per lane on
a 12.5% polyacrylamide gel. Membranes were
incubated with antibody with or without pread-
sorption with appropriate peptides for 4 h at room
temperature before use. Peptide concentrations:
0.1 ␮g/ml. *Bands compatible with physiological
proteolytic cleavage of UT-A1 (7).

noblotting was performed on whole homogenates of the ter- 7C). Preabsorption with the Ser84 phosphopeptide fully ab-
minal 80% of kidney inner medulla (the papilla). The results lated the signal (Fig. 7A). Similar results were obtained for
show that inner medullas of dDAVP-treated rats have signifi- pSer486-UT-A1 as shown in Fig. 7, D–F. Again, preabsorption
cantly higher abundances of all three phospho-UT-A proteins, with the Ser486 phosphopeptide fully ablated the signal (Fig. 7D).
pSer84-UT-A1, pSer84-UT-A3, and pSer486-UT-A1, than in- Immunogold-EM localization of phosphorylated urea chan-
ner medullas from control rats (Fig. 6A). Figure 6B graphically nel proteins. Immunogold-EM localization of pSer84- and
depicts the magnitude of UT-A phosphorylation induced by pSer486-phosphorylated urea transporters in rat inner medulla
dDAVP expressed as log2(dDAVP:control). showed no appreciable labeling in the absence of dDAVP
An in vivo dDAVP response experiment was also conducted pretreatment (not shown). After 60-min dDAVP (1 ng) treat-
in Brattleboro rats, a natural strain of rat that lacks endog- ment of rats, there was abundant labeling of IMCD cells, the
enously circulating vasopressin, to examine the immunolocal- majority of which was intracellular (Fig. 8). A large proportion
ization of phosphorylated UT-A1/3 in kidney inner medulla of the gold particles was observed in the Golgi complexes and
before and after vasopressin stimulation. Figure 7 shows a Golgi vesicles in the apical region of IMCD cells. Only a few
seemingly “all or none” pattern of pSer84-UT-A1/3 in cyto- gold particles were observed in direct association with the
plasm and the apical region of Brattleboro rat IMCD after apical plasma membrane. Some pSer-84 gold particles were
vasopressin stimulation (Fig. 7, A and B). Vasopressin-induced observed within the basolateral membrane infoldings (not
redistribution of AQP2 from intracellular domain to apical shown). Using a UT-A antibody that recognizes both UT-A1
plasma membrane was also shown as a positive control (Fig. and UT-A3 total protein, abundant plasma membrane labeling,

Fig. 4. Immunofluorescence localization of phos-


phorylated UT-A1/3 in Sprague-Dawley rat kidney
inner medulla. Phospho-Ser84 UT-A1/3 (left) and
phospho-Ser486 UT-A1 proteins (middle), as well
as total UT-A1/3 (right), are present only in col-
lecting duct cells. P7281 and P7284 antibodies
were used at a 1:200 dilution; L446 was used at a
1:100 dilution. (Preadsorption controls are shown
below in Figs. 7 and 8.)

AJP-Renal Physiol • VOL 299 • SEPTEMBER 2010 • www.ajprenal.org


UREA CHANNEL PHOSPHORYLATION AT Ser84 AND Ser486 F563
including both the apical plasma membrane and basolateral
plasma membranes, was detected.

DISCUSSION

In this paper, we used two new phospho-specific antibodies


recognizing phosphorylated serines in the collecting duct urea
transporters UT-A1 and UT-A3 to investigate vasopressin-
mediated regulation of urea transporter phosphorylation. One

