Download as pdf or txt
Download as pdf or txt
You are on page 1of 17

REVIEWS

Aquaporin 2 regulation: implications


for water balance and polycystic
kidney diseases
Emma T. B. Olesen   1,2 ✉ and Robert A. Fenton   3 ✉
Abstract | Targeting the collecting duct water channel aquaporin 2 (AQP2) to the plasma
membrane is essential for the maintenance of mammalian water homeostasis. The vasopressin V2
receptor (V2R), which is a GS protein-coupled receptor that increases intracellular cAMP levels,
has a major role in this targeting process. Although a rise in cAMP levels and activation of protein
kinase A are involved in facilitating the actions of V2R, studies in knockout mice and cell models
have suggested that cAMP signalling pathways are not an absolute requirement for V2R-mediated
AQP2 trafficking to the plasma membrane. In addition, although AQP2 phosphorylation is a
known prerequisite for V2R-mediated plasma membrane targeting, none of the known AQP2
phosphorylation events appears to be rate-limiting in this process, which suggests the
involvement of other factors; cytoskeletal remodelling has also been implicated. Notably, several
regulatory processes and signalling pathways involved in AQP2 trafficking also have a role in
the pathophysiology of autosomal dominant polycystic kidney disease, although the role of AQP2
in cyst progression is unknown. Here, we highlight advances in the field of AQP2 regulation that
might be exploited for the treatment of water balance disorders and provide a rationale for
targeting these pathways in autosomal dominant polycystic kidney disease.

Single effect
A cornerstone of internal medicine is the assessment renal outer medullary potassium channel 1 (ROMK1;
The difference in osmotic and treatment of water and electrolyte disorders, and encoded by KCNJ1) provide the single effect for the coun-
pressure that is created by the mechanisms underlying water preservation are a tercurrent multiplication mechanism3, which dilutes the
the active transport of sodium major aspect of kidney physiology. Adult human kid- urine and generates a corticomedullary osmotic gradient
chloride out of the tubular fluid in
the thick ascending limb into the
ney glomeruli filtrate on average 180 l of plasma per day in the interstitium. In combination with urea transport
interstitium; the hyperosmotic and 99% of the filtered water is reabsorbed back into the processes, this gradient provides the driving force for
gradient in the interstitium drives circulation through the epithelium of the kidney tubule regulated water reabsorption in collecting duct principal
the passive movement of water and the collecting duct. This reabsorption process occurs cells, which express AQP2, AQP3 and AQP4. The cellu-
from the tubular fluid of the
mainly through transcellular water transport, which is lar abundance of AQPs 2–4 and the presence of AQP2 in
descending limbs and collecting
ducts into the interstitium.
crucially dependent on osmosis-driven water movement the apical plasma membrane, which is crucial to water
through aquaporin (AQP) water channels (Fig. 1). The reabsorption, are regulated by the antidiuretic peptide
1
Department of Biomedical kidney proximal tubule and the adjacent initial portion hormone vasopressin (also known as arginine vasopres-
Sciences, Faculty of Health
of the long-looped thin descending limb1 are constitu- sin or antidiuretic hormone). Vasopressin is released
and Medical Sciences,
University of Copenhagen,
tively water permeable and reabsorb ~90% of filtered from the neurohypophysis in response to increases in
Copenhagen, Denmark. water. AQP1, which is present in the apical and basolat- plasma osmolality or activation of baroreceptors through
2
Department of Endocrinology eral plasma membrane in these segments, and potentially decreases in effective circulating volume (Fig. 1). In the
and Nephrology, North Zealand AQP7 in the apical plasma membrane of the proximal collecting duct, vasopressin-mediated stimulation of
Hospital, Hillerød, Denmark. straight tubule, are responsible for this water flux1,2. The the vasopressin V2 receptor (V2R; encoded by AVPR2),
3
Department of Biomedicine, remaining portion of the loop of Henle and the distal con- which is a GS protein-coupled receptor (GSPCR), increases
Faculty of Health, Aarhus voluted tubule (DCT) are impermeable to water and the cAMP levels and activates protein kinase A (PKA). In the
University, Aarhus, Denmark.
transport of ions in these segments occurs mainly through TAL, stimulation of V2R by vasopressin also enhances
✉e-mail: etbo@sund.ku.dk;
secondary active transport against the transepithelial the countercurrent multiplication and maximizes the
robert.a.fenton@
biomed.au.dk gradient. In the thick ascending limb (TAL), Na+ and Cl− osmotic gradient required for water reabsorption.
https://doi.org/10.1038/ reabsorption through Na–K–Cl cotransporter 2 (NKCC2; Defects in the vasopressin–AQP2 axis result in water
s41581-021-00447-x also known as SLC12A1 or BSC1) and K+ recycling via balance disorders (reviewed in refs4,5). The majority of

Nature Reviews | Nephrology

0123456789();:
Reviews

Key points delayed disease progression in clinical trials7–9. We dis-


cuss how advances in the field of AQP2 regulation might
• Targeting of the collecting duct water channel aquaporin 2 (AQP2) to the plasma translate to clinical benefit for patients with ADPKD
membrane is essential for the maintenance of mammalian water homeostasis. and might lead to the development of novel treatment
• Although vasopressin signalling via the Gs protein-coupled vasopressin V2 receptor options to treat ADPKD and/or alleviate polyuria, which
(V2R) is a major receptor-mediated pathway that modulates trafficking of AQP2 is a major adverse effect of tolvaptan treatment.
to the plasma membrane, this targeting can also occur independently of V2R.
• The canonical V2R signalling pathway — increased cAMP levels, activation of protein Acute regulation of water balance
kinase A (PKA) and AQP2 phosphorylation — cannot fully explain AQP2 plasma The importance of vasopressin and the V2R for
membrane targeting, and cAMP–PKA-independent pathways exist.
the maintenance of water homeostasis is evident in
• Novel regulators of AQP2 plasma membrane targeting that are independent of V2R Brattleboro rats, which lack vasopressin secretion and
and cAMP include A-kinase anchoring protein (AKAP)–PKA disruptors, Src inhibition,
develop central diabetes insipidus10, and in mice with
Wnt5a, fluconazole and epidermal growth factor receptor antagonists.
an Avpr2 deletion11, which triggers NDI (Table 1). The
• Research on the signalling networks and regulatory steps that govern AQP2
V2R is expressed in cells of the TAL, DCT and inner
trafficking has identified promising pharmacological targets for water balance
disorders.
medulla collecting duct (IMCD), where it stimulates
phosphorylation and activity of NKCC2, the NaCl
• Investigating plasma membrane targeting of AQP2 using agents that do not increase
cotransporter NCC (also known as SLC12A3) and
cellular proliferation and epithelial secretion is required to delineate the independent
role of AQP2 in cyst progression in autosomal dominant polycystic kidney disease. AQP2, respectively12–15. Of note, V2R expression is lower
in the TAL and DCT than in the collecting duct, and
vasopressin-induced cAMP levels are 15–20-fold lower
these disorders arise from inappropriately high water in the TAL than in the collecting duct12,16,17. The V2R
reabsorption despite normal extracellular osmolality, selectively activates adenylate cyclase type 6 (AC6) in the
for example, due to vasopressin secretion in response IMCD18 to produce cAMP, which is a major V2R second
to decreased effective circulating volume in the absence messenger and leads to the activation of a range of protein
of alterations in plasma osmolality. Water is conse- kinases, including PKA; V2R activation also increases
quently reabsorbed by the kidney despite an isosmotic or intracellular calcium19. Although the actions of the V2R
hypo-osmotic extracellular microenvironment. Patients are crucial for maintaining urine concentration at base-
with these disorders mainly present with oedema, which line, rats treated with a V2R antagonist could increase
occurs frequently in patients with heart failure or liver urine osmolality during water restriction20, which sug-
cirrhosis, and hyponatraemia, which is a hallmark of gests that receptors other than V2R might contribute
the syndrome of inappropriate antidiuretic hormone. to water homeostasis (water restriction experiments
Conversely, loss of vasopressin-secreting neurons, for in Avpr2-knockout mice have not been reported).
example, due to traumatic brain injury, leads to central Alternative receptors shown to be important for
diabetes insipidus, which is characterized by reduced regulation of AQP2 include the angiotensin II receptor
circulating vasopressin, reduced AQP2 and low urine type I (AT1; encoded by AGTR1), the bile acid recep-
osmolality compared with healthy individuals. Central tor (also known as farnesoid X-activated receptor), the
diabetes insipidus frequently causes dehydration and oxysterols receptor LXRβ, the oestrogen receptor (also
hypernatraemia, but responds to exogenous vasopressin known as ERα), the peroxisome proliferator-activated
administration6. Reductions in AQP2 levels or plasma receptor-γ, and the glucocorticoid and mineralocorticoid
membrane accumulation can also be caused by lithium receptors21–24. The G protein-coupled bile acid receptor 1
treatment or hypercalcaemia, or, in very rare cases, by (encoded by GPBAR1; also known as TGR5) can also
genetic disorders that disrupt the function of the V2R inc­rease cAMP levels, AQP2 protein abundance and
or AQP2. This loss of AQP2 or V2R functionality AQP2 plasma membrane targeting in IMCD cells,
causes vasopressin-resistant polyuria, which is termed and Gpbar1-knockout mice had a urine-concentrating
nephrogenic diabetes insipidus (NDI) and can lead to defect both at baseline and throughout a 36-h period
hypernatraemia and dehydration if high water intake of water restriction23. However, whether GPBAR1 has a
is not maintained. Understanding the regulation of direct role in water homeostasis in the collecting duct
water balance at the molecular level might therefore not remains unclear because Gpbar1-knockout mice also
only improve the therapeutic options for the treatment have reduced vasopressin levels. AQP2 plasma mem-
of water balance disorders, but might also clarify the brane targeting and/or cellular water permeability can
pathophysiology of diseases that originate in the kidney also be increased by the GPCR ligands calcitonin25,
tubular system. glucagon26, secretin27, angiotensin II28 or selective ago-
In this Review, we highlight new developments in nists for the prostaglandin receptors EP2 (ref.29) and EP4
the field of AQP2 regulation, with a particular focus (ref.11), which suggests that alternative GPCRs might
on the intracellular signalling pathways that link recep- have a role in water balance, especially during water
tor stimulation to the targeting of AQP2 to the plasma restriction.
membrane. In addition, we discuss the possible role
of water transport in an important kidney tubular Novel aspects of AQP2 regulation
disease — autosomal dominant polycystic kidney disease Long-term vasopressin-mediated regulation of AQP2
(ADPKD). Specific treatments that can prevent kidney involves protein synthesis and degradation, whereas
failure in high-risk patients with ADPKD are not cur- acute regulation involves apical plasma membrane inser-
rently available, although the V2R antagonist tolvaptan tion and retrieval. At baseline (that is, in non-stimulated

www.nature.com/nrneph

0123456789();:
Reviews

Stimulation of baroreceptors
or osmoreceptors
Hypothalamus

Pituitary
gland
Vasopressin
release

PCT

TAL CD

Urine Cl– Na K+
+
K+
H2O H2O H2O H2O
ROMK1

NKCC2

Apical
AQP1 AQP7 K+ AQP2
Endocytosis

PCT TAL CD
Translocation
Translocation Activation
AQP3 AQP4
Basolateral
V2R
Vasopressin
H2O H2O H2O
Blood

Fig. 1 | Kidney aquaporins and transcellular water transport. Most of the glomerular filtrate is reabsorbed iso-osmotically
through aquaporin 1 (AQP1) in the proximal convoluted tubule (PCT) and, to a lesser extent, through AQP7 in the late
proximal tubule segments; AQP1 is also expressed in the thin descending limbs and descending vasa recta (not shown).
Water reabsorption in these segments of the kidney is not regulated by vasopressin, which in response to a reduction in
circulating blood volume or an increase in plasma osmolality, is released from the pituitary gland following the activation of
baroreceptors or osmoreceptors in the hypothalamus. In the kidney, vasopressin binds to the vasopressin V2 receptor (V2R)
and enhances NaCl reabsorption in the thick ascending limb (TAL) via the concerted actions of Na–K–Cl cotransporter 2
(NKCC2) and renal outer medullary potassium channel 1 (ROMK1), which increases interstitial osmolality. Vasopressin also
enhances AQP2 levels in the apical plasma membrane of collecting duct (CD) principal cells by stimulating AQP2
translocation to the apical plasma membrane from intracellular storage vesicles and inhibiting AQP2 endocytosis. The
Recycling endosomes sustained expression of AQP2 in the apical plasma membrane, in combination with basolateral expression of AQP3 and
Organelles in the endocytic AQP4, provides a pathway for osmotic reabsorption of water.
pathway, in which plasma
membrane proteins and lipids cells), the trafficking pool of AQP2 is predominantly which renders it permeable to water. Upon removal of
that are internalized by
endocytosis are sorted and
present in recycling endosomes30. Following stimulation vasopressin, AQP2 builds up in clathrin-coated pits in
processed for export back to of the collecting duct principal cell, these vesicles traf- the plasma membrane of AQP2-transfected LLC-PK1
the cell surface for reuse. fic, dock and fuse with the apical plasma membrane, cells (AQP2–LLC-PK1; Table 1), where it undergoes

