ArticlePDF Available

Soluble (pro)renin receptor via β-catenin enhances urine concentration capability as a target of liver X receptor

Authors:

Abstract and Figures

Significance The soluble (pro)renin receptor (sPRR) is produced by protease-mediated cleavage of PRR and is elevated under certain pathological conditions. To our knowledge, no prior studies have reported the biological function of sPRR in general or the antidiuretic function of the soluble protein in particular. Here we describe a previously unreported role of sPRR in the enhancement of renal aquaporin 2 (AQP2) expression and urine-concentrating capability. We further show that sPRR acts via frizzled class receptor 8-depdendent β-catenin signaling to increase AQP2 expression in the collecting duct cells. These findings offer an unreported insight into the physiological role of sPRR in regulating fluid homeostasis. In addition, we found that liver X receptor activation by TO901317 resulted in diabetes insipidus because of the inhibition of renal PRR expression.
Content may be subject to copyright.
Soluble (pro)renin receptor via β-catenin enhances
urine concentration capability as a target of liver
X receptor
Xiaohan Lu
a,b,c,1
, Fei Wang
c,a,b,1
, Chuanming Xu
c
, Sunny Soodvilai
a,b
, Kexin Peng
a,b,c
, Jiahui Su
c
, Long Zhao
a,b
,
Kevin T. Yang
a,b
, Yumei Feng
d,e
, Shu-Feng Zhou
f
, Jan-Åke Gustafsson
g,2
, and Tianxin Yang
a,b,c,2
a
Department of Internal Medicine, University of Utah, Salt Lake City, UT 84112;
b
Veterans Affairs Medical Center, Salt Lake City, UT 84108;
c
Institute of
Hypertension, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, 510080, Peoples Republic of China;
d
Department of Pharmacology,
University of Nevada School of Medicine, Reno, NV 89557;
e
Department of Physiology and Cell Biology, University of Nevada School of Medicine, Reno,
NV 89557;
f
Department of Pharmaceutical Sciences, College of Pharmacy, University of South Florida, Tampa, FL 33620; and
g
Department of Biology
and Biochemistry, University of Houston, Houston, TX 77004
Contributed by Jan-Åke Gustafsson, February 12, 2016 (sent for review February 1, 2016; reviewed by Guanping Chen and Jia L. Zhuo)
The extracellular domain of the (pro)renin receptor (PRR) is cleaved
to produce a soluble (pro)renin receptor (sPRR) that is detected in
biological fluid and elevated under certain pathological conditions.
The present study was performed to define the antidiuretic action
of sPRR and its potential interaction with liver X receptors (LXRs),
which are known regulators of urine-concentrating capability. Wa-
ter deprivation consistently elevated urinary sPRR excretion in
mice and humans. A template-based algorithm for proteinprotein
interaction predicted the interaction between sPRR and frizzled-8
(FZD8), which subsequently was confirmed by coimmunoprecipita-
tion. A recombinant histidine-tagged sPRR (sPRR-His) in the nano-
molar range induced a remarkable increase in the abundance of
renal aquaporin 2 (AQP2) protein in primary rat inner medullary
collecting duct cells. The AQP2 up-regulation relied on sequential
activation of FZD8-dependent β-catenin signaling and cAMPPKA
pathways. Inhibition of FZD8 or tankyrase in rats induced polyuria,
polydipsia, and hyperosmotic urine. Administration of sPRR-His
alleviated the symptoms of diabetes insipidus induced in mice by
vasopressin 2 receptor antagonism. Administration of the LXR ag-
onist TO901317 to C57/BL6 mice induced polyuria and suppressed
renal AQP2 expression associated with reduced renal PRR express-
ion and urinary sPRR excretion. Administration of sPRR-His re-
versed most of the effects of TO901317. In cultured collecting duct
cells, TO901317 suppressed PRR protein expression, sPRR release, and
PRR transcriptional activity. Overall we demonstrate, for the first
time to our knowledge, that sPRR exerts antidiuretic action via
FZD8-dependent stimulation of AQP2 expression and that inhibi-
tion of this pathway contributes to the pathogenesis of diabetes
insipidus induced by LXR agonism.
soluble (pro)renin receptor
|
liver X receptor
|
aquaporin-2
|
frizzled-8
|
β-catenin
Full-length (Pro)renin receptor (PRR), a 350-amino acid
transmembrane receptor for prorenin and renin, is subjected
to protease-mediated cleavage to produce a 28-kDa protein of
the N-terminal extracellular domain, the soluble (pro)renin re-
ceptor (sPRR), and the 8.9-kDa C-terminal intracellular domain
called M8.9(1, 2). Before the cloning of full-length PRR in
mesangial cells as an integral 39-kDa membrane protein (3),
M8.9 was identified as a truncated protein associated with the
vacuolar H
+
-ATPase (V-ATPase) from bovine chromatin gran-
ules (4). The cleavage occurs in Golgi apparatus through furin
(5) or ADMA19 (6). An sPRR ELISA kit has been developed to
detect sPRR in plasma and urine samples (7)
.
With this assay,
increased serum sPRR levels have been demonstrated in patients
with heart failure (8), kidney disease (9, 10), hypertension (11),
and preeclampsia (2). Moreover, serum sPRR is positively as-
sociated with serum creatinine, blood urea nitrogen, and urine
protein and is inversely associated with the estimated glomerular
filtration rate in patients with chronic kidney disease caused by
hypertension and type 2 diabetes (9). However, serum sPRR was
not correlated with serum renin, prorenin, or aldosterone in
healthy subjects or in patients with diabetes and hypertension
(12). To our knowledge no prior studies have reported a bi-
ological function of sPRR in any condition.
Within the kidney, PRR is expressed abundantly in the distal
nephron, particularly in intercalated cells of the collecting duct
(CD) (13, 14). A functional role of PRR in regulating renal
aquaporin 2 (AQP2) expression and urine-concentrating capa-
bility has been revealed by analysis of mice with conditional
deletion of PRR in the nephron (15) and the CD (14). However,
whether the antidiuretic action of CD PRR is conferred by sPRR
remains elusive.
Liver X receptors (LXRs) are activated by oxidized choles-
terol derivatives and belong to a family of nuclear receptors that
form heterodimers with the retinoid X receptor to regulate tran-
scription of target genes governing cholesterol, fatty acid, and
glucose metabolism (16, 17) (18). LXRs consist of two isoforms,
LXRα, which is abundantly expressed in liver, small intestine,
kidney, macrophages, and adipose tissue, and LXRβ,whichis
expressed more ubiquitously (19). LXRs have an established role
in reverse cholesterol transport which leads to cholesterol efflux
from peripheral tissues to the liver (20). Interestingly, emerging
Significance
The soluble (pro)renin receptor (sPRR) is produced by protease-
mediated cleavage of PRR and is elevated under certain path-
ological conditions. To our knowledge, no prior studies have
reported the biological function of sPRR in general or the
antidiuretic function of the soluble protein in particular. Here
we describe a previously unreported role of sPRR in the en-
hancement of renal aquaporin 2 (AQP2) expression and urine-
concentrating capability. We further show that sPRR acts via
frizzled class receptor 8-depdendent β-catenin signaling to in-
crease AQP2 expression in the collecting duct cells. These find-
ings offer an unreported insight into the physiological role of
sPRR in regulating fluid homeostasis. In addition, we found that
liver X receptor activation by TO901317 resulted in diabetes
insipidus because of the inhibition of renal PRR expression.
Author contributions: S.-F.Z., J.-Å.G., and T.Y. designed research; X.L., F.W., C.X., S.S., K.P.,
J.S., L.Z., and K.T.Y. performed research; Y.F. contributed new reagents/analytic tools; and
J.-Å.G. and T.Y. wrote the paper.
Reviewers: G.C., Emory University School of Medicine; and J.L.Z., University of Mississippi.
The authors declare no conflict of interest.
1
X.L. and F.W. contributed equally to this work.
2
To whom correspondence may be addressed. Email: jgustafsson@uh.edu or Tianxin.
Yang@hsc.utah.edu.
www.pnas.org/cgi/doi/10.1073/pnas.1602397113 PNAS Early Edition
|
1of9
MEDICAL SCIENCES PNAS PLUS
evidence suggests a potential role of LXRs in the regulation of
the renin-angiotensin system (RAS) and renal transporters such
as Na-Pi transporters (21), OAT1 (22), and epithelial Na
+
channel
(ENaC) (23). In the present study we attempted to define a bi-
ological function and signaling of sPRR in the regulation of fluid
homeostasis and to test PRR/sPRR further as a target of LXRs
in the kidney.
Results
Initial Evidence for a Biological Function of sPRR. A clue indicating a
potential biological function of sPRR came from immunostain-
ing of rat kidneys using antibodies against different domains of
PRR. We examined the cellular localization of renal PRR in rats
using two different antibodies: the antibody raised against the
C-terminal domain of PRR (termed antiPRR-C antibody;
Abcam) and another antibody against the N-terminal domain in
the sPRR sequence (termed antiPRR-N antibody) (24). After
the cleavage, sPRR is recognized by antiPPR-N but not by anti
PRR-C. AntiPRR-C antibody labeled AQP2
CD cells, e.g., the
intercalated cells (Fig. 1C) (13, 25). Unexpectedly, the labeling
with antiPRR-N was found predominantly on the apical mem-
brane of CD principal cells (Fig. 1 Aand B), overlapping with
AQP2 (Fig. 1 Dand E), confirming the localization in principal
cells but not in intercalated cells. The images from colabeling
with anti-PRR antibody and anti-AQP2 antibody were merged
(Fig. 1 GI). We performed a competition assay to validate the
specificity of antiPRR-N antibody. The signal from this anti-
body disappeared after preincubation with a recombinant sPRR
protein, sPRR-His (Fig. 1 Jand K) or with the immunizing peptide
(Fig. 1L) or with the omission of primary antibody (Fig. 1M).
These immunostaining results raised the intriguing possibility
that sPRR derived from intercalated cells or other tubules may
act on an as yet unknown membrane receptor in principal cells to
regulate tubular transport function. To test this possibility, a
recombinant rat sPRR was generated using a mammalian cell-
expressing system. This protein contained sPRR with a secretion
signal in the N terminus and an eight-histidine tag in the C terminus
(termed sPRR-His) and appeared as a single 29.6-kDa band on
12% SDS/PAGE gel (Fig. 2A). Primary cultures of rat inner
medullary collecting duct (IMCD) cells in Transwells exposed to
10 nM sPRR-His for 24 h exhibited a remarkable increase in
AQP2 expression at both mRNA and protein levels (Fig. 2 Band
C). In a separate experiment, murine immortalized cortical col-
lecting duct (mpkCCD) cells were transiently transfected by a
luciferase reporter construct, pGL3-AQP2. The transfected cells
were treated for 24 h with 10 nM sPRR-His or vehicle. The
sPRR-His treatment induced a 5.5-fold increase in luciferase
activity (Fig. 2D). The in vitro findings confirmed a direct stim-
ulatory effect of sPRR-His on AQP2 expression.
sPRR Activation of the Wntβ-Catenin Pathway in the CD. Search tool
for the retrieval of interacting gene and proteins (STRING) is a
template-based algorithm for proteinprotein structure pre-
diction. We used STRING to identify proteins that interact with
sPRR. Among 10 candidate proteins, three are related to Wnt
β-catenin pathway: frizzled-8 (FZD8), low-density lipoprotein
receptor-related protein 6 (LRP6), and Wnt-3a (Fig. 2E). Be-
cause FZD8 is a receptor component in the Wntβ-catenin
pathway, subsequent studies were focused on interaction be-
tween FZD8 and sPRR. Coimmunoprecipitation experiments
using membrane fraction proteins prepared from the rat inner
medulla demonstrated that sPRR binds to FZD8 (Fig. 3A).