Downloaded from http://ajprenal.physiology.org/ at EBSCO on April 7, 2013

Fig. 5. Time course of UT-A1 and UT-A3 phosphorylation in response to


dDAVP in rat IMCD suspensions. A: representative immunoblots for pSer84-
UT-A1, pSer84-UT-A3, pSer486-UT-A1, and total UT-A1/3 in control and Fig. 6. In vivo phosphorylation of UT-A1 and UT-A3 in response to dDAVP.
dDAVP-treated IMCD suspensions prepared from Sprague-Dawley rats. At Experiment was done on 6 water-loaded Sprague-Dawley rats. Three rats
each time point, IMCD treated with 1 nM dDAVP had a substantially higher received intramuscular injection of 2 nmol dDAVP 1 h before euthanasia and
UT-A1 and UT-A3 phosphorylation level compared with its control, whereas 3 rats received saline only as control. Terminal inner medullas were dissected
the total UT-A1 or UT-A3 did not significantly change. Phosphorylation of and homogenized for immunoblotting. A: immunoblots displaying phosphor-
AQP2 at Ser256 was also measured as a positive control to demonstrate the ylation changes in response to dDAVP in whole inner medulla homogenates.
efficacy of the dDAVP. Thirty micrograms of IMCD protein were loaded per Thirty micrograms of sample protein were loaded per lane on a 12.5%
lane on a 12.5% polyacrylamide gel. B: summary line graph depicts the polyacrylamide gel. Inner medullary whole homegenates from dDAVP-treated
changes in protein abundance of each protein, expressed as log2(dDAVP: rats had significantly higher levels of pSer84-UT-A1, pSer84-UT-A3, and
control; n ⫽ 3). Band densities were quantified by drawing a single integration pSer486-UT-A1 than control rats. B: summary of quantification of immuno-
box around both bands for UT-A1 and both bands for UT-A3. blots for phospho-UT-A1 and UT-A3 presented as log2(dDAVP:control). Band
densities were quantified by drawing a single integration box around both
bands for UT-A1 and both bands for UT-A3.

AJP-Renal Physiol • VOL 299 • SEPTEMBER 2010 • www.ajprenal.org


F564 UREA CHANNEL PHOSPHORYLATION AT Ser84 AND Ser486

Downloaded from http://ajprenal.physiology.org/ at EBSCO on April 7, 2013


Fig. 7. Immunofluorescence localization of pSer84-UT-A1/3 (A–C) and pS486-UT-A1 (D–F) in Brattleboro rat kidney inner medulla– effect of dDAVP. A: test
of antibody (P7281) specificity using a peptide blocking assay (blue: nuclei, green: pSer84-UT-A1/3). Tissue sections were preincubated with antibody with or
without preadsorption with Ser84 phosphopeptide for 2 h at 4°C before use. P7281 antibody was used at a 1:50 dilution. Peptide concentration: 0.5 ␮g/ml.
Phosphorylation at Ser84 was detected in kidney sections obtained from dDAVP-treated rats but not vehicle-treated rats. Preadsorption with immunizing
phosphopeptide ablated signal. B: immunolocalization of pSer84-UT-A1/3 and total UT-A1/3 in consecutive kidney sections. pSer84-UT-A1/3 and UT-A1/3
were detected in collecting ducts only. Both antibodies were used at a 1:50 dilution. C: double labeling for pSer84-UT-A1/3 (green) and AQP2 (red). After
dDAVP treatment, AQP2 is mainly apical, whereas pSer84-UT-A1/3 is localized both apically and intracellularly. AQP2 antibody was used at a 1:100 dilution.
A–C: identical confocal microscope settings were used to visualize vehicle and dDAVP-treated samples. Immunofluorescence using pS486-UT-A1 antibody was
done in a manner similar to pS84-UT-A1/3 antibody as described above. D: test of antibody (P7284) specificity using a peptide-blocking assay (blue: nuclei,
green: pSer486-UT-A1). Phosphorylation at Ser486 was detected in kidney sections obtained from dDAVP-treated rats but not vehicle-treated rats. Preadsorption
with immunizing phosphopeptide ablated signal. E: immunolabeling of pSer486-UT-A1 and total UT-A1/3 in consecutive kidney sections. pSer486-UT-A1/3
and UT-A1/3 were detected in collecting ducts only. F: double labeling for pSer486-UT-A1 (green) and AQP2 (red). After dDAVP treatment, AQP2 is mainly
apical, whereas pSer486-UT-A1 is localized both apically and intracellularly.