Nature Reviews | Nephrology

0123456789();:
Reviews

Table 1 | Advantages and limitations of experimental models used in AQp2 research


Model Description Benefits Cautionary issues
IMCD cells Primary culture derived from Retain most features of native IMCD including Need to be stimulated with cAMP or
freshly isolated inner medulla V2R expression and (inducible) expression of vasopressin to sustain AQP2 expression;
AQP2 cAMP or vasopressin are usually removed
~16 h before experiments
Possible inter-experimental variation in
cell growth or cell purity
AQP2 traffics primarily to the basolateral
membrane following V2R stimulation
V2R is internalized in culture
mpkCCD14 Immortalized cell line derived Retain many features of native tissue Immortalized cells might differ from native
cells from mouse cortical collecting Express V2R, and V2R stimulation induces AQP2
tissue
duct expression Often cultured in media containing
hormones and/or growth factors
Form polarized monolayers on semi-permeable
supports and AQP2 is targeted to the apical Usually stimulated with dDAVP for several
plasma membrane in response to vasopressin days to induce AQP2 expression before
experiments; dDAVP is usually removed
1–4 h before experiments
MCD4 cells Cell line derived from mouse Collecting duct cell line Transfected cell line
cortical collecting duct and No previous stimulation is required for Regulation of Aqp2 transcription cannot
transfected with AQP2 constitutive AQP2 expression be investigated owing to the use of a
non-native promoter
AQP2 accumulates on the apical membrane
in response to V2R stimulation
MDCK–AQP2 Canine kidney epithelial cells Native AQP2 expression is low and cells are Variable response to V2R stimulation
cells transfected with AQP2 thus suitable for transfection with wild-type and hence studies of AQP2 membrane
AQP2 or AQP2 harbouring point mutations (for targeting are frequently performed with
example, mutations of specific phosphorylation the adenylyl cyclase stimulator forskolin
or ubiquitylation sites)
Tubular origin not completely clarified
Cells form polarized monolayers on
AQP2 reportedly traffics to apical and
semi-permeable supports
basolateral sides
Transfected cell line
Limited availability of commercial
antibodies reactive to canine epitopes
(AQP2–) Porcine proximal tubule cell line Stimulation with vasopressin increases cAMP Transfected cell line
LLC-PK1 cells transfected with AQP2 levels and AQP2 plasma membrane targeting Not a collecting duct cell line
No native AQP2 expression, thus cells are Does not consistently respond to dDAVP
suitable for transfection with wild-type or
mutated AQP2 Studies require the use of vasopressin and
might be confounded by the activity of
V1Rs
AQP2 reportedly traffics to the basolateral
membrane upon stimulation with
vasopressin
Brattleboro rats Rats with a spontaneous single- A rat model for studying vasopressin- Rats might have enhanced vasopressin-
nucleotide mutation in Avp that independent mechanisms independent V2R signalling owing to
results in defective vasopressin binding of alternative hormones such
Response to vasopressin or dAVP is higher than
secretion as oxytocin
in wild-type rats, making it a useful model for
studying vasopressin effects in general
Pkd1-knockout Mice with inducible Pkd1 Mice survive until adulthood, whereas Pkd1−/− Inducible deletion requires chemical
mice deletion or Pkd1+/− mice mice die perinatally induction
Inducible Pkd1 deletion leads to cyst Pkd1+/− mice have slow cyst progression
development within weeks of induction (occurs at 7–8 months of age)
AQP2, aquaporin 2; dDAVP, D-amino D-arginine vasopressin; IMCD, inner medulla collecting duct; MDCK, Madin–Darby canine kidney; V2R, vasopressin V2 receptor.

dynamin-dependent endocytosis31. In vivo, and in stud- of AQP2 in these experimental systems is also depend-
ies with primary IMCD cells and the mpkCCD14 cell line ent on dynamin is unclear32–35. Following endocytosis,
(Table 1), AQP2 colocalizes with clathrin, the early endo­ Rab7 and vacuolar protein sorting-associated pro­tein 35
some markers Ras-related protein Rab5A–early (Vps35) are required for sorting AQP2 within Rab5+
endosome antigen 1 (EEA1) and the lysosomal marker early endosomes, either to the lysosome for degradation36
cathepsin D at baseline; however, whether endocytosis or to the plasma membrane via recycling endosomes

www.nature.com/nrneph

0123456789();:
Reviews

Transcytosis
(Rab11+)33,37. In addition to the recycling endosome which suggests that Ser256 phosphorylation alone is not
The transport of pathway, AQP2 can traffic from the basolateral to the the rate-limiting step of AQP2 plasma membrane target-
macromolecular cargo from apical plasma membrane via microtubule-dependent ing in vivo during V2R stimulation43. By contrast, AQP2
one side of a cell to the other transcytosis38,39. Below, we provide a brief overview of phosphorylation at Ser269 markedly increases in vivo in
within a membrane-bounded
new findings in the field of AQP2 regulation, includ- the apical plasma membrane42 during V2R stimulation43.
carrier (or carriers); this
mechanism is used by ing novel insights into the role of post-translational However, although this site seems to be involved in
multicellular organisms to modifications and cytoskeletal remodelling. AQP2 accumulation in the plasma membrane (see
selectively move material below), studies of cell lines with mutations in this resi-
between two environments
AQP2 phosphorylation and ubiquitylation due suggest that it is not required for cAMP-dependent44
without altering their unique
compositions.
AQP2 exists functionally as a tetramer (Fig. 2a). Each or rapid V2R-mediated targeting of AQP2 to the plasma
monomer consists of six membrane spanning α-helices membrane45. Furthermore, the precise role of Ser269 in
Multivesicular bodies and two of the connecting loops contain Asn–Pro–Ala urine concentration and water homeostasis has not been
(MVBs). Specialized (NPA) motifs that interact in the membrane to form the addressed.
endosomes that contain
membrane-bound intraluminal
pathway for translocation of water (Fig. 2b,c). Stimulation Emerging evidence indicates that phosphoryl-
vesicles formed by budding of with vasopressin triggers a variety of phosphorylation ation of AQP2 facilitates its protein–protein bind-
the membrane into the lumen events in the collecting duct40,41, including changes in ing properties and consequently affects its cellular
of the MVB; MVBs can fuse the phosphorylation of AQP2 (ref.42) at the carboxyl distribution46,47. For example, AQP2 phosphorylation
with lysosomes, where their
terminus. In mammals, AQP2 can be phosphorylated at Ser256 reduces its binding to the lysosomal traffick-
contents are degraded, or they
can fuse with the plasma
at four sites — Ser256, Ser261, Ser264 and Ser269 (in ing regulator LYST-interacting protein 5 (LIP5; also
membrane for their contents to humans, Ser269 is conserved as Thr269) (Fig. 2d). Ser256 known as vacuolar protein sorting-associated protein
be released into the and Ser269 seem to be the most relevant to AQP2 tar- VTA1 homologue) and is predicted to reduce the sort-
extracellular space. geting and accumulation in the plasma membrane. The ing of AQP2 into multivesicular bodies (MVBs), decrease
Ser256 site is essential for vasopressin-induced accu- its lysosomal degradation and increase AQP2 protein
mulation of AQP2 in the plasma membrane in the cell half-life44,48. Ser269 is contained within the class I PDZ
models studied thus far. However, AQP2 seems to be motif of AQP2 (Fig. 2d) and might enhance its interaction
constitutively phosphorylated at this site in vivo, with with PDZ domain-containing proteins such as sorting
only minor increases following vasopressin stimulation, nexin-27, which directs AQP2 away from lysosomes;

a H2O b
AQP2 N P A
monomer
Extracellular

1 2 3 4 5 6

Intracellular
N P A
N terminus C terminus
H2O

c d
H2O

221 F
P
1 P
2

A K S L A
L S E R V
N N L
3 K
256

P P
4 A
5 RV E R E G
A V S Q R E L
E R W E
D T
L D P
6

H
S PDZ motif
6 1 P
2 G S K A Ubiquitylation
Q S
L PR Phosphorylation
269
270
264

Fig. 2 | AQp2 topology and structure. a | Aquaporin 2 (AQP2) exists as a tetramer in the plasma membrane and each
monomer contains a single pore through which water molecules pass in a single file. b | Simple topography of an
AQP2 monomer. A single monomer of an AQP2 channel consists of six membrane-spanning α-helices and short intracellular
amino and carboxy termini. Almost all AQPs have two highly conserved Asn-Pro-Ala (NPA) motifs in each monomer.
c | The NPA sequences of each monomer have been proposed to interact in the plasma membrane to form a pathway for
translocation of water across the plasma membrane. d | Carboxy-terminal tail of AQP2, highlighting the phosphorylation
sites and ubiquitylation sites that are most relevant to AQP2 trafficking. Adapted with permission from ref.152, American
Physiological Society.

Nature Reviews | Nephrology

0123456789();:
Reviews

Urine

H2O H2O H2O H2O Apical plasma H2O


AQP2 membrane

Ub P P P P P H2O P P

CHIP–NEDD4
Ubiquitylation Stabilization SIPA1-like
protein 1
P P
Endocytosis Actin
depolymerization
Ub Recycling

Ub Ub USP4 P

Ub P P
Degradation Proteasome
P
Actin
filaments
Golgi
P
cAMP–PKA
independent P
Degradation
Lysosome
Native
Misfolded AQP2 P P
AQP2 RhoA
PKA and/or
CHIP

Ub ER other kinases

AQP2
cAMP

Basolateral Ca 2+

plasma H2O H2O


AQP3 Cytosol
membrane

AQP4 V2R
Blood H2O H2O Vasopressin

Stimulation P Ser256 phosphorylation Ub Ubiquitin


Inhibition P Ser269 phosphorylation

Fig. 3 | Main mechanisms underlying the regulation of AQp2 trafficking. The figure illustrates some of the major
pathways of aquaporin 2 (AQP2) regulation, including simplified depictions of the roles of phosphorylation and
ubiquitylation. Natively folded AQP2 traffics to the Golgi apparatus, is organized into tetramers and stored in transport
vesicles. In the classical pathway, vasopressin stimulation of the vasopressin V2 receptor (V2R) causes an increase in the
second messenger cAMP, which activates protein kinase A (PKA), and leads to phosphorylation of AQP2 at Ser256 and
redistribution of AQP2-containing vesicles to the apical plasma membrane. However, activation of this pathway is not an
absolute requirement for AQP2 trafficking and cAMP–PKA-independent mechanisms exist, including the phosphorylation
of AQP2 by other protein kinases, such as AMP-activated protein kinase (AMPK), and Rho-dependent remodelling of the
actin cytoskeleton. In the apical plasma membrane, phosphorylation at Ser269 stabilizes AQP2 and limits its interaction
with the signal-induced proliferation-associated 1-like (SIPA1-like) protein 1, which reduces SIPA1-like protein 1-mediated
AQP2 endocytosis. In the plasma membrane, AQP2 can be ubiquitylated by the E3 ubiquitin ligases CHIP and NEDD4,
which promotes its endocytosis. Following AQP2 internalization, activity of the deubiquitylase ubiquitin carboxyl-terminal
hydrolase 4 (USP4) determines whether AQP2 is deubiquitylated and recycled to the plasma membrane, or targeted for
lysosomal degradation. ER, endoplasmic reticulum. Adapted with permission from ref.152, American Physiological Society.