Immunostaining showed that at low magnification (Fig. 3B),
FZD8 labeling was found predominantly in the outer and
inner medulla with sporadic labeling in the cortex. Colabeling
with AQP2 (the marker of CD principal cells) and labeling of
Fig. 1. Distinct labeling with antibodies against different domains of PRR. (AC) Immunostaining on consecutive rat kidney sections using antiPRR-N an-
tibody (magnification: 200×in A; 400×in B) recognizing the N-terminal domain of PRR (e.g., the sPRR sequence) and the antiPRR-C antibody recognizing the
C-terminal domain of PRR (C; magnification: 400×). (DF) The PRR antibodies were coincubated with anti-AQP2 antibody. (GI) Merged images. Arrows in B
and Edenote principal cells; arrows in Cand Fdenote intercalated cells. The labeling with antiPRR-N antibody was localized mostly to the apical membrane
of principal cells, contrasting with the labeling with antiPRR-C antibody in the intercalated cells. (JM) To validate the specificity of the labeling, immu-
nostaining was performed with antiPRR-N antibody preincubated with an expressed sPRR (J), sPRR-His (K), or its immunizing peptides (L) or without the
primary antibody (M). The images shown are representative of three to six animals per group.
2of9
|
www.pnas.org/cgi/doi/10.1073/pnas.1602397113 Lu et al.
consecutive sections for the electroneutral sodium, potassium and
chloride cotransporters (NKCC2), a marker of thick ascending limb
cells (Fig. 3C), confirmed FZD8 staining in the thick ascending
limb and the CD. In the CD, FZD8 staining was detected in both
principal and intercalated cells. The comparison between FZD8
and PRR labeling was made in the inner medulla where the two
signals were roughly colocalized to the CD. In principal cells,
both FZD8 and PRR were detected on the apical membrane
although FZD8 labeling was relatively diffuse (Fig. 3D).
The functional role of FZD8 in mediating sPRR signaling in
primary rat IMCD cells was assessed by using FZD8 siRNA and
an FZD8 inhibitor, OMP-54F03 (OMP). The efficacy of FZD8
siRNA was validated by immunoblotting analysis of FZD8 pro-
tein expression (Fig. 4A). Exposure of rat IMCD cells to 10 nM
sPRR-His for 24 h induced the activity of Wnt-responsive lu-
ciferase activity as assessed by using the Cignal TCF/LEF Re-
porter Assay kit (Qiagen), which was blunted by FZD siRNA
(Fig. 4B). Consistent with this result, the sPRR-His treatment
remarkably induced AQP2 protein expression, which was blun-
ted by both FZD8 siRNA (Fig. 4C) and OMP (Fig. 4D) as well as
by a tankyrase inhibitor XAV939 (XAV) (Fig. 4E) (26). Tankyrase
belongs to the poly (ADP-ribose) polymerase family responsi-
ble for the transfer of ADP ribose from NAD
+
to acceptor
proteins (27) and also for the activation of the Wntβ-catenin
pathway through the stabilization of the axinβ-catenin com-
plex (26). Our results suggest that sPRR signals through FZD8
to activate the Wnt/βcatenin pathway, leading to increased
AQP2 expression.
Arginine vasopressin (AVP) is known to induce AQP2
trafficking to the apical membrane acutely (within minutes) by
increasing phosphorylation of AQP2 and chronically (within hours)
by stimulating AQP2 transcription, both through the cAMPPKA
pathway (28, 29). We found that the rapid rise of cAMP and the
redistribution of AQP2 from the cytosol to the membrane in
response to a 30-min exposure to AVP was unaffected by XAV
treatment (Fig. 5 AC); the trafficking event was evaluated by
examining the abundance of AQP2 protein in the fractionated
cell samples. Immunoblotting detected AQP2 protein as multi-
ple bands of 3545 kDa and 29 kDa, reflecting the glycosylated
and nonglycosylated forms, respectively. In contrast, at 24 h the
increases in the cAMP level (Fig. 5D) and total protein abun-
dance (Fig. 5E) and mRNA expression (Fig. 5F) of AQP2 were
all effectively attenuated by XAV treatment. These results sug-
gest that the Wntβ-catenin pathway may specifically target
AQP2 gene transcription but not AQP2 trafficking via selective
coupling with the late but not early cAMP production after
AVP treatment.
The in Vivo Role of β-Catenin Signaling in Rats During Antidiuresis. To
probe the in vivo role of β-catenin signaling in fluid homeostasis,
we administered OMP and XAV to SpragueDawley (SD) rats
under basal conditions and during 48-h water deprivation (WD)
and evaluated their impact on water balance. Under basal con-
ditions, the administration of OMP and XAV over 48 h similarly
induced polyuria, polydipsia, and hypoosmotic urine (Fig. 6 AC).
During 48-h WD, these treatments consistently increased urine
volume and decreased urine osmolality, and these effects were
accompanied by exaggerated weight loss (Fig. 6 DF) and greater
increases in plasma osmolality and hematocrit (Hct) (Fig. 6 Gand
H). Immunoblotting showed that the abundance of β-catenin
protein was increased in the nuclear fraction from both cortex and
inner medulla following WD, indicating the activation of β-catenin
signaling (Fig. 7 Aand B). AQP2 protein abundance in both renal
cortex and inner medulla was increased remarkably following WD;
this increase was attenuated significantly by both OMP and XAV
(Fig. 7 CF).
Fig. 3. Renal FZD8 expression and its interaction with sPRR. (A) Coimmu-
noprecipitation analysis of the interaction between sPRR and FZD8. The
membrane fraction of rat renal medulla was immunoprecipitated with anti
PRR-N antibody and probed with anti-FZD8 antibody or vice versa. (B)Immu-
nolabeling of FZD8 in rat kidney at low magnification. (C) Immunolabeling of
FDZ8, AQP2, and NKCC2 in the outer medulla at high magnification. Colab-
eling for AQP2 was performed on the same section, and labeling for NKCC2
was performed on consecutive sections. CD, collecting duct; PT, proximal
tubule; TL, thick ascending limb. (D) Immunolabeling of FDZ8 and PRR in the
rat inner medulla. The consecutive sections were stained with anti-FZD8
antibody and antiPRR-N antibody. The same section was colabeled with
anti-AQP2 antibody. The images shown are representative of three to six
animals per group.
0
2
4
6
8
10
12
CTR sPRR-His
AQP2 mRNA/GAPDH
55
72
95
130
kDa
43
36
26
15
Marker E1 E2 E3
sPRR-His
29.6 kDa
E4
sPRR-His
AQP2
β-Actin
CTR
1.0f0.02 3.08f0.25 *
35~45 kDa
29 kDa
43 kDa
ABC
D
0
2
4
6
8
10
12
14
16
18
pGL3-blank pGL3-AQP2 pGL3-AQP2 + sPRR-His
Luminescence (RLU/µg protein)
E
p<0.01
Fig. 2. Characterization of sPRR function. A recombinant PRR, sPRR-His, was
expressed in a mammalian cell-expressing system as a fusion protein that con-
tained sPRR, a histidine tag in the C terminus, and a secretion signal peptide in
the N terminus. (A) sPRR-His was purified from the medium as a single
29.6-kDa band in 12% SDS/PAGE gel. E14 are sequential fractions from the
elution. (Band C) Primary rat IMCD cells grown in Transwells were exposed
to 10 nM sPRR-His for 24 h, and AQP2 expression was analyzed by quanti-
tative RT-PCR and immunoblotting (n=5 per group). (D) In a separate ex-
periment, the cultured CD cells were transfected with an AQP2-luciferase
construct, were allowed to grow to confluence, and then were treated with
vehicle or 10 nM sPRR-His for 24 h, and luciferase activity was assayed (n=5
per group). (E) STRING is a template-based algorithm for predicting protein
protein structure. We used STRING to identify proteins that interact with
sPRR. This analysis revealed 10 hits: CTSG (cathepsin G), ACE2 (angiotensin
1-converting enzyme 2), CMA1 (chymase 1), MME (membrane metallo-endo-
peptidase), ACE (angiotensin 1 converting enzyme 1), CTSZ (cathepsin Z),
ENPEP (glutamyl aminopeptidase), FZD8 (frizzled family receptor 8), WNT3A
(wingless-type MMTV integration 3A), and LRP6 (low density lipoprotein
receptor-related protein 6). CTR, control.
Lu et al. PNAS Early Edition
|
3of9
MEDICAL SCIENCES PNAS PLUS
Therapeutic Potential of sPRR-His for Treatment of Nephrogenic
Diabetes Insipidus. Nephrogenic diabetes insipidus (NDI) is
commonly caused by mutations of the vasopressin 2 receptor
(V2R) gene; a specific therapy for this disease is lacking (30, 31).
We explored the therapeutic potential of sPRR-His in a mouse
model of NDI induced with a V2R antagonist, OPC. sPRR-His
was chronically infused via a catheter placed in the jugular vein
driven by an osmotic minipump. After 7 d of sPRR-His infusion,
OPC was given via gavage at 30 mg·kg
1
·d
1
for 3 d. Adminis-
tration of the V2R antagonist resulted in symptoms of NDI,
including polydipsia, polyuria, and hypoosmotic urine, all of
which were attenuated by sPRR-His treatment (Fig. 8 AC).
This result supports the therapeutic potential of sPRR for
management of the symptoms of NDI. In the OPC-treated mice,
urinary sPRR excretion was suppressed significantly (Fig. 8D),
providing a rationale for the use of exogenous sPRR to treat
NDI. This result also suggests that the production of renal sPRR
may be under the direct control of the AVPV2R pathway. In
support of this notion, we have shown that exposure of the CD
cells to AVP stimulates the release of sPRR (14). Fig. 8Epro-
vides a schematic illustration of the mechanism of action of
sPRR in the CD principal cells. Our data suggest that sPRR
binds FDZ8, leading to the activation of β-catenin that pro-
motes chronic cAMP accumulation, ultimately enhancing
AQP2 transcription.
Induction of Diabetes Insipidus and Suppression of Renal PRR by LXR
Agonism. Besides their well-recognized role in regulating lipid
and glucose metabolism, LXRs are potential regulators of the
RAS (32) and fluid balance (33). We tested the hypothesis that
LXRs may affect renal PRR expression and local RAS and
hence fluid homeostasis. In the initial experiment, we examined
the effects of the LXR agonist TO901317 on fluid homeostasis
and on renal PRR and urinary renin levels in mice. A 7-d treat-
ment with TO901317 in C57BL6 mice reduced body weight
(32.36 ±0.63 vs. 28.61 ±0.36 g, P<0.05) and food intake (4.08 ±
0.19 vs. 3.54 ±0.15 g, P<0.05), as previously reported (34),
although to a lesser extent. This treatment led to severe polyuria
(Fig. 9A) and hypoosmotic urine (Fig. 9B), indicating a urine-
concentrating defect. The expression of renal PRR protein and
the excretion of urinary sPRR were examined by immunoblotting
and ELISA, respectively. Both renal PRR expression (Fig. 9 C
and D) and urinary sPRR excretion (Fig. 9E) were suppressed
consistently by TO901317. This experiment suggested that the
suppressed renal PRR level may be responsible for TO901317-
induced diabetes insipidus (DI).
