Anda di halaman 1dari 6

Farnesoid X receptor is essential for the survival of

renal medullary collecting duct cells under


hypertonic stress
Sujuan Xua,1, Shizheng Huangb,1, Zhilin Luana,1, Tingyue Chena, Yuanyi Weia, Miaomiao Xinga, Yaqing Lia, Chunxiu Dua,
Bing Wanga, Feng Zhenga, Nanping Wanga,c, Youfei Guana,c,2, Jan-Åke Gustafssond,e,2, and Xiaoyan Zhanga,c,2
a
Advanced Institute for Medical Sciences, Dalian Medical University, Dalian, 116044 Liaoning, China; bRenal Electrolyte and Hypertension Division,
Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104; cDepartment of Physiology and Pathophysiology, School of Basic Medical
Sciences, Dalian Medical University, Dalian, 116044 Liaoning, China; dCenter for Nuclear Receptors and Cell Signaling, Department of Biology and
Biochemistry, University of Houston, Houston, TX 77204; and eCenter for Biosciences, Department of Biosciences and Nutrition, Karolinska Institutet,
Novum, 14186 Stockholm, Sweden

Contributed by Jan-Åke Gustafsson, April 10, 2018 (sent for review March 6, 2018; reviewed by Chuanming Hao and Baoxue Yang)

Hypertonicity in renal medulla is critical for the kidney to produce state in renal medulla, which is essential for the generation of a
concentrated urine. Renal medullary cells have to survive high concentrated urine (3).
medullary osmolarity during antidiuresis. Previous study reported Renal medulla is a unique tissue in which residing cells in-
that farnesoid X receptor (FXR), a nuclear receptor transcription cluding MCDs are exposed to the harsh hypertonic and hypoxic
factor activated by endogenous bile acids, increases urine concen- environment and have to survive significant rises in NaCl and
trating ability by up-regulating aquaporin 2 expression in medul- urea concentrations during antidiuresis. Multiple osmoprotective
lary collecting duct cells (MCDs). However, whether FXR is also mechanisms have been reported to be important in maintaining
involved in the maintenance of cell survival of MCDs under the survival of renal medullary cells under hypertonic stress (4–
dehydration condition and hypertonic stress remains largely un- 7). Among them, it is generally believed that the transcription
known. In the present study, we demonstrate that 24-hours water
factor tonicity-responsive enhancer binding protein (TonEBP)
restriction selectively up-regulated renal medullary expression of
and its target osmoprotective genes including aldose reductase
FXR with little MCD apoptosis in wild-type mice. In contrast, water
(AR) and heat shock protein 70 (HSP70) represent the most
deprivation caused a massive apoptosis of MCDs in both global
FXR gene-deficient mice and collecting duct-specific FXR knockout
important protective mechanism.
mice. In vitro studies showed that hypertonicity significantly
Farnesoid X receptor (FXR), a member of the nuclear re-
increased FXR and tonicity response enhancer binding protein ceptor superfamily, is a transcription factor activated by endog-
(TonEBP) expression in mIMCD3 cell line and primary cultured enous bile acids. In addition to the liver and small intestine
MCDs. Activation and overexpression of FXR markedly increased where FXR plays an important role in bile acid, glucose, and
cell viability and decreased cell apoptosis under hyperosmotic lipid metabolism, the kidney also exhibits highly abundant FXR
conditions. In addition, FXR can increase gene expression and expression, especially in renal collecting duct cells (8). It has
nuclear translocation of TonEBP. We conclude that FXR protects been previously reported that activation of FXR increases urine
MCDs from hypertonicity-induced cell injury very likely via in- concentration by up-regulating the expression of AQP2, one of
creasing TonEBP expression and nuclear translocation. This study its direct target genes, in MCDs (9). However, whether FXR is
provides insights into the molecular mechanism by which FXR en-
hances urine concentration via maintaining cell viability of MCDs Significance
under hyperosmotic condition.
As a ligand-activated transcription factor, farnesoid X receptor
bile acid receptor | NFAT5 | hypertonicity | osmoprotection | cell viability (FXR) is abundantly expressed in the liver and small intestine,
where it plays an important role in the maintenance of bile

