You are on page 1of 13

BASIC RESEARCH www.jasn.

org

Far Upstream Element-Binding Protein 1 Binds the 39


Untranslated Region of PKD2 and Suppresses Its
Translation
Wang Zheng,*† Fan Shen,†‡ Ruikun Hu,§ Birbickram Roy,║ JungWoo Yang,† Qian Wang,†
Fan Zhang,§ Jennifer C. King,† Consolato Sergi,¶ Song-Mei Liu,‡ Emmanuelle Cordat,†
Jingfeng Tang,* Ying Cao,§ Declan Ali,║ and Xing-Zhen Chen*†
*Membrane Protein Disease and Cancer Research Centre, Provincial Innovation Center, Hubei University of
Technology, Wuhan, China; †Membrane Protein Disease Research Group, Department of Physiology, ║Department of
Biological Sciences, and ¶Department of Laboratory Medicine and Pathology, University of Alberta, Edmonton,
Alberta, Canada; ‡Medical Research Center, Zhongnan Hospital, Wuhan University, Wuhan, China; and §School of Life
Sciences and Technology, Tongji University, Shanghai, China

ABSTRACT
Autosomal dominant polycystic kidney disease pathogenesis can be recapitulated in animal models by
gene mutations in or dosage alterations of polycystic kidney disease 1 (PKD1) or PKD2, demonstrating that
too much and too little PKD1/PKD2 are both pathogenic. Gene dosage manipulation has become an
appealing approach by which to compensate for loss or gain of gene function, but the mechanisms con-
trolling PKD2 expression remain incompletely characterized. In this study, using cultured mammalian cells
and dual-luciferase assays, we found that the 39 untranslated region (39UTR) of PKD2 mRNA inhibits
luciferase protein expression. We then identified nucleotides 691–1044, which we called 3FI, as the 39UTR
fragment necessary for repressing the expression of luciferase or PKD2 in this system. Using a pull-
down assay and mass spectrometry we identified far upstream element-binding protein 1 (FUBP1) as a
3FI-binding protein. In vitro overexpression of FUBP1 inhibited the expression of PKD2 protein but not
mRNA. In embryonic zebrafish, FUBP1 knockdown (KD) by morpholino injection increased PKD2 expression
and alleviated fish tail curling caused by morpholino-mediated KD of PKD2. Conversely, FUBP1 overex-
pression by mRNA injection significantly increased pronephric cyst occurrence and tail curling in zebrafish
embryos. Furthermore, FUBP1 binds directly to eukaryotic translation initiation factor 4E-binding protein 1,
indicating a link to the translation initiation complex. These results show that FUBP1 binds 3FI in the PKD2
39UTR to inhibit PKD2 translation, regulating zebrafish disease phenotypes associated with PKD2 KD.

J Am Soc Nephrol 27: 2645–2657, 2016. doi: 10.1681/ASN.2015070836

Human autosomal dominant polycystic kidney disease PKD2 (also called polycystin-2 or transient re-
(ADPKD) is associated with renal and hepatic cysts ceptor potential polycystin-2), is a 968-amino-acid
and, to a lesser extent, pancreatic cysts and vascular (aa) integral membrane protein that acts as a cation
defects.1,2 In humans, ADPKD is caused by mutations
in the polycystic kidney disease 1 (PKD1) or PKD2 Received July 29, 2015. Accepted November 24, 2015.
genes, which encode the membrane receptor and ion
channel proteins PKD1 and PKD2, respectively. In W.Z. and F.S. contributed equally to this work.

animal models, including mouse, rat, and zebrafish, Published online ahead of print. Publication date available at
ADPKD can be recapitulated, at least in part, by loss- www.jasn.org.
or gain-of-function of PKD1 or PKD2.3–8 Therefore, Correspondence: Dr. Xing-Zhen Chen, Department of Physiol-
the protein expression, membrane localization, and ogy, University of Alberta, Edmonton, AB T6G 2H7, Canada.
Email: xzchen@ualberta.ca
function of PKD1 and PKD2 are highly regulated un-
der normal physiologic conditions. Copyright © 2016 by the American Society of Nephrology

J Am Soc Nephrol 27: 2645–2657, 2016 ISSN : 1046-6673/2709-2645 2645


BASIC RESEARCH www.jasn.org

channel permeable to calcium ions, sodium ions, and potassium 39UTR upstream/downstream of the renilla luciferase gene
ions.9 PKD2 is expressed in numerous tissues, including kidney, to form the plasmids BI16–39UTR, 59UTR–BI16, and
liver, pancreas, lung, heart, brain, intestine, and reproductive 59UTR–BI16–39UTR, and found that the luciferase activity
organs. PKD2 expression/function is regulated by a number of in HeLa cells transfected with BI16–39UTR is much lower
binding protein partners such as PKD1, TRPC1, a-actinin, than those transfected with control plasmid BI16 (Figure
mDia1, Id2, IP3R, and EGFR.10–16 PKD2 expression can also 1A). A similar inhibitory effect of 39UTR was found in the
be regulated through its 39 and 59 untranslated regions (UTRs). presence of 59UTR (by comparing between 59UTR-BI16–
RNA-binding protein bicaudal C (Bicc1) disrupts the transla- 39UTR and 59UTR-BI16). Of note, 59UTR also exhibited an
tional control of PKD2 by microRNA-17 (miR-17) by competing inhibitory effect on the luciferase activity, in agreement with
for the same binding site in PKD2 39UTR, and the lack or in- our previous report.18 To determine whether 39UTR affected
sufficiency of Bicc1 in mouse, zebrafish, and Xenopus laevis re- PKD2 mRNA stability we performed end-point and real-time
sults in renal cysts and other defects through reduced PKD2 RT-PCR assays and found that the PKD2 mRNA level is not
dosage.7,17 Cellular stress conditions and phosphorylated eIF2a significantly altered by 39UTR or 59UTR (Figure 1B), suggest-
up-regulate PKD2 translation via the 59 upstream open reading ing that 39UTR represses the protein translation of luciferase.
frame of PKD218, whereas they inhibit global protein translation. We next wanted to narrow down and identify a 39UTR
Regulation through UTRs is not as well understood as fragment that mediates the inhibition. For this we first divided
regulation through protein–protein interactions. 39UTR- 39UTR into two fragments, nt 1–1044 and nt 1025–2087
mediated regulation is usually through binding of a 39UTR- (with a 20-nt overlap), for the luciferase assays. We found
binding protein that works either by affecting RNA stability that the first half of 39UTR exhibits an inhibitory effect,
or by regulating protein translation through interacting with whereas the second half has a stimulatory effect (Figure 1C).
the translation machinery that is in contact with the 59UTR,19 Because the first fragment contains a binding site (nt 118–145)
and may also be proximate to 39UTR through the formation for miR-17 and Bicc1 we wanted to know whether the ob-
of a ‘closed-loop’ or circular’ mRNA structure.20,21 Transcript served inhibition relates to miR-17 and Bicc1. For this we
circularization can occur via the formation of an eIF4G-poly made BI16 constructs with nt 118–145 deletion from 39UTR
(A)-tail-binding protein complex that promotes recycling of and nt 1–1044, and found that the deletion has minimal or no
the 40S ribosome from the 39UTR to the 59 terminus.22 Alter- effect on luciferase activity (Figure 1D), suggesting that native
natively, circularization can occur through interaction of the miR-17 and Bicc1 do not play a significant role under these
39UTR-binding proteins with specific initiation factors conditions, although overexpressed miR-17 is reported to
thereby regulating protein translation.23,24 In either scenario, exhibit a modest inhibitory effect.26
disruption of the 59–39 interaction affects protein translation. Next we further divided nt 1–1044 and nt 1025–2087 into
In this study we first identified a PKD2 39UTR fragment and overlapping fragments and identified that nt 1–1044 and nt
its binding protein, called far upstream binding protein 1 691–1044 mediate inhibition of luciferase, whereas nt 1025–
(FUBP1), which together mediate downregulation of PKD2 1129 mediates stimulation (Figure 2). Of note, the nt 691–
translation in cultured cells. We then used zebrafish to exam- 1044 fragment had slightly less inhibitory effect on luciferase
ine the effects of FUBP1 morpholino oligonucleotide (MO) expression than nt 1–1044 (Figure 2A), although the reason
knockdown (KD) and overexpression on PKD2 translation, for this is unclear. It is possible that nt 1–1044 has a more
PKD2-dependent tail curling, and pronephric cyst formation. favorable conformation than nt 691–1044 in terms of inter-
Finally, we performed coimmunoprecipitation (co-IP) and acting with the to-be-identified inhibitory binding partner.
glutathione-S-transferase (GST) pull-down assays to reveal Because shorter fragments of nt 691–1044 and nt 1025–1129
the physical link between FUBP1 and eukaryotic initiation substantially reduced the inhibitory and stimulatory effects,
factor-4E-binding protein-1 (4EBP1). respectively (Figure 2, C and D), we called nt 691–1044
39 fragment inhibitory (3FI) and nt 1025–1129 39 fragment
stimulatory (3FS). Of note, we selected nt 691–1044, but not
RESULTS nt 1–1044, because the former is much shorter and thus would
be better for identifying specific binding partners. The inhib-
Identification of PKD2 39UTR Fragments that Regulate itory effect of 3FI on the translation of luciferase was supported
Its Protein Translation by data obtained using mutant 39UTR with 3FI deletion
We previously reported that PKD2 59UTR mediates the trans- (Figure 3, A and B). Of note, 3FI is AU-rich (68%) and has no
lational up-regulation of PKD2 under cellular stress condi- overlap with the binding site for miR-17 and Bicc1.7 We also
tions. 18 In an effort to determine whether the PKD2 performed Western blotting (WB) assays to verify the effects of
2087-nucleotide (nt) 39UTR regulates its protein expression 3FI and 3FS by directly detecting the protein level of renilla
we performed dual-luciferase assays with vector BI1625, which luciferase. We indeed found that the renilla luciferase in HeLa
we used in our previous study.18 BI16 contains a bidirectional cells, and also in HEK 293T cells, is regulated by 3FI and 3FS,
promoter that drives the transcription of the renilla and in- similar to the regulation observed in the luciferase assays (Fig-
ternal control firefly luciferases. We ligated PKD2 59UTR/ ure 3C). In addition, by replacing the coding sequence of