We performed cell-culture experiments to examine the direct
effect of TO901317 on PRR expression. mpkCCD cells were
exposed to TO901317 or vehicle for 24 h, and PRR protein ex-
pression was determined by immunoblotting. TO901317 treat-
ment reduced PRR protein expression by 85% (Fig. 9 Fand G)
and medium sPRR by 60% (Fig. 9H). In a separate experiment,
mpkCCD cells were transiently transfected by a pGL3-PRR
construct. The transfected cells were treated with TO901317 or ve-
hicle for 24 h. TO901317 treatment suppressed luciferase activity by
76% (Fig. 9I). The in vitro findings confirmed that TO901317
has a direct inhibitory effect on PRR expression.
sPRR-His Attenuates TO901317-Induced DI in Mice. In the subsequent
experiment, we performed more detailed analysis of TO901317-
induced DI and further examined the causal role of suppressed
renal PRR in this phenomenon. This experiment comprised
three groups: control, TO901317-treated, and TO901317 +sPRR-
Histreated mice. TO901317-treated mice displayed polyuria, poly-
dipsia, decreased water balance, hypoosmotic urine, and plasma
volume contraction (as reflected by the rise in Hct), confirming
the urine-concentrating defect (Fig. 10). All these parameters were
improved significantly in mice treated withTO901317 +sPRR-His
Fig. 5. The distinct role of the Wnt/ βcatenin pathway in the acute and
chronic responses to AVP in primary rat IMCD cells. The IMCD cells were
pretreated for 1 h with XAV and were treated for 30 min or 24 h with 10 nM
AVP. (A) At 30 min of AVP treatment, the medium was assayed for cAMP
using ELISA (n=6 per group). (Band C) Membrane fraction (n=6 per group)
(B) and cytosolic fraction (n=6 per group) (C) were subjected to immuno-
blotting analysis of AQP2. (DF) At 24 h, medium cAMP was determined (n=
6 per group) (D), whole-cell lysates were subjected to immunoblotting
analysis of AQP2 (n=6 per group) (E), and total RNA was subjected to
quantitative RT-PCR analysis of AQP2 mRNA (n=4 per group) (F). Data are
shown as mean ±SE; *P<0.05 vs. control;
#
P<0.05 vs. AVP alone.
Fig. 4. Analysis of the in vitro role of the Wnt/βcatenin pathway in the
regulation of AQP2 expression in response to sPRR-His treatment. (A) Vali-
dation of FZD8 knockdown by siRNA (n=3 per group). Primary rat IMCD
cells were transfected with or without FZD8 siRNA. FZD8 protein expression
was evaluated by immunoblotting. (B) Effect of sPRR-His on Wnt-response
luciferase activity in the presence or absence of FZD8 siRNA (n=12 per
group). The IMCD cells were transfected with or without FZD8 siRNA, fol-
lowed by sPRR-His treatment at 10 nM for 24 h. The Cignal reporter system
was used to evaluate the activity of the Wnt/βcatenin pathway, and the
data are presented as relative response ratio. (CE) Rat IMCD cells were
transfected with FZD8 siRNA, pretreated with OMP or XAV-939 for 1 h, and
treated with 10 nM sPRR for 24 h. AQP2 protein expression was determined
by immunoblotting analysis. Densitometric values are shown underneath
the blots. (C) Effect of FZD8 knockdown on sPRR-Induced AQP2 protein
expression (n=6 per group). (D) Effect of OMP on sPRR-His-induced AQP2
protein expression (n=6 per group). (E) Effect of XAV on sPRR-His-induced
AQP2 expression (n=6 per group). Data are shown as mean ±SE; *P<0.05
vs. control;
#
P<0.05 vs. sPRR-His.
4of9
|
www.pnas.org/cgi/doi/10.1073/pnas.1602397113 Lu et al.
(Fig. 10). Of note, water balance was determined by subtracting
urine volume from water intake. AQP2, a major water channel
on the apical membrane of the CD, plays a key role in de-
termining urine-concentrating capability. Immunoblotting dem-
onstrated that TO901317 significantly reduced renal AQP2
expression, which was partially restored by sPRR-His (Fig. 11 A
and B). We suspected that sPRR-His may affect the intrarenal
RAS, and this effect can be reflected by urinary renin activity
(35). Urinary renin activity, as assessed by measuring the gen-
eration of angiotensin I (AngI), was suppressed by TO901317,
and the suppression was completely reversed by sPRR-His (Fig.
11C). We also performed ELISA to determine the prorenin/renin
concentration in the urine. Although urinary prorenin/renin ex-
cretion was suppressed by TO901317, it was unaffected by sPRR-
His (Fig. 11D). This result is compatible with the concept that
sPRR regulates renin primarily at its activity level (3). Fig. 11E
provides a schematic illustration of the mechanism by which
TO901317 induces DI. Upon binding to TO901317, LXRs func-
tion as a transcriptional repressor to inactivate PRR transcription.
The reduced PRR/sPRR levels down-regulate AQP2 expression,
leading to DI.
Discussion
sPRR is generated by protease-mediated cleavage in the Golgi
apparatus and is released to plasma or urine. Serum sPRR levels
are elevated in various pathological states. To our knowledge, no
prior studies have reported a biological function of sPRR. We
recently used pharmacological and conditional gene-knockout
approaches to demonstrate that CD PRR has an essential role in
determining renal AQP2 expression and urine-concentrating
capability (14). In the present study, we discovered that sPRR
acts via FZD8-dependent activation of β-catenin signaling that
leads to increased AQP2 expression and thus enhanced urine-
concentrating capability. In addition, we found that LXR ago-
nism with TO901317 induced DI by inhibiting the renal PRR and
the intrarenal RAS.
Probably the most striking finding of this study was the re-
markable stimulatory effect of sPRR-His on AQP2 expression in
cultured rat IMCD cells grown in Transwells. In this experiment,
sPRR-His was used at 10 nM, which is likely a physiological
concentration. Moreover, we vigorously examined the signaling
mechanism of sPRR-His up-regulation of AQP2, revealing the
involvement of FZD8-dependent β-catenin signaling. A clue
suggesting potential interaction between sPRR and FZD8 was
obtained by a template-based algorithm for predicting protein
protein structure. The physical interaction of the two proteins in
the membrane fraction of the rat inner medulla was confirmed
by coimmunoprecipitation. The interaction also was demon-
strated at a functional level, because inhibition of FZD8 by siRNA
and pharmacological approaches effectively attenuated the stim-
ulatory effect of sPRR-His on AQP2 in cultured CD cells. The
functional role of FZD8 in regulating renal AQP2 expression and
urine-concentrating capability was confirmed in vivo by using a
FZD8 inhibitor, OMP. The antidiuretic function of FZD8 was
supported by the similar effect of a general Wnt/β-catenin in-
hibitor, XAV. Immunostaining demonstrated that FZD8 is lo-
calized to the CD and thick ascending limb, as is consistent with
the nephron-distribution pattern of PRR. Within the CD, FZD8
labeling was found in both principal and intercalated cells, a
pattern not exactly same as sPRR labeling. We suspect that
FZD8 may serve a function beyond its association with sPRR.
These results agree with a previous report that β-catenin sig-
naling mediated AVP-induced AQP2 expression in mpkCCDc14
cells (36). It is evident that β-catenin signaling is actively involved
in the physiological regulation of fluid homeostasis through cou-
pling with sPRR.
A large body of evidence has demonstrated that PRR serves as
a component of the Wnt receptor complex to regulate embryo-
genesis in low vertebrates in which the RAS does not exist (37,
38). It is evident that PRR acts in a renin-independent manner in
low vertebrates (1, 37, 39-41). PRR appears to be similarly
involved in the regulation of embryogenesis in mammals, as
evidenced by the lethal phenotype in mice with systemic or tis-
sue-specific deletion of PRR (1, 4). A large number of stud-
ies have challenged the physiological function of PRR and its
Fig. 7. The in vivo role of the Wnt/βcatenin pathway in the regulation of
renal AQP2 expression during antidiuresis in rats. (Aand B) To assess renal
β-catenin activation by WD, the nuclear fraction of renal cortex (n=4rats
per group) (A) and the inner medulla (n=4 rats per group) (B) from control
and dehydrated rats were subjected to immunoblotting analysis of β-cat-
enin. (CF) Immunoblotting analysis of AQP2 expression was performed on
the renal cortex and the inner medulla of rats treated with vehicle or WD
with (C) or without (D) OMP or with (E) or without (F) XAV (n=5 rats per
group). Data are shown as mean ±SE; *P<0.05 vs. control;
#
P<0.05 vs. WD.
CO, cortex; IM, inner medulla.
Fig. 6. The role of the Wnt/βcatenin pathway in urine-concentrating ca-
pability in rats. SD Rats were administered vehicle, OMP, or XAV and were
placed in metabolic cages for assessment of the state of water metabolism at
basal condition (AC) or after 24-h WD (DH)(n=5 rats per group). At basal
the condition, water intake (A), urine volume (B), and urine osmolality
(C) were determined. (DF) During 24-h WD, urine volume (D), urine osmo-
lality (E), and body-weight (BW) changes (F) were monitored. (Gand H)Atthe
end of the experiment plasma osmolality (G)andHct(H) were measured. *P<
0.05 vs. control;
#
P<0.05 vs. WD alone.
Lu et al. PNAS Early Edition
|
5of9
MEDICAL SCIENCES PNAS PLUS
relationship with the RAS (1, 37, 3941). Whether an intrinsic
linkage between the RAS and the developmental β-catenin path-
way occurs in mammals in settings of development or physiology is
unknown. For the first time, to our knowledge, our results link
prorenin/sPRR to the β-catenin pathway in the kidney during
physiological regulation of fluid homeostasis.
The cAMPPKA pathway is the principle mediator of AVP-
induced AQP2 trafficking and transcription (28, 42, 43). An issue
arises as to whether the Wntβ-catenin pathway interacts with
the cAMPPKA pathway during AVP-induced signaling. We
found that inhibition of the Wntβ-catenin pathway did not af-
fect AQP2 trafficking to the membrane fraction or cAMP pro-
duction induced by 30-min exposure to AVP. In contrast, this
maneuver did block AQP2 expression and cAMP production
induced by 24-h AVP treatment. These results suggest that ac-
tivation of the Wntβ-catenin pathway may be involved primarily
in sustaining the cAMP production during prolonged AVP
treatment to increase AQP2 transcription. This mechanism does
not appear to be required for the regulation of AQP2 trafficking,
which is a rapid response to AVP. There is a wealth of in-
formation regarding the role of the cAMPPKA pathway in this
acute response to AVP, but relatively little is known about this
pathway in a chronic setting of AVP treatment. To our knowl-
edge, the present study is the first to delineate a unique role for
β-catenin signaling in chronic but not acute regulation of the
cAMPPKAAQP2 axis.
Multiple previous studies, as well as this one, consistently
demonstrate that, within the CD, PRR is detected in intercalated
cells by using antibody against the C terminus of PRR (anti
PRR-C antibody) (13, 25). A question arises as to how interca-
lated cell-derived PRR up-regulates AQP2 expression in the
principal cells. This regulation may be through a paracrine mech-
anism, and there is an intriguing possibility that sPRR may serve
as a mediator of the communication between the two cell types
in the CD. In support of this possibility, an antibody against
the epitope in the sPRR, antiPRR-N antibody, stained only
principal cells but not intercalated cells or other non-CD tubules
that are known to express PRR. More direct evidence came from
cell-culture experiments showing that exposure of rat IMCD cells
to sPRR-His in the nanomolar range induced a remarkable in-
crease in AQP2 expression mimicking the effect of prorenin (44).
This result strongly suggests that sPRR has a physiological function.
The current therapy for DI is suboptimal. Although supple-
mentation of AVP is effective for central DI (45), no specific
therapy is currently available for nephrogenic DI. The present
study demonstrates, for the first time to our knowledge, the
therapeutic potential of sPRR-His in a mouse model of nephrogenic
DI induced by V2R antagonism. Because sPRR acts downstream of
V2R, it also should be effective for treatment of central DI.
A large body of evidence has established a link between energy
metabolism and fluid balance. A high-energy state, such as
obesity, is associated with disturbance of electrolytes and fluid
balance and hypertension (46, 47), whereas a low-energy state,
such as fasting, induces natriuresis and diuresis (48, 49). Along
this line, activation of PPARγ, a key regulator of glucose me-
tabolism and adipogenesis, causes body weight gain and plasma
volume expansion (50, 51). In the present study, we discovered
that LXR against TO901317 exerted a profound diuretic action
in mice as suggested by polyuria, polydipsia, hypoosmotic urine,
and contraction of plasma volume. AQP2, a major water channel
on the apical membrane of the CD, was remarkably suppressed,
likely conferring the diuretic action of TO901317.