W hole-body water homeostasis is critical for human health


and depends on the balance between water intake stimu-
lated by thirst and water excretion controlled mainly by the
acid, lipid, and glucose homeostasis. Increasing evidence is
emerging that FXR may be a critical regulatory factor in renal
physiology and pathophysiology. The present study demon-
kidneys. Each day, an adult human produces ∼1.5 L of urine strates an essential role of FXR in promoting cell survival of
despite 180 L of fluid filtered through glomerular basement medullary collecting duct cells under hypertonic stress and
membrane. Approximately, 90% of glomerular filtrate is con- identifies FXR-induced TonEBP expression and activation as an
stitutively reabsorbed in the proximal tubules and descending underlying mechanism. This work provides an insight into the
loop of Henle. The remaining filtered water is reabsorbed in role of FXR in renal urine concentration.
renal medullary collecting duct cells (MCDs), where the water
Author contributions: Y.G., J.-Å.G., and X.Z. designed research; S.X., S.H., Z.L., T.C., Y.W.,
transport is tightly regulated by arginine vasopressin (AVP), a M.X., Y.L., C.D., and B.W. performed research; N.W., Y.G., and J.-Å.G. contributed new
circulating hormone also known as antidiuretic hormone reagents/analytic tools; F.Z. and X.Z. analyzed data; and Y.G., J.-Å.G., and X.Z. wrote
(ADH) (1). the paper.
Under dehydration condition, AVP is produced by the hypo- Reviewers: C.H., Fudan University; and B.Y., Peking University.
thalamus and released from the neurohypophysis into the blood The authors declare no conflict of interest.
in response to increased plasma osmolarity or reduced blood Published under the PNAS license.
volume. It promotes water reabsorption in MCDs by increasing 1
S.X., S.H., and Z.L. contributed equally to this work.
gene expression and apical membrane targeting of aquaporin 2 2
To whom correspondence may be addressed. Email: guanyf@dmu.edu.cn, jgustafs@
(AQP2) mainly through the AVP-V2R-PKA pathway (2). How- central.uh.edu, or zhangxy@dmu.edu.cn.
ever, AVP also increases sodium chloride (NaCl) reabsorption in This article contains supporting information online at www.pnas.org/lookup/suppl/doi:10.
the thick ascending limb (TAL) and distal convolute tubule and 1073/pnas.1803945115/-/DCSupplemental.
urea permeability in the inner MCDs to establish a hyperosmotic Published online May 8, 2018.

5600–5605 | PNAS | May 22, 2018 | vol. 115 | no. 21 www.pnas.org/cgi/doi/10.1073/pnas.1803945115