however, the role of Ser269 in this interaction is ubiquitin-ligating enzymes (E3 ligases), including
unclear49. Signal-induced proliferation-associated 1-like NEDD4 and NEDD4-like (also known as NEDD4-1 and
protein 1 (SIPA1-like protein 1) also includes a PDZ NEDD4-2, respectively)51–53, and the carboxyl terminus
domain45 and its interaction with AQP2 is inhibited by of heat shock protein 70 (HSP70)-interacting protein
K63-linked polyubiquitin
Ser269 phosphorylation, which reduces SIPA1-like pro- CHIP54–56 (encoded by Stub1), plus the deubiquitylase
chain
A chain of ubiquitin molecules tein 1-mediated AQP2 endocytosis and might explain ubiquitin carboxyl-terminal hydrolase 4 (USP4)57, have
that are linked to each other on why Ser269 phosphorylation prolongs AQP2 retention been implicated in ubiquitin-dependent AQP2 degrada-
Lys63 (one of seven lysine in the plasma membrane (Fig. 3). tion (Fig. 3). In cultured mpkCCD14 cells treated with for-
residues within ubiquitin) to Ubiquitylation of AQP2 at Lys270 (Fig. 2d) with one skolin (an adenylate cyclase activator) to enhance AQP2
form a long polyubiquitin
K63-linked polyubiquitin chain enhances AQP2 endocy- membrane accumulation, removal of forskolin increased
chain; these chains can mark
the ubiquitylated protein for tosis to MVBs, after which it is either targeted to lyso- AQP2 ubiquitylation before AQP2 endocytosis50. A sim-
endocytosis. somes for degradation or excreted in exosomes50. Several ilar increase in AQP2 ubiquitylation was observed in

www.nature.com/nrneph

0123456789();:
Reviews

mpkCCD14 cells stimulated with d-amino d-arginine urine-concentrating defect. MnCl 2-induced intra-
vasopressin (dDAVP; a synthetic vasopressin ana- cellular accumulation of AQP2 was associated with
logue that acts mainly on the V2R) followed by dDAVP decreased phosphorylation (and thus increased acti-
removal58, but not in another study, where AQP2 ubiq- vation) of RhoA and increased F-actin polymerization.
uitylation levels (and AQP2 membrane accumulation) Vasopressin-induced phosphorylation of AQP2 in vitro
were highest in the presence of dDAVP59. Taken together, at Ser256, Ser264 and Ser269 was also significantly
although these studies suggest that deubiquitylation of decreased by MnCl2, but whether this effect was due to
AQP2 can reduce its endocytosis and degradation dur- effects of MnCl2 on V2R-mediated signalling pathways,
ing dDAVP stimulation, whether ubiquitylation has a role or the altered cellular distribution of AQP2 (for example,
in AQP2 internalization processes after dDAVP removal reduced Ser269 phosphorylation owing to reduced levels
is unclear. Furthermore, whether increased AQP2 ubiq- of AQP2 in the plasma membrane) remains unclear42.
uitylation is a process to remove AQP2 from the plasma The internalization of AQP2 from the plasma mem-
membrane in vivo when vasopressin levels are no longer brane involves ezrin68, which is a member of the ezrin,
raised (plasma osmolality is normal) is unclear. radixin and moesin (ERM) family of proteins that
Of note, AQP2 phosphorylation and ubiquityla- interact with both the plasma membrane and F-actin69.
tion seem to be interconnected. Phosphorylation at Ezrin is also a member of the A-kinase anchoring pro-
Ser256 and/or Ser269 seemingly counterbalances the tein (AKAP) family of proteins70. AKAPs are structurally
endocytosis-promoting effects of polyubiquitylation, diverse scaffolding proteins that tether the regulatory
whereas low Ser261 phosphorylation is associated with subunit of PKA (and other components such as phos-
low polyubiquitylation and high cellular and membrane phodiesterases) to specific cellular compartments, and
abundance of AQP2 (refs58,60). Our understanding of the thereby spatially direct cAMP signalling71 (discussed
physiological roles of V2R-mediated post-translational below). In vitro and in vivo, ezrin colocalized with AQP2
modifications of AQP2 would benefit from in vivo val- during vasopressin stimulation, and ezrin knockdown in
idation experiments using CRISPR–Cas9 approaches, cells reduced endocytosis and increased apical plasma
to eliminate one or more of the phosphorylation or membrane accumulation of AQP2 in the absence of
ubiquitylation sites. AVP stimulation, whereas no changes were observed in
cAMP or Ser256 phosphorylation levels, or in the degree
The cytoskeleton and plasma membrane targeting of AQP2 exocytosis68. The physiological role of ezrin is
Principal cells have a dense cortical actin network that unclear, as increased ezrin at the plasma membrane
limits docking of AQP2-containing vesicles; however, during vasopressin stimulation would be expected to
vasopressin stimulation causes actin depolymeriza- enhance AQP2 endocytosis. One possible explanation
tion in both the apical cell cortex and the basal stress for these seemingly paradoxical observations is that dur-
fibres61. Under baseline conditions, actin polymeriza- ing vasopressin stimulation, despite the close association
tion is maintained by the small GTPase RhoA, but this between ezrin and AQP2 at the apical plasma mem-
effect is inhibited by V2R stimulation62–64 (Fig. 3). AVPR2 brane, their interaction is limited by their phosphoryla-
mutations that render V2R constitutively active (R137L tion status. However, upon vasopressin withdrawal, their
and R137C) and increase the osmotic water permea- concomitant phosphorylation status might promote
bility of AQP2-expressing cells65 have been identified their interaction and subsequent endocytosis.
in humans66. Their effects on water permeability are Collectively, these findings suggest that cytoskeletal
PKA-independent but Rho-associated protein kinase remodelling is a key component of AQP2 targeting to the
(ROCK)-dependent, and are associated with increased plasma membrane, but several important unanswered
phosphorylation of AQP2 at Ser269 compared with questions remain. For example, whether actin depolym-
non-mutated (wild-type) V2R. Although this obser- erization is a rate-limiting step in V2R-mediated AQP2
vation suggests that Ser269 phosphorylation might be regulation, and whether cytoskeleton-mediated modu-
required for Rho-mediated and ROCK-mediated AQP2 lation of AQP2 trafficking requires Ser269 phosphoryl-
plasma membrane insertion, increased ROCK activity ation are unclear. Notably, Ser269 phosphorylation does
was not important for V2R-mediated Ser269 phosphory­ not seem to be required for V2R-mediated AQP2 tar-
lation or vasopressin-stimulated water permeability in geting to the plasma membrane. If neither cytoskeletal
wild-type cells. These observations suggest that altered remodelling nor AQP2 phosphorylation is rate-limiting
ROCK activity might only be important for AQP2 for AQP2 plasma membrane targeting, then alternative
plasma membrane targeting in pathological conditions. rate-limiting components must exist.
The trace metal manganese (Mn) can enhance the
polymerization of G-actin to form F-actin by binding Role of cAMP and PKA in plasma membrane
directly to a metal-binding site on G-actin. In vitro, targeting
exposure of LLC-PK1 cells that stably expressed AQP2 The canonical signalling pathway downstream of GSPCR
or rat IMCD cells that expressed AQP2 to MnCl2 inhib- stimulation involves the activation of adenylyl cyclases,
ited AQP2 targeting to the plasma membrane and which leads to a rise in cAMP levels and enhanced
F-actin promoted its internalization, which resulted in intra- activity of basophilic protein kinases, including PKA.
A filamentous polymer cellular accumulation67. Accordingly, MnCl2 admin- For many years, this response was considered the only
composed of soluble G-actin
monomers; it is the most
istration to mice reduced the accumulation of AQP2 mechanism underlying the effects of V2R stimulation on
abundant component of the in the plasma membrane of principal cells relative to AQP2 plasma membrane targeting72–74 and is often the
cytoskeleton of eukaryotes. vehicle-treated controls and led to a vasopressin-resistant easiest and simplest mechanism for explaining general

Nature Reviews | Nephrology

0123456789();:
Reviews

AQP2 trafficking. However, new findings have high- basophilic protein kinases. A similar increase in putat­
lighted that this model cannot explain all the molecular ive basophilic protein kinase sites in PKA-intact cells sug-
effects of vasopressin and several pathways independent gests that the V2R signals through a range of pathways,
of cAMP and/or PKA must exist, as summarized below. several of which might be involved in AQP2 plasma
membrane targeting. Of particular interest, three of the
PKA. The Ser256 site of AQP2 is located within sites in which phosphorylation increases after exposure
a PKA consensus sequence. Early studies show- to dDAVP in PKA-dKO cells are known AMPK targets;
ing that N-[2-p-bromocinnamylamino-ethyl]- other members of the AMPK-related subfamily, such as
5-isoquinolinesulphonamide (H89), which was initially serine/threonine-protein kinases Mark2 and Mark3, and
thought to be a PKA-specific inhibitor, limited AQP2 NUAK family SNF1-like kinase 2, have been proposed
membrane accumulation, suggested that PKA-mediated to phosphorylate AQP2 at Ser256 (ref.80).
phosphorylation of AQP2 at Ser256 was a key event in AMPK is an energy-sensitive kinase that is stimulated
AQP2 trafficking75. However, subsequent studies showed by high AMP and/or low ATP levels and is important
that H89 can affect the activity of protein kinases other for hypertonicity-induced increases in IMCD water
than PKA, including AMP-activated protein kinase permeability81. The AMPK s­ti­mu­la­tor 5­-a­mi­no­im­id­az­ol­e-­
(AMPK) and ROCK, and therefore responses to H89 4-­c a­r b­o xamide-1-β-d-ribofuranoside (AICAR)
should not be regarded as sufficient evidence of PKA increased the osmotic water permeability of rat IMCDs
involvement in AQP2 trafficking76. In mice expressing within 30 min compared with vehicle controls, which
dominant-negative PKA77, AQP2 abundance, Ser256 corresponded with increased phosphorylation of AQP2
phosphorylation and apical plasma membrane target- at Ser256 (ref.81). However, another study found that
ing were reduced and renal water handling was defec- AICAR inhibited dDAVP-mediated Ser256 phospho-
tive compared with wild-type controls. However, these rylation but not plasma membrane targeting of AQP2
effects could be partially rescued by dDAVP, suggesting in mpkCCD14 cells82. The difference in these findings
either a residual effect of the endogenous PKA, or the might reflect different responses to AMPK activation in
existence of a V2R-mediated but PKA-independent different cell types (that is, in cortical versus IMCD cells)
mechanism for modulation of AQP2 and urine concen- or it might be explained by time-dependent effects of
tration. By contrast, experiments that used mpkCCD14 AMPK activation (short-term activation increases AQP2
cells with a complete CRISPR–Cas9-mediated double membrane abundance and water permeability81, whereas
knockout (dKO) of the genes encoding the PKA catalytic prolonged activation has the opposite effect82). The anti-
subunits PKA-Cα and PKA-Cβ (PKA-dKO)78 suggested diabetic drug metformin, which activates AMPK by
that the dDAVP-mediated increase in AQP2 protein increasing the cellular AMP to ATP ratio, also increased
expression requires PKA activity. In these cells, AQP2 AQP2 phosphorylation and plasma membrane targeting
mRNA and protein were not detectable in whole-cell in isolated rat IMCDs (Fig. 4) and acutely increased urine
lysates following dDAVP stimulation, whereas their osmolality in AVP2R-knockout mice compared with
levels increased in control cells. Subsequent transfec- wild-type mice, and in rats treated with the V2R antag-
tion of these knockout cells with AQP2 showed that the onist tolvaptan compared with untreated controls83,84.
PKA-dKO did not significantly affect baseline Ser256 However, metformin did not increase urine concentra-
and Ser269 phosphorylation, although dDAVP-induced tion in water-loaded healthy human volunteers85 and
AQP2 plasma membrane targeting and phosphorylation hyponatraemia and oedema are not listed as common
at Ser269 seemed to be reduced compared with control side effects of metformin treatment according to the
cells using immunocytochemistry. Importantly, the sim- FDA. Overall, these observations suggest that metformin
ilar dDAVP-induced increase in AQP2 phosphorylation modulates water reabsorption predominantly in a setting
at Ser256 in PKA-dKO and wild-type cells transfected of inappropriate water diuresis (NDI models), perhaps
with AQP2 (ref.79) indicates that other protein kinases reflecting altered intracellular signalling mechanisms
can be active at this site. Of note, findings on the role under such conditions.
of PKA in cells with PKA defects should be interpreted
with caution because they might reflect the potential cAMP. levels and this process is generally believed to
role of PKA in the maintenance of a wide range of cel- be the signalling pathway that leads to AQP2 phos-
lular biological processes such as vesicular trafficking, phorylation and/or plasma membrane targeting. This
microtubule organization and actin cytoskeleton (de) hypothesis emerged mainly from observations that
polymerization, rather than a direct involvement of PKA cAMP or forskolin stimulation leads to AQP2 plasma
in dDAVP-induced AQP2 phosphorylation and plasma membrane accumulation and increases water per-
membrane targeting. meability in the collecting duct (reviewed in ref.86).
Stable-isotope labelling with amino acids in cell Importantly, studies have established that although
culture followed by quantitative phosphoproteom- cAMP is a diffusible molecule, it does not diffuse
ics demonstrated an approximately sixfold reduction freely within the cell but is instead buffered within
in the number of proteins with significant alterations in the cytosol owing to fluid-phase droplets formed by the
phosphorylation status following dDAVP stimulation cAMP-dependent protein kinase type Iα regulatory
in PKa-dKO cells compared with PKA-intact cells79. subunit; the formation of these droplets is enhanced
However, dDAVP-dependent but PKA-independent by cAMP87. However, whereas enhanced droplet for-
changes in protein phosphorylation were still observed mation is sustained during >20 min of forskolin stim-
in PKA-dKO cells at sites predicted to be targeted by ulation, it is transient (<10 min) after stimulation of

www.nature.com/nrneph

0123456789();:
Reviews

Urine

Apical plasma H2O H2O H2O H2O H2O H2O


AQP2
membrane

P P P P P P P P P P P P
Cytosol

Translocation Translocation

Actin
depolymerization
P P

P P
FMP-API-1 P P
P
A KA
Src Bosutinib Actin
filaments
A
PK
Actin P
polymerization
P P
Calcineurin
RhoA
CAM