2646 Journal of the American Society of Nephrology J Am Soc Nephrol 27: 2645–2657, 2016
www.jasn.org BASIC RESEARCH

renilla luciferase in the plasmid by that of


Flag-tagged PKD2, we found that 3FI and
3FS reduces and enhances Flag-PKD2 pro-
tein expression, respectively, in both HeLa
and HEK cells (Figure 3D), which further
confirmed the regulatory effects of 3FI and
3FS on protein translation.

Identification of 3FI-Binding Partner


FUBP1 that Suppresses PKD2 Protein
Translation
We next wanted to focus on 3FI by exam-
ining how it downregulates PKD2 trans-
lation. We reasoned that there may be a
RNA-binding protein that interacts with
3FI and mediates the inhibitory effect on
translation. For this we used PCR products
from plasmids BI16–3FI and BI16 (as a
control) as a template for in vitro transcrip-
tion using biotin-labeled uridine 59-
triphosphate to obtain biotinylated
BI16–3FI and BI16 RNA fragments that
were incubated with HeLa cell lysates, fol-
lowed by pull-down using streptavidin
beads (see Concise Methods). Proteins in
the precipitated lysates were separated by
SDS-PAGE for mass spectrometry analysis
of bands present in the 3FI lane but not in
the control lane, which identified FUBP1,
FUBP2, and eIF4G as 3FI-interacting part-
ners (see Supplemental Table 1 for a full list
of the identified partners). FUBP1 and
FUBP2 are DNA-/RNA-binding proteins
that regulate transcription, translation, or
mRNA stability of target genes, whereas
eIF4G is a ribosomal protein and a key fac-
tor in the translation initiation complex.
We then wondered whether FUBP1 and
FUBP2 actually downregulate PKD2 ex-
pression. For this we performed WB assays
using HeLa cells and found that overex-
pression of FUBP1, but not of FUBP2,
indeed reduces the PKD2 protein level to
4565% (n=3, P=0.004) (Figure 4A); how-
ever, FUBP1 had no effect on the PKD2
mRNA level. We also found that overexpres-
Figure 1. Effects of PKD2 39UTR and 59UTR on luciferase activity in HeLa cells. (A) sion of nuclear fragile X mental retardation-
Effects of 39UTR on the relative luciferase activity revealed by dual-luciferase assays in interacting protein 2, another RNA-binding
the presence or absence of 59UTR. (B) Effects of 39UTR on the mRNA level revealed by
protein identified as a 3FI-binding partner
real-time RT-PCR assays in the presence or absence of 59UTR. WBs show represen-
tative data. (C) Effects of 39UTR fragments nt 1–1044 and nt 1025–2087 on the relative
luciferase activity revealed by dual-luciferase assays in the absence of 59UTR. (D) Ef-
fects of the binding domain (nt 118–145) for miR-17 in 39UTR on the luciferase activity Control (Ctrl) indicates the absence of any
in the presence of 59UTR. 39UTRDmiR-17 and nt 1–1044DmiR-17 indicate 39UTR 39UTR fragment, but with the presence
and nt 1–1044, respectively, with deletion of miR-17-binding domain, nt 118–145. of 59UTR. NS, not significant. **P,0.01;
Statistical data are mean and SEM values from three independent experiments. ***P,0.001.

J Am Soc Nephrol 27: 2645–2657, 2016 FUBP1 Suppresses PKD2 Translation 2647
BASIC RESEARCH www.jasn.org

but had no effect in the absence of 3FI in


HEK cells (Figure 4D). FUBP1 was previ-
ously reported to bind AU-rich regions of
RNAs through a signature sequence of
AUUUA.27 Indeed, human 3FI contains
68% AU and 3FI from six different species
all contain one to three AUUUA sequences
(Supplemental Figure 1). Taken together,
these results show a specific effect of FUBP1
on PKD2 translation but not on its mRNA
stability, and suggest that FUBP1 may have
mediated, at least in part, our observed in-
hibitory effect of 3FI.
Because FUBP1 was identified in vitro
using biotin–RNA pull-down assays with
fragment 3FI, it remains to be determined
whether FUBP1 is able to bind native 3FI
inside the cell in which full length PKD2
39UTR has its distinct conformation. For
this purpose we performed RNA-protein
immunoprecipitation (RIP) assays by
transfecting HeLa cells with human influ-
enza hemagglutinin (HA)-FUBP1 or
empty vector (control) and then precipitat-
ing HA-FUBP1 whole cell extracts using
HA antibody (Figure 5A). After total
RNAs were isolated from HA-FUBP1 im-
munoprecipitates, PKD2 mRNA was in-
deed detected by RT-PCR while no PKD2
mRNA was detected in control groups (Fig-
ure 5B), indicating that FUBP1 binds the
endogenous PKD2 mRNA.

Effects of FUBP1 on PKD2 Expression


Figure 2. Effects of 39UTR fragments on luciferase activity in HeLa cells. (A) Effects of and Disease Phenotypes in Zebrafish
39UTR fragments, including 3FI, within nt 1–1044, on luciferase activity as indicated. Although zebrafish PKD2 mRNA 39UTR is
(B) Effects of 39UTR fragments within nt 1025–2087 on luciferase activity, as indicated. much shorter than that of human PKD2
(C) Effects of 39UTR fragments within 3FI on luciferase activity, as indicated. (D) Effects mRNA 39UTR (201 versus 2087 nt, respec-
of 39UTR fragments within nt 1025–1324 on luciferase activity, as indicated. Statistical
tively), the corresponding 3FI is also
data are mean and SEM values from three independent experiments. Control (Ctrl)
AU-rich (66%) and contains one AUUUA
indicates the absence of any PKD2 39UTR fragment or 59UTR in the BI16 plasmid.
*P,0.05; **P,0.01; ***P,0.001. sequence (Supplemental Figure 1). Zebra-
fish FUBP1 also shares 68% aa sequence
identity with human FUBP1. We next stud-
(Supplemental Table 1), has no effect on PKD2 expression (data ied in vivo larval zebrafish to determine the regulation of
not shown). To verify the specificity, we also used siRNA to KD PKD2 expression by FUBP1, and the associated disease phe-
FUBP1 and found that FUBP1 KD in HEK cells led to an increase notypes. For this, we injected commercially acquired MO an-
of 80611% (n=3, P=0.01) in the PKD2 protein level while having tisense oligonucleotide against PKD2 or FUBP1, or both
no significant effect on the PKD2 mRNA level (Figure 4B). In PKD2 and FUBP1, or control MO into one- to four-cell zebra-
order to determine whether translational inhibition by FUBP1 is fish embryos within 1 hour postfertilization. The same PKD2
mediated through 3FI, we cotransfected an FUBP1-expressing MO sequence and injection procedure was used as previously
plasmid and luciferase plasmid BI16 without or with 3FI into reported.28 Two days after injection, we manually dechorio-
HeLa cells and found that FUBP1 overexpression significantly nated embryos and found that fish with PKD2 MO injection,
reduces luciferase expression in the presence of 3FI, but has no but not those with control MO injection, display curled tails
effect in the absence of 3FI (Figure 4C). Consistently, FUBP1 KD (Figure 6A) and pronephric cysts, consistent with previously
by siRNA increased luciferase expression in the presence of 3FI, published results that the presence of the two phenotypes are