also involved in the maintenance of cell survival of MCDs under and primary cultured mouse MCDs were exposed to hyperto-
hypertonic stress remains largely unknown. The present studies nicity. As shown in Fig. 2, FXR was constitutively and func-
aimed to investigate whether FXR plays an important role in tionally expressed in mIMCD3 cells (Fig. 2 A and B), where its
regulating cell survival of MCDs under hyperosmotic stress expression was significantly induced by hypertonicity at mRNA
in vitro and in vivo. The underlying mechanism by which FXR (Fig. 2C) and protein levels (Fig. 2D). Similarly, in primary
affects cell viability of MCDs exposed to hypertonic condition cultured mouse MCD cells, FXR was constitutively expressed (SI
was also examined. Appendix, Fig. S1) and increased hypertonicity significantly up-
regulated FXR expression at mRNA (SI Appendix, Fig. S2A) and
Results protein (SI Appendix, Fig. S2 B and C) levels. Compared with the
Water Restriction Selectively Up-Regulated FXR Expression in Mouse levels of FXR at 800 mOsm, FXR protein expression at
Renal Medulla. Wild-type (WT) C57BL/6 mice were subjected to 900 mOsm was markedly reduced, which might be due to re-
water deprivation (WD) for 24 h. Compared with the mice with duced cell viability of MCD cells treated with 900 mOsm (SI
free access to water, water-deprived mice exhibited ∼10-fold and Appendix, Fig. S2B).
∼2-fold increase in FXR expression in renal medulla at mRNA and
protein level, respectively (Fig. 1). In contrast to renal medulla, FXR Activation and Overexpression Protected mIMCD3 Cells from
cortical expression of FXR at both mRNA and protein level Hypertonicity-Induced Cell Death. To investigate the role of FXR
remained unaltered (Fig. 1). Because renal medullas of dehydrated in cell survival under hypertonic stress, mIMCD3 cells were
mice are hypertonic, these results suggest FXR may be regulated by treated with an endogenous agonist (CDCA) and a synthetic
agonist (GW4064) of FXR or infected with an adenovirus
hypertonicity and plays an important role in urine concentration.
expressing FXRα2 (9). Cells were treated with either isotonic
Hypertonicity Induced FXR Expression in Cultured MCDs. Since FXR (300 mOsm) or hyperosmotic medium (600 mOsm), and cell
was mainly expressed in MCDs (9), we first determined whether viability was determined by the MTT assay. FXR activation by
hyperosmolarity can induce FXR expression. mIMCD3 cell line both CDCA and GW4064 significantly reduced cell death in-
duced by hypertonic stress, with little effect on cell viability un-
der the isotonic condition (Fig. 3A). Similarly, overexpression of
FXR with Ad-FXRα2 markedly promoted cell survival under
hyperosmotic challenge (Fig. 3B). Flow cytometry analysis was
further performed to examine hypertonicity-induced apoptosis of
mIMCD3 cells. FXR activation by CDCA and GW4064 signifi-
cantly reduced hypertonicity (600 mOsm)-induced cell apoptosis
(Fig. 3C). Consistently, CDCA and GW4064 treatment blocked
hypertonicity-induced cleavage of procaspase-3 (Fig. 3D). These

PHYSIOLOGY
findings suggest that FXR may promote cell survival of MCDs
under dehydration state possibly by blocking caspase-3 activation-
associated apoptosis.

FXR Gene Deficiency Potentiated MCD Cell Apoptosis in Dehydrated


Mice in Vivo. To address whether FXR promotes the survival of
MCDs under dehydrated condition, WT, global FXR gene-
deficient mice (FXR−/−), and collecting duct-specific FXR gene
knockout mice (CD-FXR−/−) were generated and were deprived
of water for 24 h. As shown in Fig. 4 A and B, the TUNEL assay
demonstrated that water restriction induced a small amount
apoptosis of MCDs in WT mice but resulted in a massive apo-
ptosis in FXR−/− mice. FXR−/− mice exhibited much more ap-
optotic cells than WT mice after 24-h water restriction. The
majority of the apoptotic cells were the MCDs (Fig. 4 A and B).
To further confirm the role of collecting duct FXR, collecting
duct-specific FXR gene knockout mice (CD-FXR−/−) were
generated by crossing FXR flox/flox mice with AQP2-Cre mice
as previously reported (10–13) and were subjected to 24-h WD.
Compared with WT mice, CD-FXR−/− mice also displayed a signif-
icant increase in apoptotic cells of medullary collecting ducts after
water restriction (SI Appendix, Fig. S3). Together, these findings
clearly demonstrate that FXR can protect MCDs from hypertonicity-
induced injury and promote their survival under hypertonic stress.