antibody recognizes the phosphorylated Ser84 site present in Ser84 and Ser486. It is at present unclear whether UT-A1 and
both UT-A1 and UT-A3, which was demonstrated by LC- UT-A3 function independently as separate transporters or are
MS/MS analysis of rat collecting ducts by Bansal et al. (2). The part of a more complex structure combining UT-A1 and
second antibody recognizes the phosphorylated Ser486 site, UT-A3 in a single heteromeric transporter. However, both can
present in UT-A1 but not UT-A3, which was demonstrated function independently as homomeric transporters with heter-
originally by Hoffert et al. (16) and has been extensively ologous expression in oocytes (6, 13, 24) or cultured cells (6,
studied by Blount et al. (6). The latter study used mutational 36). UT-A1 and UT-A3 have been shown to be coregulated, at
analysis to infer a role for the Ser486 phosphorylation in the least in long-term regulation of transporter abundance. Chronic
regulation of UT-A1. conditions associated with downregulation of both UT-A1 and
UT-A1 and UT-A3 are proteins of 929 and 460 amino acids UT-A3 include potassium depletion (19), ureteral obstruction
(Fig. 1) that are derived from the same gene (Slc14a2) as a (21), extracellular fluid volume expansion (41), and glucocor-
result of incorporation of alternative exons that contain either ticoid excess (22). Also, Dahl salt-sensitive rats show increased
a stop codon in the case of the shorter UT-A3 (exon 12) or a levels of both UT-A1 and UT-A3 compared with control rats
full open reading frame that allows splicing to exon 14 in the (11). However, studies of different hydration states in animals
case of the longer UT-A1 (23 exons) (28, 33, 35). Since exons showed that UT-A1 and UT-A3 expression can be regulated
1–11 are the same in UT-A1 and UT-A3, the corresponding differently (1, 23).
amino acid sequence is the same for the first 459 amino acids Since Ser84 is present in both UT-A1 and UT-A3, it was of
of both proteins. Our L446 antibody was made to recognize the particular interest to determine whether the increased abun-
NH2-terminal tail of both proteins allowing assessment of both dance of phosphorylated Ser84 demonstrated by mass spec-
UT-A1 and UT-A3 abundances on one immunoblot. The two trometry (2) was due to vasopressin-induced phosphorylation
are discriminated on the basis of size (Fig. 3). Figure 1 of either UT-A1, UT-A3, or both. The results shown in Figs. 3
illustrates the relationship between the two proteins and labels and 5 demonstrated that indeed phosphorylation of this site is
the two phosphorylation sites under study in this paper, viz markedly increased in both urea transporters in response to the

AJP-Renal Physiol • VOL 299 • SEPTEMBER 2010 • www.ajprenal.org


UREA CHANNEL PHOSPHORYLATION AT Ser84 AND Ser486 F565

Fig. 8. Immunogold electron microscopy dem-

Downloaded from http://ajprenal.physiology.org/ at EBSCO on April 7, 2013


onstrates that pSer84 UT-A and pSer486 UT-A
are predominantly associated with intracellular
compartments. A: overview of an IMCD cell in
midregion of inner medulla labeled with
pSer84 UT-A. B: higher magnification of the
area highlighted in A. Few gold particles are
directly associated with the apical plasma
membrane (apm). Abundant labeling is ob-
served in the subapical Golgi apparatus (g) and
intracellular vesicles. C: further example of the
association of pSer84 UT-A with the subapical
Golgi apparatus. D: overview of an IMCD cell
in midregion of inner medulla labeled with
pSer486 UT-A. E: higher magnification of the
area highlighted in D. Few gold particles are
directly associated with the apm. Abundant
labeling is observed in subapical vesicles.
F: pSer486 UT-A is also abundant in the sub-
apical Golgi apparatus. G: overview of a prin-
cipal cell in IMCD labeled with L446 (UT-
A1/3 specific antibody). Inset: area highlighted
in box 1 and demonstrates labeling of the
subapical Golgi apparatus. H: area corresponds
to box 2 in G and demonstrates abundant la-
beling of the apical plasma membrane. I: area
corresponds to box 3 in G and demonstrates
that some gold particles are also observed in
the basolateral plasma membrane. In all im-
ages, gold particles are 10 nm in diameter.