In light of sPRRs action as a key regulator of AQP2 expression
and urine-concentrating capability, we suspected that PRR/sPRR
Fig. 8. Antidiuretic action of sPRR-His in a mouse model of NDI. Male C57/
BL6 mice were infused for 7 d with sPRR-His via a catheter implanted in the
jugular vein and then received oral administration of either vehicle or the
V2R antagonist OPC31260 (OPC) for 3 d. Mice were placed in metabolic cages
for assessment of daily water intake and urine output. (A) Daily water intake
(n=4 mice per group). (B) Daily urine output (n=4 mice per group).
(C) Urine osmolality (n=4 mice per group). (D) Urinary sPRR excretion (n=4
mice per group). Data are shown as mean ±SE. In AC,
#
P<0.05 vs. OPC
alone; in D*P<0.05 vs. control. (E) Schematic illustration of the mechanism
of action of sPRR. In the lumen of the distal nephron, sPRR binds FZD8 in a
receptor complex on the apical membrane of principal cells, resulting in the
activation of β-catenin, which promotes cAMP accumulation, ultimately
leading to increased AQP2 transcription and enhanced urine concentration.
43 kDa
TO901317CTR
PRR
β-Actin 43 kDa
Luminescence
(RLU/µg protein)
0
0.2
0.4
0.6
Cell medium sPRR content
(pg/μg protein)
CTR TO901317
p<0.05
400
800
1200
1600
Urinary sPRR excretion
(pg/24h)
0
CTR TO901317
p<0.05
Urinary osmolality
(mOsmo/kg.H2O)
Urine volume (ml/24h)
0
0.4
0.8
1.2
CTR TO901317
PRR densitometry
TO901317CTR
PRR
β-Actin
43 kDa
43 kDa
0
0.4
0.8
1.2
CTR TO901317
PRR densitometry
p<0.05
p<0.05
A
D
G
B
E
H
C
F
I
0
1000
2000
3000
CTR TO901317
p<0.05
4000
0
1.0
2.0
3.0
pGL3-blank pGL3-PRR pGL3-PRR +
TO901317
4.0
0
1.0
2.0
CTR TO901317
p<0.05
3.0
p<0.05
Fig. 9. Initial characterization of diuretic and PRR-inhibitory actions of
TO901317 in vivo and in vitro. Male C57/BL7 mice received oral administra-
tion of TO901317 with or without i.v. infusion of sPRR-His for 7 d. The mice
receiving vehicle treatment served as a control. At the end of experiment,
24-h urine collection was performed, followed by analysis of urinary sPRR
excretion by ELISA and renal PRR expression by immunoblotting. (A) Urine
output (n=30 mice per group). (B) Urinary osmolality (n=15 mice per
group). (C) Immunoblotting analysis of renal PRR expression (n=15 mice per
group). (D) Densitometric analysis of the immunoblot in C.(E) ELISA analysis
of urinary sPRR (n=8 mice per group). (FI) The effect of TO901317 on PRR
expression in cultured CD cells. mpkCCD cells were exposed to vehicle or
10 μM TO901317 for 24 h. The cell lysates were subjected to immunoblotting
analysis of PRR protein expression, and the medium was assayed for sPRR
concentration by using ELISA and normalized by protein content. In a sep-
arate experiment, the cells were transfected with a PRR-luciferase construct,
allowed to grown to confluence, and then were treated with vehicle or
10 μM TO901317 for 24 h. (F) Immunoblotting analysis of PRR (n=15 per
group). (G) Densitometric analysis of the immunoblot in A.(H) ELISA analysis
of medium sPRR (n=10 per group). (I) The luciferase assay (n=5 per group).
6of9
|
www.pnas.org/cgi/doi/10.1073/pnas.1602397113 Lu et al.
may be a molecular target of LXRs in the kidney. In vivo data
showed a significant reduction of renal PRR expression and
urinary sPRR excretion in TO901317-treated mice, and the
administration of sPRR-His completely rescued TO901317-induced
DI. Consistent with this observation, in vitro data demonstrated
that TO901317 had a direct inhibitory effect on PRR expression
and PPR-luciferase activity in cultured CD cells. These results
demonstrate, for the first time to our knowledge, that LXRs may
function as a transcriptional repressor of the PRR gene. It is
interesting that LXRβ
/
mice display central DI caused by the
impairment of AVP production (33). This phenotype suggests an
antidiuretic action of LXRβ, which is the opposite of the diuretic
action of TO901317. Therefore we speculate that diuretic action
of TO901317 may be conferred mainly by LXRα. This possibility
needs to be validated in future studies using LXRα-null mice.
It has been shown that acute LXR activation induces a tran-
sient increase in renin transcription in the juxtaglomerular cells
(32), but its chronic activation remarkably suppresses the ex-
pression of renin, AT1R, and angiotensin 1-converting enzyme 1
(ACE) in the heart and kidney following isoproterenol treatment
(52). In agreement with the inhibitory effect of LXRs on local
RAS, we found that TO901317 treatment remarkably suppressed
urinary renin, an index of the intrarenal RAS (35, 53). Although
LXRs may function as a negative regulator of renin expression at
the juxtaglomerular apparatus in the acute setting, they appear
primarily to suppress PRR and the local RAS to elicit a diuretic
response.
In summary, the present study reports, for the first time to our
knowledge, a biological function of sPRR in regulating fluid
homeostasis. sPRR is associated with FZD8 to activate β-catenin,
which interacts with the cAMPPKA pathway to induce AQP2
expression and enhance the urine-concentrating capability. Be-
cause intercalated cells are the potential source of sPRR, and
principal cells are the site of its action, it seems reasonable to
speculate that sPRR mediates the communication between the
two cell types in the CD. Last, we discovered the diuretic action
of the LXR agonist TO901317, which is conferred by inhibition
of the renal PRR/sPRR system.
Methods
Animals. Male 10- to 12-wk-old SD rats and C57BL6 mice were purchased from
Charles River Laboratories and the Jackson Laboratory, respectively. All an-
imals were maintained in a temperature-controlled room with a 12:12-h light:
dark cycle, with free access to tap water and standar d rat chow. Animals were
randomized into different experimental groups. Animal protocols were
approved by the Animal Care and Use Committees at the University of Utah
and the Sun Yat-sen University.
Rat Experiments. Under isoflurane inhalation, rats were s.c. implanted with an
osmotic minipump delivering a tankyrase inhibitor, XAV939 (XAV; Med-
chemExpress) at 5 mg·kg
1
·d
1
or received a FZD8 inhibitor, OMP54F03
(OMP) (a gift from John Lewicki, OncoMed Pharmaceuticals, Redwood City,
CA), at 25 mg/kg via daily i.p. injection. All animals were acclimatized to
metabolic cages for 7 d. After collection of baseline data for 2 d, rats were
water deprived for 48 h but had free access to chow diets. At the end of the
experiment, under isoflurane anesthesia, blood was withdrawn from the
vena cava, one kidney was cut into cortex and inner medulla and snap-
frozen, and the other kidney was fixed and paraffin embedded.
Plasmid Construction and sPRR Protein Purification. The cDNA of PRR (Gen-
Bank accession no. NM_001007091.1; also known as ATP6AP2)was
subcloned into the pMD-18T vector (Takara). sPRR, a solubilized form of
sPRR (residues 17274) lacking the transmembrane domain at the C termi-
nus, was combined with an eight-histidine tag in the C terminus (sPRR-His),
was generated by PCR from the PRR expression construct, and then was
cloned into pcDNA3.1. sPRR-His was generated by using a mammalian 293 cell
system and was purified by binding to IDA-Sephadex G-25 (GE Healthcare)
(XBIO, Shanghai, China).
Mouse Experiments. Male C57/BL6 mice received chronic i.v. infusion of vehicle
or sPRR-His at 30 μg·kg
1
·d
1
as described above. After the surgery, the mice
were acclimatized to metabolic cages for 7 d and then were randomly di-
vided to receive vehicle or a V2R antagonist, OPC-31260 (Sigma), at
30 mg·kg
1
·d
1
via gavage for 3 d. Water intake, urine volume, and urine
osmolality were recorded daily during the entire experimental period. In
another experiment to test the effects of sPRR-His on TO901317-induced DI,
male C57/BL6 mice were treated for 7 d with vehicle or with TO901317 alone
or in combination with sPRR-His. TO901317 was added to the chow diet at
Fig. 10. Effect of sPRR-His on TO901317-induced DI in mice. Male C57/BL6
mice were treated with vehicle or with TO901317 alone or in combination
with sPRR-His for 7 d. (A) Urine output (n=8 mice per group). (B) Water
intake (n=8 mice per group). (C) Water balance (n=8 mice per group).
(D) Urinary osmolality (n=8 mice per group). (E) Hct (n=8 mice per group).
AQP2
β-Actin
29 kDa
43 kDa
35~45 kDa
Urinary renin activity (ng/24h)
0
0.05
0.10
0.15
0.20
CTR TO901317 TO901317 + sPRR-His
p<0.05 p<0.05
0.25
AQP2 densitometry
0
0.2
0.4
0.6
0.8
1.0
1.2
CTR TO901317 TO901317 + sPRR-His
p<0.05 p<0.05
Urinary total prorenin/ renin
excretion (ng/24h)
p<0.05
0
0.3
0.6
0.9
CTR TO901317 TO901317 + sPRR-His
A
C
E
B
D
CTR TO901317 TO901317 + sPRR-His
Repressor
PRR
Inactive transcription
LXR
TO
PRR/sPRR
AQP2
DI
Fig. 11. Effect of sPRR-His on renal AQP2 expression and urinary renin in
TO901317-treated mice. Male C57/BL6 mice were treated with vehicle or
with TO901317 alone or in combination with sPRR-His for 7 d. AQP2 ex-
pression was analyzed by immunoblotting and immunostaining. Urinary
renin activity was determined by measuring AngI generation, and urinary
prorenin/renin concentration was determined by ELISA. (A) Immunoblotting
analysis of AQP2 expression (n=15 mice per group). (B) Densitometric
analysis of the immunoblots in A.(C) Urinary renin activity (n=8 mice per
group). (D) Urinary prorenin/renin excretion (n=8 mice per group).
(E) Schematic illustration of the mechanism by which the LXR agonist
TO091317 (TO) suppresses PRR transcription and induces DI. LXRs bound to
TO091317 function as a transitional repressor for the PRR gene, leading to a
reduced PRR/sPRR level that decreases AQP2 expression, ultimately causing DI.
Lu et al. PNAS Early Edition
|
7of9
MEDICAL SCIENCES PNAS PLUS
an estimated level of 50 mg·kg body weight
1
·d
1
. All mice were placed in
metabolic cages, and 24-h water intake and urine output were recorded and
collected at the end of the experiments.
Cell-Culture Experiments. For sPRR signaling studies, primary IMCD cells were
prepared from 4-wk-old SD rats as previously described (35). The cells were
grown in Transwell plates (catalog no. 29442-074; VWR International) with
DMEM/F-12 medium containing 10% (vol/vol) FBS, 0.5 μM 8-Br-cAMP,
130 mM NaCl, and 80 mM urea. Upon confluence, the cells were serum deprived
for 12 h and pretreated with an inhibitor (10 μMXAVor10μMOMP)fol-
lowed by 24-h treatment with AVP (10 nM) or sPRR-His (10 nM). At the end
of the experiments, the medium was collected for biochemical assays.
The effect of TO901337 on PRR expression was tested in mpkCCD cells.
These cells were grown to confluence in six-well plates. After 12-h serum
deprivation, the cells were treated with vehicle or 10 μM TO901317 for 24 h
and then were harvested for analysis of PRR expression and sPRR release. In
a separate experiment, mpkCCD cells at 50% density were transiently
transfected with a construct containing the luciferase gene under the con-
trol of the 2,016-bp 5flanking region of the PRR gene. Upon confluence,
the transfected cells were treated for 24 h with vehicle or 10 μM TO901317
and then were harvested for analysis of luciferase activity.
