Download as pdf or txt
Download as pdf or txt
You are on page 1of 13

The FASEB Journal article fj.201601087RR. Published online November 14, 2016.

THE

JOURNAL • REVIEW • www.fasebj.org

Role of post-transcriptional regulation of mRNA


stability in renal pathophysiology: focus on chronic
kidney disease
Eva Feigerlová*,†,‡,1 and Shyue-Fang Battaglia-Hsu§
*Service d’Endocrinologie, Centre Hospitalier Universitaire de Poitiers, Pôle DUNE, Poitiers, France; †Université de Poitiers, Unité de
Formation et de Recherche Médecine Pharmacie, Poitiers, France; ‡Clinical Investigation Centre 1402, Unité 1082, INSERM, Poitiers, France;
and §Nutrition Génétique et Exposition aux Risques Environnementaux, INSERM Unité 954, Université de Lorraine et Centre Hospitalier
Régional Universitaire de Nancy, Vandœuvre les Nancy, France

ABSTRACT: Chronic kidney disease (CKD) represents an important public health problem. Its progression to end-
stage renal disease is associated with increased morbidity and mortality. The determinants of renal function
decline are not fully understood. Recent progress in the understanding of post-transcriptional regulation of
mRNA stability has helped the identification of both the trans- and cis-acting elements of mRNA as potential
markers and therapeutic targets for difficult-to-diagnose and -treat diseases, including CKDs such as diabetic
nephropathy. Human antigen R (HuR), a trans-acting element of mRNA, is an RNA binding factor (RBF) best
known for its ability to stabilize AU-rich-element-containing mRNAs. Deregulated HuR subcellular localization
or expression occurs in a wide range of renal diseases, such as metabolic acidosis, ischemia, and fibrosis. Besides
RBFs, recent evidence revealed that noncoding RNA, such as microRNA and long noncoding RNA, participates
in regulating mRNA stability and that aberrant noncoding RNA expression accounts for many pathologic re-
nal conditions. The goal of this review is to provide an overview of our current understanding of the post-
transcriptional regulation of mRNA stability in renal pathophysiology and to offer perspectives for this class of
diseases. We use examples of diverse renal diseases to illustrate different mRNA stability pathways in specific
cellular compartments and discuss the roles and impacts of both the cis- and trans-activating factors on the
regulation of mRNA stability in these diseases.—Feigerlová, E., Battaglia-Hsu, S.-F. Role of post-transcriptional
regulation of mRNA stability in renal pathophysiology: focus on chronic kidney disease. FASEB J. 31, 000–000 (2017).
www.fasebj.org
KEY WORDS: RNA binding factor • noncoding RNA • P-body • stress granule • RNA-exosome

The progression of chronic kidney disease (CKD) to matrix (ECM). The TGF-b is a master regulator of ECM
end-stage renal disease results in increased morbidity and one of the major mediators of fibrotic events (1).
and mortality. Diabetes and hypertension are major Abnormalities in several signaling pathways have been
causes of CKD in Western countries. The determinants demonstrated—among them, the renin–angiotensin
of renal function decline are not fully understood. system, reactive oxygen species, endoplasmic reticu-
Tubulointerstitial fibrosis is the hallmark of disease lum stress, and proinflammatory cytokines (2). The
progression caused by an accumulation of extracellular pathophysiological mechanisms leading to these changes

ABBREVIATIONS: z-cryst, z-crystallin/NADPH:quinone reductase; AngII, angiotensin II; ARE, AU-rich element; AUF1, ARE/poly(U)-binding/degradation
factor 1; BicC, bicaudal C protein; CCD, cortical collecting duct; CKD, chronic kidney disease; Col1a2, collagen type I a2; COX, cyclooxygenase; DN,
diabetic nephropathy, Dvl, dishevelled; ECM, extracellular matrix; GA, glutaminase; HNF, hepatocyte nuclear factor; hnRNP K, heterogeneous ribo-
nucleoprotein K; HuR, human antigen R; KSRP, K-homology splicing regulator protein; lncRNA, long noncoding RNA; miRNA, microRNA; MMP,
metalloproteinase; MR, mineralocorticoid receptor; NMD, nonsense-mediated mRNA decay; NOD, nucleotide-binding oligomerization domain-containing
protein; NOX, hydrogen peroxide-producing NADPH oxidase; NSD, nonstop mRNA decay; PAI, plasminogen activator inhibitor; P-body, processing
body; PEPCK, phosphoenolpyruvate carboxykinase; PGE2, prostaglandin E2; pHRE, pH response element; Pin, peptidyl-prolyl cis-trans isomerase; PKD,
polycystic kidney disease; PTC, premature termination codon; PTH, parathyroid hormone; RISC, RNA-induced silencing complex; RBF, RNA binding
factor; SG, stress granule; SNAT3, System N and A transporter 3; TIAR, T-cell–restricted intracellular antigen-1-related protein; Tis, tetradecanoyl phorbol
acetate–inducible sequence; tonEBP, tonicity-responsive enhancer-binding protein; Tsc, TGF-b-stimulated clone; TTP, tristetraprolin; V-ATPase, vacuolar
H+-translocating ATPase; Ybx, Y-box binding protein
1
Correspondence: Centre Hospitalier Universitaire de Poitiers, Service d’Endocrinologie, Pôle DUNE, Poitiers, France. E-mail: eva.feigerlova@
fulbrightmail.org
doi: 10.1096/fj.201601087RR

0892-6638/17/0031-0001 © FASEB 1
Downloaded from www.fasebj.org to IP 139.80.123.48. The FASEB Journal Vol., No. , pp:, November, 2016
have not been clearly established. Similarly, specific Role of lncRNA, P-body, and stress granule in
biomarkers are not currently known. Recently, post- the regulation of mRNA stability
transcriptional regulation of mRNA stability has emerged
as a major mechanism modulating gene expression, and its Experimental evidence has suggested that both miRNA
involvement in renal diseases has been evidenced. and lncRNA participate in the regulation of mRNA sta-
bility. miRNAs the short single-stranded RNAs bearing
fully or partially complimentary sequences to target genes,
POST-TRANSCRIPTIONAL REGULATION OF can be incorporated into RNA-induced silencing complex
mRNA TURNOVER (RISC) for interaction with their target mRNA, mainly at
its 39-UTR. A typical miRNA-mediated mRNA decay re-
The major mRNA surveillance pathways in mammalian quires Argonaute proteins, GW182 from P-body, CCR4:
cells include nonsense-mediated mRNA decay (NMD) and NOT deadenylase, and DCP1:DCP2 decapping complexes
nonstop mRNA decay (NSD) (3). NMD is mediated by (19, 20). The incorporation of GW182 appears to mark the
either the RNA exosome in the 39–59 direction (4) or by transcript for degradation by deadenylation and decapp-
processing bodies (P-bodies) in the 59–39 direction after an ing (21, 22). For the degradation of ARE-mRNAs mediated
initial removal of poly(A) tail by deadenylases (5). NMD through RISC, current evidence suggests that miRNA
regulates the degradation of both normal mRNAs, like within RISC recognizes the ARE sequences through the
those with alternatively spliced exons in 39-UTR, and ab- help of ARE binding proteins (AUBPs) such as triste-
normal mRNAs, like those with premature termination traprolin (TTP) (23). In fact, AUBPs act as linkers between
codon (PTC) (6). It is estimated that about one-third of hu- ARE-mRNA and degradation machinery, and the binding
man diseases are caused by PTC (7). One such example of of AUBPs to mRNA target activates deadenylation as well
renal disease is Liddle syndrome, elicited by a PTC in the as decapping, triggering its decay via both RNA exosome
SCNN1B gene (8). Unlike NMD, NSD is activated only in and Xrn1-containing decapping complex [see Carpenter
the presence of mRNA transcripts missing a stop codon. In et al. (24)]. Several AUBPs have actually been identified;
mammalian cells, eRF3-like factor Ski7 appears to detect the these include K homology splicing regulatory protein
presence of stalled RNA-ribosome complex at the 39 end of (KSRP), TTP, and butyrate response factor-1, all capable of
RNA transcripts that lack a stop codon, targeting them to interacting with poly(A) ribonuclease and RNA exosome
the RNA exosome for elimination. This pathway, however, complex (25–27). The relevance of AUBPs in kidney func-
is not well characterized in humans. tion can be appreciated in an experimental model of CKD
associated with hyperparathyroidism, where an increased
Role of cis-elements and trans-regulatory parathyroid hormone (PTH) mRNA level can be attributed
factors in mRNA stability to a decreased interaction between the 39-UTR of PTH
mRNA and KSRP and causes decreased mRNA decay via
The stability of mRNA depends on its interactions with both the RNA exosome (28, 29) (Table 1). The relevance of
cis- and trans-regulatory factors. Cis-regulatory elements lncRNA in mRNA stability has been discussed only very
reside within mRNA transcript, including not only the recently. For example, Sirt1 antisense lncRNA is found to
untranslated regions but also the coding region (9). stabilize Sirt1 transcripts via a direct interaction with Sirt1
Adenylate-uridylate-rich elements (AREs) are the best- 39-UTR, thus rescuing the suppression mediated by
studied cis-elements localized to the 39-UTR of many miRNA-34a (30). Similar base-pairing interaction be-
mRNAs. In silico analyses predicted their occurrence in tween various lncRNAs and cognate mRNA has also
5–8% of human mRNAs encoding functionally important been found to affect the stability of target transcripts
proteins (10). The presence of unique ARE sequences im- with developmental function (31, 32). Its contribution to
plies complex regulatory mechanisms involving interac- renal diseases, however, is not yet clear.
tions, even between different RNA binding factors (RBFs) P-bodies and stress granules (SGs) are cytoplasmic
for competitive binding to mRNAs (3). RBFs and non- structures containing a network of proteins necessary for
coding RNAs are examples of trans-regulatory elements. mRNA regulation and silencing (33, 34). In renal epithelial
The interactions of RBF, with its target transcripts can often cells, sequestration of mRNA-protein complex in SGs was
be modulated by both the subcellular localization and the evidenced during osmotic stress (35). P-bodies have been
post-translational modification of the RBF itself. The RBF shown to participate in the formation of planar cell polarity
interacts mainly with 39-UTR of the target transcript (11). via an RBF-dependent uncoupling of dishevelled (Dvl)
HuR, a member of the embryonic lethal abnormal vision- signaling from the canonical Wnt pathway (36) and are
like family of RNA-binding proteins (12), is among the RBFs also implicated in TGF-b-driven fibrosis in mouse kidney
most studied in renal pathophysiology. Depending on cells (37) (Table 1).
the type of cell, the pathophysiological condition, and the
presence of interacting RBFs, HuR can either promote the
stability or the decay of its mRNA target (13, 14). Non- POST-TRANSCRIPTIONAL REGULATION OF mRNA
coding RNAs such as microRNA (miRNA) and long non- STABILITY IN CKD
coding RNA (lncRNA), have recently been implicated in
diverse human diseases; the particular involvement of CKD and progressive renal fibrosis remain a therapeutic
miRNA in renal pathology has been the subject of recent challenge. Fibrotic kidney has a limited ability to recover
reviews (15–18). from such injuries as ischemia–reperfusion and is associated