FXR Gene Knockout Mice Exhibited Attenuated Renal TonEBP Expression


After 24-h Water Restriction. Tonicity-responsive enhancer binding
protein (TonEBP) is a transcription factor promoting cellular ac-
cumulation of organic osmolytes in the hypertonic renal medulla by
stimulating expression of its target genes including aldose reductase
Fig. 1. Water restriction selectively induced FXR expression in renal me-
(AR) and heat shock protein 70 (HSP70) (7). To elucidate the
dullas of C57BL/6 mice. Male C57BL/6 mice were water-deprived for 24 h. (A)
FXR mRNA expression was selectively induced in renal medulla. FXR mRNA
underlying mechanism by which FXR protects MCDs from hyper-
levels were analyzed by qRT-PCR, *P < 0.05 vs. control (n = 4). (B) FXR protein tonic stress, renal expression of TonEBP and its target genes AR
expression was significantly increased in renal medulla as assessed by and HSP70 was determined. As expected, 24-h dehydration
Western blot. Lamin A/C was used as protein loading control. (C) Statistical markedly increased renal TonEBP expression in WT mice at
analysis of Western blot shown in B, **P < 0.01 vs. control (n = 4). both mRNA (Fig. 4C) and protein level (Fig. 4D). However,

Xu et al. PNAS | May 22, 2018 | vol. 115 | no. 21 | 5601


Fig. 2. Hypertonicity-induced FXR expression in
mouse MCDs. (A) Constitutive expression of FXR in
mouse inner MCDs (mIMCD3). Cultured mIMCD3 cells
were transfected with an EGFP-AQP2 expression vec-
tor and then exposed to hypertonic stress (600 mOsm)
for 6 h. Note that FXR was mainly expressed in the
nucleus (red) and AQP2 was predominantly located in
cell membrane (green). (Scale bar, 25 uM.) (B) Lucif-
erase reporter assay demonstrating that hypertonicity
at 600 mOsm significantly induced FXR transcrip-
tion activity. ***P < 0.001 vs. 300 mOsm (n = 12).
(C) Induction of FXR mRNA expression by hyper-
tonicity in a dose-dependent manner. Cultured
mIMCD3 cells were incubated with hypertonic so-
lutions for 6 h. *P < 0.05 vs. 300 mOsm (n = 4). (D)
Western blot assay showing that hypertonicity
significantly induced FXR protein expression in
mIMCD3 cells. Cells were exposed to various hy-
pertonic stress (400, 500, and 600 mOsm) for 6 h.

in FXR − /− mice, 24-h water restriction just slightly induced FXR Induced TonEBP Expression and Nuclear Translocation in Cultured
TonEBP mRNA and protein expression (Fig. 4 C and D). MCDs. To confirm the effect of FXR on TonEBP expression and
These results suggest that FXR may exert osmoprotective activity, primary cultured mouse MCDs were treated with two
effect on MCDs via increasing TonEBP expression and FXR agonists or infected with Ad-FXRα2. As shown in Fig. 5 A
transcriptional activity in renal medulla. and B, CDCA and GW4064 treatment markedly up-regulated

Fig. 3. FXR protected mIMCD3 cells from hypertonic stress-induced cell apoptosis. The mIMCD3 cells were subjected to hypertonicity (600 mOsm) for 6 h in
the presence or absence of FXR selective agonist GW4064 and CDCA. (A) MTT cell viability analysis showing that hypertonicity-induced cell death was sig-
nificantly attenuated by GW4064 and CDCA treatment. **P < 0.01 vs. 300 mOsm+DMSO, ##P < 0.01 vs. 600 mOsm+DMSO (n = 10). (B) Overexpression of FXR-
ameliorated mIMCD3 cell death under hypertonic stress. Cells were infected with Ad-FXRα2 or Ad-VP16. After 36 h of infection, the cells were challenged with
hypertonic stress (600 mOsm) for 6 h. Cell viability was determined by the MTT assay. ***P < 0.001 vs. 300 mOsm+Ad-VP16, ###P < 0.001 vs. 600 mOsm+Ad-
VP16 (n = 6). (C) FXR activation blocked hypertonicity-induced apoptosis of mIMCD3 cells. The mIMCD3 were treated with isotonic (300 mOsm) or hypertonic
medium (600 mOsm) for 6 h, and apoptotic cells were analyzed by the flow cytometry. Treatment of cells with GW4064 (2.5 μM) and CDCA (50 μM) markedly
reduced hypertonicity-induced cell apoptosis. ***P < 0.001 vs. 300 mOsm+DMSO, and #P < 0.05 and ##P < 0.01 vs. 600 mOsm+DMSO (n = 3). (D) FXR ac-
tivation with GW4064 and CDCA markedly reduced hypertonicity-induced activation of the caspase-3 apoptotic cascade. Note that protein expression of the
apoptotic marker, cleaved caspase-3, was markedly attenuated by GW4064 and CDCA. The results are representative of three independent experiments.