V2 receptor vasopressin analog dDAVP. Furthermore, this of both Ser84 and Ser486 of UT-A1, and Ser84 of UT-A3, in
result demonstrates regulated phosphorylation of UT-A3, IMCD suspensions indicates a very rapid response, within 1
which has not been known previously to be a target for min of vasopressin addition. This finding is coherent with time
regulated phosphorylation. The time course of phosphorylation course studies of urea transport in isolated, perfused IMCD

AJP-Renal Physiol • VOL 299 • SEPTEMBER 2010 • www.ajprenal.org


F566 UREA CHANNEL PHOSPHORYLATION AT Ser84 AND Ser486

segments showing the initial rise in urea permeability ⬃40 s T)-␾, where ␾ is a hydrophobic amino acid (25). MLCK has
after vasopressin exposure (40). It is also consistent with only one known target, viz. myosin regulatory light chain A
UT-A1 phosphorylation studies using [32P] incorporation to and B isoforms where Thr18 and Ser19 are phosphorylated
show increased phosphorylation in the same time frame (44). (sequence PQRATSNVFA) (17, 18). The signature for Erk1/
The time course was similar for both sites and for Ser84 in both 2-mediated phosphorylation has been reported to be x-P-x-(S/
UT-A1 and UT-A3, suggesting that the same signaling path- T)-P-x (proline moieties both up- and downstream from target
ways were rate limiting in each case. In addition, the time Ser or Thr) (25). No ERK1/2 candidate sites have been iden-
course of AQP2 phosphorylation at Ser256 has been shown to tified in UT-A1 or UT-A3 by mass spectrometry as of this
be similar (8, 15). Previous studies suggested that this rate- writing, although other critical sites may remain undiscovered
limiting process for the vasopressin response is at or before the (42). In addition, because inhibitors of calmodulin block some
level of cAMP generation (29, 40). aspects of vasopressin signaling (10), it is worthwhile to
We also confirmed the strong regulation of phosphorylation consider CaM-kinase II which is strongly expressed in the
at Ser84 and Ser486 using immunohistochemistry. In general, IMCD (39) as a candidate for phosphorylation at Ser84 and
with confocal immunofluorescence, only IMCD cells were Ser486 of UT-A1/3. The CaM-kinase II target signature has
labeled and labeling included both the cell periphery and been reported to be x-R-x-x-(S/T)-x-x. Both Ser84 and Ser486
intracellular regions. Phosphorylated UT-A1/3 is not restricted have a basic amino acid in the -3 position, making them