2 Vol. 31 February 2017 The FASEB Journal x www.fasebj.org FEIGERLOVÁ AND BATTAGLIA-HSU
Downloaded from www.fasebj.org to IP 139.80.123.48. The FASEB Journal Vol., No. , pp:, November, 2016
TABLE 1. Post-transcriptional regulation of mRNA in renal pathophysiology and disease

RNA binding factor Target Effect site Model Mechanism Reference

Metabolic acidosis
z-cryst, AUF-1 GA 3UTR Rat renal cortex pH-responsive stabilization of 14
HuR (proximal GA mRNA through binding
convoluted of z-cryst, AUF1, and HuR to
tubule) pHRE
Not indicated SNAT3 3UTR LLC-PK1-F+-cells pH-responsive stabilization of 47
SNAT3 mRNA via binding
to pHRE
AUF-1 PEPCK 3UTR LLC-PK1-F+-9C cells pH- responsive stabilization of 43, 48
HuR PEPCK mRNA via HuR and
AUF1
Osmotic stress
TIA-1 Osmolytes SG NRK 52E Sequestration of mRNA-protein 35
complex in SGs
Tis11b MR 3UTR CCD cell line Tis11b favors degradation of Mr 52
B6D2 mice mRNA under hypertonic
conditions
ATP depletion and
ischemia/reperfusion
injury
HuR V-ATPase 3UTR Porcine proximal HuR maintains stability of V-ATPase 55
tubule, LLC-PK1 mRNAs during ATP depletion
cells
HuR Bcl-2, Hsp70 3UTR Rat kidneys HuR protects proximal tubule cells 56
LLC-PK1 cells during and after I/R through
modulation of Bcl-2 and Hsp70
expression
Inflammation and
fibrosis
HuR COX-2 3UTR Human MC AngII via activation of HuR 61
shuttling causes an increase in
COX-2 mRNA stability with
a rise in PGE2
HuR COX-2 3UTR Sprague-Dawley AngII via HuR shuttling stabilizes 62
rats kidney Pai1 and Cox2 mRNAs
PAI-1 Rat MC
HuR cyclin D1 3UTR Human MC AngII stabilizes cyclin D1 mRNA 65
via HuR
HuR MMP-9 3UTR Rat MC HuR-mediated Mmp9 mRNA 67
stabilization
TIAR MMP-13 3UTR Human MC Inhibition of MMP-13 translation 69
by TIAR bound to MMP-13 mRNA
Diabetic nephropathy
HuR NOD2 3UTR Human kidney HuR mediates hyperglycemia 70
STZ-diabetic enhanced NOD2 expression
rat kidney and mRNA stability
NRK-52E
Rat MC
Ybx1 Tsc-22 P-bodies Mouse MC TGF-b induced increase in miR-216a 37
inhibits Ybx1 leading to a release
of Tsc-22 mRNA from the P-body
with an increase in Tsc-22
translation
Ybx1 TGF-b 5UTR Human proximal TGF-b1 translation requires Ybx1
tubular binding to a high-affinity site
epithelial within the 59-UTR TGF-b-mRNA
cell line HK-2
hnRNPK VEGF 3UTR Murine proximal PKC-d positively regulates Vegf 75, 76
tubular epithelial mRNA translation through
cells activation of hnRNPK
Diabetic db/db
mice
(continued on next page)

REGULATION OF mRNA STABILITY IN RENAL DISEASE 3


Downloaded from www.fasebj.org to IP 139.80.123.48. The FASEB Journal Vol., No. , pp:, November, 2016
TABLE 1. (continued)

RNA binding factor Target Effect site Model Mechanism Reference

Polycystic kidney
disease
BicC Dvl P-bodies MDCK cell line Uncoupling of Dvl2 36
BicC1 2/2 mouse signaling from the
canonical Wnt
pathway leads to
accumulation of
BicC and Dvl2 in
P-bodies
miR-17–92 PKD1, PKD2 3UTR mIMCD3 cells Pkd2 mRNA and 86
Pkd1 mRNA
repression by
binding of miR-17
miR-17– 92 HNF-1b 3UTR mIMCD3 cells Hnf1b mRNA 86
repression by
binding
of miR-92a
miR-200 PKD1 3UTR mIMCD3 cells Repression of 87
Pkd1 mRNA by
binding of miR-200
Hyperparathyroidism
KSRP PTH 3UTR RNA HEK293 cells, Decrease in PTH 28, 29
exosome rat parathyroid mRNA levels
glands involves PTH
mRNA 39-UTR
ARE, KSRP and
the RNA exosome
Pin 1 PTH 3UTR HEK293 cells, Pin1 activation 78
rat parathyroid increases
glands KSRP–PTH
mRNA interactions,
decreasing PTH
mRNA levels

MC, mesangial cell; mIMCD3, mouse inner medullary collecting duct cells; NRK-52E, rat renal proximal tubular cells; ns, not specified; R,
reperfusion; STZ, streptozotocin

with a progressive renal function decline that ultimately tubule (Fig. 1) (39). In the mitochondria of renal tubular
leads to end-stage renal failure. The recent discoveries point cells, glutamine is first deaminated by glutaminase
to the important role of mRNA stability during the patho- (GA), then oxidized by glutamate dehydrogenase (40)
logical process. In this section, we first summarize our actual to produce ammonium ion and a-ketoglutarate (41).
knowledge of the role of post-transcriptional regulation of The latter either produces HCO32 and H+ ions or is
mRNA turnover in the kidney in pathophysiological states, oxidized by cytosolic phosphoenolpyruvate carbox-
such as chronic metabolic acidosis, osmotic stress, and ykinase (PEPCK) to generate phosphoenolpyruvate
ischemia–reperfusion injury. Further, we detail its role (42). The combined reactions also lead to the production
in diabetic nephropathy, chronic kidney fibrosis, and of HCO32 and H+ ions, which are used for the conver-
inflammation. Finally, we discuss how mRNA stability sion of phosphoenolpyruvate to glucose or for its oxi-
affects the physiology and pathology of the kidney in dation to CO2.
the context of planar cell polarity and hyperparathy- Several RBFs mediate the stabilization or the degrada-
roidism to illustrate the emerging role of miRNA on tion of certain mRNAs involved in the chronic adaptive
mRNA decay through exosome and P-bodies. metabolic response, including GA, PEPCK and SNAT3
(43, 44). RBFs such as z-cryst (45), AU-factor 1 (AUF1) (13),
Chronic metabolic acidosis and HuR (12) can all interact with the cis pH-response
element (pHRE) (direct repeat or a single copy of an 8 nt
During chronic metabolic acidosis, enzymes and ion AU-sequence: UUAAAAUA) of the GA mRNA (46) and
transporters involved in the synthesis and production modify its stability—for example, binding to z-crystallin/
of ammonium and bicarbonates are up-regulated (38). NADPH:quinone reductase (z-cryst) accelerates its decay,
Glutamine is the precursor needed for the formation and binding to HuR increases its stability. It is thought
of ammonium. Metabolic acidosis thus activates the that, in response to metabolic acidosis, z-cryst is recruited
transport of excess plasma glutamine across the baso- to the cytoplasmic SGs, and HuR is translocated into cy-
lateral membrane of the epithelium via glutamine Sys- toplasm as the result of endoplasmic reticulum stress.
tem N and A transporter 3 (SNAT3) in the renal proximal Because cytoplasmic HuR helps to stabilize GA mRNA,