2648 Journal of the American Society of Nephrology J Am Soc Nephrol 27: 2645–2657, 2016
www.jasn.org BASIC RESEARCH

We next examined PKD2 and FUBP1


expression in these fish by WB, using
antibodies against zebrafish PKD2 and
FUBP1. We found that MO injection effec-
tively reduces PKD2 and FUBP1 expres-
sion, and that PKD2 expression increases in
fish with FUBP1 MO injection (Figure 6B),
in agreement with our in vitro data (Figure
4). These data indicate that reduced PKD2
dosage in larval zebrafish results in tail
curling that is rescued or alleviated by
FUBP1 KD through up-regulating PKD2 ex-
pression. Consistently, we found that com-
bined injection of in vitro transcribed human
FUBP1 mRNA and PKD2 MO, but not single
injection of either of the two, results in fish tail
curling (Figure 7, A and B), presumably due to
reduction in the PKD2 expression to below a
threshold value by the coinjection. Further, we
wanted to determine whether FUBP1 also reg-
ulates PKD2 KD-associated pronephric cyst
formation. For this we employed FUBP1 over-
expression through mRNA injection and
found that FUBP1 mRNA injection signifi-
cantly increases the occurrence rate of pro-
Figure 3. Effects of 3FI and 3FS on luciferase activity and protein expression of lu- nephric cysts induced by PKD2 MO KD in
ciferase and PKD2 in HeLa and HEK cells. (A) Effect of 3FI (39UTRD3FI) or 3FS zebrafish (Figure 7, C–E). Thus, taken to-
(39UTRD3FS) deletion from 39UTR on luciferase activity in HeLa cells. (B) Effect of 3FI or gether, our data demonstrate that FUBP1 reg-
3FS deletion from 39UTR on luciferase activity in HEK cells. (C) Representative WB data ulates PKD2-associated disease phenotypes
showing the effects of 3FI and 3FS on the expression of renilla luciferase protein in (tail curling and pronephric cyst) in larval ze-
HeLa and HEK cells, as indicated. Firefly luciferase blots serve as an internal control.
brafish, presumably through regulating PKD2
(D) Representative WB data showing the effects of 3FI and 3FS on the expression of
protein dosage.
Flag-PKD2 in HeLa and HEK cells, as indicated. Firefly luciferase blots serve as an
internal control. Statistical data are mean and SEM values from three independent
experiments. Control (Ctrl) indicates the absence of any PKD2 39UTR fragment or Interaction between FUBP1 and
59UTR in the BI16 plasmid. *P,0.05; **P,0.01. Translation Regulator 4EBP1
We next investigated whether 3FI and
FUBP1 are physically linker to the initiation
significantly correlated to each other.29,30 Additionally, tail of translation, which may be the basis for the downregulation of
curling angle strongly correlated with PKD2 expression level PKD2 translation by 3FI and FUBP1. It is well known that
(Figure 6D). We thus decided to use ‘tail curling’ as a valid 4EBP1 binds to eukaryotic initiation factor-4E (eIF4E) to
readout to examine the functional implications of regulation disrupt the eIF4E–eIF4G initiation complex thereby reducing
of PKD2 expression by FUBP1. Although FUBP1 MO injec- the translation initiation activity. Several RNA-binding pro-
tion alone did not lead to tail curling, fish with coinjection of teins are known to bind 39UTR and reduce the formation of
FUBP1 and PKD2 MOs showed substantially reduced tail initiation complex eIF4E–eIF4G through interacting with
curling compared to PKD2 MO injection alone, indicating 4EBPs.19,31 We thus first used co-IP assays to see whether
that FUBP1 KD rescues the phenotype caused by PKD2 KD. FUBP1 interacts with 4EBP1. In both HEK and HeLa cells,
After 2 dpf, tail curling gradually increased in fish with PKD2 we found that FUBP1 indeed precipitates 4EBP1 (Figure 8A),
MO injection alone, while in fish with FUBP1 MO coinjection indicating that they are in the same protein complex in these
tail curling gradually decreased (Figure 6, A and C). On aver- cells. To determine whether they associate directly with each
age, the angle of tail curling with PKD2 KD alone increased other, we performed GST pull-down assays. For this purpose,
from 154623° at 2 dpf to 224632° at 5 dpf (n=55), showing we divided human FUBP1 into three overlapping fragments,
increased severity with time, whereas the angle of tail curling the N-terminus (NT; aa 1–112), central domain (CD; aa 100–
with PKD2 and FUBP1 MO coinjection decreased from 586 447) and C-terminus (CT; aa 442–644), and purified them
11° at 2 dpf to 1868° at 5 dpf (n=43), indicating a rescue effect from Escherichia coli. We found that the CD, but not the two
that increases with increasing time. other fragments, is able to precipitate 4EBP1 protein purified

J Am Soc Nephrol 27: 2645–2657, 2016 FUBP1 Suppresses PKD2 Translation 2649
BASIC RESEARCH www.jasn.org

DISCUSSION

We previously reported that PKD2 is trans-


lationally upregulated under cellular stress
conditions and that this regulation is related
to the presence of a conserved short up-
stream open reading frame fragment in the
PKD2 mRNA 59UTR.18 In this study, we
investigated the role of PKD2 mRNA
39UTR in the regulation of PKD2 pro-
tein level. Using dual-luciferase assays and
RT-PCR we have shown that human PKD2
39UTR represses the protein translation of
luciferase. We then identified the inhibi-
tory nt fragment 3FI (nt 691–1044) and,
by biotin-labeled RNA pull-down assays,
its binding protein FUBP1. We have shown
that FUBP1 does exhibit an inhibitory ef-
fect on PKD2 translation, but not tran-
scription, in cultured cells. Finally, we
have shown that, in embryonic zebrafish,
FUBP1 regulates the severity of tail curling
and pronephric cystogenesis due to PKD2
MO KD, presumably through regulating
PKD2 expression in a similar way to that
observed in cultured cells.
A “two hit” model was first proposed in
1992 to illustrate a possible mechanism un-
derlying the cystogenesis of ADPKD. 32
This hypothesis suggested that the occur-
rence of a somatic mutation in the remain-
ing normal allele of some cells initiated cyst
formation in the affected cells. In other
words, ADPKD is recessive at the cellular
Figure 4. Effect of FUBP1 on PKD2 protein and mRNA levels. (A) Effect of FUBP1 level. This model received strong support
overexpression on PKD2 in HeLa cells. Left panel shows representative WB data. from the identification of somatic muta-
Center panel shows quantified WB data averaged from n=3 independent experiments. tions that inactivated the normal alleles in
Right panel shows PKD2 mRNA expression by RT-PCR. (B) Effect of FUBP1 siRNA KD 20%–43% of the analyzed PKD1 or PKD2
on PKD2 expression in HEK cells. Left panel shows representative WB data. Center
renal cysts.33–36 However, no somatic mu-
panel shows quantified WB data averaged from three independent experiments. Right
tation in the normal allele was found in the
panel shows PKD2 mRNA expression obtained by RT-PCR. (C) Effect of FUBP1
overexpression on activity of luciferase without (BI16) or with 3FI (BI16–3FI) as 39UTR in
remaining cysts. Over the past decades, ev-
HeLa cells. (D) Effect of FUBP1 siRNA KD on activity of luciferase without (BI16) or with idence has been increasing in support of a
3FI (BI16–3FI) as 39UTR in HEK cells. Data were obtained from n=3 independent ex- “dosage” model that suggests renal cysto-
periments. *P,0.05; **P,0.01. genesis can be initiated when the wild-type
PKD1 or PKD2 protein level is reduced
from E. coli. (Figure 8B). Further, we found that addition of below a threshold level in the absence of a somatic muta-
3FI RNA, but not a control RNA fragment, significantly in- tion.37–39 Therefore, it became important to elucidate how
creases the binding between the FUBP1 CD and purified PKD1 and PKD2 protein expression is regulated to identify new
4EBP1 (Figure 8C). Interestingly, FUBP1 CD was found to therapeutic targets, especially as the outcomes of previous clinical
have four K-homology (KH) domains for DNA and RNA trials are rather negative. FUBP1 identified in this study is a novel
binding. These data suggest that the inhibitory effect of 3FI translational regulator of PKD2 and indeed regulated PKD2-
and FUBP1 on PKD2 translation is mediated by the FUBP1– associated zebrafish tail curvature and pronephric cystogenesis,
4EBP1 binding strength. In summary, these experiments to- which can be accounted for by changes in the PKD2 protein dosage.
gether demonstrate that FUBP1 is indeed physically linked to The fact that both loss- and gain-of-function of PKD2 are
4EBP1, a regulator of the initiation of translation. pathogenic indicates that the PKD2 protein level must be