Ca2+ AMPK
Basolateral
plasma EGFR
membrane

Blood AG-490
V2R FZD Fluconazole
Wnt5a
Metformin Statin
Erlotinib

Stimulation P Ser256 phosphorylation Drug


Inhibition P Ser269 phosphorylation

Fig. 4 | Alternative regulators of AQp2 trafficking. The figure illustrates regulators of aquaporin 2 (AQP2) plasma
membrane targeting that are independent of the vasopressin V2 receptor (V2R; encoded by Avpr2) and/or cAMP–protein
kinase A (PKA)-independent. The A-kinase anchoring protein (AKAP)–PKA inhibitor FMP-API-1 increases PKA activity and
plasma membrane targeting of AQP2, and increases urine osmolality in mice with tolvaptan-induced nephrogenic diabetes
insipidus (NDI)98. The tyrosine-protein kinase Src inhibitor bosutinib increases Ser269 phosphorylation and plasma
membrane targeting of AQP2 independently of Ser256 phosphorylation103. The epidermal growth factor receptor (EGFR)
inhibitor erlotinib increases AQP2 plasma membrane targeting and urine concentration in mice with lithium-induced
NDI110. The EGFR–protein kinase JAK2 inhibitor A­G-­490 increases V2R-dependent and V­­­­­­­­­­­­­­­­­­­­­­­­­­­­­­­­­­­­­­­­­­­­­­­­­­­­­­­­­­­­­­­­­­­­­­­­­­­­­­­­­­­­­­­­­­­­­­­­­­­­­­­­­­­­­­­­­­­­­­­­­­­­­­­­­­­­­­­­­­­2­­­­­­­­­­­­­­­­­­­­­­­­­­­­­­­­­­­­­­­­­­­­­­­­­­­­­­­­­­­­­­­­­­­­­R­­­­­­­­­­­­­­­­­­­­­­­­­­­­­­­­­­­­­­­­­­­­­­­­­­­­­­­­­­­­­­­­­­­­­­­­­­­­­­­­­­­­­­­­­­-­­­­­­­­­­­­­­­­­­­i­­­­­­­­­­­­­­­­­­­­­n­­­­­­­d­­­­­ep­­endent t­­­­­­­­­­­­­­­­­­­­­­­­­­a­­­­­­­­­­­­­r­­­­­­­­­­­­­­­­­g­­­e­­­­ting o­­­­f
A­­­­Q­­P­2 t­­­­o t­­h­e p­­l­a­­sma membrane in rats108. Wnt5a acts via Frizzled receptors (FZDs) to increase intracellular Ca2+ and induces
calmodulin (CAM) and calcineurin-dependent AQP2 plasma membrane targeting, as well as urine concentration in mice
with tolvaptan-induced NDI105. Metformin increases Ser256 phosphorylation via AMP-activated protein kinase (AMPK) and
plasma membrane targeting of AQP2 and increases urine concentration in rats with tolvaptan-induced NDI and in
Avpr2-knockout mice83,84. Statins107 and fluconazole60 inhibit RhoA activity and actin polymerization, which enables AQP2
plasma membrane targeting. The drugs depicted in the figure have shown promise in the treatment of autosomal dominant
polycystic kidney disease in animal studies or in clinical trials.

the β2 adrenergic receptor, a GSPCR coupled to ade- be important for V2R-mediated AQP2 plasma mem-
nylyl cyclase87. These observations highlight that the brane targeting because in resting conditions (AQP2
effects of administering either cell-permeable cAMP present in intracellular vesicles), localized effects of
or the general adenylyl cyclase activator forskolin do the cAMP-specific 3′,5′-cyclic phosphodiesterase 4D
not completely mimic the effect of GSPCR signalling, might prevent inappropriate translocation of AQP2 to
as there are differences in constraining the diffusion the plasma membrane as a result of small changes in
of cAMP throughout the cell when using the vari- intracellular cAMP levels89. The cellular localization
ous approaches. The low cytosolic concentration of and specificity of cAMP–PKA signalling are further
cAMP achieved by this ‘buffering’ of cAMP within ensured by the actions of various scaffolding proteins,
droplets allows for phosphodiesterase-mediated including AKAPs90,91, which are also associated with
compartmentalization 87,88. This mechanism might AQP2-bearing vesicles (see below).

Nature Reviews | Nephrology

0123456789();:
Reviews

Conceptual advances in GPCR signalling, such receptors, activation of these receptors can have dif-
as the discovery of β-arrestin and various alternative ferent and sometimes opposing roles in physiology
pathways that can relay information to specific targets and, occasionally, within the same cell71,94,95. Several
within the cell (reviewed in ref.92) have brought the investigations have been aimed at uncovering how
widely accepted view of the role of cAMP in AQP2 phos- compartmentalization of cAMP is achieved, includ-
phorylation and trafficking into question (reviewed in ing the role of more than 30 canonical AKAPs87,88,91.
ref.86). One example of how AQP2 plasma membrane The modulation of specific AKAPs to target selected
targeting can be achieved in the absence of cAMP PKA-regulated domains is an area of interest in the field
was demonstrated using mice with global AC6 defi- of precision pharmacology91. In the kidney, AKAP18δ
ciency in which cAMP does not increase in response is mainly expressed in IMCDs and traffics to the plasma
to dDAVP stimulation18. Compared with control mice, membrane along with AQP2 (ref.96), suggesting that it
AC6-deficient mice have impaired AQP2 phosphoryla- anchors PKA to AQP2-containing vesicles and controls
tion at Ser256 and Ser269 and reduced apical membrane cAMP-mediated AQP2 phosphorylation at Ser256 and
AQP2 accumulation in IMCD cells, which results in a plasma membrane targeting. In line with this hypoth-
urine-concentrating defect. However, although urine esis, the general AKAP inhibitor Ht31 prevented
osmolality remained lower than that observed in con- forskolin-mediated AQP2 plasma membrane target-
trol animals after water restriction, the fold change in ing in primary cultured rat IMCD cells97, but it did not
urine osmolality in AC6-deficient mice in response to inhibit PKA activity in an in vitro assay, suggesting that
water restriction was greater (~2.5-fold) than in con- the catalytic activity of PKA is not sufficient for AQP2
trols (~1.5-fold). Urine osmolality increased ~10-fold in translocation. By contrast, another study showed that
response to dDAVP in knockout mice, almost match- Ht31 led to AQP2 plasma membrane accumulation in
ing the ~12-fold increase observed in wild-type con- mpkCCD14 cells independently of V2R activation98. The
trols. Furthermore, both dDAVP and water restriction discrepancies between the two studies might be due to
induced Ser256 phosphorylation in the inner medulla. cell type-specific effects and further highlight that the
AC6-deficient mice were able to concentrate their urine role of PKA in AQP2 plasma membrane targeting is not
to >2,000 mOsm/kg, which cannot be explained by fully understood. Exposure to another AKAP inhibitor,
residual AC6 activity in the cortex and must therefore FMP-API-1 (ref.99) (a low-molecular-weight compound
occur through cAMP-independent AQP2 regulation in with enhanced plasma membrane permeability), also
the IMCD18. Overall, this study highlights that AC6 and increased apical AQP2 expression in mpkCCD14 cells
cAMP are important for AQP2 phosphory­lation to similar levels to vasopressin alone98. Furthermore,
and membrane abundance at baseline but are not FMP-API-1 or its derivative FMP-API-1/27 potently
required for acute regulation of AQP2, including apical increased urine osmolality and reduced urine output in
plasma membrane targeting and Ser256 phosphoryla- tolvaptan-treated mice, highlighting them as a potential
tion. This conclusion is supported by the observation novel treatment approach for NDI.
that stimulation of the V2R, or the prostaglandin recep- The effects of the inhibitors of the AKAP–PKA
tors EP2 or EP4, during pharmacological inhibition interaction, Ht31 or FMP-API-1, on the biological
of cAMP generation, enhances AQP2 targeting to the activity of PKA are complex. On the one hand, Ht31
plasma membrane93. decreases certain cAMP–PKA-dependent events, such as
In summary, cAMP and PKA are not an absolute forskolin-induced phosphorylation of cAMP-responsive
requirement for Ser256 phosphorylation or AQP2 element-binding proteins (CREBs)100 and PKA-induced
plasma membrane targeting. Although several alternative inhibition of Rho activity101. Therefore, in some cases,
kinases have been identified, their roles and the pathways Ht31 inhibits local PKA activity, presumably by dis-
that lead to their activation are unclear. Furthermore, placing PKA from its anchored sites and substrates,
whether cAMP or PKA activity is necessary for actin regardless of the intracellular concentration of active
depolymerization is unknown. PKA. Similarly, although FMP-API-1 increased PKA
activity in cardiac myocytes, as assessed by a PKA assay
Novel regulators of AQP2 membrane targeting and increased phosphorylation of selected targets, it did
New insights into the complex signalling networks and not induce phosphorylation of PKA substrates globally99.
regulatory steps that govern AQP2 trafficking have By contrast, Ht31 and FMP-API-1 increased the gen-
identified promising pharmacological targets for the eral phosphorylation of PKA substrates in mouse corti-
treatment of water balance disorders (reviewed in ref.5). cal collecting duct cells98, but despite this global effect,
Several of these novel compounds increase AQP2 plasma Ht31 prevented AQP2 plasma membrane targeting in
membrane targeting independently of known V2R sig- response to forskolin stimulation97. This observation
nalling pathways such as cAMP–PKA and extracellular suggests that PKA targets of relevance to cAMP-induced
signal-regulated kinase (ERK; also known as MAPK), and AQP2 plasma membrane targeting are inhibited by
not only represent potential therapeutics but might also Ht31. Collectively, although these studies provide evi-
help to determine the role of AQP2 in ADPKD. Some of dence for V2R-independent AQP2 plasma membrane
the newest approaches are detailed below (Fig. 4). targeting and a tool for studying PKA involvement, they
do not directly support a role for PKA in AQP2 plasma
Inhibitors of AKAP-mediated events. Although the membrane targeting in general.
intracellular levels of cAMP are increased by stim- Another AKAP shown to have an indirect role in
ulation of a variety of different hormone-coupled AQP2 plasma membrane targeting through cytoskeletal

www.nature.com/nrneph

0123456789();:
Reviews

remodelling is AKAP13 (also known as lymphoid blast Treatment of rat primary IMCD cultures with 10 μM
crisis oncogene)102. This AKAP has guanine nucleo­ fluconazole prevented forskolin-mediated AQP2 plasma
tide exchange factor (GEF) activity and activates RhoA. membrane targeting106. However, in another study,
Thus, AKAP13 inhibitors might increase AQP2 plasma 50 μM of fluconazole increased AQP2 plasma mem-
membrane targeting through inhibition of RhoA and brane accumulation in IMCD cultures to levels similar
consequent actin depolymerization. A small-molecule to those observed using forskolin or vasopressin, and
screening approach identified Scaff10-8 as a compound increased collecting duct water permeability60. The
that, by binding to RhoA and inhibiting AKAP13- reason for the contrasting effects of fluconazole at the
mediated RhoA activation, promotes actin depolym- two concentrations is unknown. The effects of 50 μM
erization102. However, although treatment of primary fluconazole were not accompanied by increases in PKA
IMCD cultures with Scaff10-8 resulted in redistribution activity or AQP2 phosphorylation at Ser256 or Ser269,
of AQP2 towards the plasma membrane, AQP2 was which suggests that they are independent of canonical
not inserted into the plasma membrane. These studies signalling pathways (cAMP–PKA)60. Compared with
suggest that, although actin depolymerization facilitates vehicle-treated controls, treatment of mice with flucona-
AQP2-containing vesicular transport to the plasma zole for 96 h (at a dose that resulted in an extracellular
membrane, this process alone is not sufficient to increase fluconazole concentration similar to that observed in
collecting duct water transport, and that other regulatory patients treated for fungal infections) decreased urine
processes, such as AQP2 phosphorylation, are required. output and increased urine osmolality60. Fluconazole
treatment of mice for 96 h also ameliorated the polyuric
Src inhibition. Phosphorylation of the non-receptor effect of tolvaptan60. These effects of fluconazole were
tyrosine kinase Src was enhanced in IMCD cells follow- associated with decreased RhoA activity and depoly­
ing dDAVP stimulation of the V2R40. Src inhibition using merization of F-actin, but this can only partially explain
dasatinib or bosutinib increased AQP2 plasma mem- the observed functional effects because RhoA inhibition
brane targeting in AQP2–LLC-PK1 cells and in fresh alone does not have similar effects102.
rat kidney tissue independently of changes in cAMP Fluconazole functions as an antimycotic agent by
levels or PKA activity103. Dasatinib increased AQP2 blocking the fungal lanosterol 14a-demethylase (LDM;
Ser269 phosphorylation in AQP2–LLC-PK1 cells inde- encoded by CYP51A1) and inhibiting ergosterol produc-
pendently of phosphorylation at Ser256, and resulted in tion, which causes membrane defects. Native mamma-
increased AQP2 exocytosis and decreased endocytosis lian LDM, which has an important role in the last steps of
compared with vehicle-treated AQP2–LLC-PK1 cells. cholesterol synthesis, is also inhibited by fluconazole.
This increased AQP2 trafficking with dasatinib was not Of note, statins, which are cholesterol-lowering drugs
apparent in cells expressing an AQP2-S269A mutant, that act at an early step of cholesterol synthesis (conver-
which indicates that Ser269 phosphorylation has an sion of 3-hydroxy-3-methylglutaryl-CoA to mevalonic
important role in this effect. Future work should inves- acid), also increase AQP2 plasma membrane targeting
tigate the potential role of RhoA and/or ROCK in this and can ameliorate NDI107. Similar to statins, fluconazole
process. These results might also explain why hypon- might interfere with mevalonate conversion and there-
atraemia has been reported in patients treated with Src fore interrupt the formation of isoprenoids, which are
inhibitors104. important for the membrane tethering and activation of
small GTPases such as Rho. Hence, fluconazole might
Wnt5a. In mpkCCD14 cells, the endogenous frizzled alter AQP2 function by altering both RhoA activity and
receptor ligand Wnt5a increased calcium signalling and its subcellular distribution.
AQP2 protein expression, plasma membrane target-
ing and Ser269 phosphorylation compared with vehi- EGF receptor antagonists. AG-490 is an epidermal
cle controls105. Unlike dDAVP, Wnt5a did not increase growth factor receptor (EGFR) antagonist and a tyros-
cAMP or PKA activity, and the increase in AQP2 phos- ine protein kinase JAK2 inhibitor that was identified in
phorylation induced by Wnt5a (but not dDAVP) could an LLC-PK1 cell-based screening assay as an enhancer
be prevented by the calcineurin inhibitor cyclosporine A. of AQP2 plasma membrane targeting108. These results
Furthermore, Wnt5a-induced AQP2 protein expression were verified in AQP2-transfected Madin–Darby canine
was not inhibited by H89, indicating that the underlying kidney cells (MDCK–AQP2; Table 1) and in kidney tis-
mechanism is not dependent on H89-sensitive kinases. sue slices from Sprague–Dawley rats cultured in vitro.
In mice with tolvaptan-induced polyuria, Wnt5a treat- In vasopressin-deficient Brattleboro rats (Table  1) ,
ment for 2 h also caused a minor increase in urine osmo- AG-490 increased AQP2 plasma membrane targeting
lality compared with vehicle-treated mice. Collectively, in cortical but not medullary collecting ducts108. This
these results indicate that the signalling pathway acti- observation suggests that in the IMCD, EGFR might
vated by Wnt5a differs from that downstream of V2R inhibit V2R-induced AQP2 plasma membrane tar-
stimulation. geting, which is in accordance with the observation
that EGF enhances diuresis109. However, in the corti-
Fluconazole. Triazolpropenon, which was identified as cal collecting duct, AG-490 might have an additional
a modulator of AQP2 plasma membrane targeting in vasopressin-independent effect on AQP2 plasma mem-
screening assays that used kidney collecting duct cells brane targeting. The involvement of EGFR in AQP2
transfected with AQP2 (MCD4)106 (Table 1), is struc- plasma membrane targeting was further supported by
turally similar to the antimycotic agent fluconazole. experiments showing that the FDA-approved EGFR