Enzyme Immunoassay. Urinary or medium sPRR was determined using a com-
mercially sPRR enzyme immunoassay (EIA) kit (catalog no. JP27782; Immuno-
Biological Laboratories) according to the manufacturers instructions.
Immunofluorescence Staining. The tissues were fixed in 10% neutral buffered
formalin for 24 h and then were embedded in paraffin. After deparaffini-
zation, thin sections (4 μm) were processed for double-labeling with im-
munofluorescence. For antigen recovery, the slides were immersed in
Tris·HCl EDTA buffer (pH 9.0) at a high temperature (98 °C) for 12 min. The
slides were blocked in 1% (wt/vol) BSA for 1 h and then were incubated
overnight with primary antibody at 4 °C. After the primary antibody was
washed off, sections were incubated for 1 h at room temperature with
donkey antigoat-IgG-FITC (Santa Cruz) or donkey anti-rabbit IgG-TRITC
(Life Technologies). Rabbit anti-PRR antibody from Abcam was raised
against residues 335350 in the C terminus (termed antiPRR-C antibody).
A second anti-PRR antibody used in the present study raised against residues
218235 in the N terminus of PRR (termed antiPRR-N antibody) was
generated in Y.F.s laboratory (24). Goat anti-AQP2 antibody was purchased
from Santa Cruz. Rabbit anti-NKCC2 antibody was purchased from Stress-
Marq Biosciences Inc.
Immunoblotting. Renal tissues were lysed and subsequently sonicated. Protein
concentrations were determined using Coomassie reagent. Forty micrograms
of protein from each sample were denatured in boiling water, separated by
SDS/PAGE, and transferred onto nitrocellulose membranes. Blots were blocked
1 h with 5% nonfat dry milk in Tris-buffered saline (TBS), followed by overnight
incubation with primary antibody. After washing with TBS, blots were incubated
with goat anti-rabbit/mouse HRP-conjugated secondary antibody and visualized
using ECL. The blots were quantitated by using Image-Pro Plus (Media Cyber-
netics). The primary antibodies were goat anti-AQP2 antibody, rabbit antiPRR-N
antibody, and rabbit anti-FZD8 antibody (all from Santa Cruz), rabbit anti-V2R
antibody (Abcam), and goat antiβ-catenin antibody (Novus).
Quantitative RT-PCR. Total RNA was isolated from renal tissues and reverse
transcribed to cDNA. Oligonucleotides were designed using Primer3 software
(bioinfo.ut.ee/primer3-0.4.0/). Primers of AQP2 were 5-gctgtcaatgctctccacaa-3
(sense) and 5-ggagcaaccggtgaaataga-3(antisense); primers for GAPDH were
5-gtcttcactaccatggagaagg-3(sense) and 5-tcatggatgaccttggccag-3(antisense).
Cell Membrane and Cytoplasmic Protein Fraction Isolation. The membrane and
cytosolic fractions of proteins were extracted using a kit according to the
manufacturers instructions (catalog no. BSP002; Bio Basic Inc.).
Coimmunoprecipitation. For coimmunoprecipitation with PRR and FZD8, the
membrane fraction of rat renal inner medullary was performed using a kit
(catalog no. BSP002; Bio Basic Inc.). AntiPRR-N (from Y.F.) or anti-FZD8 (Santa
Cruz) antibody was cross-linked with the magnetic beads (catalog no. 88805;
Pierce) and then incubated for 30 min with the renal medullary membrane
proteins. The beads were collected, washed, and eluted. The immunoprecipi-
tated samples were analyzed for the binding partner by immunoblotting.
siRNA or Plasmid Transfection in Primary Cultured IMCD Cells. IMCD cells were
transfected with FZD8 siRNA oligonucleotides (Invitrogen) or the luciferase
reporter plasmid (catalog no. CCS-018L; Qiagen) at a final concentration of
5 nM using HiPerFect transfection reagent (catalog no. 301702; Qiagen) for
72 h. The efficiency of FZD8 knockdown was validated by immunoblotting of
FZD8. For the luciferase assay, each sample consisted of a positive control, a
negative control, and a target luciferase construct, and the reporter activity
was calculated according to the manufacturers instruction.
Preparation of Luciferase Constructs. Genomic DNA was extracted from rat tail
using a Tissue DNA kit (D3396-01; Promega). A 2,016-bp fragment of the 5
flanking region of the PRR gene (GenBank accession no. NM_001007091;
1,941 ±75 bp) was amplified from the rat genomic DNA by PCR and
subcloned to the pGL3-Basic reporter vector (Promega) using NheI and BgIII
restriction sites; this construct was termed pGL3-PRR.A 2,084-bp frag-
ment of the 5flanking region of the AQP2 gene (GenBank accession no.
NM_012909; 2,000 ±84 bp) was cloned to the pGL3-Basic reporter vector by
a similar strategy; this construct was termed pGL3-AQP2.The identity of
these constructs was validated by sequencing.
Luciferase Assay. The mpkCCD cells were transfected with pGL3-AQP2 plasmid
or empty vector by using HiPerFect Transfection Reagent (catalog no. 301702;
Qiagen). Upon confluence, all cells were starved for 12 h; then pGL3-PRR- and
pGL3AQP2-transfected cells were treated for 24 h with TO901317 (10 μM) or
sPRR-His (10 nM), respectively. The vehicle-treated group served as a control.
The luciferase activities were measured using a luciferase assay system
(Promega), and luminescence was detected by using an illuminometer (BMG
FLUOstar OPTIMA).
Statistical Analysis. Data are summarized as means ±SE. Statistical analysis
was performed using ANOVA with the Bonferroni test for multiple com-
parisons or paired or unpaired Studentsttest for two comparisons. P<0.05
was considered statistically significant.
ACKNOWLEDGMENTS. We thank Dr. John Lewicki (OncoMed Pharmaceuti-
cals) for providing the FZD8 inhibitor OMP-54F03. This work was supported
by National Natural Science Foundation of China Grants 91439205 and
31330037; NIH Grants DK094956 and DK104072; National Basic Research
Program of China 973 Program 2012CB517600 Grant 2012CB517602; and
by the Veterans Administration Merit Review. T.Y. is a Research Career Sci-
entist in the Department of Veterans Affairs. J.-Å.G. was supported by
the Swedish Science Council and by Grant E-0004 from the Robert A.
Welch Foundation. Y.F. was supported by Grant NHLBI/R01HL122770
from the NIH.
1. Nguyen G (2011) Renin and prorenin receptor in hypertension: Whats new? Curr
Hypertens Rep 13(1):7985.
2. Watanabe N, et al. (2012) Soluble (pro)renin receptor and blood pressure during
pregnancy: A prospective cohort study. Hypertension 60(5):12501256.
3. Nguyen G, et al. (2002) Pivotal role of the renin/prorenin receptor in angiotensin II
production and cellular responses to renin. J Clin Invest 109(11):14171427.
4. Kinouchi K, et al. (2010) The (pro)renin receptor/ATP6AP2 is essential for vacuolar
H+-ATPase assembly in murine cardiomyocytes. Circ Res 107(1):3034.
5. Cousin C, et al. (2009) Soluble form of the (pro)renin receptor generated by in-
tracellular cleavage by furin is secreted in plasma. Hypertension 53(6):10771082.
6. Yoshikawa A, et al. (2011) The (pro)renin receptor is cleaved by ADAM19 in the
Golgi leading to its secretion into extracellular space. Hypertens Res 34(5):
599605.
7. Maruyama N, Segawa T, Kinoshita N, Ichihara A (2013) Novel sandwich ELISA for
detecting the human soluble (pro)renin receptor. Front Biosci (Elite Ed) 5:
583590.
8. Fukushima A, et al. (2013) Increased plasma soluble (pro)renin receptor levels are
correlated with renal dysfunction in patients with heart failure. Int J Cardiol 168(4):
43134314.
9. Hamada K, et al. (2013) Serum level of soluble (pro)renin receptor is modulated in
chronic kidney disease. Clin Exp Nephrol 17(6):848856.
10. Watanabe N, et al. (2013) Prediction of gestational diabetes mellitus by soluble
(pro)renin receptor during the first trimester. J Clin Endocrinol Metab 98(6):25282535.
11. Morimoto S, et al. (2014) Serum soluble (pro)renin receptor levels in patients with
essential hypertension. Hypertens Res 37(7):642648.
12. Nguyen G, et al. (2014) Plasma soluble (pro)renin receptor is independent of plasma
renin, prorenin, and aldosterone concentrations but is affected by ethnicity.
Hypertension 63(2):297302.
13. Advani A, et al. (2009) The (Pro)renin receptor: Site-specific and functional linkage to
the vacuolar H+-ATPase in the kidney. Hypertension 54(2):261269.
14. Wang FLX, et al. (2016) Antidiuretic action of collecting duct (pro)renin receptor
downstream of vasopressin/EP4 receptor. J Am Soc Nephrol, in press.
8of9
|
www.pnas.org/cgi/doi/10.1073/pnas.1602397113 Lu et al.
15. Ramkumar N, et al. (2015) Nephron-specific deletion of the prorenin receptor causes a
urine concentration defect. Am J Physiol Renal Physiol 309(1):F48F56.
16. Berkenstam A, Gustafsson JA (2005) Nuclear receptors and their relevance to diseases
related to lipid metabolism. Curr Opin Pharmacol 5(2):171176.
17. Alberti S, Steffensen KR, Gustafsson JA (2000) Structural characterisation of the
mouse nuclear oxysterol receptor genes LXRalpha and LXRbeta. Gene 243(1-2):
93103.
18. Zelcer N, Hong C, Boyadjian R, Tontonoz P (2009) LXR regulates cholesterol uptake
through Idol-dependent ubiquitination of the LDL receptor. Science 325(5936):
100104.
19. Repa JJ, Mangelsdorf DJ (2000) The role of orphan nuclear receptors in the regulation
of cholesterol homeostasis. Annu Rev Cell Dev Biol 16:459481.
20. Repa JJ, et al. (2000) Regulation of absorption and ABC1-mediated efflux of choles-
terol by RXR heterodimers. Science 289(5484):15241529.
21. Caldas YA, et al. (2011) Liver X receptor-activating ligands modulate renal and in-
testinal sodium-phosphate transporters. Kidney Int 80(5):535544.
22. Kittayaruksakul S, Soodvilai S, Asavapanumas N, Muanprasat C, Chatsudthipong V
(2012) Liver X receptor activation downregulates organic anion transporter 1 (OAT1)
in the renal proximal tubule. Am J Physiol Renal Physiol 302(5):F552F560.
23. Soodvilai S, Jia Z, Fongsupa S, Chatsudthipong V, Yang T (2012) Liver X receptor
agonists decrease ENaC-mediated sodium transport in collecting duct cells. Am J
Physiol Renal Physiol 303(12):F1610F1616.
24. Li W, et al. (2012) Brain-targeted (pro)renin receptor knockdown attenuates angio-
tensin II-dependent hypertension. Hypertension 59(6):11881194.
25. Gonzalez AA, Luffman C, Bourgeois CR, Vio CP, Prieto MC (2013) Angiotensin
II-independent upregulation of cyclooxygenase-2 by activation of the (Pro)renin re-
ceptor in rat renal inner medullary cells. Hypertension 61(2):443449.
26. Huang SM, et al. (2009) Tankyrase inhibition stabilizes axin and antagonizes Wnt
signalling. Nature 461(7264):614620.
27. Smith S, Giriat I, Schmitt A, de Lange T (1998) Tankyrase, a poly(ADP-ribose) poly-
merase at human telomeres. Science 282(5393):14841487.
28. Nielsen S, et al. (2002) Aquaporins in the kidney: From molecules to medicine. Physiol
Rev 82(1):205244.