2650 Journal of the American Society of Nephrology J Am Soc Nephrol 27: 2645–2657, 2016
www.jasn.org BASIC RESEARCH

and plays important roles in organ development, including


lung, brain, and neural network development.27,43,44 In con-
trast, FUBP1 was known to transcriptionally downregulate the
expression of TNFa.45 Interestingly, c-Myc is elevated in cystic
tissues of various PKD models and TNFa is elevated in PKD2+/2
mice.46–48 Moreover, c-Myc transgenic mice and in vitro appli-
cation of TNFa to kidney in culture developed cysts.46,47 Thus,
although FUBP1 altered the expression of three cystogenic pro-
teins, PKD2 (down), TNFa (down), and c-Myc (up), whether
and how FUBP1 is implicated in cystogenesis remains to be
determined.
Although FUBP1 was originally identified as a transcription
factor of c-Myc, recent reports demonstrated that FUBP1 also
functions as a RNA-binding protein for translational regula-
tion of its bound genes, including GAP43, p21, Cox-2, and
NPM.49–52 Similar to bicaudal C that contains three RNA-
binding KH domains and binds to the 39UTR of PKD2,
Figure 5. Interaction between FUBP1 and native PKD2 mRNA. FUBP1 has four KH domains. Sequence analysis found that
HeLa cells were transfected with HA-FUBP1 or empty vector. FUBP1 binds to AU-rich regions in mRNA 39UTR, which is
Forty-eight hours after transfection, cell lysates were im- supported by the fact that the FUBP1-binding 3FI contains
munoprecipitated with HA antibody or nonimmune rabbit IgG 68% AU. FUBP1 is present in multiple tissues including kid-
(control). (A) WB analysis of cell lysates (Inputs) and immunopre- ney, liver, pancreas, thymus, ovary, and skin.53,54 In kidney
cipitates with antibodies indicated. (B) RT-PCR analysis of RNAs FUBP1 is expressed at various locations, including podocytes
isolated from immunoprecipitates. +RT and –RT represent RT and proximal and distal tubules (Supplemental Figure 2), and
reactions carried out in the presence and absence of reverse
developmentally decreases53,54, whereas the PKD2 protein
transcription, respectively.
level increases in the kidney during development,40 consistent
with downregulation of PKD2 translation by FUBP1.
strictly regulated within a narrow range.3,6 Consistent with As a transcription regulator, FUBP1 was reported to be
this, it is known that PKD2 protein expression is developmen- localized primarily in the nucleus.55–57 However, to function
tally regulated40, whereas generally the protein level is regu- as a translational regulator, FUBP1 is also required in the
lated transcriptionally or translationally. By computational cytoplasm. It was known that, under normal physiologic con-
analyses, putative binding sites for transcription factors E2F, ditions, FUBP1 primarily localizes in the nucleus, and that
EGRF, ETS, SP1, and ZBP-89 were identified in the promoter under stress conditions, such as in the presence of heat shock,
region of both human and mouse PKD2.41 To date, however, viral infection, and oxidative stress, FUBP1 either stops enter-
there have been little experimental data available regarding ing the nucleus or translocates from the nucleus to the cyto-
transcription factors that regulate PKD2 transcription, sug- plasm, resulting in cytoplasmic accumulation.27 Interestingly,
gesting that translational regulation of PKD2 plays an impor- TGF-b1, which is upregulated in ADPKD, was also reported to
tant role. Indeed, a short upstream open reading frame in induce translocation of FUBP1 from the nucleus to the
PKD2 mRNA 59UTR is implicated in the translational up- cytoplasm.58
regulation of PKD2 by cellular stresses18, and microRNA-17 The presence of a physical interaction between FUBP1 and
post-transcriptionally downregulates PKD2 expression by eIF4E-binding protein 4EBP1 suggests the complex 3FI–
binding to its mRNA 39UTR thereby promoting cell prolifera- FUBP1 located at the 39UTR of PKD2 mRNA is indeed con-
tion.26 Furthermore, it was reported that the RNA-binding nected, via 4EBP1, to the initiation complex located at the
protein bicaudal C upregulates PKD2 expression by antagoniz- 59UTR of PKD2 mRNA. Thus, our study may have
ing the repressive activity of miR-17 through binding compet- identified a novel example of a ‘closed-loop’ or ‘circular’ model
itively to the same site on the PKD2 mRNA 39UTR.7 Of note, of mRNAs. However, it remains to be determined whether 3FI
the 3FI and 3FS identified in this study do not overlap with the and FUBP1 enhance 4EBP1–eIF4E binding, thereby reducing
miR-17 binding site. Taken together, these data show that the eIF4E–eIF4G binding in the proximity of PKD2 59UTR.
39UTR possesses multiple elements important for complex In summary, we found that FUBP1 binds with the AU-rich
regulation of the PKD2 protein level. FUBP1 was first identified 3FI in the PKD2 mRNA 39UTR and negatively regulates PKD2
as a single-strand DNA-binding protein that upregulates the protein translation in cultured cell lines. In embryonic zebra-
transcription of proto-oncogen c-Myc through binding to the fish, decreased FUBP1 by MO injection and increased FUBP1
far upstream element in the promoter region 1.5 kb upstream by mRNA injection alleviates and aggravates, respectively, the
of the transcription start site.42 FUBP1 promotes cell prolif- tail curling and/or pronephric cyst occurrence due to reduced
eration, survival and metastasis through up-regulating c-Myc PKD2 level by MO injection, presumably through affecting

J Am Soc Nephrol 27: 2645–2657, 2016 FUBP1 Suppresses PKD2 Translation 2651
BASIC RESEARCH www.jasn.org

unveiled a novel mechanism of dosage regu-


lation that is important for ADPKD patho-
genesis and might enable the discovery of
new therapeutic targets in future studies.