Nature Reviews | Nephrology

0123456789();:
Reviews

CFTR corrector
antagonist erlotinib increased plasma membrane target- this simplified mechanism and is likely to involve a range
A family of compounds that ing of AQP2 in LLC-PK1 cells110 and in mice with lithium- of mechanisms, including changes in cellular prolifera-
facilitate the transport of induced NDI; these mice also had increased urine tion and transformation of the epithelia from a reabsorp-
misfolded CFTR proteins osmolality and reduced urine volume after erlotinib tive to a secretory phenotype123. A detailed description
(produced owing to mutations
treatment110. of the intricate pathophysiology and cellular mechanism
in CFTR) to the cell membrane
without being degraded. underlying ADPKD is beyond the scope of this article
AQP2 regulation and ADPKD therapy (but is available elsewhere124,125).
The prevalence of ADPKD is 1 in 500–1,000 live births
and at least 45% of patients progress to kidney failure111,112. Cell proliferation and epithelial transport
The competitive V2R antagonist tolvaptan delayed the Targeting a broad range of proteins, including the YAP
onset of kidney failure in clinical trials7–9,113 and is now transcriptional coactivators, MYC, RhoA–ROCK and
approved by, among others, the FDA and the EMA for the mechanistic target of rapamycin (mTOR), to con-
this purpose. However, polyuria is a major side effect of trol cellular proliferation in ADPKD has shown promise
tolvaptan and leads to treatment discontinuation in ~8% in animal studies126–128. Of note, PC1 has been shown
of patients7; moreover, treatment with tolvaptan requires to modulate RhoA activation and ROCK signalling129.
an intake of >2–3 l of water per day113. Importantly, However, randomized controlled clinical trials using
in the tolvaptan initiation phase, the drug should be mTOR inhibitors in patients with ADPKD have been
titrated to sustain a urine osmolality of <300 mOsm/kg generally disappointing130,131. The discrepancies between
(normal range 400–900 mOsm/kg) and ensure suffi- the effects observed in mouse models and in humans
cient inhibition of V2R signalling113. Consequently, tol- might be related to drug dosage, because the doses tested
vaptan use might become one of the leading causes of in animals were 35–70-fold higher than those that were
NDI in the future and novel discoveries in the field administered clinically (owing to potential side effects).
of AQP2 research might have direct clinical benefits in Moreover, mTOR inhibitors were primarily tested in
patients with ADPKD receiving tolvaptan treatment. rodent models of early-onset or rapidly progressive dis-
In this section, we provide a brief and focused overview of ease at early stages of cyst development, which might not
ADPKD pathophysiology and discuss treatment options reflect the complex and heterogeneous pathogenesis of
for tolvaptan-induced polyuria based on novel insights ADPKD in humans.
into AQP2 regulation. A focus is placed on those that Another strategy for treating ADPKD involves tar-
could be tested in clinical trials of patients treated with geting the secretory phenotype of the epithelia because
tolvaptan, and we propose the novel hypothesis that formation of fluid-filled cysts from epithelial cells is the
AQP2 dysregulation is an overlooked aspect of ADPKD hallmark of ADPKD, a process that is fuelled at least in
pathophysiology and requires investigation. part by cAMP-mediated Cl− secretion through CFTR123.
Accordingly, treatment of neonatal, kidney-specific
The pathophysiology of ADPKD Pkd1-knockout mice with small-molecule CFTR inhibi-
ADPKD is an adult-onset ciliopathy114 that is character- tors for 7 days slowed kidney enlargement and cyst expan-
ized by diffuse cyst development, and ~95% of ADPKD sion, and partially preserved kidney function compared
cases are associated with mutations in PKD1 (encoding with untreated knockout mice121. In Pkd1-knockout
polycystin 1 (PC1)) or PKD2 (encoding PC2). PC1 and mice (a model of ADPKD), CFTR was predominantly
PC2 can form a complex that traffics to the primary detected in the apical plasma membrane (consistent with
cilia of kidney collecting duct cells where it functions its role in cAMP-dependent fluid secretion), which con-
as a cation channel115. Normally, bending the primary trasts with the intracellular, and apical and basolateral
cilium alters flow-induced calcium signalling116, which membrane localization observed in wild-type mice132.
modulates various signalling pathways, including induc- However, the CFTR corrector VX-809 redirected CFTR
ing alterations in cAMP. In the collecting duct, these sig- towards the basolateral plasma membrane, which pro-
nalling responses are amplified by ATP release through moted a fluid absorption cyst phenotype, rather than
connexin 30 channels situated on the apical plasma mem- the fluid secretion phenotype observed in untreated
brane of β-intercalated cells, and P2Y2 receptors on the Pkd1-knockout mice. VX-809 also reduced the cyst
apical plasma membrane of principal cells117. However, in index and serum creatinine in Pkd1-knockout mice,
ADPKD, flow-induced calcium signalling is impaired118, although a prominent cystic phenotype remained133.
which leads to disproportionate GPCR signalling and a Compared with control mice, Pkd1-knockout mice also
myriad of cellular effects, including increased intracel- have reduced levels of the α-subunit of the epithelial Na+
lular cAMP and increased activity of the cystic fibrosis channel (αENaC), which is highly associated with the
transmembrane regulator (CFTR). These alterations endoplasmic reticulum, rather than being expressed at
result in progressive fluid secretion into the cyst lumen, the apical plasma membrane as normal. Surprisingly,
which is widely thought to be a major pathophysiological VX-809 rescued the subcellular location defect of αENaC
mechanism for cyst growth. Supporting this mechanism, but not its expression levels132. In line with these studies,
conditional inactivation of PC1 (ref.119) and PC2 (ref.120) patients with ADPKD have higher fractional excretion
in mice leads to the development of kidney cysts and of Na+ than healthy individuals134, which is consistent
small-molecule CFTR inhibitors slow cyst growth121 (dis- with lower apical expression of ENaC. These observa-
cussed below). By contrast, mice deficient for connexin tions suggest that ADPKD is characterized by increased
30 or P2Y2 do not develop kidney cysts117,122. Thus, the cellular proliferation, increased epithelial Cl− secretion
pathophysiology of ADPKD is not readily explained by and deficient epithelial Na+ reabsorption in the kidney.

www.nature.com/nrneph

0123456789();:
Reviews

Urine and microcysts as early as 1 week postpartum135; induc-


ible Pkd1-knockout mice have a similar phenotype
Cl–
H2O H2O Cl– Cl– 4 weeks after induction of knockout when the mice are
Apical plasma membrane ‘pre-cystic’ (ref.136). However, whether Cl− secretion into
the lumen of cysts severely affects the osmotic gradient
and favours water transport into cysts, or whether water
Cytosol AQP2 CFTR is retained in the lumen owing to AQP2 dysregulation
remains unclear. The role of AQP2 in the pathophysiol-
Translocation ogy of ADPKD therefore requires thorough investiga-
tion. Below, we discuss the potential of novel regulators
of AQP2 that have slowed the progression of ADPKD in
animal models or clinical trials.

Multipronged approach to prevent kidney failure in


PKA patients with ADPKD. Patients with ADPKD might
Src Pravastatin
benefit from a multipronged treatment approach that
Bosutinib
targets cellular proliferation and transformation of the
Metformin epithelia from a reabsorptive to a secretory phenotype123
Cell proliferation simultaneously (Fig. 5). Currently, tolvaptan is the only
RhoA cAMP drug approved by the FDA for ADPKD treatment and
is predicted to delay the progression to kidney failure by
ERK up to 7.3 years7–9,113. The rationale for the use of tolvaptan
is that its inhibition of V2R signalling should reduce
cAMP-mediated CFTR-induced Cl − secretion and
MYC
ERK-mediated epithelial cell proliferation137. Animal
mTOR
models suggest that cAMP formation in ADPKD occurs
Nucleus through the calcium-sensitive adenylyl cyclases AC5
Basolateral
plasma membrane (ref.138) and AC6 (ref.139), which enhance cyst expansion,
whereas in vitro studies have also suggested a role for
AC3 (refs140,141). In general, V2R has a preference for AC6
V2R (refs18,141) and we would predict that tolvaptan inhibits
Blood Tolvaptan cAMP formation mediated by AC6 but not AC5, and
Vasopressin
only has an effect in portions of the nephron and the
Stimulation Inhibition Drug collecting duct where V2R is highly expressed. Given
its aforementioned side effects, the use of tolvaptan in
Fig. 5 | Drug targets and novel treatment options for ADpKD. Cell proliferation and ADPKD might benefit from treatment in parallel with
cystic fibrosis transmembrane regulator (CFTR)-mediated Cl− secretion are established agents that increase AQP2 plasma membrane targeting
contributors to cyst progression in autosomal dominant polycystic kidney disease and urine concentration, independently of the detri-
(ADPKD). Several novel drugs that target these processes have been tested in patients mental signalling pathways of V2R. Intriguingly, several
with ADPKD. Metformin inhibits mechanistic target of rapamycin (mTOR), which is a of these potential pathways can be targeted by drugs
major regulator of MYC transcription and reduces cyst progression, as well as glomerular
shown to delay cyst progression in preliminary clinical or
filtration rate decline in ADPKD mouse models. A retrospective study of metformin in
preclinical trials.
patients with ADPKD supported these findings147; randomized controlled clinical trials
are ongoing. The tyrosine-protein kinase Src inhibitor bosutinib prevented cyst Bosutinib was administered to patients with ADPKD
progression in a randomized controlled clinical trial in patients with ADPKD142. Statins during a 24-month phase II randomized controlled clini-
can inhibit RhoA and pravastatin inhibited cyst progression in young adults with cal trial142. Increases in total kidney volume were reduced
ADPKD143. Statins, bosutinib and metformin also regulate aquaporin 2 (AQP2) plasma to 0.84% in patients treated with bosutinib (200–400 mg
membrane targeting (Fig. 3). Thus, in contrast to ADPKD drugs that target only cell daily) compared with 4.74% in patients who received
proliferation (such as everolimus), these newer and more promising drug options also a placebo. Although the rationale for using Src inhib-
enhance the absorptive phenotype of the collecting duct epithelium. Tolvaptan, which is itors to treat ADPKD was to target their effect on cell
a vasopressin V2 receptor (V2R) antagonist, inhibits extracellular signal-regulated kinase proliferation, Src inhibition might have also caused
(ERK)-mediated cell proliferation and cAMP-mediated CFTR activity, and delays kidney
vasopressin-independent targeting of AQP2 towards
failure in ADPKD7–9,113. PKA, protein kinase A.
the apical plasma membrane (discussed earlier), thereby
correcting the concentrating defect and restoring
Potential role for AQP2 in ADPKD epithelial fluid reabsorption.
Compared with healthy individuals, urine osmolal- Patients with ADPKD aged 8–22 years that were
ity is lower and urine volume higher in patients with being treated with the angiotensin-converting enzyme
ADPKD, despite increased vasopressin levels. This inhibitor lisinopril were randomly assigned to receive
observation is consistent with the hypothesis that the placebo or the statin pravastatin in a small randomized
pathophysiology of ADPKD is partially due to reduced controlled trial143. At the 3-year follow-up, the percen­
absorptive capacity in cystic kidneys that do not respond tage change in total kidney volume (adjusted for height,
sufficiently to vasopressin123. In line with this hypothesis, age, sex and hypertension status) was significantly
Aqp2-knockout mice display collecting duct expansion decreased in the pravastatin group versus placebo