Downloaded from http://ajprenal.physiology.org/ at EBSCO on April 7, 2013


to the apical plasma membrane, contrary to what was seen compatible with CaM-kinase II targets. Furthermore, Rinschen
previously in the case of phosphorylated AQP2 at Ser269 (14, et al. (32) recently reported that vasopressin activates CaM
27). Thus, the results do not provide evidence pointing to a kinase II in collecting duct cells. Finally, Stewart et al. (37)
specific role of phosphorylation of UT-A1 or UT-A3 in plasma proposed that urea transport by UT-A3 may be regulated via
membrane retention or regulation of endocytosis as for Ser269 phosphorylation by an acidophilic kinase casein kinase II. Its
phosphorylation of AQP2 (26). Supporting this view were our target motif signature is x-x-(S/T)-x-x-(E/D)-x (25). The only
findings from immunogold electron microscopy using the two demonstrated phosphorylation sites in UT-A1/3 that are com-
antibodies that showed virtually exclusive presence of phos- patible with this motif are Ser-62 and Ser-63 (DLRSSDEDS),
phorylated UT-A in intracellular compartments, with much of although this site has not been found to show an increase in
the intracellular labeling associated with the Golgi apparatus phosphorylation in response to vasopressin (2).
and Golgi vesicles in the apical region of the cells (Fig. 8). Our PKA is capable of phosphorylating both Ser84 and Ser486
data highlight the need for high-resolution imaging techniques in vitro (2) but such a demonstration does not imply that PKA
for confirmation of confocal microscopy studies, especially is the only or even the chief kinase responsible for phosphor-
when drawing conclusions regarding subcellular distribution. It ylation at these sites. However, Zhang et al. (44) demonstrated
remains possible that the phosphorylation at either site plays a that the moderately specific PKA inhibitor H-89 blocked va-
role in exocytosis of UT-A1 or UT-A3 as proposed by Blount sopressin-induced phosphorylation of UT-A1, pointing to a
et al. (6) and that the phosphorylation is rapidly removed on role for PKA in regulation of sites present in UT-A1, presum-
delivery of the urea channels to the plasma membrane. Another ably Ser486 and/or Ser499.
possibility is that some portion of plasma membrane UT
channels is phosphorylated, but that the phosphorylation is ACKNOWLEDGMENTS
masked by proteins that bind to the phosphorylation sites or We acknowledge the professional skills and advice of Dr. C. A. Combs
due to conformational changes in channel structure. [Light Microscopy Core Facility, National Heart, Lung, and Blood Institute
The amino acid sequence surrounding the Ser84 phosphoryla- (NHLBI)] regarding light microscopy-related data in this paper. E. Merete
Locke is thanked for excellent technical assistance in immunogold electron
tion site is SKRRESELPRRA (Ser84 is underlined). As can be microscopy.
seen, the Ser84 site is in a basic region of the protein (surrounded
by arginine and lysine moieties up- and downstream). Thus, it is GRANTS
likely that Ser84 is phosphorylated by one or more “basophilic” This study was supported by the Intramural Budget of the NHLBI (Project
kinases (25). The amino acid sequence surrounding the Ser486 Z01-HL-001285). S. Hwang was supported by an American Physiological
phosphorylation site is FPRRKSVFHIE (Ser486 is underlined). Society Undergraduate Summer Research Fellowship. M. M. Rinschen was
Thus, Ser486 is also in a basic region with arginines and supported by the Biomedical Exchange Program, Hannover and the Braun
Foundation, Melsungen. R. A. Fenton is funded by the Danish Medical
lysines upstream from the phosphorylation site. This again Research Council, The Lundbeck Foundation, and the Novo Nordisk Founda-
points to basophilic kinases as candidates for phosphorylation tion.
of Ser486. Vasopressin is known to signal in part through
activation of the basophilic kinase protein kinase A (PKA). DISCLOSURES
The phosphorylation motif signature for PKA is x-(R/K)-(R/ No conflicts of interest, financial or otherwise, are declared by the author(s).
K)-x-(S/T)-x (25). (This terminology gives the amino acid
sequence surrounding the target serine or threonine where R/K REFERENCES
means either arginine or lysine.) In collecting ducts, vasopres- 1. Bagnasco SM, Peng T, Nakayama Y, Sands JM. Differential expression
sin has also been found to signal via pathways involving of individual UT-A urea transporter isoforms in rat kidney. J Am Soc
several additional kinases including myosin light chain kinase Nephrol 11: 1980 –1986, 2000.
(MLCK) (8), Akt or protein kinase B (31), and ERK1/2 (31). 2. Bansal AD, Hoffert JD, Pisitkun T, Hwang S, Chou CL, Boja ES,
Among these, only Akt has the phosphorylation motif signa- Wang G, Knepper MA. Phosphoproteomic profiling reveals vasopressin-
regulated phosphorylation sites in collecting duct. J Am Soc Nephrol 21:
ture to be a candidate for the two phosphorylation sites under 303–315, 2010.
study in this paper, i.e., Ser84 and Ser486 of urea transporters. 3. Barile M, Pisitkun T, Yu MJ, Chou CL, Verbalis MJ, Shen RF,
The Akt phosphorylation target signature is R-x-R-x-x-(S/ Knepper MA. Large scale protein identification in intracellular aqua-