4 Vol. 31 February 2017 The FASEB Journal x www.fasebj.org FEIGERLOVÁ AND BATTAGLIA-HSU
Downloaded from www.fasebj.org to IP 139.80.123.48. The FASEB Journal Vol., No. , pp:, November, 2016
Figure 1. Role of RNA-stabilizing factors in renal response to chronic metabolic acidosis. Selective stabilization of GA mRNA and
PEPCK mRNA in maintaining adaptive metabolic response. A pHRE containing a direct repeat of 8-nt AU sequences
(UUAAAUUAUUU), within the GA mRNA binds RNA-binding proteins, including z-cryst and HuR. In response to metabolic
acidosis, stabilization of the GA mRNA by z-cryst and HuR produces an increase in GA mRNA. The concurrent binding of AUF1
and HuR to the AU-rich sequences within the 39-UTR of the PEPCK mRNA mediates pH-responsive stabilization of PEPCK mRNA
(adapted from refs. 43–45, 47).

higher GA protein expression alleviates acidosis (14). On heightened SG formation (35), suggesting that the
the contrary, under physiologic pH, binding of z-cryst to shutdown of global protein synthesis (via mRNA se-
pHRE of GA mRNA promotes the degradation of GA questration in SGs) favors the eventual production and
mRNA, leading to lower but normal GA protein expres- transport of the osmolytes. Another example is the post-
sion (14). Similarly, a pHRE-dependent stabilization by transcriptional regulation of mineralocorticoid recep-
RBFs of the SNAT3 and PEPCK transcripts also helps to tor (MR) by a RNA binding and destabilizing protein,
up-regulate the cognate protein products to eventually tetradecanoyl phorbol acetate-inducible-sequence 11b
eliminate H+ (43, 47). The function of specific RBFs, such (Tis11b). Indeed, when cortical collecting duct (CCD)
as AUF1, in mRNA metabolism seems to change under cells are exposed to hypertonic milieu, Tis11b expression
some still ill-defined conditions, in that its knockdown is increased, leading to greater Mr mRNA degradation
appears to increase the basal PEPCK mRNA level and (52). Because MR is known to regulate aldosterone-
hence the protein product (48). stimulated Na+ reabsorption in the renal cortex, the post-
transcriptional regulation of MR by Tis11b can contribute to
the pathophysiology of hypertension or renal insufficiency.
Osmotic stress It is notable that besides hosting the binding sites for sev-
eral RBFs, the 39-UTR of TonEBP mRNA contains several
In response to chronic hypertonic stress, the tonicity-
miRNA recognition sites based on in silico analyses; the
responsive enhancer-binding protein (TonEBP), known
presence of such sites is a likely explanation for why the
as nuclear factor of activated T cells 5 (NFAT5), activates
overexpression of miR-200b and miR-717 significantly di-
certain enzymes and membrane proteins to synthesize
minishes the level of TonEBP mRNA and its protein (53).
and transport osmolytes needed to restore cell volume
and to maintain intracellular ionic strength (49–51). The
broader roles played by NFAT5 in kidney pathophysiol- ATP depletion and ischemia/reperfusion injury
ogy can be illustrated by the renal atrophy in Nfat5
knockout mice (50). In rat kidney epithelial NRK-52E cells, Vacuolar H+-translocating (V)-ATPases are transporters
acute hypertonic stress (.200 mOsmol/kg) caused responsible for the acidification of membrane-bound

REGULATION OF mRNA STABILITY IN RENAL DISEASE 5


Downloaded from www.fasebj.org to IP 139.80.123.48. The FASEB Journal Vol., No. , pp:, November, 2016
Figure 2. HuR-mediated mRNA stability in renal fibrosis and inflammation. Overview of AngII and cytokine-induced stabiliza-
tion of target mRNAs. Treatments with AngII or IL-1b induce PKCd-dependent phosphorylation of HuR, triggering the
nucleocytoplasmic shuttling of HuR to facilitate HuR binding to its AU-rich target mRNAs in the cytoplasm. This results in the
stabilization of COX-2 mRNA, PAI-1 mRNA, CCND1 mRNA, and MMP-9 mRNA and the consequent higher protein syntheses that
eventually lead to more inflammation and ECM accumulation (adapted from refs. 61, 62, 67).

intracellular compartments (54). As ATP depletion acti- Vasodilatory prostaglandins participate in regulation of
vates the nucleocytoplasmic shuttling of HuR and in- renal blood flow, glomerular filtration rate (57), and so-
creases HuR mRNA translatability, ATP depletion can dium reabsorption at the thick ascending limb of the loop
increase V-ATPase expression through HuR-mediated of Henley (58) and are essential to maintain proper kidney
mRNA stabilization (55). In energy-depleted LLC-PK1 function. Cyclooxygenase (COX)- 2 is a prerequisite for the
cells, HuR suppression via RNA interference leads to conversion of arachidonic acid to prostaglandins; its ac-
reduced expression of antiapoptotic proteins Bcl-2 and tivity is needed for renal protection against vasoactive
Hsp70, and a higher rate of cellular apoptosis, indicat- Angiotensin (Ang)II (59). The expression of COX-2 mostly
ing that the cellular stress caused by ATP insufficiency occurs through transcriptional activation induced by
in kidney cells can be eased by the presence of HuR. The proinflammatory cytokines and tumor promoters (60). In
protective function of HuR was also observed in rat renal mesangial cells, PKC responds to vasoactive factors,
proximal tubular cells exposed to ischemia–reperfusion and its activation has been shown to control HuR export
events, given that these cellular insults lead to both (Fig. 2). Doller et al. (61) demonstrated that in human
higher HuR expression and HuR export into the cyto- mesangial cells, AngII can trigger an intracellular prosta-
plasm to stabilize the mRNAs of antistress genes (56). glandin E2 (PGE2) increase via HuR-mediated COX2
These pieces of evidence support the role of HuR in mRNA stabilization and that the enhanced HuR binding
protecting renal epithelia from injury. to COX-2 ARE was the result of an increased nucleocyto-
plasmic HuR shuttling consequent to PKC-d mediated
Diabetic nephropathy, inflammation, phosphorylation. Moreover, in AngII-treated rats, greater
and fibrosis nucleocytoplasmic HuR shuttling as well as stronger HuR
binding to Pai-1 mRNA and Cox2 mRNA were observed in
Early events involved in renal fibrosis are related to the cytoplasmic fractions of renal homogenate, confirming
proliferation of mesangial cells and ECM accumulation. the participation in vivo of HuR in the AngII-induced

6 Vol. 31 February 2017 The FASEB Journal x www.fasebj.org FEIGERLOVÁ AND BATTAGLIA-HSU
Downloaded from www.fasebj.org to IP 139.80.123.48. The FASEB Journal Vol., No. , pp:, November, 2016
Figure 3. Renal damage and fibrosis in diabetic conditions. In rat glomerular mesangial cells, hyperglycemic conditions can
enhance the production of ROS via the NOX4 signaling pathway and help to stabilize the NOD2 mRNA via HuR binding to its
39-UTR. In another model, treatment with TGF-b under diabetic conditions leads to increased miR-216a level, which, in turn,
inhibits its target, Ybx1, an RNA binding protein. In parallel, miR-216a enhances the expressions of Tsc-22 and Col1a2. Ybx1
colocalizes with P-body and forms a ribonucleoprotein complex with Tsc-22 mRNA (adapted from refs. 37, 70, 101).