5602 | www.pnas.org/cgi/doi/10.1073/pnas.1803945115 Xu et al.


Fig. 4. FXR deficiency accelerated MCD cell apoptosis and attenuated TonEBP expression in dehydrated mice. C57BL/6 mice were water-deprived for 24 h.
Apoptotic cells were identified as dark brown staining by the TUNEL assay. Mice with free access to water were used as control. (A) TUNEL assay dem-
onstrating increased apoptotic cells were observed in the kidneys of FXR−/− mice with 24-h water restriction. Immunohistochemical staining of AQP2 (light
blue) was used to indicate renal-collecting ducts. Twenty-four-hour WD barely induced apoptosis of renal medullary cells in WT mice (FXR+/+) but resulted
in a massive apoptosis in FXR−/− mice (arrows). (Scale bar, 10 uM.) (B) Quantitative analysis of apoptotic cells in renal medullas of FXR+/+ and FXR−/− mice.
*P < 0.05 vs. FXR+/+ control mice, ##P < 0.01 vs. FXR+/+ with WD, &&&P < 0.001 vs. FXR−/− control mice (n = 3). (C and D) WD failed to induce TonEBP
expression at mRNA (C) and protein level (D) in the kidneys of FXR−/− mice. FXR +/+ and FXR−/− mice were water-deprived for 24 h. TonEBP mRNA and
protein expression was measured by real-time PCR and immunoblot, respectively. *P < 0.05 vs. FXR+/+ control mice, #P < 0.05 vs. FXR−/− with WD (n = 6).
CD, collecting duct.

TonEBP mRNA and protein expression. FXR activation also tissues with active cholesterol metabolism including the liver,

PHYSIOLOGY
induced protein expression of FXR and TonEBP target genes intestine, and adrenal gland, where FXR plays an important role
including AR and Hsp70 (Fig. 5B). Similarly, overexpression of in bile acid biosynthesis and transport, cholesterol metabolism,
FXRα2 via an adenovirus-based approach increased TonEBP and glucocorticoid production (17–19). It has been previously
expression at both mRNA and protein levels (Fig. 5 C and D). reported that FXR is also highly expressed in the kidney and is
Expression of FXR as well as AR and Hsp70 was also signifi- critical for maintaining renal function (9). Disruption of FXR
cantly induced (Fig. 5D). These findings demonstrate that FXR gene causes a significant defect in urine concentrating ability,
can promote TonEBP expression at both mRNA and protein level. resulting in a polyuria phenotype in mice. In addition, FXR
It is clear that transcriptional activity of TonEBP depends on dysfunction is associated with a worsened renal injury in diabetic
its nuclear translocation (14, 15). To further test whether FXR mice (20). The present study further demonstrates that FXR
can increase TonEBP transcriptional activity, primary MCDs gene deficiency significantly increases cell death of MCDs under
were treated with the FXR agonist CDCA and GW4064 or hypertonic conditions, while FXR activation and overexpression
overexpressed with FXRα2. Both FXR activation and over- markedly reduces hypertonicity-induced apoptosis of MCDs.
expression markedly promoted TonEBP nuclear translocation These findings provide evidence that FXR is a key factor in pro-
(Fig. 5 E and F). Importantly, in primary cultured MCDs from moting cell survival of MCDs in hypertonic renal medulla, thereby
FXR−/− mice, Ad-FXRα2 infection not only rescued FXR ex- maintaining renal urine concentrating ability during antidiuresis.
pression, but also caused a significant translocation of TonEBP TonEBP, also called nuclear factor of activated T-cell 5
from the cytoplasm to the nucleus (SI Appendix, Fig. S4). To- (NFAT5), is a transcription factor critical for osmoadaptive re-
gether, these findings demonstrate that FXR indeed increases sponse of renal medullary cells to hyperosmolar stress (14). It
TonEBP activity by enhancing its nuclear translocation. exerts an osmoprotective effect on renal medullary cells mainly
via up-regulating gene transcription of many of its target genes
Discussion including AR, HSP70, and aquaporins (AQPs) (15). Genetically
This study demonstrates a critical role of FXR in the survival of modified animals with deficient TonEBP activity in the kidney
MCDs under dehydration condition. Twenty-four-hour water suffer from severe medullary atrophy in association with cell
restriction selectively up-regulates renal medullary expression of death, demonstrating that TonEBP is essential for the survival of
FXR with little MCD apoptosis in WT mice. In contrast, WD the renal medullary cells (7). Although it is well accepted that
causes a massive apoptosis of MCDs in both global FXR gene- TonEBP is a key transcription factor in osmoprotection, mech-
deficient mice and collecting duct-specific FXR knockout mice. anisms involved in TonEBP expression and activation remain
In cultured MCDs, activation and overexpression of FXR largely unknown. In the present study, we found that renal
markedly promote cell viability and increase TonEBP expression TonEBP expression was significantly reduced in FXR−/− mice
and activity under hyperosmotic stress. FXR may represent a with WD, while FXR activation markedly up-regulated expres-
factor protecting the MCDs from hypertonic stress-induced in- sion of TonEBP and its target genes. These findings raise a
jury via increasing TonEBP activity, thereby maintaining the ability possibility that FXR may be a transcription factor important for
of renal medulla to concentrate urine under hydration status. driving TonEBP expression. However, the underlying mecha-
FXR belongs to the superfamily of nuclear receptor tran- nism warrants further investigation.
scription factors and can be activated by many naturally occurred The present study also provides clear evidence that FXR can
endogenous bile acids (16). It is abundantly expressed in the promote nuclear translocation of TonEBP protein in MCDs. It is