AJP-Renal Physiol • VOL 299 • SEPTEMBER 2010 • www.ajprenal.org


UREA CHANNEL PHOSPHORYLATION AT Ser84 AND Ser486 F567
porin-2 vesicles from renal inner medullary collecting duct. Mol Cell 24. Maciver B, Smith CP, Hill WG, Zeidel ML. Functional characterization
Proteomics 4: 1095–1106, 2005. of mouse urea transporters UT-A2 and UT-A3 expressed in purified
4. Berliner RW, Levinsky NG, Davidson DG, Eden M. Dilution and Xenopus laevis oocyte plasma membranes. Am J Physiol Renal Physiol
concentration of the urine and the action of antidiuretic hormone. Am J 294: F956 –F964, 2008.
Med 27: 730 –744, 1958. 25. Miller ML, Jensen LJ, Diella F, Jorgensen C, Tinti M, Li L, Hsiung
5. Blount MA, Klein JD, Martin CF, Tchapyjnikov D, Sands JM. M, Parker SA, Bordeaux J, Sicheritz-Ponten T, Olhovsky M, Pas-
Forskolin stimulates phosphorylation and membrane accumulation of culescu A, Alexander J, Knapp S, Blom N, Bork P, Li S, Cesareni G,
UT-A3. Am J Physiol Renal Physiol 293: F1308 –F1313, 2007. Pawson T, Turk BE, Yaffe MB, Brunak S, Linding R. Linear motif
6. Blount MA, Mistry AC, Frohlich O, Price SR, Chen G, Sands JM, atlas for phosphorylation-dependent signaling. Sci Signal 1: ra2, 2008.
Klein JD. Phosphorylation of UT-A1 urea transporter at serines 486 and 26. Moeller HB, Praetorius J, Rutzler MR, Fenton RA. Phosphorylation of
499 is important for vasopressin-regulated activity and membrane accu- aquaporin-2 regulates its endocytosis and protein-protein interactions.
mulation. Am J Physiol Renal Physiol 295: F295–F299, 2008. Proc Natl Acad Sci USA 107: 424 –429, 2010.
7. Bradford AD, Terris JM, Ecelbarger CA, Klein JD, Sands JM, Chou 27. Moeller HB, Knepper MA, Fenton RA. Serine 269 phosphorylated
CL, Knepper MA. 97- And 117-kDa forms of collecting duct urea aquaporin-2 is targeted to the apical membrane of collecting duct principal
transporter UT-A1 are due to different states of glycosylation. Am J cells. Kidney Int 75: 295–303, 2008.
Physiol Renal Physiol 281: F133–F143, 2001. 28. Nakayama Y, Naruse M, Karakashian A, Peng T, Sands JM, Bag-
8. Chou CL, Christensen BM, Frische S, Vorum H, Desai RA, Hoffert nasco SM. Cloning of the rat Slc14a2 gene and genomic organization of
JD, de Lanerolle P, Nielsen S, Knepper MA. Nonmuscle myosin II and the UT-A urea transporter. Biochim Biophys Acta 1518: 19 –26, 2001.
myosin light chain kinase are downstream targets for vasopressin signaling 29. Nielsen S, Knepper MA. Vasopressin activates collecting duct urea
in the renal collecting duct. J Biol Chem 279: 49026 –49035, 2004. transporters and water channels by distinct physical processes. Am J