expression of the proinflammatory and profibrotic serine Human MMP-13 is also known for its involvement in in-
protease inhibitor renal plasminogen activator inhibitor-1 flammation and angiogenesis.
(PAI-1) and COX-2 (62). In addition, AngII was shown to Our understanding of the role of post-transcriptional
promote a fibrotic process (63) through the overexpression RNA regulation in DN was recently advanced by several
of the cell cycle regulator cyclinD1, which is known to studies. For example, Shang and collaborators (70) found
shorten the progression of the G1 phase (64). CCND1 that an abnormally high HuR expression was present in
mRNA contains specific AREs within its 39-UTR. The in- the kidney biopsies of patients with DN, and it appeared
creased cyclin D1 expression in mesangial cells after AngII to correlate with the level of nucleotide-binding oligo-
treatment observed by Che et al. (65) is thus the likely merization domain-containing protein (NOD)-2. Because
result of increased HuR binding to the cytoplasmic hyperglycemic conditions have been associated with
CCND1 mRNA; such an increase can lead to abnormal enhanced production of ROS via hydrogen peroxide-
proliferation of the mesangial cells. producing NADPH oxidase (NOX4) signaling in rat
Further insights into the post-transcriptional regulation glomerular mesangial cells, it has been proposed that
of ECM have been provided by studies of the matrix higher NOD2 mRNA stability is mediated by higher
metalloproteinases (MMPs). MMP-9 has been implicated HuR-dependent NOD2 mRNA stabilization as a result
in both the remodeling of ECM and the progression of of the increased presence of cytoplasmic HuR conse-
chronic inflammatory renal diseases (66). In response to quent to ROS-mediated cell signaling (Fig. 3). The in-
inflammatory cytokines such as IL-1b, mesangial cells vestigators also evidenced that the expression and the
treated with ATP displayed increased HuR nucleocyto- translocation of HuR, and the stability of NOD2 mRNA
plasmic shuttling and MMP-9 expression resulting from were related to NADPH oxidase-mediated redox sig-
HuR-mediated mRNA stabilization (67). In contrary, naling. In parallel, an amelioration of renal injury and a
neutralization of HuR after treatment with NO reduced reduction of NOD2 expression were observed after
the stability of MMP-9 mRNA (68). Moreover, an alter- HuR gene silencing in diabetic rats. Kato et al. (37)
natively spliced form of T-cell–restricted intracellular suggested that post-transcriptional regulation partici-
antigen-1-related protein (TIAR), an RBF, has been shown pates in TGF-b-induced renal fibrosis in DN. They
to inhibit the translation of human MMP-13 mRNA showed that TGF-b treatment leads to miR-216a in-
through its binding to the 39-UTR of MMP-13 mRNA (69). crease in mouse mesangial cells and that this increase in

REGULATION OF mRNA STABILITY IN RENAL DISEASE 7


Downloaded from www.fasebj.org to IP 139.80.123.48. The FASEB Journal Vol., No. , pp:, November, 2016
miR-216a, on the one hand, inhibits the RNA target replace the binding of KSRP to PTH mRNA ARE, leading
binding of a RBF called Y-box binding protein 1 (Ybx1) to increased PTH mRNA stability and hence high PTH
(37, 71, 72), and, on the other hand, enhanced the ex- protein levels. Furthermore, the peptidyl-prolyl cis-trans
pression of TGF-b-stimulated clone 22 (Tsc-22) and of isomerase (Pin)-1 was demonstrated to decrease PTH
collagen type I a-2 (Col1a2). Mechanistically, it turned mRNA by enhancing KSRP–PTH mRNA interactions (78).
out that Ybx1 colocalizes with P-body and forms a ri- On the contrary, when Pin-1 was not active, KSRP was
bonucleoprotein complex with Tsc-22 mRNA in the phosphorylated, which led to its inactivation and facilitated
mouse mesangial cells. The TGF-b-induced increase in the binding of AUF1 to the 39-UTR of PTH mRNA, resulting
miR216 thus causes the release of Tsc-22 mRNA from in increased PTH mRNA stability.
the P-body and promotes its translation by inhibiting
the binding of Ybx1 to Tsc-22 mRNA. Last, in a series of
studies, the role of VEGF and its regulation in diabetes Polycystic kidney disease and planar
was examined. It was found that VEGF induces hy- cell polarity
pertrophy in murine proximal tubular epithelial cells
and that its expression increases in the renal cortex of The primary cilia of the renal epithelial cells are the me-
diabetic mice (73). In addition, the application of AngII chanical sensors, transducing the urinary flow information
in the proximal tubular epithelial cells leads to enhanced into biologic signal via the activation of Ca2+ channels (79).
Vegf synthesis via heterogeneous ribonucleoprotein Kidney cilia express regulators of planar cell polarity,
(hnRNP K)-mediated mRNA stabilization (74, 75). which are crucial for well-oriented renal cell divisions and
Mechanistically, the activation of tyrosine/protein ki- elongation of tubules (80, 81). The propagation of planar
nase c/src by AngII plays a crucial role in the activation cell polarity between cells depends on several principal
of hnRNP K binding to Vegf mRNA, mediated through proteins, among them the Dvl-1 protein (Drosophila)/Dvl
a PKC-d-dependent hnRNP K Ser302 phosphorylation (mammals) (82). The loss of planar cell polarity is associ-
(75, 76). Likewise, VEGF expression was found to be ated with polycystic kidney disease (PKD) (80). Recently,
increased in db/db diabetic mice as a result of a greater the observations of Maisonneuve et al. (36) have shed new
Vegf mRNA stability made possible by hnRNP K binding insights into the physiopathology of PKD, revealing that
to Vegf mRNA (76). bicaudal C (BicC), a conserved RNA-binding protein, can
uncouple Dvl2 signaling from the Wnt pathway. The Wnt
pathway controls the planar cell polarity and the activa-
Hyperparathyroidism in CKD tion of cell proliferation via b-catenin signaling. The tar-
geted inactivation of BicC produced abnormalities in the
In CKD, low calcium and high phosphate levels lead to planar alignment of motile cilia, with a consequent accu-
hyperparathyroidism characterized by increased PTH mulation of BicC in P-bodies. Evidence further suggested
gene expression (77). Nechama et al. (28, 29) demonstrated that miRNAs are involved in PKD. For example, in
that PTH mRNA is more stable in parathyroid glands from BicC12/2 mice, an increased Pkd2 gene expression (the
CKD or calcium-depleted animals comparing to para- PC2 protein) was associated with the presence of bi-
thyroid glands from control and phosphorus-depleted lateral polycystic kidneys (83, 84). Additional experi-
rats. Low serum phosphate was shown to facilitate the mentation revealed that BicC1 specifically up-regulates
interaction between PTH mRNA and KSRP at 39-UTR of PC2 expression through blocking the mRNA destabili-
the PTH mRNA. The binding of KSRP appeared to pro- zation caused by the binding of miR-17 to the 39-UTR of
mote the binding of RNA exosome to PTH mRNA in Pkd2 mRNA (85). Moreover, in renal epithelial cells,
parathyroid extracts, leading to a decrease in PTH mRNA miR-17 ;92 cluster and miR-200 have been shown to
stability and thus to reduced PTH protein level. In the down-regulate the expression of the key cystic genes
same animal model, in parathyroid glands from calcium- Pkd1, Pkd2, and Pkhd1 via the repression of HNF-1b (86,
depleted or CKD rats, AUF1 and N-ras appeared to 87). In fact, Hnf-1b directly regulates the transcription of

TABLE 2. Pharmaceutical agents modulating mRNA stability

Pharmaceutical
agent/molecule Mechanism of action Effect Model Reference

DHTS Inhibition of HuR–mRNA Down-regulation of TNFa Human breast cancer cells 98


interaction expression
Indoline Inhibition of nuclear–cytoplasmic Destabilization of HIF-1a Human cancer cells and 99
sulfonamide HuR shuttling mRNA normal endothelial cells
MPT0B098a
Latrunculin Ab Inhibition of nuclear–cytoplasmic Decreased stability of HuR Human hepatocellular 100
Blebbistatinc HuR shuttling targets COX-2, cyclin A, carcinoma cells
cyclin D mRNAs

Model represents clinical indication. DHTS, 15,16-dihydrotanshinone-I; HIF-1a, hypoxia-inducible factor-1a. aInhibitor of microtubule
polymerization. bActin inhibitor. c Myosin II inhibitor.