Nature Reviews | Nephrology

0123456789();:
Reviews

(23% ± 3% versus 31% ± 3%; P = 0.02). The patients in PKA-mediated Rho inhibition 101 and, importantly,
these trials were young and, on average, normotensive cAMP-induced CFTR activation151. We predict that
and normocholesterolaemic; therefore, the effect of identifying specific AKAPs involved in PKA-mediated
pravastatin is unlikely to be secondary to vascular ben- cyst progression could lead to the development of
efits. As discussed earlier, statins have a mechanism of AKAP–PKA disrupters that target such selected AKAPs.
action similar to fluconazole; thus, they might not only In contrast to tolvaptan, these would function inde-
inhibit RhoA–ROCK-mediated MYC transcription and pendently of specific receptors and tubular structures.
cellular proliferation during ADPKD but they might also As discussed above, specific AKAP–PKA interaction
enhance AQP2 plasma membrane targeting. inhibitors might also rescue the concentrating defect
In addition to its aforementioned effects on AQP2, caused by tolvaptan treatment in ADPKD, as observed
which are probably due to its effects on AMPK, met- with FMP-API-1 (ref.98).
formin inhibits the activity of CFTR and mTOR, and
has shown promise in preventing and/or reducing cyst Conclusions
growth in miniature pig models of ADPKD and in some, Much is known about the molecular regulation of
but not all, mouse models of ADPKD144–146. Randomized AQP2, but several of the signalling pathways that link
controlled clinical trials are currently underway to inves- receptor stimulation to AQP2 plasma membrane tar-
tigate the effects of metformin in patients with ADPKD, geting remain elusive. Furthermore, how these signal-
but preliminary retrospective studies have indicated ling pathways interact or their potential crosstalk, and
a slower decline in glomerular filtration rate over a whether one or more of these pathways is dominant
3-year period in patients with diabetes and ADPKD over another, are unknown. Nonetheless, the canonical
who received metformin, compared with patients with pathway of AQP2 targeting to the plasma membrane
ADPKD without diabetes147. Whether this reduction in — cAMP–PKA-mediated Ser256 phosphorylation — is
glomerular filtration rate decline is due to the ability of insufficient to explain multiple research findings. For
metformin to reduce blood sugar levels in patients with example, Ser256 phosphorylation is not a rate-limiting
diabetes or any direct beneficial effect on ADPKD is step for plasma membrane targeting and can occur with-
unknown. out cAMP-stimulated PKA activity; cAMP and PKA
Erlotinib has been approved by the FDA for the activity are important for AQP2 protein abundance, but
treatment of non-small-cell lung cancer. This drug AQP2 plasma membrane targeting and urine osmolality
reduces mTOR and ERK signalling and reduced cyst can increase independently from their activity; AMPK
growth in Pkd1-knockout mice through an unidenti- might be a novel signalling node of PKA-independent
fied mechanism148. As discussed previously, erlotinib V2R signalling and AQP2 plasma membrane target-
also increases AQP2 plasma membrane targeting and, ing, but the mechanism underlying AMPK activation
although clinical trials in ADPKD are not available, it is downstream of V2R activation is unknown.
another candidate tolvaptan companion drug. In ADPKD, despite the known urine-concentrating
defect and the generation of large fluid-filled cysts that
New treatment options for ADPKD derive from the tubular epithelium, the pathophysiolog-
The aforementioned effects of fluconazole60 — enhancing ical role of AQP2 has never been investigated in detail.
AQP2 plasma membrane targeting without increasing This gap in knowledge is probably due to the complexity
cAMP or PKA levels, and decreasing activity of RhoA — of separating known factors that occur downstream of
might be beneficial in ADPKD. Furthermore, this anti- V2R activation and that are known to be detrimental
mycotic is generally well tolerated during long-term in ADPKD, such as increased cAMP and ERK activa-
administration, for example, in patients with recurrent tion, from a direct role of AQP2. However, the emer-
candidiasis149, and its urine-concentrating effects seem gence of novel signalling pathways and compounds that
to be sustained over several days60. Understanding the cause AQP2 plasma membrane targeting independently
effects of other AQP2 regulators such as Wnt5a, or other from cAMP–PKA provides an opportunity to investi-
frizzled receptors in ADPKD would be informative and gate in detail the pathophysiological role of AQP2 in
might uncover further alternative treatment options for ADPKD, potentially expanding the breadth of current
ADPKD. Finally, AKAP–PKA disrupters might also be treatment options. Furthermore, novel compounds
beneficial in ADPKD. Although the general AKAP– that increase AQP2 plasma membrane targeting inde-
PKA interaction inhibitors Ht31 and FMP-API-1 pendently of V2R, cAMP or PKA might be ideal com-
increased PKA activity in collecting duct cells98, which panion drugs to tolvaptan in the treatment of ADPKD
is a known detrimental factor in ADPKD150, certain because they might ameliorate both cyst progression and
physiological effects of PKA are attenuated in the con- tolvaptan-induced polyuria. Clinical trials are warranted
text of AKAP–PKA disruption; the general AKAP–PKA to develop such a multipronged approach.
interaction inhibitor Ht31 decreases forskolin-induced
phosphorylation of CREB (a sensor for PKA activity)100, Published online xx xx xxxx

1. Nawata, C. M. & Pannabecker, T. L. Mammalian urine from AQP7 knockout mice. Biochim. Biophys. Acta 4. Kortenoeven, M. L. & Fenton, R. A. Renal aquaporins
concentration: a review of renal medullary architecture 1758, 1106–1110 (2006). and water balance disorders. Biochim. Biophys. Acta
and membrane transporters. J. Comp. Physiol. B 188, 3. Fenton, R. A. & Knepper, M. A. Mouse models and 1840, 1533–1549 (2014).
899–918 (2018). the urinary concentrating mechanism in the 5. Cheung, P. W., Bouley, R. & Brown, D. Targeting the traff­
2. Sohara, E., Rai, T., Sasaki, S. & Uchida, S. new millennium. Physiol. Rev. 87, 1083–1112 icking of kidney water channels for therapeutic benefit.
Physiological roles of AQP7 in the kidney: lessons (2007). Annu. Rev. Pharmacol. Toxicol. 60, 175–194 (2020).