Xu et al. PNAS | May 22, 2018 | vol. 115 | no. 21 | 5603


Fig. 5. FXR up-regulated TonEBP expression and nuclear translocation in primary cultured mouse IMCD cells. Primary MCD cells were treated with GW4064
(2.5 μM) and CDCA (50 μM) for 6 h or infected with Ad-FXRα2. (A) Real-time PCR analysis demonstrating mRNA expression of TonEBP in primary MCD cells
isolated from WT mice. **P < 0.01, ***P < 0.001 vs. DMSO (n = 4). (B) Western blot assay showing that treatment of MCDs with GW4064 and CDCA markedly
induced protein expression of FXR, TonEBP, and osmoprotective genes HSP70 and AR. (C) Quantitative PCR analysis showing that overexpression of FXR
significantly up-regulated TonEBP mRNA levels in MCDs. *P < 0.05 vs. Ad-VP16 (n = 8). (D) Immunoblot assay demonstrating overexpression of FXR markedly
induced FXR, TonEBP, HSP70, and AR expression. (E) FXR activation promoted nuclear translocation of TonEBP. Primary MCDs were treated with GW4064 and
CDCA for 6 h. Confocal immunofluorescence was performed to detect TonEBP (red) nuclear translocation. The nucleus was visualized by staining with DAPI
(blue). (Scale bar, 5 uM.) (F) FXR overexpression with Ad-FXRα2 markedly facilitated nuclear translocation of TonEBP. Cells infected with a control virus (Ad-
VP16) had little effect on TonEBP nuclear localization. (Scale bar, 5 uM.)