8 Vol. 31 February 2017 The FASEB Journal x www.fasebj.org FEIGERLOVÁ AND BATTAGLIA-HSU
Downloaded from www.fasebj.org to IP 139.80.123.48. The FASEB Journal Vol., No. , pp:, November, 2016
Pkhd1, which plays a crucial role in the development of affected—in particular, renal diseases—and examine
autosomal recessive PKD (88). their interacting noncoding RNAs by using either RNA
immunoprecipitation or microarray technology (94).
Equally, one may also search, at either the cellular or tissue
CONCLUSIONS AND PERSPECTIVES level, for the involvement of noncoding RNAs using
comparative profiling approaches. Take, for example, the
Recent data in the literature suggest that post- RNA-binding factor BicC, which has been shown to reg-
transcriptional regulation of mRNA stability plays im- ulate the mRNA stability of Wnk1, V-ATPase B1 and
portant roles in CKD. It underlies the capacity of renal adenylate cyclase6 (95–97), it is not known whether non-
cells to form transient protein assemblies like stress coding RNAs can alter the expression of BicC via modu-
granules SGs and P-bodies, which functionally interact lating its mRNA stability. If so, can these noncoding RNAs
with both translational and degradation machinery to be used as targets for treatment? Also, it is known that in
modulate the expression of genes contributing to disease the 39-UTR of certain key pathogenic genes, such as PKD1
phenotypes. Given that the presently available thera- and TonEBP, there are conserved miRNA binding sites for
peutic options are of rather limited efficacy, potential in- miR-200 (87) for PKD1, and miR-200b and miR-717 for
terventions designed to modulate the metabolism of TonEBP (53); however, the involvement of these miRNAs
relevant RNA species through influencing their decay in the stability of the PKD1 and TonEBP mRNA remains
may offer new opportunities for the treatment of CKD. to be answered and their potential as therapeutic targets
Nonetheless, the current development for such ap- investigated.
proaches is still in its infancy, despite the many exciting Altogether, deregulated mRNA stability appears to be a
experimental results that are being reported. For example, significant mechanism controlling gene expression in renal
the RNA interference approach has been in the forefront disease. It enables rapid and specialized local control of
of such strategies and was successfully used for treating gene expression in the special cellular compartments and
small experimental animals with diseases such as in- opens new diagnostic and therapeutic possibilities.
flammatory bowel disease (89). Small molecules have also
been found to modulate the function of RNA binding
proteins such as HuR (90) and RNA terminal uridylyl ACKNOWLEDGMENTS
transferase (91) for potential treatment of diseases ranging
The authors thank Dr. Battaglia (Laboratoire Interdisci-
from cancer to infectious disease. These findings represent plinaire des Environnements Continentaux, UMR 7360
the broader research efforts devoted to identifying po- CNRS/Université de Lorraine) for critical reading of the
tential targets for post-transcriptional modification of manuscript.
mRNA stability. Such progress is particularly advanced
for cancer therapy (92). The application of chemical in-
hibitors to disrupt cytoskeleton dependent RBF-mRNA AUTHOR CONTRIBUTIONS
transport (93) represents a potential new strategy for
controlling renal fibrosis, given that (1) the components of E. Feigerlova wrote the manuscript, and S.-F Battaglia-Hsu
cytoskeleton are implicated in ECM formation, and (2) the participated in writing in all sections.
actin/cytoskeleton-dependent HuR transport has been
shown to regulate hypertension-induced fibrosis and in-
flammation through stabilization of COX-2 mRNA (61). REFERENCES
Examples of recent advances in this field are summarized
1. Schnaper, H. W., Hayashida, T., Hubchak, S. C., and Poncelet, A. C.
in Table 2. It is our hope that the lessons learned from the (2003) TGF-beta signal transduction and mesangial cell fibrogenesis.
treatment strategies aimed at altering RNA stability in the Am. J. Physiol. Renal Physiol. 284, F243–F252
field of oncology can be applied in the near future to CKD, 2. Forbes, J. M., and Cooper, M. E. (2013) Mechanisms of diabetic
complications. Physiol. Rev. 93, 137–188
given that similar mechanisms participate in modulating 3. Wu, X., and Brewer, G. (2012) The regulation of mRNA stability in
the expression of renal disease–causing genes, such as the mammalian cells: 2.0. Gene 500, 10–21
TGF-b cytokine. 4. Schmid, M., and Jensen, T. H. (2008) The exosome: a multipurpose
It is worth noting that the involvement of noncoding RNA-decay machine. Trends Biochem. Sci. 33, 501–510
5. Garneau, N. L., Wilusz, J., and Wilusz, C. J. (2007) The highways and
RNA in the regulation of mRNA stability of the disease byways of mRNA decay. Nat. Rev. Mol. Cell Biol. 8, 113–126
genes remains to be explored in the field of renal disease. 6. Mendell, J. T., Sharifi, N. A., Meyers, J. L., Martinez-Murillo, F., and
For instance, in the experimental model of CKD associated Dietz, H. C. (2004) Nonsense surveillance regulates expression of
diverse classes of mammalian transcripts and mutes genomic noise.
with hyperparathyroidism, the altered intracellular PTH Nat. Genet. 36, 1073–1078
mRNA level was proposed to be caused by disrupted in- 7. Kuzmiak, H. A., and Maquat, L. E. (2006) Applying nonsense-
teractions of the 39-UTR of PTH mRNA with both KSRP mediated mRNA decay research to the clinic: progress and chal-
and RNA exosome (28, 29). Are there any noncoding lenges. Trends Mol. Med. 12, 306–316
8. Shimkets, R. A., Warnock, D. G., Bositis, C. M., Nelson-Williams, C.,
RNAs that participate in the decay of PTH mRNA? If so, Hansson, J. H., Schambelan, M., Gill, J. R., Jr., Ulick, S., Milora, R. V.,
what are the noncoding RNAs involved in regulating and Findling, J. W., et al. (1994) Liddle’s syndrome: heritable human
the stability of the particular gene transcripts relevant to hypertension caused by mutations in the beta subunit of the
individual kidney pathology? To fully appreciate the in- epithelial sodium channel. Cell 79, 407–414
9. Grosset, C., Chen, C. Y., Xu, N., Sonenberg, N., Jacquemin-Sablon,
volvement of specific noncoding RNAs in kidney dys- H., and Shyu, A. B. (2000) A mechanism for translationally
function, one can begin with the identification of genes coupled mRNA turnover: interaction between the poly(A) tail