Downloaded from http://ajprenal.physiology.org/ at EBSCO on April 7, 2013


9. Chou CL, Rapko SI, Knepper MA. Phosphoinositide signaling in rat Physiol Renal Fluid Electrolyte Physiol 265: F204 –F213, 1993.
inner medullary collecting duct. Am J Physiol Renal Physiol 274: F564 – 30. Nielsen S, Terris J, Smith CP, Hediger MA, Ecelbarger CA, Knepper
F572, 1998. MA. Cellular and subcellular localization of the vasopressin-regulated
10. Chou CL, Yip KP, Knepper MA. Role of Ca/calmodulin in vasopressin- urea transporter in rat kidney. Proc Natl Acad Sci USA 93: 5495–5500,
stimulated aquaporin-2 trafficking in rat collecting duct. J Am Soc Nephrol 1996.
10: 13A, 1999. 31. Pisitkun T, Jacob V, Schleicher SM, Chou CL, Yu MJ, Knepper MA.
11. Fenton RA, Chou CL, Ageloff S, Brandt W, Stokes JB, Knepper MA. Akt and ERK1/2 pathways are components of the vasopressin signaling
Increased collecting duct urea transporter expression in Dahl salt-sensitive network in rat native IMCD. Am J Physiol Renal Physiol 295: F1030 –
rats. Am J Physiol Renal Physiol 285: F143–F151, 2003. F1043, 2008.
12. Fenton RA, Knepper MA. Urea and renal function in the 21st century: 32. Rinschen MM, Yu MJ, Wang G, Boja ES, Hoffert JD, Pisitkun T,
insights from knockout mice. J Am Soc Nephrol 18: 679 –688, 2007. Knepper MA. Quantitative phosphoproteomic analysis reveals vasopres-
13. Fenton RA, Stewart GS, Carpenter B, Howorth A, Potter EA, Cooper sin V2 receptor-dependent signaling pathways in renal collecting duct
GJ, Smith CP. Characterization of mouse urea transporters UT-A1 and cells. Proc Natl Acad Sci USA 107: 3882–3887, 2010.
UT-A2. Am J Physiol Renal Physiol 283: F817–F825, 2002. 33. Sands JM. Molecular approaches to urea transporters. J Am Soc Nephrol
14. Hoffert JD, Fenton RA, Moeller HB, Simons B, Tchapyjnikov D, 13: 2795–2806, 2002.
McDill BW, Yu MJ, Pisitkun T, Chen F, Knepper MA. Vasopressin- 34. Sands JM, Nonoguchi H, Knepper MA. Vasopressin effects on urea and
stimulated increase in phosphorylation at Ser269 potentiates plasma mem- H2O transport in inner medullary collecting duct subsegments. Am J
brane retention of aquaporin-2. J Biol Chem 283: 24617–24627, 2008. Physiol Renal Fluid Electrolyte Physiol 253: F823–F832, 1987.
15. Hoffert JD, Nielsen J, Yu MJ, Pisitkun T, Schleicher SM, Nielsen S, 35. Smith CP, Fenton RA. Genomic organization of the mammalian
Knepper MA. Dynamics of aquaporin-2 serine-261 phosphorylation in SLC14a2 urea transporter genes. J Membr Biol 212: 109 –117, 2006.
response to short-term vasopressin treatment in collecting duct. Am J 36. Stewart GS, King SL, Potter EA, Smith CP. Acute regulation of
Physiol Renal Physiol 292: F691–F700, 2007. mUT-A3 urea transporter expressed in a MDCK cell line. Am J Physiol
16. Hoffert JD, Pisitkun T, Wang G, Shen RF, Knepper MA. Quantitative Renal Physiol 292: F1157–F1163, 2007.
phosphoproteomics of vasopressin-sensitive renal cells: regulation of 37. Stewart GS, Thistlethwaite A, Lees H, Cooper GJ, Smith C. Vasopres-
aquaporin-2 phosphorylation at two sites. Proc Natl Acad Sci USA 103: sin regulation of the renal UT-A3 urea transporter. Am J Physiol Renal