CONCISE METHODS

Plasmid Constructs, Cell Culture, and


Gene Transfection
Human PKD2 mRNA 39UTR composed of
2087 nt was amplified from HeLa cells
w ith the following primers: forward: 59-
GTCGGGCCCTATGTGTGTTTCAGTATGTG-39;
reverse: 59-CGCCGTTTAAACTTTCTGCTACTA-
TATCAAG-39, and subcloned into luciferase vector
BI16,25 a kind gift from Dr. Ed Grabczyk (Louisiana
State University, Baton Rouge, LA), at the ApaI and
PmeI restriction sites. Fragments of human PKD2
mRNA 39UTR were also cloned into BI16 at the
same sites. Human FUBP1 coding sequence was am-
plified from plasmid pOTB7–FUBP1 purchased
from Open Biosystems (Thermo Fisher Scientific,
Waltham, MA) and inserted into pcDNA3.1(+).
Flag tag was inserted at the 59 of the FUBP1 coding
region. All mutations were made with the Quik-
Change Lighting Site-Directed Mutagenesis kit (Agi-
lent Technologies, Santa Clara, CA) and confirmed
by sequencing. HeLa and HEK cells were cultured in
DMEM supplemented with 10% FBS and 1%
penicillin/streptomycin (Sigma-Aldrich, St. Louis,
MO). Cells of fewer than 25 passages were cultured
to full confluence before collection. Transient trans-
fection was performed on cells cultured to 70%–90%
Figure 6. Effect of FUBP1 KD on PKD2 expression and tail curling in embryonic ze- confluence using Lipofectamine 2000 (Invitrogen,
brafish. (A) Representative pictures of 2–5 dpf zebrafish. Embryos were injected with Carlsbad, CA) according to the manufacturer’s
MO(s) of control (Ctrl; 2 ng), FUBP1 (1.5 ng), PKD2 (0.5 ng), or PKD2+FUBP1 (0.5+1.5 ng) instructions.
within 1 hour postfertilization. Ctrl, Ctrl MO. (B) Representative WB data from
5-dpf larvae obtained using PKD2 and FUBP1 antibodies (Cat#: OAEB01480 and
Antibodies
ARP37732_T100, Aviva System Biology). (C) Averaged tail curling angle for PKD2
Rabbit antibodies against PKD2 (H-280) and FLAG
(n=55) and PKD2+FUBP1 (n=43) MO injections. The angles for FUBP1 (n=76) and Ctrl
(D-8), goat antibody against FUBP2 (D-12), and
(n=60) MOs were both zero and thus not plotted here. **P,0.01; ***P,0.001. (D) Tail
mouse antibodies against firefly luciferase (Luci 21 1–
curling angle plotted against PKD2 expression level from 12 individual fish at 5 dpf,
showing correlation between tail curling angle and PKD2 expression level. PKD2 WB 107), b-actin (C-4) green fluorescent protein (GFP;
bands were quantified and divided by the corresponding b-actin bands using ImageJ B-2), GST (B-14), and 4EBP1 (P-1) were purchased
and normalized to the fish showing no tail curvature. The curve is an empirical ex- from Santa Cruz Biotechnology (Santa Cruz, CA).
ponential decay fit that generated a correlation coefficient of R=0.93. Mouse antibody against renilla luciferase was pur-
chased from Chemicon International (Temecula,
CA). Goat antibody against PKD2 and rabbit anti-
PKD2 dosage. In fact, for human patients whose PKD2 mutant body against FUBP1 were from Aviva System Biology (San Diego, CA).
(s) remains partially functional, a peptide of FUBP1, a RNA frag- Secondary antibodies were purchased from GE Healthcare (Waukesha, WI).
ment of human PKD2 3FI, or chemical that disrupts the FUBP1–
39UTR binding are potential drug candidates because they would KD by siRNA
abolish (or significantly reduce) the downregulation of PKD2 KD of human FUBP1 by siRNAwas performed using oligonucleotides at a
translation by FUBP1, thereby increasing PKD2 dosage and com- final concentration of 20 nM with transfection reagent Lipofectamine
pensating for partial loss of PKD2 function. Our study thus 2000 according to the manufacturer’s instructions. siRNA

2652 Journal of the American Society of Nephrology J Am Soc Nephrol 27: 2645–2657, 2016
www.jasn.org BASIC RESEARCH

as described, 28 for zebrafish PKD2 KD. A


translation-blocking fubp1 MO (Gene Tools
LLC) was also used. The MO sequences were
as follows: pkd2, 59-AGGACGAACGCGA-
CTGGAGCTCATC-39; fubp1, 59- GGCCA-
TGTCTGCACGAACAGTCTTC-39; gli2 (an
antisense mismatch morpholino, used as a neg-
ative control), 59-CCTCTTACCTCAGTTA-
CAATTTATA-39. 61 Capped human fubp1
mRNA was synthesized using the mMessage
mMachine T7 kit (Ambion, Austin, TX) and in-
jected into fertilized embryos (at 200 pg each).
Embryos at 3 dpf were fixed in 1.5% glutaralde-
hyde, 1% paraformaldehyde, 70 mM sodium phos-
phate, pH 7.2, and 3% sucrose overnight at 4°C.
After being washed in PBS and taken through an
ethanol dehydration series, the embryos were em-
bedded in glycolmethacrylate (JB-4; Polyscience,
Warrington, PA) according to the manufacturer’s
instructions and sectioned at 4 mm. Slides were
stained with methylene blue and azure II,62 moun-
ted, and examined under a microscope.
The study was approved by the Ethical Com-
mittee for Animal Experiments of the University of
Alberta, and was carried out in accordance with the
Guidelines for Research with Experimental Ani-
mals of the University of Alberta and the Guide for
the Care and Use of Laboratory Animals (National
Institutes of Health Guide) revised in 1996.

RT-PCR
Total cellular RNA was prepared using TRIzol
reagent (Invitrogen), according to the manufac-
Figure 7. Effect of FUBP1 overexpression on tail curling and pronephric cystogenesis
turer’s manual. Contaminating DNA was digested
of larval zebrafish. (A) Representative pictures showing tails of 3 dpf zebrafish with
injection of water (control; Ctrl), PKD2 MO, FUBP1 mRNA, or both PKD2 MO+FUBP1 with RNase-free DNase (Promega, Madison, WI).
mRNA within 1 hour postfertilization. (B) Average percentages of fish having a curled Single-strand cDNA synthesis was carried out using
tail under different conditions: PKD2 MO injection alone (n=112), FUBP1 mRNA in- Superscript III reverse transcription (Invitrogen),
jection alone (n=152), and coinjection (n=95). *P,0.05; **P,0.01. (C) Ctrl (water- according to the manufacturer’s instructions. End-
injected) larva at 3 dpf with straight tail and histologically normal glomerulus and point PCR was performed using 28-cycle protocol
adjacent tubules. (D) PKD2 MO-injected larva at 3 dpf showing curly tail and pro- with Taq DNA polymerase (Invitrogen). The oligo-
nephric cyst formation (arrows), which is confirmed by a histologic section that also nucleotide primers for each gene were as follows:
displayed dilated pronephric tubules (★). (E) Average percentages of fish exhibiting b-actin, sense 59- CCTGGCACCCAGCACAAT-39
pronephric cysts under different conditions: PKD2 MO (0.15 ng) injection alone
and antisense 59- GGGCCGGACTCGTCATACT-
(n=257), FUBP1 mRNA (200 pg) injection alone (n=157), and coinjection (n=248). G,
39; firefly luciferase, sense 59- CGTTCGTCA-
glomerulus; Pt/Pd, pronephric tubule/duct; Nc, notochord. **P,0.01.
CATCTCATCTACCTCC-39 and antisense
59- GCAGAGCGACACCTTTAGGCAGACC-39;
oligonucleotides were purchased from Santa Cruz Biotechnology (Cat renilla luciferase, sense 59- CATTCAAGGAGAAGGGCGAGGTTAG
no.: sc-43760) and control siRNA from Gene Pharma (Shanghai, China). and antisense 59- TGTAGTTGCGGACAATCTGGACGAC-39; pkd2,
The efficiency of the siRNA KD was assessed by WB. sense 59- GTATGACGGCTCACGCCTGTAATCC-39 and antisense
59- AGAGATGGAGTTTCGCCACATTGCC-39; fubp1, sense
Zebrafish Experiments 59- CATAGAAGAAAAGATTGGTGGC-39 and antisense 59- AGGATTA-
Wild-type zebrafish AB strain was maintained and staged as previ- TAAGGTGCAGGGTTG-39. Real-time PCR was performed using a
ously described.59,60 Embryos were kept in E3 solution. A translation- 7900HT Fast Real-time PCR System (Applied Biosystems, Foster
blocking antisense MO (Gene Tools LLC, Philomath, OR) City, CA) with the same primers. Fast SYBR Green Master Mix (In-
was injected at the one-cell stage within 1 hour postfertilization, vitrogen) was used. 7900HT Fast Real-Time PCR System 2.4.1