REGULATION OF mRNA STABILITY IN RENAL DISEASE 9


Downloaded from www.fasebj.org to IP 139.80.123.48. The FASEB Journal Vol., No. , pp:, November, 2016
and a c-fos RNA coding determinant via a protein complex. Cell 34. Stoecklin, G., and Kedersha, N. (2013) Relationship of GW/P-bodies
103, 29–40 with stress granules. Adv. Exp. Med. Biol. 768, 197–211
10. Bakheet, T., Williams, B. R., and Khabar,K. S. (2003) ARED 2.0: an update 35. Bounedjah, O., Hamon, L., Savarin, P., Desforges, B., Curmi, P. A.,
of AU-rich element mRNA database. Nucleic Acids Res. 31, 421–423 and Pastré, D. (2012) Macromolecular crowding regulates assembly
11. Lovci, M. T., Bengtson, M. H., and Massirer, K. B. (2016) Post- of mRNA stress granules after osmotic stress: new role for compatible
translational modifications and RNA-binding proteins. Adv. Exp. osmolytes. J. Biol. Chem. 287, 2446–2458
Med. Biol. 907, 297–317 36. Maisonneuve, C., Guilleret, I., Vick, P., Weber, T., Andre, P., Beyer,
12. Cherry, J., Karschner, V., Jones, H., and Pekala, P. H. (2006) HuR, an T., Blum, M., and Constam, D. B. (2009) Bicaudal C, a novel
RNA-binding protein, involved in the control of cellular differenti- regulator of Dvl signaling abutting RNA-processing bodies, controls
ation. In Vivo 20, 17–23 cilia orientation and leftward flow. Development 136, 3019–3030
13. White, E. J., Brewer, G., and Wilson, G. M. (2013) Post-transcriptional 37. Kato, M., Wang, L., Putta, S., Wang, M., Yuan, H., Sun, G., Lanting, L.,
control of gene expression by AUF1: mechanisms, physiological Todorov, I., Rossi, J. J., and Natarajan, R. (2010) Post-transcriptional
targets, and regulation. Biochim. Biophys. Acta 1829, 680–688 up-regulation of Tsc-22 by Ybx1, a target of miR-216a, mediates TGF-
14. Ibrahim, H., Lee, Y. J., and Curthoys, N. P. (2008) Renal response to beta-induced collagen expression in kidney cells. J. Biol. Chem. 285,
metabolic acidosis: role of mRNA stabilization. Kidney Int. 73, 11–18 34004–34015
15. Trionfini, P., Benigni, A., and Remuzzi, G. (2015) MicroRNAs in 38. Curthoys, N. P., and Gstraunthaler, G. (2001) Mechanism of
kidney physiology and disease. Nat. Rev. Nephrol. 11, 23–33 increased renal gene expression during metabolic acidosis. Am. J.
16. Wei, Q., Mi, Q. S., and Dong, Z. (2013) The regulation and function Physiol. Renal Physiol. 281, F381–F390
of microRNAs in kidney diseases. IUBMB Life 65, 602–614 39. Solbu, T. T., Boulland, J. L., Zahid, W., Lyamouri Bredahl, M. K.,
17. Bhatt, K., Kato, M., and Natarajan, R. (2016) Mini-review: emerging Amiry-Moghaddam, M., Storm-Mathisen, J., Roberg, B. A., and
roles of microRNAs in the pathophysiology of renal diseases. Am. J. Chaudhry, F. A. (2005) Induction and targeting of the glutamine
Physiol. Renal Physiol. 310, F109–F118 transporter SN1 to the basolateral membranes of cortical kidney
18. Lorenzen, J. M., and Thum, T. (2016) Long noncoding RNAs tubule cells during chronic metabolic acidosis suggest a role in pH
in kidney and cardiovascular diseases. Nat. Rev. Nephrol. 12, regulation. J. Am. Soc. Nephrol. 16, 869–877
360–373 40. Sastrasinh, S., and Sastrasinh, M. (1989) Glutamine transport in
19. Filipowicz, W., Jaskiewicz, L., Kolb, F. A., and Pillai, R. S. (2005) Post- submitochondrial particles. Am. J. Physiol. 257, F1050–F1058
transcriptional gene silencing by siRNAs and miRNAs. Curr. Opin. 41. Gerich, J. E., Meyer, C., Woerle, H. J., and Stumvoll, M. (2001) Renal
Struct. Biol. 15, 331–341 gluconeogenesis: its importance in human glucose homeostasis.
20. Behm-Ansmant, I., Rehwinkel, J., Doerks, T., Stark, A., Bork, P., and Diabetes Care 24, 382–391
Izaurralde, E. (2006) mRNA degradation by miRNAs and GW182 42. Yang, J., Kalhan, S. C., and Hanson, R. W. (2009) What is the
requires both CCR4:NOT deadenylase and DCP1:DCP2 decapping metabolic role of phosphoenolpyruvate carboxykinase? J. Biol. Chem.
complexes. Genes Dev. 20, 1885–1898 284, 27025–27029
21. Ding, L., and Han, M. (2007) GW182 family proteins are crucial for 43. Gummadi, L., Taylor, L., and Curthoys, N. P. (2012) Concurrent
microRNA-mediated gene silencing. Trends Cell Biol. 17, 411–416 binding and modifications of AUF1 and HuR mediate the pH-
22. Eulalio, A., Huntzinger, E., and Izaurralde, E. (2008) GW182 responsive stabilization of phosphoenolpyruvate carboxykinase
interaction with Argonaute is essential for miRNA-mediated trans- mRNA in kidney cells. Am. J. Physiol. Renal Physiol. 303, F1545–F1554
lational repression and mRNA decay. Nat. Struct. Mol. Biol. 15, 44. Hansen, W. R., Barsic-Tress, N., Taylor, L., and Curthoys, N. P.
346–353 (1996) The 39-nontranslated region of rat renal glutaminase mRNA
23. Jing, Q., Huang, S., Guth, S., Zarubin, T., Motoyama, A., Chen, J., contains a pH-responsive stability element. Am. J. Physiol. 271,
Di Padova, F., Lin, S. C., Gram, H., and Han, J. (2005) Involvement of F126–F131
microRNA in AU-rich element-mediated mRNA instability. Cell 120, 45. Tang, A., and Curthoys, N. P. (2001) Identification of zeta-crystallin/
623–634 NADPH:quinone reductase as a renal glutaminase mRNA pH re-
24. Carpenter, S., Ricci, E. P., Mercier, B. C., Moore, M. J., and sponse element-binding protein. J. Biol. Chem. 276, 21375–21380
Fitzgerald, K. A. (2014) Post-transcriptional regulation of gene ex- 46. Laterza, O. F., and Curthoys, N. P. (2000) Specificity and functional
pression in innate immunity. Nat. Rev. Immunol. 14, 361–376 analysis of the pH-responsive element within renal glutaminase
25. Tao, X., and Gao, G. (2015) Tristetraprolin recruits eukaryotic mRNA. Am. J. Physiol. Renal Physiol. 278, F970–F977
initiation factor 4E2 to repress translation of AU-rich element- 47. Balkrishna, S., Bröer, A., Welford, S. M., Hatzoglou, M., and Bröer, S.
containing mRNAs. Mol. Cell. Biol. 35, 3921–3932 (2014) Expression of glutamine transporter Slc38a3 (SNAT3)
26. Lykke-Andersen, J., and Wagner, E. (2005) Recruitment and during acidosis is mediated by a different mechanism than tissue-
activation of mRNA decay enzymes by two ARE-mediated decay ac- specific expression. Cell. Physiol. Biochem. 33, 1591–1606
tivation domains in the proteins TTP and BRF-1. Genes Dev. 19, 48. Mufti, J., Hajarnis, S., Shepardson, K., Gummadi, L., Taylor, L.,
351–361 and Curthoys, N. P. (2011) Role of AUF1 and HuR in the pH-
27. Gherzi, R., Lee, K. Y., Briata, P., Wegmüller, D., Moroni, C., Karin, responsive stabilization of phosphoenolpyruvate carboxykinase
M., and Chen, C. Y. (2004) A KH domain RNA binding protein, mRNA in LLC-PK₁-F⁺ cells. Am. J. Physiol. Renal Physiol. 301,
KSRP, promotes ARE-directed mRNA turnover by recruiting the F1066–F1077
degradation machinery. Mol. Cell 14, 571–583 49. Ito, T., Fujio, Y., Hirata, M., Takatani, T., Matsuda, T., Muraoka,
28. Nechama, M., Ben-Dov, I. Z., Briata, P., Gherzi, R., and Naveh-Many, S., Takahashi, K., and Azuma, J. (2004) Expression of taurine
T. (2008) The mRNA decay promoting factor K-homology splicing transporter is regulated through the TonE (tonicity-responsive
regulator protein post-transcriptionally determines parathyroid element)/TonEBP (TonE-binding protein) pathway and con-
hormone mRNA levels. FASEB J. 22, 3458–3468 tributes to cytoprotection in HepG2 cells. Biochem. J. 382,
29. Nechama, M., Peng, Y., Bell, O., Briata, P., Gherzi, R., Schoenberg, 177–182
D. R., and Naveh-Many, T. (2009) KSRP-PMR1-exosome association 50. López-Rodrı́guez, C., Antos, C. L., Shelton, J. M., Richardson, J. A.,
determines parathyroid hormone mRNA levels and stability in Lin, F., Novobrantseva, T. I., Bronson, R. T., Igarashi, P., Rao, A., and
transfected cells. BMC Cell Biol. 10, 70 Olson, E. N. (2004) Loss of NFAT5 results in renal atrophy and lack
30. Wang, G. Q., Wang, Y., Xiong, Y., Chen, X. C., Ma, M. L., Cai, R., Gao, of tonicity-responsive gene expression. Proc. Natl. Acad. Sci. USA 101,
Y., Sun, Y. M., Yang, G. S., and Pang, W. J. (2016) Sirt1 AS lncRNA 2392–2397
interacts with its mRNA to inhibit muscle formation by attenuating 51. Burg, M. B., Ferraris, J. D., and Dmitrieva, N. I. (2007) Cellular
function of miR-34a. Sci. Rep. 6, 21865 response to hyperosmotic stresses. Physiol. Rev. 87, 1441–1474
31. Fatica, A., and Bozzoni, I. (2014) Long non-coding RNAs: new 52. Viengchareun, S., Kamenicky, P., Teixeira, M., Butlen, D., Meduri,
players in cell differentiation and development. Nat. Rev. Genet. 15, G., Blanchard-Gutton, N., Kurschat, C., Lanel, A., Martinerie, L.,
7–21 Sztal-Mazer, S., Blot-Chabaud, M., Ferrary, E., Cherradi, N., and
32. Angrand, P. O., Vennin, C., Le Bourhis, X., and Adriaenssens, E. Lombès, M. (2009) Osmotic stress regulates mineralocorticoid
(2015) The role of long non-coding RNAs in genome formatting receptor expression in a novel aldosterone-sensitive cortical col-
and expression. Front. Genet. 6, 165 lecting duct cell line. Mol. Endocrinol. 23, 1948–1962
33. Sen, G. L., and Blau, H. M. (2005) Argonaute 2/RISC resides in sites 53. Huang, W., Liu, H., Wang, T., Zhang, T., Kuang, J., Luo, Y., Chung,
of mammalian mRNA decay known as cytoplasmic bodies. Nat. Cell S. S., Yuan, L., and Yang, J. Y. (2011) Tonicity-responsive microRNAs
Biol. 7, 633–636 contribute to the maximal induction of osmoregulatory transcription