www.nature.com/nrneph

0123456789();:
Reviews

6. Verbalis, J. G. Acquired forms of central diabetes 29. Olesen, E. T., Rutzler, M. R., Moeller, H. B., 51. Trimpert, C. et al. NDFIP allows NEDD4/
insipidus: Mechanisms of disease. Best Pract. Res. Praetorius, H. A. & Fenton, R. A. Vasopressin- NEDD4L-induced AQP2 ubiquitination and
Clin. Endocrinol. Metab. 34, 101449 (2020). independent targeting of aquaporin-2 by selective degradation. PLoS ONE 12, e0183774 (2017).
7. Torres, V. E. et al. Tolvaptan in patients with autosomal E-prostanoid receptor agonists alleviates nephrogenic 52. Lee, Y. J. et al. E3 ubiquitin-protein ligases in rat
dominant polycystic kidney disease. N. Engl. J. Med. diabetes insipidus. Proc. Natl Acad. Sci. USA 108, kidney collecting duct: response to vasopressin
367, 2407–2418 (2012). 12949–12954 (2011). stimulation and withdrawal. Am. J. Physiol. Renal
8. Torres, V. E. et al. Multicenter, open-label, extension 30. Jung, H. J. & Kwon, T. H. Molecular mechanisms Physiol. 301, F883–F896 (2011).
trial to evaluate the long-term efficacy and safety of regulating aquaporin-2 in kidney collecting duct. 53. Medvar, B., Raghuram, V., Pisitkun, T., Sarkar, A. &
early versus delayed treatment with tolvaptan in Am. J. Physiol. Renal Physiol. 311, F1318–F1328 Knepper, M. A. Comprehensive database of human E3
autosomal dominant polycystic kidney disease: (2016). ubiquitin ligases: application to aquaporin-2
the TEMPO 4:4 Trial. Nephrol. Dial. Transpl. 33, 31. Sun, T. X. et al. Aquaporin-2 localization in regulation. Physiol. Genomics 48, 502–512 (2016).
477–489 (2018). clathrin-coated pits: inhibition of endocytosis by 54. Wu, Q. et al. CHIP regulates aquaporin-2 quality
9. Torres, V. E. et al. Tolvaptan in later-stage autosomal dominant-negative dynamin. Am. J. Physiol. Renal control and body water homeostasis. J. Am. Soc.
dominant polycystic kidney disease. N. Engl. J. Med. Physiol. 282, F998–F1011 (2002). Nephrol. 29, 936–948 (2018).
377, 1930–1942 (2017). 32. Moeller, H. B., Knepper, M. A. & Fenton, R. A. Serine 55. Centrone, M. et al. AQP2 abundance is regulated by
10. Valtin, H., Schroeder, H. A., Benirschke, K. & 269 phosphorylated aquaporin-2 is targeted to the the E3-ligase CHIP Via HSP70. Cell Physiol. Biochem.
Sokol, H. W. Familial hypothalamic diabetes insipidus apical membrane of collecting duct principal cells. 44, 515–531 (2017).
in rats. Nature 196, 1109–1110 (1962). Kidney Int. 75, 295–303 (2009). 56. Dema, A. et al. Cyclin-dependent kinase 18 controls
11. Li, J. H. et al. A selective EP4 PGE2 receptor agonist 33. Wang, W. L. et al. Rab7 involves Vps35 to mediate trafficking of aquaporin-2 and its abundance through
alleviates disease in a new mouse model of X-linked AQP2 sorting and apical trafficking in collecting duct ubiquitin ligase STUB1, which functions as an AKAP.
nephrogenic diabetes insipidus. J. Clin. Invest. 119, cells. Am. J. Physiol. Renal Physiol. 318, F956–F970 Cells 9, 673 (2020).
3115–3126 (2009). (2020). 57. Murali, S. K., Aroankins, T. S., Moeller, H. B. &
12. Rieg, T. et al. Adenylyl cyclase 6 enhances NKCC2 34. Vossenkamper, A. et al. Microtubules are needed for Fenton, R. A. The deubiquitylase USP4 interacts
expression and mediates vasopressin-induced the perinuclear positioning of aquaporin-2 after its with the water channel AQP2 to modulate its apical
phosphorylation of NKCC2 and NCC. Am. J. Pathol. endocytic retrieval in renal principal cells. Am. J. membrane accumulation and cellular abundance.
182, 96–106 (2013). Physiol. Cell Physiol. 293, C1129–C1138 (2007). Cells 8, 265 (2019).
13. Mutig, K. et al. Demonstration of the functional 35. Tajika, Y. et al. Aquaporin-2 is retrieved to the apical 58. Moeller, H. B., Aroankins, T. S., Slengerik-Hansen, J.,
impact of vasopressin signaling in the thick ascending storage compartment via early endosomes and Pisitkun, T. & Fenton, R. A. Phosphorylation and
limb by a targeted transgenic rat approach. Am. J. phosphatidylinositol 3-kinase-dependent pathway. ubiquitylation are opposing processes that regulate
Physiol. Renal Physiol. 311, F411–F423 (2016). Endocrinology 145, 4375–4383 (2004). endocytosis of the water channel aquaporin-2.
14. Pedersen, N. B., Hofmeister, M. V., Rosenbaek, L. L., 36. Lee, M. S., Choi, H. J., Park, E. J., Park, H. J. J. Cell Sci. 127, 3174–3183 (2014).
Nielsen, J. & Fenton, R. A. Vasopressin induces & Kwon, T. H. Depletion of vacuolar protein 59. Moeller, H. B. et al. Regulation of the water channel
phosphorylation of the thiazide-sensitive sodium sorting-associated protein 35 is associated with aquaporin-2 via 14-3-3theta and -zeta. J. Biol. Chem.
chloride cotransporter in the distal convoluted tubule. increased lysosomal degradation of aquaporin-2. 291, 2469–2484 (2016).
Kidney Int. 78, 160–169 (2010). Am. J. Physiol. Renal Physiol. 311, F1294–F1307 60. Vukicevic, T. et al. Fluconazole increases osmotic water
15. Mutig, K. et al. Vasopressin V2 receptor expression (2016). transport in renal collecting duct through effects on
along rat, mouse, and human renal epithelia with 37. Nedvetsky, P. I. et al. A role of myosin Vb and aquaporin-2 trafficking. J. Am. Soc. Nephrol. 30,
focus on TAL. Am. J. Physiol. Renal Physiol. 293, Rab11-FIP2 in the aquaporin-2 shuttle. Traffic 8, 795–810 (2019).
F1166–F1177 (2007). 110–123 (2007). 61. Loo, C. S. et al. Quantitative apical membrane
16. Welker, P. et al. Renal Na+-K+-Cl− cotransporter activity 38. Yui, N. et al. Basolateral targeting and microtubule- proteomics reveals vasopressin-induced actin
and vasopressin-induced trafficking are lipid dependent transcytosis of the aquaporin-2 water dynamics in collecting duct cells. Proc. Natl Acad. Sci.
raft-dependent. Am. J. Physiol. Renal Physiol. 295, channel. Am. J. Physiol. Cell Physiol. 304, C38–C48 USA 110, 17119–17124 (2013).
F789–F802 (2008). (2013). 62. Simon, H., Gao, Y., Franki, N. & Hays, R. M.
17. Sakuma, Y. et al. Differential effects of hyperosmolality 39. Bouley, R., Yui, N., Terlouw, A., Cheung, P. W. & Vasopressin depolymerizes apical F-actin in rat inner
on Na-K-ATPase and vasopressin-dependent cAMP Brown, D. Chlorpromazine induces basolateral medullary collecting duct. Am. J. Physiol. 265,
generation in the medullary thick ascending limb and aquaporin-2 accumulation via F-actin depoly­ C757–C762 (1993).
outer medullary collecting duct. Hypertens. Res. 28, merization and blockade of endocytosis in renal 63. Chou, C. L. et al. Non-muscle myosin II and myosin
671–679 (2005). epithelial cells. Cells 9, 1057 (2020). light chain kinase are downstream targets for
18. Rieg, T. et al. Adenylate cyclase 6 determines cAMP 40. Deshpande, V. et al. Phosphoproteomic identification vasopressin signaling in the renal collecting duct.
formation and aquaporin-2 phosphorylation and of vasopressin V2 receptor-dependent signaling in the J. Biol. Chem. 279, 49026–49035 (2004).
trafficking in inner medulla. J. Am. Soc. Nephrol. 21, renal collecting duct. Am. J. Physiol. Renal Physiol. 64. Klussmann, E. et al. An inhibitory role of Rho in the
2059–2068 (2010). 317, F789–F804 (2019). vasopressin-mediated translocation of aquaporin-2
19. Star, R. A., Nonoguchi, H., Balaban, R. & 41. Hoffert, J. D., Pisitkun, T., Wang, G., Shen, R. F. & into cell membranes of renal principal cells. J. Biol.
Knepper, M. A. Calcium and cyclic adenosine Knepper, M. A. Quantitative phosphoproteomics Chem. 276, 20451–20457 (2001).
monophosphate as second messengers for of vasopressin-sensitive renal cells: regulation of 65. Ranieri, M. et al. The vasopressin receptor 2 mutant
vasopressin in the rat inner medullary collecting duct. aquaporin-2 phosphorylation at two sites. Proc. Natl R137L linked to the nephrogenic syndrome of
J. Clin. Invest. 81, 1879–1888 (1988). Acad. Sci. USA 103, 7159–7164 (2006). inappropriate antidiuresis (NSIAD) signals through an
20. Marples, D., Christensen, B. M., Frokiaer, J., 42. Hoffert, J. D. et al. Vasopressin-stimulated increase alternative pathway that increases AQP2 membrane
Knepper, M. A. & Nielsen, S. Dehydration reverses in phosphorylation at Ser269 potentiates plasma targeting independently of S256 phosphorylation.
vasopressin antagonist-induced diuresis and membrane retention of aquaporin-2. J. Biol. Chem. Cells 9, 1354 (2020).
aquaporin-2 downregulation in rats. Am. J. Physiol. 283, 24617–24627 (2008). 66. Feldman, B. J. et al. Nephrogenic syndrome of
275, F400–F409 (1998). 43. Xie, L. et al. Quantitative analysis of aquaporin-2 inappropriate antidiuresis. N. Engl. J. Med. 352,
21. Su, W., Cao, R., Zhang, X. Y. & Guan, Y. Aquaporins phosphorylation. Am. J. Physiol. Renal Physiol. 298, 1884–1890 (2005).
in the kidney: physiology and pathophysiology. F1018–F1023 (2010). 67. Lei, L. et al. Manganese promotes intracellular
Am. J. Physiol. Renal. Physiol. 318, F193–F203 44. Moeller, H. B., Praetorius, J., Rutzler, M. R. & accumulation of AQP2 via modulating F-actin
(2019). Fenton, R. A. Phosphorylation of aquaporin-2 polymerization and reduces urinary concentration in
22. Zhang, X. Y., Wang, B. & Guan, Y. F. Nuclear receptor regulates its endocytosis and protein-protein mice. Am. J. Physiol. Renal Physiol. 314, F306–F316
regulation of aquaporin-2 in the kidney. Int. J. Mol. interactions. Proc. Natl Acad. Sci. USA 107, 424–429 (2018).
Sci. 17, 1105 (2016). (2010). 68. Li, W. et al. Ezrin directly interacts with AQP2
23. Li, S. et al. Bile acid G protein-coupled membrane 45. Wang, P. J. et al. Vasopressin-induced serine 269 and promotes its endocytosis. J. Cell Sci. 130,
receptor TGR5 modulates aquaporin 2-mediated phosphorylation reduces Sipa1l1 (signal-induced 2914–2925 (2017).
water homeostasis. J. Am. Soc. Nephrol. 29, proliferation-associated 1 like 1)-mediated 69. Neisch, A. L. & Fehon, R. G. Ezrin, radixin and moesin:
2658–2670 (2018). aquaporin-2 endocytosis. J. Biol. Chem. 292, key regulators of membrane-cortex interactions
24. Li, C. et al. Molecular mechanisms of angiotensin II 7984–7993 (2017). and signaling. Curr. Opin. Cell Biol. 23, 377–382
stimulation on aquaporin-2 expression and trafficking. 46. Roche, J. V., Nesverova, V., Olsson, C., Deen, P. M. & (2011).
Am. J. Physiol. Renal Physiol. 300, F1255–F1261 Tornroth-Horsefield, S. Structural insights into AQP2 70. Dransfield, D. T. et al. Ezrin is a cyclic AMP-dependent
(2011). targeting to multivesicular bodies. Int. J. Mol. Sci. 20, protein kinase anchoring protein. EMBO J. 16,
25. Bouley, R. et al. Calcitonin has a vasopressin-like effect 5351 (2019). 35–43 (1997).
on aquaporin-2 trafficking and urinary concentration. 47. Roche, J. V. & Tornroth-Horsefield, S. Aquaporin 71. Brunton, L. L., Hayes, J. S. & Mayer, S. E. Functional
J. Am. Soc. Nephrol. 22, 59–72 (2011). protein-protein interactions. Int. J. Mol. Sci. 18, compartmentation of cyclic AMP and protein kinase
26. Yano, Y. et al. Aquaporin 2 expression increased by 2255 (2017). in heart. Adv. Cyclic Nucleotide Res. 14, 391–397
glucagon in normal rat inner medullary collecting 48. Roche, J. V. et al. Phosphorylation of human (1981).
ducts. Am. J. Physiol. Renal Physiol. 296, F54–F59 aquaporin 2 (AQP2) allosterically controls its 72. Knepper, M. A., Kwon, T. H. & Nielsen, S. Molecular
(2009). interaction with the lysosomal trafficking protein LIP5. physiology of water balance. N. Engl. J. Med. 373,
27. Chu, J. Y. et al. Phenotypes developed in secretin J. Biol. Chem. 292, 14636–14648 (2017). 196 (2015).
receptor-null mice indicated a role for secretin in 49. Choi, H. J. et al. Sorting nexin 27 regulates the 73. Nedvetsky, P. I. et al. in Aquaporins. Handbook of
regulating renal water reabsorption. Mol. Cell Biol. lysosomal degradation of aquaporin-2 protein in Experimental Pharmacology Vol. 190 (ed. Beitz, E.)
27, 2499–2511 (2007). the kidney collecting duct. Cells 9, 1208 (2020). (Springer, 2009).
28. Lee, Y. J. et al. Increased AQP2 targeting in primary 50. Kamsteeg, E. J. et al. Short-chain ubiquitination 74. Noda, Y. & Sasaki, S. Trafficking mechanism of water
cultured IMCD cells in response to angiotensin II mediates the regulated endocytosis of the channel aquaporin-2. Biol. Cell 97, 885–892 (2005).
through AT1 receptor. Am. J. Physiol. Renal Physiol. aquaporin-2 water channel. Proc. Natl Acad. Sci. USA 75. Katsura, T., Gustafson, C. E., Ausiello, D. A. & Brown,
292, F340–F350 (2007). 103, 18344–18349 (2006). D. Protein kinase A phosphorylation is involved in