7159 –7164, 2006. Physiol 296: F642–F648, 2009.
17. Ikebe M, Hartshorne DJ. Phosphorylation of smooth muscle myosin at 38. Terris JM, Knepper MA, Wade JB. UT-A3: localization and character-
two distinct sites by myosin light chain kinase. J Biol Chem 260: ization of an additional urea transporter isoform in the IMCD. Am J
10027–10031, 1985. Physiol Renal Physiol 280: F325–F332, 2001.
18. Ikebe M, Hartshorne DJ, Elzinga M. Identification, phosphorylation, 39. Uawithya P, Pisitkun T, Ruttenberg BE, Knepper MA. Transcriptional
and dephosphorylation of a second site for myosin light chain kinase on profiling of native inner medullary collecting duct cells from rat kidney.
the 20,000-dalton light chain of smooth muscle myosin. J Biol Chem 261: Physiol Genomics 32: 229 –253, 2008.
36 –39, 1986. 40. Wall SM, Han JS, Chou CL, Knepper MA. Kinetics of urea and water
19. Jung JY, Madsen KM, Han KH, Yang CW, Knepper MA, Sands JM, permeability activation by vasopressin in rat terminal IMCD. Am J Physiol
Kim J. Expression of urea transporters in potassium-depleted mouse Renal Fluid Electrolyte Physiol 262: F989 –F998, 1992.
kidney. Am J Physiol Renal Physiol 285: F1210 –F1224, 2003. 41. Wang XY, Beutler K, Nielsen J, Nielsen S, Knepper MA, Masilamani
20. Knepper MA, Mindell JA. Structural biology: molecular coin slots for S. Decreased abundance of collecting duct urea transporters UT-A1 and
urea. Nature 462: 733–734, 2009. UT-A3 with ECF volume expansion. Am J Physiol Renal Physiol 282:
21. Li C, Klein JD, Wang W, Knepper MA, Nielsen S, Sands JM, Frokiær F577–F584, 2002.
J. Altered expression of urea transporters in response to ureteral obstruc- 42. Wang Y, Klein JD, Blount MA, Martin CF, Kent KJ, Pech V, Wall
tion. Am J Physiol Renal Physiol 286: F1154 –F1162, 2004. SM, Sands JM. Epac regulates UT-A1 to increase urea transport in inner
22. Li C, Wang W, Summer SN, Falk S, Schrier RW. Downregulation of medullary collecting ducts. J Am Soc Nephrol 20: 2018 –2024, 2009.
UT-A1/UT-A3 is associated with urinary concentrating defect in glucocor- 43. Yu MJ, Pisitkun T, Wang G, Shen RF, Knepper MA. LC-MS/MS
ticoid-excess state. J Am Soc Nephrol 19: 1975–1981, 2008. analysis of apical and basolateral plasma membranes of rat renal collecting
23. Lim SW, Han KH, Jung JY, Kim WY, Yang CW, Sands JM, Knepper duct cells. Mol Cell Proteomics 5: 2131–2145, 2006.
MA, Madsen KM, Kim J. Ultrastructural localization of UT-A and UT-B 44. Zhang C, Sands JM, Klein JD. Vasopressin rapidly increases the
in rat kidneys with different hydration status. Am J Physiol Regul Integr phosphorylation of the UT-A1 urea transporter in rat IMCDs through
Comp Physiol 290: R479 –R492, 2006. PKA. Am J Physiol Renal Physiol 282: F85–F90, 2002.

AJP-Renal Physiol • VOL 299 • SEPTEMBER 2010 • www.ajprenal.org


Copyright of American Journal of Physiology: Renal Physiology is the property of American
Physiological Society and its content may not be copied or emailed to multiple sites or posted
to a listserv without the copyright holder's express written permission. However, users may
print, download, or email articles for individual use.

You might also like