J Am Soc Nephrol 27: 2645–2657, 2016 FUBP1 Suppresses PKD2 Translation 2653
BASIC RESEARCH www.jasn.org

synthesized using the mMessage mMachine T7


kit (Ambion) in the presence of 14Biotin- uridine
59-triphosphate (Roche, Basel, Switzerland) and
purified using phenol:chlorophorm extraction
and MegaClean columns (Ambion). Biotin–RNA
pull-down assays were carried out as described
previously.63 Briefly, the biotinylated RNA was
bound to streptavidin-coated Dynabeads (Invitro-
gen) according to the manufacturer’s instructions.
The cytoplasmic extracts of HeLa cells were pre-
cleared by incubation with RNA-affinity resin
containing a control fragment. The unbound frac-
tion was incubated with RNA-affinity resin con-
taining human PKD2 mRNA 39UTR fragments in
the binding buffer (40 mM potassium chloride,
1.5 mM magnesium chloride, 0.2 mM EDTA, 1
mM dithiothreitol, 6% glycerol, 2 mg/ml heparin,
20 mM HEPES, and pH 8.0) supplemented with
15 mg/ml yeast tRNA (Sigma-Aldrich) and
200 U/ml RNaseOUT (Invitrogen). After 2 hours
of incubation at 4°C, the unbound proteins were
removed and, after extensive washes with the bind-
ing buffer, the proteins were eluted by boiling in
Laemmli buffer and resolved on SDS-PAGE. Pro-
tein bands were cut for mass-spectrometric anal-
ysis (University of Alberta, Edmonton, AB).

RIP
RIP assay was carried out as described pre-
viously.64 Briefly, HeLa cells were transfected
with pCGNM2 harboring human HA–FUBP1
(a kind gift from Dr. David Levens, National
Institutes of Health). Forty-eight hours after
transfection, cells were collected using RIP
Figure 8. Interaction between FUBP1 and 4EBP1, and effect of 3FI. (A) Co-IP experiments buffer (150 mM potassium chloride, 25 mM
using native HEK and HeLa cells. FUBP1 antibody was used for precipitation and 4EBP1 Tris, pH 7.4, 5 mM EDTA, 0.5 mM dithiothrei-
antibody for immunoblotting. (B) GST pull-down experiments using purified GST-tagged tol, 0.5% NP-40, 100 U/ml RNAase inhibitor
human NT (aa 1–112), CD (aa 100–447), and CT (aa 442–644) peptides and GFP-tagged [Invitrogen], proteinase inhibitor mixture),
human 4EBP1 protein from E. coli. (C) Effect of 3FI on in vitro binding between purified and cell debris pelleted by centrifugation at
GST–CD and GFP–4EBP1. Left panel shows representative WB data. The reaction system 13,000 rpm for 10 minutes. HA tag antibody
containing purified GST–CD and GFP–4EBP1 was supplemented with 3FI RNA, an un- (Santa Cruz Biotechnology; catalog no.:
related RNA (Ctrl RNA), or none. GST pull-down assays were then performed, with GST sc-805) or control IgG (Abcam, Inc., Cambridge,
and GFP antibodies for immunoblotting. Right panel shows quantified and normalized WB
MA; catalogue no.: ab27478) was added to the
data averaged from three independent experiments. NS, not significant. *P=0.03.
supernatant and incubated for 4 hours at 4°C
with gentle rotation. A total of 50 ml protein
software was used to perform quantification and to generate cycle G-Sepharose beads (GE Healthcare) were then added and incu-
threshold values. bated for 2 hours at 4°C with gentle rotation. Beads were pelleted
at 2500 rpm for 30 seconds and washed three times with RIP
Biotin–RNA Pull-Down buffer, followed by one wash in PBS. Coprecipitated RNAs were
Templates for in vitro synthesis of biotinylated RNA were generated then isolated with TRIzol and RT-PCR assays were performed with
by PCR from the BI16 vector with or without human PKD2 mRNA following primers: pkd2, sense 59-GTATGACGGCTCACGCCTG-
39UTR fragments. Forward and reverse primers are as follows: forward, TAATCC-39 and antisense 59-AGAGATGGAGTTTCGCCA-
59-AAATTAATACGACTCACTATAGGGACGAGCAGTAATTC- CATTGCC-39; b-actin, sense 59-CCTGGCACCCAGCACAAT-39 and
TAGAGG-39 (the T7 promoter sequence is underlined), reverse, antisense 59-GGGCCGGACTCGTCATACT-39. Protein isolated by the
59- GGGCAAACAACAGATGGCTGGCAAC-39. Biotinylated RNA was beads was detected by WB.