well known that nuclear import of TonEBP is essential for its Similar to TonEBP, FXR may also represent a hypertonicity-
transcriptional activity. Multiple mechanisms have been reported responsive gene. Twenty-four-hour water restriction selectively up-
to contribute to the nucleocytoplasmic shuttling of TonEBP regulated FXR expression in renal medulla at both mRNA and
protein, which requires the presence of the nuclear export signal protein levels. Similarly, hypertonic stress significantly induced
and nuclear localization signal at its N terminus (21). The sig- FXR mRNA and protein levels and its transcriptional activity in
naling cascades of Fyn, a shrinkage-activated tyrosine kinase, and cultured MCDs. These findings demonstrate that FXR gene is
p38, a subgroup of the mitogen-activated kinases, have been transcriptionally regulated in response to hypertonicity. However,
reported to be major signaling pathways for hypertonic activation it is currently unclear whether there is a hypertonicity-responsive
of TonEBP (22). In contrast, transcriptional coactivator with element in the promoter region of FXR gene. It is also uncertain
PDZ-binding motif (TAZ) can suppress TonEBP activity
whether FXR is under the transcriptional regulation of TonEBP.
through tyrosine phosphorylation (23). We report here that FXR
Addressing these important issues may significantly advance our
activation and overexpression can dramatically diminish cyto-
plasmic TonEBP levels and increase its nuclear localization, knowledge in understanding the mechanism by which FXR is regu-
providing a mechanism for TonEBP transactivation. Currently, lated and the importance of FXR in renal physiology.
the mechanism by which FXR drives nuclear import of TonEBP In summary, the present studies demonstrate that dehydration
is not clear. One possibility worth addressing is that FXR may and hypertonicity increase FXR expression and its transcrip-
form a transcriptional complex with TonEBP, which increases tional activity in the MCDs. FXR gene deficiency significantly
both TonEBP nuclear translocation and its transactivation. potentiates MCD death in mice with water restriction. FXR
Given a ubiquitous distribution pattern of both FXR and activation and overexpression markedly promote cell survival of
TonEBP, they may act in concert in regulating many other bi- cultured MCDs. FXR-driven TonEBP expression and nuclear
ological processes than those involved in the response to hy- translocation may contribute to its osmoprotective role in
pertonicity in renal medulla. renal medulla.

5604 | www.pnas.org/cgi/doi/10.1073/pnas.1803945115 Xu et al.


Materials and Methods protocols were reviewed and approved by the Animal Care and Use Review
Male C57BL/6 WT mice (8-10 wk old) were purchased from the Experimental Committee of Dalian Medical University.
Animal Center at Peking University Health Science Center. FXR knockout Chemicals and reagents, methods of cell viability assay, real-time PCR,
mice were purchased from the Jackson Laboratory. Renal collecting duct- Western blot, immunostaining, flow cytometry, primary culture of mouse
specific FXR gene knockout mice were generated by crossing FXRflox/flox IMCD cells, and luciferase assay, TUNEL assay, immunofluorescence
mice with an aquaporin 2 gene promoter driven-Cre transgenic mouse and staining, and statistical analyses are described in SI Appendix, Materials
validated as previously described (10–13). Eight- to 10-week-old male ani- and Methods.
mals used in the study were housed on a daily 12-h light/black cycle under
controlled temperature and humidity in the animal facility of Peking Uni- ACKNOWLEDGMENTS. We thank Dr. Frank Gonzalez for providing FXRflox/
versity Health Science Center. All animals had free access to food. Mice were flox mice. This work was supported by the National Natural Science
allowed free access to water or subjected to water restriction for 24 h. After Foundation of China Grants 81570636, 81722010 (to X.Z.), 81390351, and
water restriction, mice were killed and perfused with cold PBS. The left 91639201 (to Y.G.); Dalian High-level Talent Innovation Support Pro-
kidneys were fixed and paraffin-embedded, while the right kidneys were gram 2016RD13; and Peacock Plan of Shenzhen city Grant KQTD20140630100746562.
used to dissect renal cortex and medulla. The use of animals and the study J.Å.G. was supported by Robert A. Welch Foundation Grant E-0004.