29. Knepper MA, et al. (1996) Renal aquaporins. Kidney Int 49(6):17121717.
30. Fujiwara TM, Bichet DG (2005) Molecular biology of hereditary diabetes insipidus.
J Am Soc Nephrol 16(10):28362846.
31. Wesche D, Deen PM, Knoers NV (2012) Congenital nephrogenic diabetes insipidus:
The current state of affairs. Pediatr Nephrol 27(12):21832204.
32. Morello F, et al. (2005) Liver X receptors alpha and beta regulate renin expression in
vivo. J Clin Invest 115(7):19131922.
33. Gabbi C, et al. (2012) Central diabetes insipidus associated with impaired renal
aquaporin-1 expression in mice lacking liver X receptor β.Proc Natl Acad Sci USA
109(8):30303034.
34. Tachibana H, et al. (2012) Activation of liver X receptor inhibits osteopontin and
ameliorates diabetic nephropathy. J Am Soc Nephrol 23(11):18351846.
35. Wang F, et al. (2014) Prostaglandin E-prostanoid4 receptor mediates angiotensin II-
induced (pro)renin receptor expression in the rat renal medulla. Hypertension 64(2):
369377.
36. Jung HJ, et al. (2015) Tankyrase-mediated β-catenin activity regulates vasopressin-
induced AQP2 expression in kidney collecting duct mpkCCDc14 cells. Am J Physiol
Renal Physiol 308(5):F473F486.
37. Cruciat CM, et al. (2010) Requirement of prorenin receptor and vacuolar H+-ATPase-
mediated acidification for Wnt signaling. Science 327(5964):459463.
38. Oshima Y, Morimoto S, Ichihara A (2014) Roles of the (pro)renin receptor in the
kidney. World J Nephrol 3(4):302307.
39. Rousselle A, Sihn G, Rotteveel M, Bader M (2014) (Pro)renin receptor and V-ATPase:
From Drosophila to humans. Clin Sci (Lond) 126(8):529536.
40. Krop M, Lu X, Danser AH, Meima ME (2013) The (pro)renin receptor. A decade of
research: What have we learned? Pflugers Arch 465(1):8797.
41. Song R, Preston G, Ichihara A, Yosypiv IV (2013) Deletion of the prorenin receptor
from the ureteric bud causes renal hypodysplasia. PLoS One 8(5):e63835.
42. Yasui M, Zelenin SM, Celsi G, Aperia A (1997) Adenylate cyclase-coupled vasopressin
receptor activates AQP2 promoter via a dual effect on CRE and AP1 elements. Am J
Physiol 272(4 Pt 2):F443F450.
43. Zharkikh L, et al. (2002) Renal principal cell-specific expression of green fluorescent
protein in transgenic mice. Am J Physiol Renal Physiol 283(6):F1351F1364.
44. Lu X, et al. (2015) Activation of ENaC in collecting duct cells by prorenin and its re-
ceptor PRR: Involvement of Nox4-derived hydrogen peroxide. Am J Physiol Renal
Physiol, in press.
45. Kim RJ, Malattia C, Allen M, Moshang T, Jr, Maghnie M (2004) Vasopressin and
desmopressin in central diabetes insipidus: adverse effects and clinical considerations.
Pediatr Endocrinol Rev 2(Suppl 1):115123.
46. Granger JP, West D, Scott J (1994) Abnormal pressure natriuresis in the dog model of
obesity-induced hypertension. Hypertension 23(1, Suppl):I8I11.
47. West DB, Wehberg KE, Kieswetter K, Granger JP (1992) Blunted natriuretic response
to an acute sodium load in obese hypertensive dogs. Hypertension 19(1, Suppl):
I96I100.
48. Spark RF, Arky RA, Boulter PR, Saudek CD, OBrian JT (1975) Renin, aldosterone and
glucagon in the natriuresis of fasting. N Engl J Med 292(25):13351340.
49. Weinsier RL (1971) Fastinga review with emphasis on the electrolytes. Am J Med
50(2):233240.
50. Zhang H, et al. (2005) Collecting duct-specific deletion of peroxisome proliferator-
activated receptor gamma blocks thiazolidinedione-induced fluid retention. Proc Natl
Acad Sci USA 102(26):94069411.
51. Guan Y, et al. (2005) Thiazolidinediones expand body fluid volume through PPARgamma
stimulation of ENaC-mediated renal salt absorption. Nat Med 11(8):861866.
52. Kuipers I, et al. (2010) Activation of liver X receptor-alpha reduces activation of the
renal and cardiac renin-angiotensin-aldosterone system. Laboratory Investigation
90(4):630636.
53. Wang F, et al. (2015) Renal medullary (pro)renin receptor contributes to angiotensin
II-induced hypertension in rats via activation of the local renin-angiotensin system.
BMC Med 13:278.
Lu et al. PNAS Early Edition
|
9of9
MEDICAL SCIENCES PNAS PLUS
... While recombinant s(P)RR altered markers of endothelial function in vitro, the physiological relevance of the s(P)RR concentrations used in this study is a potential limitation. Several in vitro mechanistic studies of s(P)RR function have used treatment concentrations between 10 and 100 nM [12,[41][42][43] to determine the functional role s(P)RR pathways in pathological conditions. However, studies assessing maternal circulating s(P)RR levels in preeclamptic women have shown that concentrations are between 30 and 40 ng/ml (approximately 1.1-1.5 nM) [39,44]. ...
Article
Full-text available
Preeclampsia is classified as new-onset hypertension coupled with gross endothelial dysfunction. Placental (pro)renin receptor ((P)RR) and plasma soluble (P)RR (s(P)RR) are elevated in patients with preeclampsia. Thus, we aimed to interrogate the role (P)RR may play in the pathogenesis of preeclampsia. Human uterine microvascular endothelial cells (HUtMECs, n = 4) were cultured with either; vehicle (PBS), 25–100 nM recombinant s(P)RR, or 10 ng/ml TNF-a (positive control) for 24 h. Conditioned media and cells were assessed for endothelial dysfunction markers via qPCR, ELISA, and immunoblot. Angiogenic capacity was assessed through tube formation and adhesion assays. Additionally, pregnant rats were injected with an adenovirus overexpressing s(P)RR from mid-pregnancy (day 8.5), until term ( n = 6–7 dams/treatment). Maternal and fetal tissues were assessed. HUtMECs treated with recombinant s(P)RR displayed increased expression of endothelial dysfunction makers including vascular cell adhesion molecule-1, intracellular adhesion molecule-1, and endothelin-1 mRNA expression ( P = 0.003, P = 0.001, P = 0.009, respectively), along with elevated endothelin-1 protein secretion ( P < 0.001) compared with controls. Recombinant s(P)RR impaired angiogenic capacity decreasing the number of branches, total branch length, and mesh area ( P < 0.001, P = 0.004, and P = 0.009, respectively), while also increasing vascular adhesion ( P = 0.032). +ADV rats exhibited increased systolic ( P = 0.001), diastolic ( P = 0.010), and mean arterial pressures ( P = 0.012), compared with -ADV pregnancies. Renal arteries from +ADV-treated rats had decreased sensitivity to acetylcholine-induced relaxation ( P = 0.030), compared with -ADV pregnancies. Our data show that treatment with s(P)RR caused hypertension and growth restriction in vivo and caused marked endothelial dysfunction in vitro. These findings demonstrate the significant adverse actions of s(P)RR on vascular dysfunction that is characteristic of the preeclamptic phenotype.
... After cleavage, s(P)RR could be released into the extracellular fluid, such as plasma and urine [9][10][11][12]. Similar to the regulatory mission of f(P)RR, s(P)RR has also confirmed its role in controlling RAS activation, vessel function, and electrolyte balance and confirmed its clinical correlation in the development of various diseases [13][14][15][16][17]. Furthermore, although it still has controversy, the residual transmembrane and intracellular domains after (P)RR cleavage might be defined as M8.9 fragment. ...
Article
Full-text available
The (pro)renin receptor ((P)RR) is an essential component of the renin–angiotensin system (RAS) as a specific single-pass transmembrane receptor for prorenin and renin and has now emerged as a multifunctional protein implicated in a wide variety of developmental and physio-pathological processes and pathways. The (P)RR may be of pathological significance in metabolic syndrome. The (P)RR has received much consideration; substantial efforts have been made to understand the localization, regulation, and function of the (P)RR at both a molecular and system level. (P)RR regulation of cell function depends on whether it is intact or cleaved into its constituent forms. Therefore, the present chapter describes immunohistochemical approaches to examine the expression of (P)RR in various organs. It was shown that different molecular forms of (P)RR could be present in different tissue compartments in almost all organs. Among them, the liver has high PRR activity. Our findings could elucidate more detailed distribution of different (P)RR molecular forms in different organs, which could provide useful information to further investigate the pathophysiological mechanisms of the development of various diseases in the future.
... The fPRR is consumed by proteases to produce sPRR, which contains the N-terminal extracellular domains of the PRR [22,[52][53][54], a C-terminal intracellular domain of 8.9 kDa, and an ATPase-associated truncated protein [55]. Like fPRR, sPRR has a crucial role in various physiological and pathological processes [56], such as the activation of tissue RAS [57], regulation of water balance [58], hypertension [59], and tubular injury [60]. It has been demonstrated that excess albumin can activate the expression of sPRR in HK-2 renal proximal tubular epithelial cells [16]. ...
Article
Full-text available
Chronic kidney disease (CKD) is associated with advanced oxidation protein products (AOPPs). A recent study has shown that AOPP-induced renal tubular injury is mediated by the (pro)renin receptor (PRR). However, it is unclear whether the PRR decoy inhibitor PRO20 can protect against renal damage related to AOPPs in vivo. In this study, we examined the role of the PRR in rats with AOPP-induced renal oxidative damage. Male SD rats were subjected to unilateral nephrectomy, and after a four-day recuperation period, they were randomly divided into four groups (n = 6/group) for four weeks: control (CTR), unmodified rat serum albumin (RSA, 50 mg/kg/day via tail-vein injection), AOPPs-RSA (50 mg/kg/day via tail-vein injection), and AOPPs-RSA + PRO20 (50 mg/kg/day via tail-vein injection + 500 μg/kg/day via subcutaneous injection) groups. PRO20 was administered 3 days before AOPPs-RSA injection. Renal histopathology evaluation was performed by periodic acid–Schiff (PAS) staining, and biochemical parameters related to renal injury and oxidative stress biomarkers were evaluated. The expression of related indicators was quantified by RT-qPCR and immunoblotting analysis. In the results, rats in the AOPPs-RSA group exhibited higher levels of albuminuria, inflammatory cell infiltration, and tubular dilation, along with upregulation of oxidative stress, profibrotic and proinflammatory factors, and elevation of AOPP levels. Meanwhile, in the PRO20 group, these were significantly reduced. Moreover, the levels of almost all components of the renin-angiotensin system (RAS) and Nox4-dependent H2O2 production in urine and the kidneys were elevated by AOPPs-RSA, while they were suppressed by PRO20. Furthermore, AOPPs-RSA rats showed elevated kidney expression of the PRR and soluble PRR (sPRR) and increased renal excretion of sPRR. In summary, these findings suggest that PRR inhibition may serve as a protective mechanism against AOPP-induced nephropathy by inhibiting the intrarenal RAS and Nox4-derived H2O2 mechanisms.
... The truncated membrane-bound form remains inside the cell and interacts with vacuolar H + -ATPase (V-ATPase) as a component of the multisubunit membrane-bound proton pump [2,9]. s(P)RR, which comprises most of the ECD, is secreted extracellularly and interacts with Frizzled-8 (FZD8) on the surface of the renal collecting duct principal cells, enhancing the urineconcentrating capability via FZD8-dependent β-catenin signaling [11,12]. The full-length (P)RR is a component of the Wnt receptor complex [13]. ...