2654 Journal of the American Society of Nephrology J Am Soc Nephrol 27: 2645–2657, 2016
www.jasn.org BASIC RESEARCH

Dual Luciferase DISCLOSURES


HeLa and HEK cells were cultured and transfection was performed as None.
described above with plasmids BI16 with or without a human PKD2
mRNA 39UTR fragment. At 12 hours after transfection, cells were
collected and dual-luciferase assays were carried out with Dual- REFERENCES
Luciferase Reporter Assay System (Promega) according to the man-
ufacturer’s instructions. Relative luciferase activity was assessed as the 1. Qian F, Noben-Trauth K: Cellular and molecular function of mucolipins
ratio of renilla to firefly luciferase units (relative light units). (TRPML) and polycystin 2 (TRPP2). Pflugers Arch 451: 277–285, 2005
2. Bichet D, Peters D, Patel AJ, Delmas P, Honoré E: Cardiovascular
polycystins: insights from autosomal dominant polycystic kidney
Co-IP
disease and transgenic animal models. Trends Cardiovasc Med 16:
HEK or HeLa cell monolayers in 100-mm dishes were washed twice
292–298, 2006
with PBS and solubilized in ice-cold CellLytic-M lysis buffer (Sigma- 3. Wu G, D’Agati V, Cai Y, Markowitz G, Park JH, Reynolds DM, Maeda Y, Le
Aldrich) supplemented with proteinase inhibitor mixture. Superna- TC, Hou H Jr, Kucherlapati R, Edelmann W, Somlo S: Somatic inactivation
tants were collected after centrifugation at 16,000 3 g for 15 minutes. of Pkd2 results in polycystic kidney disease. Cell 93: 177–188, 1998
Equal amounts of total proteins from postnuclear supernatants were 4. Lu W, Peissel B, Babakhanlou H, Pavlova A, Geng L, Fan X, Larson C,
Brent G, Zhou J: Perinatal lethality with kidney and pancreas defects in
precleared for 1 hour with protein G-Sepharose, and then incubated
mice with a targetted Pkd1 mutation. Nat Genet 17: 179–181, 1997
for 4 hours at 4°C with the antibody against FUBP1. After the addi- 5. Kurbegovic A, Côté O, Couillard M, Ward CJ, Harris PC, Trudel M: Pkd1
tion of 100 ml of 50% protein G-Sepharose, the mixture was incu- transgenic mice: adult model of polycystic kidney disease with extra-
bated overnight with gentle shaking at 4°C. The immune complexes renal and renal phenotypes. Hum Mol Genet 19: 1174–1189, 2010
absorbed to protein G-Sepharose were washed five times with Non- 6. Park EY, Sung YH, Yang MH, Noh JY, Park SY, Lee TY, Yook YJ, Yoo KH,
idet P-40 lysis buffer (50 mM Tris, pH 7.5, 150 mM sodium chloride Roh KJ, Kim I, Hwang YH, Oh GT, Seong JK, Ahn C, Lee HW, Park JH:
Cyst formation in kidney via B-Raf signaling in the PKD2 transgenic
[NaCl], 1% Nonidet P-40) with proteinase inhibitor and eluted by
mice. J Biol Chem 284: 7214–7222, 2009
SDS loading buffer. Precipitated proteins were analyzed by WB using 7. Tran U, Zakin L, Schweickert A, Agrawal R, Döger R, Blum M, De
the antibodies against FUBP1 and 4EBP1. Robertis EM, Wessely O: The RNA-binding protein bicaudal C regu-
lates polycystin 2 in the kidney by antagonizing miR-17 activity. De-
GST Pull-Down velopment 137: 1107–1116, 2010
Two micrograms of purified GFP-tagged human 4EBP1 from E. coli was 8. Dutcher SK, Lin H: Tying TAZ and Nek1 into polycystic kidney disease
through polycystin 2 levels. J Am Soc Nephrol 22: 791–793, 2011
incubated with 2 mg purified GST-tagged human FUBP1 fragment from
9. González-Perrett S, Kim K, Ibarra C, Damiano AE, Zotta E, Batelli M,
E. coli in the binding buffer (50 mM Tris, pH 7.5, 150 mM NaCl, 1 mM Harris PC, Reisin IL, Arnaout MA, Cantiello HF: Polycystin-2, the protein
calcium chloride). The mixture was incubated at room temperature for mutated in autosomal dominant polycystic kidney disease (ADPKD), is a
1 hour with gentle shaking, followed by another hour of incubation after Ca2+-permeable nonselective cation channel. Proc Natl Acad Sci U S A
addition of 100 ml 50% glutathione-agarose beads (GE Healthcare). The 98: 1182–1187, 2001
10. Li Q, Montalbetti N, Shen PY, Dai XQ, Cheeseman CI, Karpinski E, Wu
beads were then washed several times with 140 mM NaCl, 10 mM
G, Cantiello HF, Chen XZ: Alpha-actinin associates with polycystin-2
Na2HPO4, 1.8 mM KH2PO4 pH 7.5, and the remaining proteins and regulates its channel activity. Hum Mol Genet 14: 1587–1603, 2005
were eluted using 13 SDS loading buffer and resolved by SDS-PAGE 11. Li X, Luo Y, Starremans PG, McNamara CA, Pei Y, Zhou J: Polycystin-1
and transferred to a nitrocellulose membrane (Bio-Rad, Hercules, CA). and polycystin-2 regulate the cell cycle through the helix-loop-helix
The membrane was then immunoblotted with GFP and GSTantibodies. inhibitor Id2. Nat Cell Biol 7: 1202–1212, 2005
12. Li Y, Wright JM, Qian F, Germino GG, Guggino WB: Polycystin 2 in-
teracts with type I inositol 1,4,5-trisphosphate receptor to modulate
Statistical Analyses intracellular Ca2+ signaling. J Biol Chem 280: 41298–41306, 2005
Data were analyzed and plotted using Sigmaplot 12 (Systat Software, 13. Ma R, Li WP, Rundle D, Kong J, Akbarali HI, Tsiokas L: PKD2 functions as
San Jose, CA), and expressed as mean6SEM (N), where N indicates an epidermal growth factor-activated plasma membrane channel. Mol
the number of experimental repeats. Paired or unpaired t tests were Cell Biol 25: 8285–8298, 2005
used to compare two sets of data. A probability value (P) of less than 14. Rundle DR, Gorbsky G, Tsiokas L: PKD2 interacts and co-localizes with
mDia1 to mitotic spindles of dividing cells: role of mDia1 IN PKD2 lo-
0.05, 0.01, and 0.001 was considered statistically significant and in-
calization to mitotic spindles. J Biol Chem 279: 29728–29739, 2004
dicated by *, **, and ***, respectively. 15. Tsiokas L, Kim E, Arnould T, Sukhatme VP, Walz G: Homo- and heter-
odimeric interactions between the gene products of PKD1 and PKD2.
Proc Natl Acad Sci U S A 94: 6965–6970, 1997
16. Tsiokas L, Arnould T, Zhu C, Kim E, Walz G, Sukhatme VP: Specific
ACKNOWLEDGMENTS association of the gene product of PKD2 with the TRPC1 channel. Proc
Natl Acad Sci U S A 96: 3934–3939, 1999
W.Z. is a recipient of the Alberta Innovates-Doctoral Graduate Stu- 17. Bouvrette DJ, Sittaramane V, Heidel JR, Chandrasekhar A, Bryda EC:
dent Scholarship. This work was supported by the Natural Sciences Knockdown of bicaudal C in zebrafish (Danio rerio) causes cystic kid-
neys: a nonmammalian model of polycystic kidney disease. Comp Med
and Engineering Research Council of Canada, the Kidney Foundation
60: 96–106, 2010
of Canada, the Canadian Institutes of Health Research (to X.Z.C.), and 18. Yang J, Zheng W, Wang Q, Lara C, Hussein S, Chen X-Z: Translational
the National Natural Science Foundation of China (Grant 81570648, up-regulation of polycystic kidney disease protein PKD2 by endo-
to X.Z.C.; Grant 31171412, to Y.C.). plasmic reticulum stress. FASEB J 27: 4998–5009, 2013

J Am Soc Nephrol 27: 2645–2657, 2016 FUBP1 Suppresses PKD2 Translation 2655
BASIC RESEARCH www.jasn.org