1. Knepper MA, Kwon TH, Nielsen S (2015) Molecular physiology of water balance. N 13. Gao M, et al. (2015) Disruption of prostaglandin E2 receptor EP4 impairs urinary
Engl J Med 372:1349–1358. concentration via decreasing aquaporin 2 in renal collecting ducts. Proc Natl Acad Sci
2. Li Y, Wei Y, Zheng F, Guan Y, Zhang X (2017) Prostaglandin E2 in the regulation of USA 112:8397–8402.
water transport in renal collecting ducts. Int J Mol Sci 18:E2539. 14. Ferraris JD, Burg MB (2006) Tonicity-dependent regulation of osmoprotective genes
3. Bachmann S, Mutig K (2017) Regulation of renal Na-(K)-Cl cotransporters by vaso- in mammalian cells. Contrib Nephrol 152:125–141.
pressin. Pflugers Arch 469:889–897. 15. Burg MB, Ferraris JD, Dmitrieva NI (2007) Cellular response to hyperosmotic stresses.
4. Hao CM, et al. (2000) Dehydration activates an NF-kappaB-driven, COX2-dependent Physiol Rev 87:1441–1474.
survival mechanism in renal medullary interstitial cells. J Clin Invest 106:973–982. 16. Shapiro H, Kolodziejczyk AA, Halstuch D, Elinav E (2018) Bile acids in glucose me-
5. Han Q, et al. (2011) AMPK potentiates hypertonicity-induced apoptosis by suppress- tabolism in health and disease. J Exp Med 215:383–396.
ing NFκB/COX-2 in medullary interstitial cells. J Am Soc Nephrol 22:1897–1911. 17. Inagaki T, et al. (2005) Fibroblast growth factor 15 functions as an enterohepatic
6. Hao CM, Redha R, Morrow J, Breyer MD (2002) Peroxisome proliferator-activated
signal to regulate bile acid homeostasis. Cell Metab 2:217–225.
receptor delta activation promotes cell survival following hypertonic stress. J Biol
18. Hoekstra M, et al. (2012) FXR agonist GW4064 increases plasma glucocorticoid levels
Chem 277:21341–21345.
in C57BL/6 mice. Mol Cell Endocrinol 362:69–75.
7. Lee SD, et al. (2011) TonEBP stimulates multiple cellular pathways for adaptation to
19. Houten SM, Volle DH, Cummins CL, Mangelsdorf DJ, Auwerx J (2007) In vivo imaging
hypertonic stress: Organic osmolyte-dependent and -independent pathways. Am J
of farnesoid X receptor activity reveals the ileum as the primary bile acid signaling
Physiol Renal Physiol 300:F707–F715.
8. Zhang XY, Wang B, Guan YF (2016) Nuclear receptor regulation of aquaporin-2 in the tissue. Mol Endocrinol 21:1312–1323.
kidney. Int J Mol Sci 17:E1105. 20. Herman-Edelstein M, Weinstein T, Levi M (2018) Bile acid receptors and the kidney.
9. Zhang X, et al. (2014) Farnesoid X receptor (FXR) gene deficiency impairs urine con- Curr Opin Nephrol Hypertens 27:56–62.
centration in mice. Proc Natl Acad Sci USA 111:2277–2282. 21. Cheung CY, Ko BC (2013) NFAT5 in cellular adaptation to hypertonic stress–Regula-
10. Nelson RD, et al. (1998) Expression of an AQP2 Cre recombinase transgene in kidney tions and functional significance. J Mol Signal 8:5.
and male reproductive system of transgenic mice. Am J Physiol 275:C216–C226. 22. Ko BC, et al. (2002) Fyn and p38 signaling are both required for maximal hypertonic

PHYSIOLOGY
11. Sinal CJ, et al. (2000) Targeted disruption of the nuclear receptor FXR/BAR impairs bile activation of the osmotic response element-binding protein/tonicity-responsive en-
acid and lipid homeostasis. Cell 102:731–744. hancer-binding protein (OREBP/TonEBP). J Biol Chem 277:46085–46092.
12. Guan Y, et al. (2005) Thiazolidinediones expand body fluid volume through PPAR- 23. Jang EJ, et al. (2012) TAZ suppresses NFAT5 activity through tyrosine phosphorylation.
gamma stimulation of ENaC-mediated renal salt absorption. Nat Med 11:861–866. Mol Cell Biol 32:4925–4932.

Xu et al. PNAS | May 22, 2018 | vol. 115 | no. 21 | 5605

Anda mungkin juga menyukai