Article
Full-text available
We have previously reported that monoclonal antibodies against the (pro)renin receptor [(P)RR] can reduce the Wnt/β-catenin-dependent development of pancreatic ductal adenocarcinoma (PDAC), the most common pancreatic cancer. Antibodies against two (P)RR regions (residues 47–60 and 200–213) located in the extracellular domain (ECD) reduced the proliferation of human PDAC cells in vitro. Although these regions probably participate in the activation of Wnt/β-catenin signaling, their functional significance remains unclear. Moreover, the (P)RR ECD is predicted to possess an intrinsically disordered region (IDR), which allows multiple protein interactions because of its conformational flexibility. In this study, we investigated the significance of the two regions and the IDR by in silico 3D structural analysis using the AlphaFold2 program and evolutionary sequence conservation profile. The model showed that ECD adopted a folded domain (residues 17–269) and had an IDR (residues 270–296). The two regions mapped onto the structural model formed a continuous surface patch comprising evolutionarily conserved hydrophobic residues. The homodimeric structure predicted by AlphaFold2 showed that full-length (P)RR comprising the ECD, single-span transmembrane, and cytoplasmic domains formed a twofold symmetric dimer via the ECD, which explains the experimentally proven homodimerization. The dimer model possessed two hand-shaped grooves with residues 47–60 and 200–213 in their palms and the IDR as their fingers. Based on these findings, we propose that the IDR-containing hydrophobic grooves act as a binding site for (P)RR and perform multiple functions, including Wnt signaling activation.
Article
Soluble prorenin receptor (sPRR), a component of the renin angiotensin system (RAS), has been identified as a plasma biomarker for hypertension and cardiovascular diseases in humans. Despite studies showing that sPRR in kidney is produced by renal collecting duct (CD) tubular cells, its biological actions modulating cardiorenal function remains unknown. Therefore, this study aimed to create a new murine model to investigate how expressing human sPRR (HsPRR) in CD influence the intrarenal RAS status and renal hemodynamics in male and female mice fed a regular chow. CD-HsPRR mice were generated by breeding human sPRR-Myc-tag transgenic mice with transgenic mice expressing Hoxb7/Cre, which showed increased renal sPRR expression but unchanged circulating levels compared to CTL mice in both sexes. Plasma RAS levels were not influenced by the expression of HsPRR in CD as well. CD-HsPRR expression showed only in female littermates: 1) increased 24-hour blood pressure, due to elevated daytime mean and systolic values, 2) impaired blood pressure response to an acute dose of losartan and reduced chronic angiotensin II (AngII)-hypertension, 3) reduced ACE activity and Ang II content in renal cortex, 4) decreased GFR, with no changes in natriuresis and kaliuresis despite the upregulation of β-ENaC in renal cortex. Taken together, these data suggest that expression of HsPRR in CD could interact with Ang II type 1 receptor mediating a sex-specific, Ang II-independent renal dysfunction and pro-hypertensive phenotype.
Article
Full-text available
Accumulating evidence has demonstrated the existence of extra-adrenal aldosterone in various tissues, including the brain, heart, vascular, adipocyte, and kidney, mainly based on the detection of the CYP11B2 (aldosterone synthase, cytochrome P450, family 11, subfamily B, polypeptide 2) expression using semi-quantitative methods including reverse transcription-polymerase chain reaction and antibody-based western blotting, as well as local tissue aldosterone levels by antibody-based immunosorbent assays. This mini-review highlights the current evidence and challenges in extra-adrenal aldosterone, focusing on intrarenal aldosterone. A narrative review. Locally synthesized aldosterone may play a vital role in various physio-pathological processes, especially cardiovascular events. The site of local aldosterone synthesis in the kidney may include the mesangial cells, podocytes, proximal tubules, and collecting ducts. The synthesis of renal aldosterone may be regulated by (pro)renin receptor/(pro)renin, angiotensin II/Angiotensin II type 1 receptor, wnt/β-catenin, cyclooxygenase-2/prostaglandin E2, and klotho. Enhanced renal aldosterone release promotes Na+ reabsorption and K+ excretion in the distal nephron and may contribute to the progress of diabetic nephropathy and salt-related hypertension. Inhibition of intrarenal aldosterone signaling by aldosterone synthase inhibitors or mineralocorticoid receptor antagonists may be a hopeful pharmacological technique for the therapy of diabetic nephropathy and saltrelated hypertension. Yet, current reports are often conflicting or ambiguous, leading many to question whether extra-adrenal aldosterone exists, or whether it is of any physiological and pathophysiological significance.
Article
Full-text available
Purpose Investigate whether local angiotensin II (AngII) and its AngII type 1 and 2 receptors (AT1R, AT2R) in the endometrium are different and correlate with microvessel density in women with reproductive failure and pregnancy outcomes. Methods Endometrium during the window of implantation from 40 women with recurrent miscarriage (RM) and 40 with recurrent implantation failure (RIF) were compared with 27 fertile women. Peri-implantation endometrium from 54 women prior to euploid embryo transfer were collected and compared in women with successful pregnancy and unsuccessful pregnancy. Results Compared with fertile women, expression of AT2R was significantly lower, while AT1R/AT2R expression ratio was significantly higher in the stroma of the RIF group. Endometrium arteriole MVD was significantly lower and negatively correlated with the AT1R/AT2R expression ratio in the stroma of the RIF group. No significant differences and correlations were found in the RM group. Compared with the pregnancy group, expression of AT1R and AT2R were significantly lower in all compartments, but only AT1R/AT2R ratio was significantly higher in the stroma of the non-pregnancy group. Similarly, endometrium arteriole MVD was also significantly lower and negatively correlated with the AT1R/AT2R ratio in the stroma of the non-pregnancy group. Conclusion Local renin-angiotensin system is dysregulated in peri-implantation endometrium and associated with abnormal angiogenesis in RIF and poor implantation outcome after embryo transfer.
Article
Full-text available
Chronic kidney disease (CKD) is associated with advanced oxidation protein products 8 (AOPPs). A recent study has shown that AOPPs-induced renal tubular injury is mediated by the 9 (pro)renin receptor (PRR). However, it is unclear whether the PRR decoy inhibitor PRO20 can pro-10 tect against renal damage related to AOPPs in vivo. In this study, we examined the role of PRR in 11 rats with AOPPs-induced renal oxidative damage. Male SD rats were subjected to unilateral ne-12 phrectomy, after a four-day recuperation period, they were randomly divided into four groups (n = 13 6/group) for four weeks: control (CTR), unmodified rat serum albumin (RSA, 50 mg/kg/day via tail-14 vein injection), AOPPs-RSA (50 mg/kg/day via tail-vein injection), and AOPPs-RSA + PRO20 (50 15 mg/kg/day via tail-vein injection + 500 μg/kg/day via subcutaneous injection) groups, PRO20 was 16 administered 3 days before AOPPs-RSA injection. Renal histopathology evaluation was performed 17 by periodic acid Schiff (PAS) staining, and biochemical parameters related to renal injury and oxi-18 dative stress biomarkers were evaluated. The expression of related indicators was quantified by RT-19 qPCR and Immunoblotting analysis. In the results, rats in the AOPPs-RSA group exhibited higher 20 levels of albuminuria, inflammatory cell infiltration, and tubular dilation, along with upregulation 21 of oxidative stress, profibrotic and proinflammatory factors, and elevation of AOPPs levels. 22 Whereas in the PRO20 group, these were significantly reduced. Moreover, the levels of almost all 23 components of the renin-angiotensin system (RAS) and Nox4-dependent H2O2 production in urine 24 and kidneys were elevated by AOPPs-RSA, while they were suppressed by PRO20. Furthermore, 25 AOPPs-RSA rats showed elevated kidney expression of PRR and soluble PRR (sPRR), and increased 26 renal excretion of sPRR. In summary, these findings suggest that PRR inhibition may serve as a 27 protective mechanism against AOPPs induced nephropathy by inhibiting the intrarenal RAS and 28 Nox4-derived H2O2 mechanisms. 29
Article
Final urine volume and concentration are defined by water reabsorption through the water channel proteins aquaporin (AQP)-2, -3 and -4 in the collecting duct. However, the transcriptional regulation of these AQPs is not well understood. The Hippo/Yes-associated protein 1 (YAP) pathway plays an important role in organ size control and tissue homeostasis. When the Hippo pathway including the Mst1/Mst2 kinases is inhibited, YAP is activated and functions as a transcription co-activator. Our previous work revealed a pathological role of tubular YAP activation in chronic kidney disease, but the physiological role of YAP in the kidney remains to be established. Here, we found that tubule-specific Yap knockout mice showed increased urine output and decreased urinary osmolality. Decreases in Aqp2, -3 and -4 mRNA and protein abundance in the kidney were evident in Yap knockout mice. Analysis of Mst1/Mst2 double knockout and Mst1/Mst2/Yap triple knockout mice showed that expression of Aqp2 and Aqp4 but not Aqp3 was dependent on YAP. Furthermore, YAP was recruited to the promoters of the Aqp2 and Aqp4 genes and stimulated their transcription. Interestingly, YAP was found to interact with transcription factors GATA2, GATA3 and NFATc1. These three factors promoted Aqp2 transcription in a YAP dependent manner in collecting duct cells. These three factors also promoted Aqp4 transcription whereas only GATA2 and GATA3 enhanced Aqp3 transcription. Thus, our results suggest that YAP promotes Aqp2 and Aqp4 transcription, interacts with GATA2, GATA3 and NFATc1 to control Aqp2 expression, while Aqp-2, -3 and -4 exploit overlapping mechanisms for their baseline transcriptional regulation.
Article
Increasing evidence showed the generation of aldosterone in the collecting duct (CD), a major site for the synthesis and action of prostaglandin E2 (PGE2). Both aldosterone and PGE2 are stimulated by Na⁺ depletion or K⁺ loading or angiotensin II. However, what happens first and the correlation between PGE2 and aldosterone in the CD remains obscure. PGE2 stimulates aldosterone release in the adrenal zona glomerulosa cells via E-prostanoid (EP) receptors by activating the cAMP/cAMP-responsive element-binding pathway. Here, we further tested the hypothesis that the synthesis of aldosterone is stimulated by PGE2 in CD cells and the underlying mechanism. In primary mouse inner medullary CD cells, PGE2 incubation significantly stimulated aldosterone release in parallel with an increase of CYP11B2 (cytochrome P-450, family 11, subfamily B, polypeptide 2), (pro)renin receptor (PRR), and prorenin protein expression and upregulation of medium soluble PRR (sPRR), prorenin/renin, and cAMP levels, both of which were attenuated by the EP1 antagonist SC-19220 and the protein kinase C (PKC) inhibitor calphostin C. PGE2-induced release of aldosterone, prorenin/renin, and sPRR was also abolished by the protein kinase A inhibitor H89. Both a PRR antagonist PRO20 and β-catenin inhibitor ICG001 blunted PGE2 or sPRR-induced aldosterone and prorenin/renin secretion. Exogenous sPRR protein incubation not only stimulated the release of aldosterone and prorenin/renin but also reversed PF429242 (an inhibitor of subtilisin kexin isozyme-1, which can block endogenous sPRR generation)-attenuated aldosterone and prorenin/renin secretion. Therefore, we conclude that PGE2 increases CD aldosterone synthesis through the EP1 receptor via the PKC/cAMP/sPRR/β-catenin pathway.