19. Jackson RJ, Hellen CU, Pestova TV: The mechanism of eukaryotic 39. Jiang ST, Chiou YY, Wang E, Lin HK, Lin YT, Chi YC, Wang CK, Tang MJ,
translation initiation and principles of its regulation. Nat Rev Mol Cell Li H: Defining a link with autosomal-dominant polycystic kidney disease
Biol 11: 113–127, 2010 in mice with congenitally low expression of Pkd1. Am J Pathol 168:
20. Gallie DR: A tale of two termini: a functional interaction between the 205–220, 2006
termini of an mRNA is a prerequisite for efficient translation initiation. 40. Markowitz GS, Cai Y, Li L, Wu G, Ward LC, Somlo S, D’Agati VD: Pol-
Gene 216: 1–11, 1998 ycystin-2 expression is developmentally regulated. Am J Physiol 277:
21. Varani G: Delivering messages from the 39 end. Proc Natl Acad Sci U S A F17–F25, 1999
98: 4288–4289, 2001 41. Lantinga-van Leeuwen IS, Leonhard WN, Dauwerse H, Baelde HJ, van
22. Gingras AC, Raught B, Sonenberg N: eIF4 initiation factors: effectors of Oost BA, Breuning MH, Peters DJ: Common regulatory elements in the
mRNA recruitment to ribosomes and regulators of translation. Annu polycystic kidney disease 1 and 2 promoter regions. Eur J Hum Genet
Rev Biochem 68: 913–963, 1999 13: 649–659, 2005
23. Ostareck DH, Ostareck-Lederer A, Shatsky IN, Hentze MW: Lipoxygenase 42. Duncan R, Bazar L, Michelotti G, Tomonaga T, Krutzsch H, Avigan M,
mRNA silencing in erythroid differentiation: The 3’UTR regulatory complex Levens D: A sequence-specific, single-strand binding protein activates
controls 60S ribosomal subunit joining. Cell 104: 281–290, 2001 the far upstream element of c-myc and defines a new DNA-binding
24. Mazumder B, Seshadri V, Imataka H, Sonenberg N, Fox PL: Trans- motif. Genes Dev 8: 465–480, 1994
lational silencing of ceruloplasmin requires the essential elements of 43. Kim MJ, Park BJ, Kang YS, Kim HJ, Park JH, Kang JW, Lee SW, Han JM,
mRNA circularization: poly(A) tail, poly(A)-binding protein, and eukaryotic Lee HW, Kim S: Downregulation of FUSE-binding protein and c-myc by
translation initiation factor 4G. Mol Cell Biol 21: 6440–6449, 2001 tRNA synthetase cofactor p38 is required for lung cell differentiation.
25. Sammarco MC, Grabczyk E: A series of bidirectional tetracycline-in- Nat Genet 34: 330–336, 2003
ducible promoters provides coordinated protein expression. Anal Bi- 44. Irwin N, Baekelandt V, Goritchenko L, Benowitz LI: Identification of two
ochem 346: 210–216, 2005 proteins that bind to a pyrimidine-rich sequence in the 39-untranslated
26. Sun H, Li QW, Lv XY, Ai JZ, Yang QT, Duan JJ, Bian GH, Xiao Y, Wang region of GAP-43 mRNA. Nucleic Acids Res 25: 1281–1288, 1997
YD, Zhang Z, Liu YH, Tan RZ, Yang Y, Wei YQ, Zhou Q: MicroRNA-17 45. Rabenhorst U, Beinoraviciute-Kellner R, Brezniceanu ML, Joos S,
post-transcriptionally regulates polycystic kidney disease-2 gene and Devens F, Lichter P, Rieker RJ, Trojan J, Chung HJ, Levens DL, Zörnig
promotes cell proliferation. Mol Biol Rep 37: 2951–2958, 2010 M: Overexpression of the far upstream element binding protein 1 in
27. Zhang J, Chen QM: Far upstream element binding protein 1: a com- hepatocellular carcinoma is required for tumor growth. Hepatology 50:
mander of transcription, translation and beyond. Oncogene 32: 2907– 1121–1129, 2009
2916, 2013 46. Li X, Magenheimer BS, Xia S, Johnson T, Wallace DP, Calvet JP, Li R: A
28. Sun Z, Amsterdam A, Pazour GJ, Cole DG, Miller MS, Hopkins N: A tumor necrosis factor-alpha-mediated pathway promoting autosomal
genetic screen in zebrafish identifies cilia genes as a principal cause of dominant polycystic kidney disease. Nat Med 14: 863–868, 2008
cystic kidney. Development 131: 4085–4093, 2004 47. Lanoix J, D’Agati V, Szabolcs M, Trudel M: Dysregulation of cellular
29. Giamarchi A, Feng S, Rodat-Despoix L, Xu Y, Bubenshchikova E, proliferation and apoptosis mediates human autosomal dominant
Newby LJ, Hao J, Gaudioso C, Crest M, Lupas AN, Honoré E, polycystic kidney disease (ADPKD). Oncogene 13: 1153–1160, 1996
Williamson MP, Obara T, Ong AC, Delmas P: A polycystin-2 (TRPP2) 48. Ricker JL, Mata JE, Iversen PL, Gattone VH: c-myc antisense oligonu-
dimerization domain essential for the function of heteromeric poly- cleotide treatment ameliorates murine ARPKD. Kidney Int 61[Suppl]:
cystin complexes. EMBO J 29: 1176–1191, 2010 S125–S131, 2002
30. Feng S, Okenka GM, Bai CX, Streets AJ, Newby LJ, DeChant BT, 49. Kusik BW, Hammond DR, Udvadia AJ: Transcriptional regulatory re-
Tsiokas L, Obara T, Ong AC: Identification and functional character- gions of gap43 needed in developing and regenerating retinal gan-
ization of an N-terminal oligomerization domain for polycystin-2. J Biol glion cells. Dev Dyn 239: 482–495, 2010
Chem 283: 28471–28479, 2008 50. Wang W, Furneaux H, Cheng H, Caldwell MC, Hutter D, Liu Y, Holbrook
31. Kong J, Lasko P: Translational control in cellular and developmental N, Gorospe M: HuR regulates p21 mRNA stabilization by UV light. Mol
processes. Nat Rev Genet 13: 383–394, 2012 Cell Biol 20: 760–769, 2000
32. Reeders ST: Multilocus polycystic disease. Nat Genet 1: 235–237, 1992 51. Dean JL, Sully G, Clark AR, Saklatvala J: The involvement of AU-rich
33. Cornec-Le Gall E, Audrézet MP, Le Meur Y, Chen JM, Férec C: Genetics element-binding proteins in p38 mitogen-activated protein kinase path-
and pathogenesis of autosomal dominant polycystic kidney disease: 20 way-mediated mRNA stabilisation. Cell Signal 16: 1113–1121, 2004
years on. Hum Mutat 35: 1393–1406, 2014 52. Olanich ME, Moss BL, Piwnica-Worms D, Townsend RR, Weber JD: Identi-
34. Pei Y, Watnick T, He N, Wang K, Liang Y, Parfrey P, Germino G, St fication of FUSE-binding protein 1 as a regulatory mRNA-binding protein that
George-Hyslop P: Somatic PKD2 mutations in individual kidney and represses nucleophosmin translation. Oncogene 30: 77–86, 2011
liver cysts support a “two-hit” model of cystogenesis in type 2 auto- 53. Weber A, Kristiansen I, Johannsen M, Oelrich B, Scholmann K, Gunia S,
somal dominant polycystic kidney disease. J Am Soc Nephrol 10: May M, Meyer HA, Behnke S, Moch H, Kristiansen G: The FUSE binding
1524–1529, 1999 proteins FBP1 and FBP3 are potential c-myc regulators in renal, but not
35. Koptides M, Hadjimichael C, Koupepidou P, Pierides A, Constantinou in prostate and bladder cancer. BMC Cancer 8: 369, 2008
Deltas C: Germinal and somatic mutations in the PKD2 gene of renal 54. Lu JY, Schneider RJ: Tissue distribution of AU-rich mRNA-binding
cysts in autosomal dominant polycystic kidney disease. Hum Mol Genet proteins involved in regulation of mRNA decay. J Biol Chem 279:
8: 509–513, 1999 12974–12979, 2004
36. Brasier JL, Henske EP: Loss of the polycystic kidney disease (PKD1) 55. Jang M, Park BC, Kang S, Chi SW, Cho S, Chung SJ, Lee SC, Bae KH,
region of chromosome 16p13 in renal cyst cells supports a loss-of- Park SG: Far upstream element-binding protein-1, a novel caspase
function model for cyst pathogenesis. J Clin Invest 99: 194–199, 1997 substrate, acts as a cross-talker between apoptosis and the c-myc on-
37. Lantinga-van Leeuwen IS, Dauwerse JG, Baelde HJ, Leonhard WN, van cogene. Oncogene 28: 1529–1536, 2009
de Wal A, Ward CJ, Verbeek S, Deruiter MC, Breuning MH, de Heer E, 56. Liu J, Chung HJ, Vogt M, Jin Y, Malide D, He L, Dundr M, Levens D:
Peters DJ: Lowering of Pkd1 expression is sufficient to cause polycystic JTV1 co-activates FBP to induce USP29 transcription and stabilize p53
kidney disease. Hum Mol Genet 13: 3069–3077, 2004 in response to oxidative stress. EMBO J 30: 846–858, 2011
38. Kim I, Li C, Liang D, Chen XZ, Coffy RJ, Ma J, Zhao P, Wu G: Polycystin-2 57. Bazar L, Harris V, Sunitha I, Hartmann D, Avigan M: A transactivator of
expression is regulated by a PC2-binding domain in the intracellular c-myc is coordinately regulated with the proto-oncogene during cellular
portion of fibrocystin. J Biol Chem 283: 31559–31566, 2008 growth. Oncogene 10: 2229–2238, 1995

2656 Journal of the American Society of Nephrology J Am Soc Nephrol 27: 2645–2657, 2016
www.jasn.org BASIC RESEARCH

58. Milosevic J, Bulau P, Mortz E, Eickelberg O: Subcellular fractionation of W, Fishman MC: Early development of the zebrafish pronephros and
TGF-beta1-stimulated lung epithelial cells: a novel proteomic approach for analysis of mutations affecting pronephric function. Development 125:
identifying signaling intermediates. Proteomics 9: 1230–1240, 2009 4655–4667, 1998
59. Roy B, Ferdous J, Ali DW: NMDA receptors on zebrafish Mauthner cells 63. Takagi M, Absalon MJ, McLure KG, Kastan MB: Regulation of p53
require CaMKII-a for normal development. Dev Neurobiol 75: 145– translation and induction after DNA damage by ribosomal protein L26
162, 2015 and nucleolin. Cell 123: 49–63, 2005
60. Brewster DL, Ali DW: Expression of the voltage-gated potassium 64. Rinn JL, Kertesz M, Wang JK, Squazzo SL, Xu X, Brugmann SA,
channel subunit Kv1.1 in embryonic zebrafish Mauthner cells. Neurosci Goodnough LH, Helms JA, Farnham PJ, Segal E, Chang HY: Functional
Lett 539: 54–59, 2013 demarcation of active and silent chromatin domains in human HOX loci
61. Karlstrom RO, Tyurina OV, Kawakami A, Nishioka N, Talbot WS, Sasaki by noncoding RNAs. Cell 129: 1311–1323, 2007
H, Schier AF: Genetic analysis of zebrafish gli1 and gli2 reveals di-
vergent requirements for gli genes in vertebrate development. De-
velopment 130: 1549–1564, 2003
62. Drummond IA, Majumdar A, Hentschel H, Elger M, Solnica-Krezel L, This article contains supplemental material online at http://jasn.asnjournals.
Schier AF, Neuhauss SC, Stemple DL, Zwartkruis F, Rangini Z, Driever org/lookup/suppl/doi:10.1681/ASN.2015070836/-/DCSupplemental.

J Am Soc Nephrol 27: 2645–2657, 2016 FUBP1 Suppresses PKD2 Translation 2657

You might also like