10 Vol. 31 February 2017 The FASEB Journal x www.fasebj.org FEIGERLOVÁ AND BATTAGLIA-HSU
Downloaded from www.fasebj.org to IP 139.80.123.48. The FASEB Journal Vol., No. , pp:, November, 2016
factor OREBP in response to high-NaCl hypertonicity. Nucleic Acids 73. Senthil, D., Choudhury, G. G., McLaurin, C., and Kasinath, B. S.
Res. 39, 475–485 (2003) Vascular endothelial growth factor induces protein synthesis
54. Forgac, M. (1999) Structure and properties of the vacuolar in renal epithelial cells: a potential role in diabetic nephropathy.
(H+)-ATPases. J. Biol. Chem. 274, 12951–12954 Kidney Int. 64, 468–479
55. Jeyaraj, S., Dakhlallah, D., Hill, S. R., and Lee, B. S. (2005) HuR 74. Paziewska, A., Wyrwicz, L. S., Bujnicki, J. M., Bomsztyk, K., and
stabilizes vacuolar H+-translocating ATPase mRNA during cellular Ostrowski, J. (2004) Cooperative binding of the hnRNP K three KH
energy depletion. J. Biol. Chem. 280, 37957–37964 domains to mRNA targets. FEBS Lett. 577, 134–140
56. Ayupova, D. A., Singh, M., Leonard, E. C., Basile, D. P., and Lee, B. S. 75. Feliers, D., Lee, M. J., Ghosh-Choudhury, G., Bomsztyk, K., and
(2009) Expression of the RNA-stabilizing protein HuR in ischemia- Kasinath, B. S. (2007) Heterogeneous nuclear ribonucleoprotein K
reperfusion injury of rat kidney. Am. J. Physiol. Renal Physiol. 297, contributes to angiotensin II stimulation of vascular endothelial
F95–F105 growth factor mRNA translation. Am. J. Physiol. Renal Physiol. 293,
57. Navar, L. G., Inscho, E. W., Majid, S. A., Imig, J. D., Harrison-Bernard, F607–F615
L. M., and Mitchell, K. D. (1996) Paracrine regulation of the renal 76. Sataranatarajan, K., Lee, M. J., Mariappan, M. M., and Feliers, D.
microcirculation. Physiol. Rev. 76, 425–536 (2008) PKCdelta regulates the stimulation of vascular endothelial
58. Romero, J. C., Bentley, M. D., Vanhoutte, P. M., and Knox, F. G. factor mRNA translation by angiotensin II through hnRNP K. Cell.
(1989) Intrarenal mechanisms that regulate sodium excretion in Signal. 20, 969–977
relationship to changes in blood pressure. Mayo Clin. Proc. 64, 77. Kidney Disease: Improving Global Outcomes (KDIGO) CKD-MBD
1406–1424 Work Group. (2009) KDIGO clinical practice guideline for the di-
59. Jaimes, E. A., Tian, R. X., Pearse, D., and Raij, L. (2005) Up- agnosis, evaluation, prevention, and treatment of chronic kidney
regulation of glomerular COX-2 by angiotensin II: role of reactive disease-mineral and bone disorder (CKD-MBD). Kidney Int. Suppl.
oxygen species. Kidney Int. 68, 2143–2153 113 (Suppl), S1–S130
60. Pfeilschifter, J., and Mühl, H. (1990) Interleukin 1 and tumor 78. Nechama, M., Uchida, T., Mor Yosef-Levi, I., Silver, J., and
necrosis factor potentiate angiotensin II- and calcium ionophore- Naveh-Many, T. (2009) The peptidyl-prolyl isomerase Pin1 de-
stimulated prostaglandin E2 synthesis in rat renal mesangial cells. termines parathyroid hormone mRNA levels and stability in rat
Biochem. Biophys. Res. Commun. 169, 585–595 models of secondary hyperparathyroidism. J. Clin. Invest. 119,
61. Doller, A., Akool, S., Huwiler, A., Müller, R., Radeke, H. H., 3102–3114
Pfeilschifter, J., and Eberhardt, W. (2008) Posttranslational 79. Praetorius, H. A., and Spring, K. R. (2003) The renal cell primary
modification of the AU-rich element binding protein HuR by pro- cilium functions as a flow sensor. Curr. Opin. Nephrol. Hypertens. 12,
tein kinase Cdelta elicits angiotensin II-induced stabilization and 517–520
nuclear export of cyclooxygenase 2 mRNA. Mol. Cell. Biol. 28, 80. Saburi, S., Hester, I., Fischer, E., Pontoglio, M., Eremina, V., Gessler,
2608–2625 M., Quaggin, S. E., Harrison, R., Mount, R., and McNeill, H. (2008)
62. Doller, A., Gauer, S., Sobkowiak, E., Geiger, H., Pfeilschifter, J., and Loss of Fat4 disrupts PCP signaling and oriented cell division and
Eberhardt, W. (2009) Angiotensin II induces renal plasminogen leads to cystic kidney disease. Nat. Genet. 40, 1010–1015
activator inhibitor-1 and cyclooxygenase-2 expression post- 81. Luyten, A., Su, X., Gondela, S., Chen, Y., Rompani, S., Takakura, A.,
transcriptionally via activation of the mRNA-stabilizing factor and Zhou, J. (2010) Aberrant regulation of planar cell polarity in
human-antigen R. Am. J. Pathol. 174, 1252–1263 polycystic kidney disease. J. Am. Soc. Nephrol. 21, 1521–1532
63. Boffa, J. J., Lu, Y., Placier, S., Stefanski, A., Dussaule, J. C., and 82. Devenport, D. (2014) The cell biology of planar cell polarity. J. Cell
Chatziantoniou, C. (2003) Regression of renal vascular and Biol. 207, 171–179
glomerular fibrosis: role of angiotensin II receptor antagonism 83. Zhang, P., Luo, Y., Chasan, B., González-Perrett, S., Montalbetti, N.,
and matrix metalloproteinases. J. Am. Soc. Nephrol. 14, Timpanaro, G. A., Cantero, Mdel. R., Ramos, A. J., Goldmann, W. H.,
1132–1144 Zhou, J., and Cantiello, H. F. (2009) The multimeric structure of
64. Lang, S., Hartner, A., Sterzel, R. B., and Schöcklmann, H. O. (2000) polycystin-2 (TRPP2): structural-functional correlates of homo- and
Requirement of cyclin D1 in mesangial cell mitogenesis. J. Am. Soc. hetero-multimers with TRPC1. Hum. Mol. Genet. 18, 1238–1251
Nephrol. 11, 1398–1408 84. Wessely, O., Tran, U., Zakin, L., and De Robertis, E. M. (2001)
65. Che, Y., Yi, L., Akhtar, J., Bing, C., Ruiyu, Z., Qiang, W., and Rong, W. Identification and expression of the mammalian homologue of
(2014) AngiotensinII induces HuR shuttling by post-transcriptional Bicaudal-C. Mech. Dev. 101, 267–270
regulated CyclinD1 in human mesangial cells. Mol. Biol. Rep. 41, 85. Tran, U., Zakin, L., Schweickert, A., Agrawal, R., Döger, R., Blum, M.,
1141–1150 De Robertis, E. M., and Wessely, O. (2010) The RNA-binding protein
66. Lenz, O., Elliot, S. J., and Stetler-Stevenson, W. G. (2000) Matrix bicaudal C regulates polycystin 2 in the kidney by antagonizing miR-
metalloproteinases in renal development and disease. J. Am. Soc. 17 activity. Development 137, 1107–1116
Nephrol. 11, 574–581 86. Patel, V., Williams, D., Hajarnis, S., Hunter, R., Pontoglio, M., Somlo,
67. Huwiler, A., Akool, S., Aschrafi, A., Hamada, F. M., Pfeilschifter, J., S., and Igarashi, P. (2013) miR-17~92 miRNA cluster promotes kid-
and Eberhardt, W. (2003) ATP potentiates interleukin-1 beta- ney cyst growth in polycystic kidney disease. Proc. Natl. Acad. Sci. USA
induced MMP-9 expression in mesangial cells via recruitment of the 110, 10765–10770
ELAV protein HuR. J. Biol. Chem. 278, 51758–51769 87. Patel, V., Hajarnis, S., Williams, D., Hunter, R., Huynh, D., and
68. Akool, S., Kleinert, H., Hamada, F. M., Abdelwahab, M. H., Igarashi, P. (2012) MicroRNAs regulate renal tubule maturation
Förstermann, U., Pfeilschifter, J., and Eberhardt, W. (2003) Nitric through modulation of Pkd1. J. Am. Soc. Nephrol. 23, 1941–1948
oxide increases the decay of matrix metalloproteinase 9 mRNA by 88. Hiesberger, T., Bai, Y., Shao, X., McNally, B. T., Sinclair, A. M., Tian,
inhibiting the expression of mRNA-stabilizing factor HuR. Mol. Cell. X., Somlo, S., and Igarashi, P. (2004) Mutation of hepatocyte nuclear
Biol. 23, 4901–4916 factor-1beta inhibits Pkhd1 gene expression and produces renal
69. Yu, Q., Cok, S. J., Zeng, C., and Morrison, A. R. (2003) Translational cysts in mice. J. Clin. Invest. 113, 814–825
repression of human matrix metalloproteinases-13 by an alterna- 89. Peer, D., Park, E. J., Morishita, Y., Carman, C. V., and Shimaoka, M.
tively spliced form of T-cell-restricted intracellular antigen-related (2008) Systemic leukocyte-directed siRNA delivery revealing cyclin
protein (TIAR). J. Biol. Chem. 278, 1579–1584 D1 as an anti-inflammatory target. Science 319, 627–630
70. Shang, J., Wan, Q., Wang, X., Duan, Y., Wang, Z., Wei, X., Zhang, Y., 90. Wang, Z., Bhattacharya, A., and Ivanov, D. N. (2015) Identification
Wang, H., Wang, R., and Yi, F. (2015) Identification of NOD2 as a of small-molecule inhibitors of the HuR/RNA interaction using a
novel target of RNA-binding protein HuR: evidence from NADPH fluorescence polarization screening assay followed by NMR valida-
oxidase-mediated HuR signaling in diabetic nephropathy. Free Radic. tion. PLoS One 10, e0138780
Biol. Med. 79, 217–227 91. Cording, A., Gormally, M., Bond, P. J., Carrington, M.,
71. Evdokimova, V., Ruzanov, P., Anglesio, M. S., Sorokin, A. V., Balasubramanian, S., Miska, E. A., and Thomas, B. (2016)
Ovchinnikov, L. P., Buckley, J., Triche, T. J., Sonenberg, N., and Selective inhibitors of trypanosomal uridylyl transferase RET1
Sorensen, P. H. (2006) Akt-mediated YB-1 phosphorylation activates establish druggability of RNA post-transcriptional modifications.
translation of silent mRNA species. Mol. Cell. Biol. 26, 277–292 [E-pub ahead of print] RNA Biol. DOI: 10.1080/15476286.2015.
72. Capowski, E. E., Esnault, S., Bhattacharya, S., and Malter, J. S. (2001) 1137422
Y box-binding factor promotes eosinophil survival by stabilizing 92. Jimbo, M., Blanco, F. F., Huang, Y. H., Telonis, A. G., Screnci, B. A.,
granulocyte-macrophage colony-stimulating factor mRNA. J. Immunol. Cosma, G. L., Alexeev, V., Gonye, G. E., Yeo, C. J., Sawicki, J. A.,
167, 5970–5976 Winter, J. M., and Brody, J. R. (2015) Targeting the mRNA-binding