Article
Full-text available
Within the kidney, the (pro)renin receptor (PRR) is predominantly expressed in the collecting duct (CD), particularly in intercalated cells, and it is regulated by the PGE2 receptor EP4. Notably, EP4 also controls urinary concentration through regulation of aquaporin 2 (AQP2). Here, we tested the hypothesis that sequential activation of EP4 and PRR determines AQP2 expression in the CD, thus mediating the antidiuretic action of vasopressin (AVP). Water deprivation (WD) elevated renal PRR expression and urinary soluble PRR excretion in rats. Intrarenal infusion of a PRR decoy peptide, PRO20, or an EP4 antagonist partially prevented the decrease in urine volume and the increase in urine osmolality and AQP2 expression induced by 48-hour WD. In primary cultures of rat inner medullary CD cells, AQP2 expression induced by AVP treatment for 24 hours depended on sequential activation of the EP4 receptor and PRR. Additionally, mice lacking PRR in the CD exhibited increased urine volume and decreased urine osmolality under basal conditions and impaired urine concentrating capability accompanied by severe volume loss and a dangerous level of plasma hyperosmolality after WD. Together, these results suggest a previously undescribed linear AVP/PGE2/EP4/PRR pathway in the CD for regulation of AQP2 expression and urine concentrating capability.
Article
Full-text available
The collecting duct (CD) has been recognized as an important source of prorenin/renin and it also expresses (pro)renin receptor (PRR). The goal of this study was to examine the hypothesis that prorenin or renin via PRR regulates epithelial Na(+) channel (ENaC) activity in mpkCCD cells. Transepithelial Na(+) transport was measured by using a conventional epithelial volt-ohmmeter and was expressed as the calculated equivalent current (Ieq). Amiloride-inhibitable Ieq was used as a reflection of ENaC activity. Administration of prorenin in the nanomolar range induced a significant increase in Ieq that was detectable as early as 1 min, peaked at 5 min, and gradually returned to baseline within 15 min. These changes in Ieq were completely prevented by a newly developed PRR decoy inhibitor, PRO20. Prorenin-induced Ieq was inhibitable by amiloride. As compared with prorenin, renin was less effective in stimulating Ieq. Prorenin-induced Ieq was attenuated by apocynin but enhanced by Tempol, the latter effect being prevented by catalase. In response to prorenin treatment, the levels of total reactive oxygen species and H2O2 were both increased, as detected by spin-trap analysis and ROS-Glo™ H2O2 Assay, respectively. Both siRNA-mediated Nox4 knockdown and the dual Nox1/4 inhibitor GKT137892 attenuated prorenin-induced Ieq. Overall, our results demonstrate that activation of PRR by prorenin stimulates ENaC activity in CD cells via Nox4-derived H2O2.
Article
Full-text available
Background: (Pro)renin receptor (PRR) is a new component of the renin-angiotensin system and regulates renin activity in vitro. Within the kidney, PRR is highly expressed in the renal medulla where its expression is induced by angiotensin II infusion. The objective of the present study was to test a potential role of renal medullary PRR during angiotensin II-induced hypertension. Methods: A rat AngII infusion model (100 ng/kg/min) combined with renal intramedullary infusion of PRO20, a specific inhibitor of PRR, was builded. And the intravenous PRO20 infusion serve as control. Mean arterial pressure was recorded by radiotelemetry for one week. Further anaylsis of kidney injury, inflammation, biochemical indices and protein localization were perrformed in vivo or in vitro. Results: Radiotelemetry demonstrated that AngII infusion elevated the mean arteria pressure from 108 ± 5.8 to 164.7 ± 6.2 mmHg. Mean arterial pressure decreased to 128.6 ± 5.8 mmHg (P < 0.05) after intramedullary infusion of PRO20, but was only modestly affected by intravenous PRO20 infusion. Indices of kidney injury, including proteinuria, glomerulosclerosis, and interstitial fibrosis, inflammation, and increased renal medullary and urinary renin activity following angiotensin II infusion were all remarkably attenuated by intramedullary PRO20 infusion. Following one week of angiotensin II infusion, increased PRR immunoreactivity was found in vascular smooth muscle cells. In cultured rat vascular smooth muscle cells, angiotensin II induced parallel increases in soluble PRR and renin activity, and the latter was significantly reduced by PRO20. Conclusion: Renal medullary PRR mediates angiotensin II-induced hypertension, likely by amplifying the local renin response.
Article
Full-text available
Aquaporin-2 (AQP2) mediates arginine vasopressin (AVP)-induced water reabsorption in the kidney collecting duct. AVP regulates AQP2 expression primarily via Gsα/cAMP/PKA signaling. Tankyrase, a member of PARP family, is known to mediate Wnt/β-catenin signaling-induced gene expression. We examined whether tankyrase plays a role in AVP-induced AQP2 regulation via ADP-ribosylation of G protein-alpha (Gα) and/or β-catenin-mediated transcription of AQP2. RT-PCR and immunoblotting analysis revealed the mRNA and protein expression of tankyrase in mouse kidney and mouse collecting duct mpkCCDc14 cells. dDAVP-induced AQP2 upregulation was attenuated in mpkCCDc14 cells under the tankyrase inhibition by XAV939 treatment or siRNA knockdown. FRET image analysis, however, revealed that XAV939 treatment did not affect dDAVP- or forskolin-induced PKA activation. Inhibition of tankyrase decreased dDAVP-induced phosphorylation of β-catenin (S552) and nuclear translocation of phospho-β-catenin. siRNA-mediated knockdown of β-catenin decreased forskolin-induced AQP2 transcription and dDAVP-induced AQP2 expression. Moreover, inhibition of PI3K/Akt, which was associated with decreased nuclear translocation of β-catenin, diminished dDAVP-induced AQP2 upregulation, further indicating that β-catenin mediates AQP2 expression. Taken together, tankyrase plays a role in AVP-induced AQP2 regulation, which is likely via β-catenin-mediated transcription of AQP2, but not ADP-ribosylation of Gα. The results provide novel insights on vasopressin-mediated urine concentration and homeostasis of body water metabolism. Copyright © 2014, American Journal of Physiology - Renal Physiology.
Article
Full-text available
Prorenin receptor (PRR) is a multi-functioning protein possessing at least four different roles: (1) working as a receptor for renin and prorenin producing angiotensin I from angiotensinogen thus enhancing the tissue renin-angiotensin system; (2) inducing intracellular signals when a ligand binds to PRR; (3) participating in the functions of vacuolar proton ATPase; and (4) constituting the Wnt signaling receptor complex. Here, the roles of PRR in kidney physiology and diabetic conditions as well as recent findings regarding a soluble form of PRR are discussed. We also propose the possible mechanism concerning diabetic nephropathy as "trade-off hypothesis" from a PRR point of view. In brief, under hyperglycemic conditions, injured podocytes degrade degenerated proteins and intracellular organelles which require V-ATPase and PRR for vesicle internal acidification. Sustained hyperglycemia overproduces PRR molecules, which are transported to the transmembrane and bind to increased serum prorenin in the diabetic condition. This enhances tissue renin-angiotensin system and PRR-mediated mitogen-activated protein kinase signals, resulting in increased injurious molecules such as transforming growth factor-β, cyclooxygenase2, interleukin-1β, and tumor necrosis factor-α ending in diabetic nephropathy progression. Although many findings led us to better PRR understanding, future works should elucidate which PRR functions, of the four discussed here, are dominant in each cell and kidney disease context.
Article
The prorenin receptor (PRR), a recently discovered component of the renin angiotensin system, is expressed in the nephron in general and the collecting duct in particular. However, the physiological significance of nephron PRR remains unclear, partly due to developmental abnormalities associated with global or renal-specific PRR gene knockout. Therefore, we developed mice with inducible nephron-wide PRR deletion using the Pax8-rtTA and LC-1 transgenes and loxP flanked PRR alleles such that ablation of PRR occurs in adulthood, after induction with doxycycline. Nephron specific PRR knockdown (KO) mice have normal survival to ~1 year of age and no renal histological defects. Compared to controls, PRR KO mice had 65% lower medullary PRR mRNA and protein levels and markedly diminished renal PRR immunofluorescence. During both normal water intake and mild water restriction, PRR KO mice had significantly lower urine osmolality, higher water intake and higher urine volume compared to controls. No differences were seen in urine vasopressin excretion, urine Na+ and K+ excretion, plasma Na+ or plasma osmolality between the two groups. However, PRR KO mice had reduced medullary AQP-2 levels and AVP-stimulated cAMP accumulation in isolated renal medulla as compared to controls. Taken together, these results suggest nephron PRR can potentially modulate renal water excretion. Copyright © 2015, American Journal of Physiology - Renal Physiology.
Article
Angiotensin II (Ang II) stimulates (pro) renin receptor (PRR) expression in the renal collecting duct, triggering the local renin response in the distal nephron. Our recent study provided evidence for involvement of cyclooxygenase-2-prostaglandin E-2 pathway in Ang II-dependent stimulation of PRR expression in the collecting duct. Here, we tested the role of E-prostanoid (EP) subtypes acting downstream of cyclooxygenase-2 in this phenomenon. In primary rat inner medullary collecting duct cells, Ang II treatment for 12 hours induced a 1.8-fold increase in the full-length PRR protein expression. To assess the contribution of EP receptor, the cell was pretreated with specific EP receptor antagonists: SC-51382 (for EP1), L-798106 (for EP3), L-161982 (for EP4), and ONO-AE3-208 (ONO, a structurally distinct EP4 antagonist). The upregulation of PRR expression by Ang II was consistently abolished by L-161982 and ONO and partially suppressed by SC-51382 but was unaffected by L-798106. The PRR expression was also significantly elevated by the EP4 agonist CAY10598 in the absence of Ang II. Sprague-Dawley rats were subsequently infused for 1 or 2 weeks with vehicle, Ang II alone, or in combination with ONO. Ang II infusion induced parallel increases in renal medullary PRR protein and renal medullary and urinary renin activity and total renin content, all of which were blunted by ONO. Both tail cuff plethysmography and telemetry demonstrated attenuation of Ang II hypertension by ONO. Overall, these results have established a crucial role of the EP4 receptor in mediating the upregulation of renal medullary PRR expression and renin activity during Ang II hypertension.
Article
The (pro)renin receptor ((P)RR) is expressed in several tissues including kidney, heart and brain, and is thought to regulate the tissue renin-angiotensin system (RAS) through the non-proteolytic activation of prorenin. (P)RR is cleaved by furin to generate soluble (P)RR (s(P)RR), which is secreted into the extracellular space. s(P)RR is a candidate biomarker reflecting the status of the tissue RAS. Here, we investigated the relationship between background factors and serum s(P)RR levels. We measured s(P)RR levels in 122 patients with essential hypertension (EH) and assessed the relationships between background factors and s(P)RR levels. Serum s(P)RR levels were 19.0±4.9 ng ml(-1). Single regression analyses showed that age (r=0.251, P<0.01), serum creatinine levels (r=0.229, P<0.05) and urinary angiotensinogen excretion (r=0.196, P<0.05) were positively correlated with s(P)RR levels, whereas estimated glomerular filtration rate (eGFR; r=-0.337, P<0.001) were negatively correlated. Multiple regression analyses of age, blood pressure (BP), hemoglobin A1c (HbA1c) and s(P)RR levels revealed that age and s(P)RR levels were negatively correlated with the eGFR (P<0.05). In patients with EH, serum s(P)RR levels correlated positively with renal function independent of age, BP and HbA1c. These findings support s(P)RR as a useful biomarker that reflects the status of the tissue RAS.Hypertension Research advance online publication, 20 March 2014; doi:10.1038/hr.2014.46.
Article
A decade ago, the (P)RR [(pro)renin receptor] was discovered and depicted as a potential activator of the tissue renin-angiotensin system. For this reason, the role of the (P)RR in cardiovascular diseases and diabetes has been particularly studied. However, the discovery of embryonic lethality after (P)RR gene deletion in mouse and zebrafish paved the way for additional roles of (P)RR in cell homoeostasis. Indeed, the (P)RR has been shown to associate with vacuolar H+-ATPase, hence its other name ATP6ap2. Developmental studies in Xenopus and Drosophila have revealed an essential role of this association to promote the canonical and non-canonical Wnt signalling pathways, whereas studies with tissue-specific gene deletion have pointed out a role in autophagy. The present review aims to summarize recent findings on the cellular functions of (P)RR emerging from various mutated and transgenic animal models.