Nature Reviews | Nephrology

0123456789();:
Reviews

regulated exocytosis of aquaporin-2 in transfected nephrogenic diabetes insipidus. Nat. Commun. 9, 123. Sullivan, L. P., Wallace, D. P. & Grantham, J. J.
LLC-PK1 cells. Am. J. Physiol. 272, F817–F822 (1997). 1411 (2018). Epithelial transport in polycystic kidney disease.
76. Limbutara, K., Kelleher, A., Yang, C. R., Raghuram, V. 99. Christian, F. et al. Small molecule AKAP-protein kinase Physiol. Rev. 78, 1165–1191 (1998).
& Knepper, M. A. Phosphorylation changes in A (PKA) interaction disruptors that activate PKA 124. Menezes, L. F. & Germino, G. G. The pathobiology of
response to kinase inhibitor H89 in PKA-null cells. interfere with compartmentalized cAMP signaling in polycystic kidney disease from a metabolic viewpoint.
Sci. Rep. 9, 2814 (­20­19­). cardiac myocytes. J. Biol. Chem. 286, 9079–9096 Nat. Rev. Nephrol. 15, 735–749 (2019).
77. Gilbert, M. L., Yang, L., Su, T. & McKnight, G. S. (2011). 125. Cornec-Le Gall, E., Alam, A. & Perrone, R. D.
Expression of a dominant negative PKA mutation 100. Friedrich, M. W., Aramuni, G., Mank, M., Autosomal dominant polycystic kidney disease. Lancet
in the kidney elicits a diabetes insipidus phenotype. Mackinnon, J. A. & Griesbeck, O. Imaging CREB 393, 919–935 (2019).
Am. J. Physiol. Renal Physiol. 308, F627–F638 (2015). activation in living cells. J. Biol. Chem. 285, 126. Cai, J. et al. A RhoA-YAP-c-Myc signaling axis
78. Isobe, K. et al. Systems-level identification of 23285–23295 (2010). promotes the development of polycystic kidney
PKA-dependent signaling in epithelial cells. Proc. Natl 101. Wang, Y., Chen, Y., Chen, M. & Xu, W. AKAPs disease. Genes Dev. 32, 781–793 (2018).
Acad. Sci. USA 114, E8875–E8884 (2017). competing peptide HT31 disrupts the inhibitory effect 127. Shillingford, J. M., Piontek, K. B., Germino, G. G. &
79. Datta, A. et al. PKA-independent vasopressin of PKA on RhoA activity. Oncol. Rep. 16, 755–761 Weimbs, T. Rapamycin ameliorates PKD resulting from
signaling in renal collecting duct. FASEB J. 34, (2006). conditional inactivation of Pkd1. J. Am. Soc. Nephrol.
6129–6146 (2020). 102. Schrade, K. et al. An AKAP-Lbc-RhoA interaction 21, 489–497 (2010).
80. Bradford, D. et al. Use of LC-MS/MS and Bayes’ inhibitor promotes the translocation of aquaporin-2 128. Shillingford, J. M. et al. The mTOR pathway is
theorem to identify protein kinases that to the plasma membrane of renal collecting duct regulated by polycystin-1, and its inhibition reverses
phosphorylate aquaporin-2 at Ser256. Am. J. Physiol. principal cells. PLoS ONE 13, e0191423 (2018). renal cystogenesis in polycystic kidney disease.
Cell Physiol. 307, C123–C139 (2014). 103. Cheung, P. W., Terlouw, A., Janssen, S. A., Brown, D. Proc. Natl Acad. Sci. USA 103, 5466–5471 (2006).
81. Liwang, J. K. et al. Role of PKC and AMPK in & Bouley, R. Inhibition of non-receptor tyrosine 129. Streets, A. J., Prosseda, P. P. & Ong, A. C. Polycystin-1
hypertonicity-stimulated water reabsorption in rat kinase Src induces phosphoserine 256-independent regulates ARHGAP35-dependent centrosomal RhoA
inner medullary collecting ducts. Am. J. Physiol. Renal aquaporin-2 membrane accumulation. J. Physiol. activation and ROCK signaling. JCI Insight 5,
Physiol. 316, F253–F262 (2019). 597, 1627–1642 (2019). e135385 (2020).
82. Al-Bataineh, M. M. et al. Activation of the metabolic 104. Hill, J., Shields, J. & Passero, V. Tyrosine kinase 130. Serra, A. L. et al. Sirolimus and kidney growth in
sensor AMP-activated protein kinase inhibits inhibitor-associated syndrome of inappropriate autosomal dominant polycystic kidney disease.
aquaporin-2 function in kidney principal cells. Am. J. secretion of anti-diuretic hormone. J. Oncol. Pharm. N. Engl. J. Med. 363, 820–829 (2010).
Physiol. Renal Physiol. 311, F890–F900 (2016). Pract. 22, 729–732 (2016). 131. Walz, G. et al. Everolimus in patients with autosomal
83. Klein, J. D. et al. Metformin, an AMPK activator, 105. Ando, F. et al. Wnt5a induces renal AQP2 expression dominant polycystic kidney disease. N. Engl. J. Med.
stimulates the phosphorylation of aquaporin 2 and by activating calcineurin signalling pathway. 363, 830–840 (2010).
urea transporter A1 in inner medullary collecting Nat. Commun. 7, 13636 (2016). 132. Yanda, M. K., Cha, B., Cebotaru, C. V. & Cebotaru, L.
ducts. Am. J. Physiol. Renal Physiol. 310, 106. Bogum, J. et al. Small-molecule screening identifies Pharmacological reversal of renal cysts from secretion
F1008–F1012 (2016). modulators of aquaporin-2 trafficking. J. Am. Soc. to absorption suggests a potential therapeutic
84. Efe, O., Klein, J. D., LaRocque, L. M., Ren, H. & Nephrol. 24, 744–758 (2013). strategy for managing autosomal dominant polycystic
Sands, J. M. Metformin improves urine concentration 107. Mortensen, L. A., Bistrup, C., Jensen, B. L. & kidney disease. J. Biol. Chem. 294, 17090–17104
in rodents with nephrogenic diabetes insipidus. Hinrichs, G. R. A mini-review of pharmacological (2019).
JCI Insight 1, e88409 (2016). strategies used to ameliorate polyuria associated with 133. Yanda, M. K., Liu, Q. & Cebotaru, L. A potential
85. Bech, A. P., Wetzels, J. F. M. & Nijenhuis, T. X-linked nephrogenic diabetes insipidus. Am. J. strategy for reducing cysts in autosomal dominant
Effects of sildenafil, metformin, and simvastatin on Physiol. Renal Physiol. 319, F746–F753 (2020). polycystic kidney disease with a CFTR corrector.
ADH-independent urine concentration in healthy 108. Nomura, N. et al. High-throughput chemical screening J. Biol. Chem. 293, 11513–11526 (2018).
volunteers. Physiol. Rep. 6, e13665 (2018). identifies AG-490 as a stimulator of aquaporin 2 134. Graffe, C. C., Bech, J. N., Lauridsen, T. G. &
86. Olesen, E. T. & Fenton, R. A. Aquaporin-2 membrane membrane expression and urine concentration. Am. J. Pedersen, E. B. Urinary excretion of AQP2 and ENaC
targeting: still a conundrum. Am. J. Physiol. Renal Physiol. Cell Physiol. 307, C597–C605 (2014). in autosomal dominant polycystic kidney disease
Physiol. 312, F744–F747 (2017). 109. Gow, C. B. & Phillips, P. A. Epidermal growth factor during basal conditions and after a hypertonic saline
87. Zhang, J. Z. et al. Phase separation of a PKA as a diuretic in sheep. J. Physiol. 477, 27–33 (1994). infusion. Am. J. Physiol. Renal Physiol. 302,
regulatory subunit controls cAMP compartmentation 110. Cheung, P. W. et al. EGF receptor inhibition by F917–F927 (2012).
and oncogenic signaling. Cell 182, 1531–1544.e15 erlotinib increases aquaporin 2-mediated renal water 135. Chen, Y. et al. Aquaporin 2 promotes cell migration
(2020). reabsorption. J. Am. Soc. Nephrol. 27, 3105–3116 and epithelial morphogenesis. J. Am. Soc. Nephrol.
88. Bock, A. et al. Optical mapping of cAMP signaling (2016). 23, 1506–1517 (2012).
at the nanometer scale. Cell 182, 1519–1530.e17 111. Gabow, P. A. Autosomal dominant polycystic kidney 136. Verschuren, E. H. J. et al. Polycystin-1 dysfunction
(2020). disease. N. Engl. J. Med. 329, 332–342 (1993). impairs electrolyte and water handling in a renal
89. Stefan, E. et al. Compartmentalization of 112. Hajji, M. et al. Clinical study on autosomal dominant precystic mouse model for ADPKD. Am. J. Physiol.
cAMP-dependent signaling by phosphodiesterase-4D polycystic kidney disease among North Tunisians. Renal Physiol. 315, F537–F546 (2018).
is involved in the regulation of vasopressin-mediated Saudi J. Kidney Dis. Transpl. 30, 175–184 (2019). 137. Reif, G. A. et al. Tolvaptan inhibits ERK-dependent cell
water reabsorption in renal principal cells. J. Am. Soc. 113. Chebib, F. T. et al. A practical guide for treatment of proliferation, Cl− secretion, and in vitro cyst growth of
Nephrol. 18, 199–212 (2007). rapidly progressive ADPKD with tolvaptan. J. Am. Soc. human ADPKD cells stimulated by vasopressin. Am. J.
90. Liu, S. et al. Phosphodiesterases coordinate Nephrol. 29, 2458–2470 (2018). Physiol. Renal Physiol. 301, F1005–F1013 (2011).
cAMP propagation induced by two stimulatory G 114. Hildebrandt, F., Benzing, T. & Katsanis, N. 138. Wang, Q. et al. Adenylyl cyclase 5 deficiency reduces
protein-coupled receptors in hearts. Proc. Natl Acad. Ciliopathies. N. Engl. J. Med. 364, 1533–1543 renal cyclic AMP and cyst growth in an orthologous
Sci. USA 109, 6578–6583 (2012). (2011). mouse model of polycystic kidney disease. Kidney Int.
91. Omar, M. H. & Scott, J. D. AKAP signaling islands: 115. Douguet, D., Patel, A. & Honore, E. Structure and 93, 403–415 (2018).
venues for precision pharmacology. Trends Pharmacol. function of polycystins: insights into polycystic kidney 139. Rees, S. et al. Adenylyl cyclase 6 deficiency
Sci. 41, 933–946 (2020). disease. Nat. Rev. Nephrol. 15, 412–422 (2019). ameliorates polycystic kidney disease. J. Am. Soc.
92. Wingler, L. M. & Lefkowitz, R. J. Conformational basis 116. Leipziger, J. & Praetorius, H. A. Renal autocrine and Nephrol. 25, 232–237 (2014).
of G protein-coupled receptor signaling versatility. paracrine signaling: a story of self-protection. 140. Pinto, C. S., Reif, G. A., Nivens, E., White, C. &
Trends Cell Biol. 30, 736–747 (2020). Physiol. Rev. 100, 1229–1289 (2020). Wallace, D. P. Calmodulin-sensitive adenylyl cyclases
93. Olesen, E. T., Moeller, H. B., Assentoft, M., MacAulay, N. 117. Svenningsen, P., Burford, J. L. & Peti-Peterdi, J. mediate AVP-dependent cAMP production and Cl−
& Fenton, R. A. The vasopressin type 2 receptor and ATP releasing connexin 30 hemichannels mediate secretion by human autosomal dominant polycystic
prostaglandin receptors EP2 and EP4 can increase flow-induced calcium signaling in the collecting duct. kidney cells. Am. J. Physiol. Renal Physiol. 303,
aquaporin-2 plasma membrane targeting through a Front. Physiol. 4, 292 (2013). F1412–F1424 (2012).
cAMP-independent pathway. Am. J. Physiol. Renal 118. Xu, C. et al. Human ADPKD primary cyst epithelial 141. Sherpa, R. T. et al. Sensory primary cilium is a
Physiol. 311, F935–F944 (2016). cells with a novel, single codon deletion in the PKD1 responsive cAMP microdomain in renal epithelia.
94. Brunton, L. L., Hayes, J. S. & Mayer, S. E. Hormonally gene exhibit defective ciliary polycystin localization Sci. Rep. 9, 6523 (2019).
specific phosphorylation of cardiac troponin I and and loss of flow-induced Ca2+ signaling. Am. J. Physiol. 142. Tesar, V. et al. Bosutinib versus placebo for autosomal
activation of glycogen phosphorylase. Nature 280, Renal Physiol. 292, F930–F945 (2007). dominant polycystic kidney disease. J. Am. Soc.
78–80 (1979). 119. Shibazaki, S. et al. Cyst formation and activation Nephrol. 28, 3404–3413 (2017).
95. Hayes, J. S., Brunton, L. L. & Mayer, S. E. Selective of the extracellular regulated kinase pathway after 143. Cadnapaphornchai, M. A. et al. Effect of pravastatin
activation of particulate cAMP-dependent protein kidney specific inactivation of Pkd1. Hum. Mol. Genet. on total kidney volume, left ventricular mass index,
kinase by isoproterenol and prostaglandin E1. 17, 1505–1516 (2008). and microalbuminuria in pediatric autosomal
J. Biol. Chem. 255, 5113–5119 (1980). 120. Ma, M., Tian, X., Igarashi, P., Pazour, G. J. & Somlo, S. dominant polycystic kidney disease. Clin. J. Am. Soc.
96. Henn, V. et al. Identification of a novel A-kinase Loss of cilia suppresses cyst growth in genetic models Nephrol. 9, 889–896 (2014).
anchoring protein 18 isoform and evidence for of autosomal dominant polycystic kidney disease. 144. Leonhard, W. N. et al. Salsalate, but not metformin
its role in the vasopressin-induced aquaporin-2 Nat. Genet. 45, 1004–1012 (2013). or canagliflozin, slows kidney cyst growth in an
shuttle in renal principal cells. J. Biol. Chem. 279, 121. Yang, B., Sonawane, N. D., Zhao, D., Somlo, S. & adult-onset mouse model of polycystic kidney disease.
26654–26665 (2004). Verkman, A. S. Small-molecule CFTR inhibitors slow EBioMedicine 47, 436–445 (2019).
97. Klussmann, E., Maric, K., Wiesner, B., Beyermann, M. cyst growth in polycystic kidney disease. J. Am. Soc. 145. Lian, X. et al. The combination of metformin and
& Rosenthal, W. Protein kinase A anchoring proteins Nephrol. 19, 1300–1310 (2008). 2-deoxyglucose significantly inhibits cyst formation
are required for vasopressin-mediated translocation 122. Zhang, Y. et al. Genetic deletion of P2Y2 receptor in miniature pigs with polycystic kidney disease.
of aquaporin-2 into cell membranes of renal principal offers long-term (5 months) protection against Br. J. Pharmacol. 176, 711–724 (2019).
cells. J. Biol. Chem. 274, 4934–4938 (1999). lithium-induced polyuria, natriuresis, kaliuresis, and 146. Takiar, V. et al. Activating AMP-activated protein
98. Ando, F. et al. AKAPs-PKA disruptors increase AQP2 collecting duct remodeling and cell proliferation. kinase (AMPK) slows renal cystogenesis. Proc. Natl
activity independently of vasopressin in a model of Front. Physiol. 9, 1765 (2018). Acad. Sci. USA 108, 2462–2467 (2011).

www.nature.com/nrneph

0123456789();:
Reviews

147. Pisani, A., Riccio, E., Bruzzese, D. & Sabbatini, M. to CFTR by AKAPs. Am. J. Physiol. Cell Physiol. 278, manuscript, and reviewed or edited the manuscript before
Metformin in autosomal dominant polycystic kidney C417–C422 (2000). submission.
disease: experimental hypothesis or clinical fact? BMC 152. Fenton, R. A., Murali, S. K. & Moeller, H. B.
Nephrol. 19, 282 (2018). Advances in aquaporin-2 trafficking mechanisms and Competing interests
148. Nikonova, A. S. et al. Opposing effects of inhibitors of their implications for treatment of water balance The authors declare no competing interests.
aurora-A and EGFR in autosomal-dominant polycystic disorders. Am. J. Physiol. Cell Physiol. 319, C1–C10
kidney disease. Front. Oncol. 5, 228 (2015). (2020). Peer review information
149. Collins, L. M., Moore, R. & Sobel, J. D. Prognosis and Nature Reviews Nephrology thanks T.-H. Kwon, V. Torres and
long-term outcome of women with idiopathic recurrent Acknowledgements the other, anonymous, reviewer(s) for their contribution to the
vulvovaginal candidiasis caused by candida albicans. The authors’ research is supported by the Danish Medical peer review of this work.
J. Low. Genit. Tract Dis. 24, 48–52 (2020). Research Council, The Novo Nordisk Foundation, The Leducq
150. Ye, H. et al. The regulatory 1alpha subunit of protein Foundation and the Carlsberg Foundation. Publisher’s note
kinase A modulates renal cystogenesis. Am. J. Physiol. Springer Nature remains neutral with regard to jurisdictional
Renal Physiol. 313, F677–F686 (2017). Author contributions claims in published maps and institutional affiliations.
151. Huang, P., Trotter, K., Boucher, R. C., Milgram, S. L. & All authors researched data for the article, made substantial
Stutts, M. J. PKA holoenzyme is functionally coupled contributions to discussions of the content, wrote the © Springer Nature Limited 2021

Nature Reviews | Nephrology

0123456789();:

You might also like