REGULATION OF mRNA STABILITY IN RENAL DISEASE 11


Downloaded from www.fasebj.org to IP 139.80.123.48. The FASEB Journal Vol., No. , pp:, November, 2016
protein HuR impairs malignant characteristics of pancreatic ductal H. (2012) ASK3 responds to osmotic stress and regulates blood
adenocarcinoma cells. Oncotarget 6, 27312–27331 pressure by suppressing WNK1-SPAK/OSR1 signaling in the kidney.
93. Eberhardt, W., Badawi, A., Biyanee, A., and Pfeilschifter, J. (2016) Nat. Commun. 3, 1285
Cytoskeleton-Dependent Transport as a Potential Target for In- 98. D’Agostino, V. G., Lal, P., Mantelli, B., Tiedje, C., Zucal, C.,
terfering with Post-transcriptional HuR mRNA Regulons. Front. Thongon, N., Gaestel, M., Latorre, E., Marinelli, L., Seneci, P.,
Pharmacol. 7, 251 Amadio, M., and Provenzani, A. (2015) Dihydrotanshinone-I inter-
94. Van Nostrand, E. L., Pratt, G. A., Shishkin, A. A., Gelboin-Burkhart, feres with the RNA-binding activity of HuR affecting its post-
C., Fang, M. Y., Sundararaman, B., Blue, S. M., Nguyen, T. B., Surka, transcriptional function. Sci. Rep. 5, 16478
C., Elkins, K., Stanton, R., Rigo, F., Guttman, M., and Yeo, G. W. 99. Cheng, Y. C., Liou, J. P., Kuo, C. C., Lai, W. Y., Shih, K. H., Chang,
(2016) Robust transcriptome-wide discovery of RNA-binding C. Y., Pan, W. Y., Tseng, J. T., and Chang, J. Y. (2013) MPT0B098, a
protein binding sites with enhanced CLIP (eCLIP). Nat. Methods novel microtubule inhibitor that destabilizes the hypoxia-inducible
13, 508–514 factor-1a mRNA through decreasing nuclear-cytoplasmic trans-
95. Karet, F. E., Finberg, K. E., Nelson, R. D., Nayir, A., Mocan, H., location of RNA-binding protein HuR. Mol. Cancer Ther. 12,
Sanjad, S. A., Rodriguez-Soriano, J., Santos, F., Cremers, C. W., 1202–1212
Di Pietro, A., Hoffbrand, B. I., Winiarski, J., Bakkaloglu, A., Ozen, S., 100. Doller, A., Badawi, A., Schmid, T., Brauss, T., Pleli, T., zu Heringdorf,
Dusunsel, R., Goodyer, P., Hulton, S. A., Wu, D. K., Skvorak, A. B., D. M., Piiper, A., Pfeilschifter, J., and Eberhardt, W. (2015) The
Morton, C. C., Cunningham, M. J., Jha, V., and Lifton, R. P. (1999) cytoskeletal inhibitors latrunculin A and blebbistatin exert
Mutations in the gene encoding B1 subunit of H+-ATPase cause antitumorigenic properties in human hepatocellular carcinoma
renal tubular acidosis with sensorineural deafness. Nat. Genet. 21, cells by interfering with intracellular HuR trafficking. Exp. Cell Res.
84–90 330, 66–80
96. Piazzon, N., Maisonneuve, C., Guilleret, I., Rotman, S., and Constam, 101. Fraser, D. J., Phillips, A. O., Zhang, X., van Roeyen, C. R.,
D. B. (2012) Bicc1 links the regulation of cAMP signaling in Muehlenberg, P., En-Nia, A., and Mertens, P. R. (2008) Y-box
polycystic kidneys to microRNA-induced gene silencing. J. Mol. Cell protein-1 controls transforming growth factor-beta1 translation in
Biol. 4, 398–408 proximal tubular cells. Kidney Int. 73, 724–732
97. Naguro, I., Umeda, T., Kobayashi, Y., Maruyama, J., Hattori, K.,
Shimizu, Y., Kataoka, K., Kim-Mitsuyama, S., Uchida, S., Vandewalle, Received for publication September 26, 2016.
A., Noguchi, T., Nishitoh, H., Matsuzawa, A., Takeda, K., and Ichijo, Accepted for publication November 7, 2016.

12 Vol. 31 February 2017 The FASEB Journal x www.fasebj.org FEIGERLOVÁ AND BATTAGLIA-HSU
Downloaded from www.fasebj.org to IP 139.80.123.48. The FASEB Journal Vol., No. , pp:, November, 2016
Role of post-transcriptional regulation of mRNA stability in renal
pathophysiology: focus on chronic kidney disease
Eva Feigerlová and Shyue-Fang Battaglia-Hsu

FASEB J published online November 14, 2016


Access the most recent version at doi:10.1096/fj.201601087RR

Subscriptions Information about subscribing to The FASEB Journal is online at


http://www.faseb.org/The-FASEB-Journal/Librarian-s-Resources.aspx

Permissions Submit copyright permission requests at:


http://www.fasebj.org/site/misc/copyright.xhtml

Email Alerts Receive free email alerts when new an article cites this article - sign up at
http://www.fasebj.org/cgi/alerts

© FASEB

Downloaded from www.fasebj.org to IP 139.80.123.48. The FASEB Journal Vol., No. , pp:, November, 2016

You might also like