ArticlePDF Available

Key contribution of CPEB4mediated translational control to cancer progression

Authors:
  • Institut Hospital del Mar d'Investigacions Mèdiques (IMIM)

Abstract and Figures

Malignant transformation, invasion and angiogenesis rely on the coordinated reprogramming of gene expression in the cells from which the tumor originated. Although deregulated gene expression has been extensively studied at genomic and epigenetic scales, the contribution of the regulation of mRNA-specific translation to this reprogramming is not well understood. Here we show that cytoplasmic polyadenylation element binding protein 4 (CPEB4), an RNA binding protein that mediates meiotic mRNA cytoplasmic polyadenylation and translation, is overexpressed in pancreatic ductal adenocarcinomas and glioblastomas, where it supports tumor growth, vascularization and invasion. We also show that, in pancreatic tumors, the pro-oncogenic functions of CPEB4 originate in the translational activation of mRNAs that are silenced in normal tissue, including the mRNA of tissue plasminogen activator, a key contributor to pancreatic ductal adenocarcinoma malignancy. Taken together, our results document a key role for post-transcriptional gene regulation in tumor development and describe a detailed mechanism for gene expression reprogramming underlying malignant tumor progression.
In vitro characterization of the effects of CPEB4 knockdown in RWP-1 cells.(a) CPEB4 mRNA expression in parental RWP-1 cells and in cells transfected with shCtrl or four different CPEB4-targeting shRNAs (shCPEB4_1 through shCPEB4_4). Values correspond to three independent experiments. Data are mean ± s.e.m. The P values (determined by Student's t test) are relative to shCtrl cells. *P ≤ 0.05. shCPEB4_2 and shCPEB4_4 showed the greatest reduction of CPEB4 concentrations and were selected for further experiments. (b) Downregulation of CPEB4 protein concentrations in control cells and in cells transfected with shCPEB4, as analyzed by western blot. Tubulin concentrations are shown as the loading control. One representative experiment (n = 3) is shown. (c) Proliferation rate of parental RWP-1 cells or cells harboring shCtrl or shCPEB4 analyzed using a 3-(4.5-dimethylthiazol-2-yl)-2.5-diphenyltetrazolium bromide (MTT) assay (three independent experiments). Data are mean ± s.e.m. Abs 570, absorbance at 570 nm. (d) Matrigel invasion of parental, shCtrl and shCPEB4 RWP-1 cells. Invasion (after 72 h) was measured by absorbance at 590 nm (Abs 590) (three independent experiments). Data are mean ± s.e.m. The P values (determined by Student's t test) are relative to shCtrl cells. ***P ≤ 0.001. (e) Soft agar colony formation of parental RWP-1 cells and cells expressing shCtrl or shCPEB4. The graph shows the number of colonies (mean ± s.e.m.) after 4 weeks of three independent experiments. The P values (determined by Student's t test) are relative to shCtrl cells. **P ≤ 0.01. Shown at the right are representative images of the colonies formed by each type of cell.
… 
Effects of CPEB4 knockdown in vivo on tumorigenicity in nude mice.(a) Tumorigenicity of untransfected, shCtrl, shCPEB4_2 and shCPEB4_4 RWP-1 cells (1 × 106) after subcutaneous injection in the flanks of nude mice (n = 10). One representative mouse and the tumors it formed are shown. The quantification of tumor weight and volume is shown on the right. Box and whisker plots show the mean, quartiles and tenth and ninetieth percentiles of the data. The P values (determined by Student's t test) are relative to tumors formed by shCtrl cells. *P ≤ 0.05, **P ≤ 0.01, ***P ≤ 0.001. Scale bars, 1 cm. (b) Quantification of the tumor weight and volume of untransfected, shCtrl, shCPEB4_2 and shCPEB4_4 Capan-1 cells (1 × 106) after subcutaneous injection in the flanks of nude mice (n = 8). Box and whisker plots show the mean, quartiles and tenth and ninetieth percentiles of the data. The P values (determined by Student's t test) are relative to tumors formed by shCtrl cells. *P ≤ 0.05, **P ≤ 0.01. (c) RWP-1 LUC cells were injected intraperitoneally, and bioluminescent activity was measured weekly. Two representative mice from each group (n = 10) after 3 weeks of injection are shown. Scale bars, 1 cm. (d) Kaplan-Meier survival curves in tumor-bearing nude mice injected with control or knockdown CPEB4 (shCPEB4) RWP-1 LUC cells. Control denotes the group of mice injected with untransfected or shCtrl RWP-1 LUC cells (n = 19), and shCPEB4 denotes the group of mice injected with shCPEB4_2 and shCPEB4_4 RWP-1 LUC cells (n = 19). The P values (determined by log-rank test) are relative to mice from the control group. *P ≤ 0.05.
… 
PA expression is regulated at the translational level by CPEB4 in normal and tumoral pancreatic tissues.(a) Relative tPA mRNA levels, compared to HPRT, detected by qRT-PCR in normal human acinar and ductal pancreatic cells, PDA and endocrine tumors. Data are mean ± s.e.m. (b) Analysis of the polyadenylation of tPA transcripts from mouse normal pancreas and ductal tumor from Ela-myc mice (top) or normal human pancreas and RWP-1 and SK-PC-1 cells (bottom). At the top, the complementary DNA was detected by labeling with [32P] γ-dATP followed by autoradiography. 1 indicates the internal control of tPA mRNA expression, and 2 indicates polyadenylation. A20 and A90 indicate the respective length of poly(A) tail. (c) A schematic representation of the 3′ UTR of human tPA mRNA (top) and luciferase Renilla activity (bottom) of synthetic chimeric mRNAs containing the firefly luciferase coding sequence fused upstream from the indicated tPA 3′ UTRs injected into oocytes. The intact 3′ UTR of cyclin B1 and mutants of the three CPEs therein were used as controls. Translation of the nonrepressed control mRNA was adjusted to 100%. (d) Polyadenylation analysis of tPA transcripts from untransfected, shCtrl or shCPEB4 RWP-1 cells. (e) tPA RNA and protein expression after CPEB4 knockdown in RWP-1 cells. HPRT (for RNA) or tubulin (protein) was used for normalization. Data are mean ± s.e.m. (f) tPA immunohistochemistry in tumors from nude mice after subcutaneous or intraperitoneal injection of RWP-1 untransfected, shCtrl or shCPEB4 cells. Scale bars, 100 μm. At the bottom is quantification of tPA knockdown in tumors, calculated by the percentage of tPA staining intensity and compared to shCtrl tumors. The P values (determined by Student's t test) relative to shCtrl are shown. ***P ≤ 0.001. Data are mean ± s.e.m. One representative experiment (n = 3) is shown (a–d).
… 
Content may be subject to copyright.
articles
nature medicineVOLUME 18 | NUMBER 1 | JANUARY 2012 83
Reprogramming of gene expression is a key step during cancer develop-
ment in which the aberrant control of genes primarily related to cell
proliferation, apoptosis, metabolism and invasion sets up the basis for
malignant transformation1. This phenomenon has been extensively
studied at the level of transcription, but it is becoming evident that
the post-transcriptional regulation of specific mRNA subpopulations
contributes substantially to the broad expression changes of the genes
responsible for tumoral properties of cells2–5. Coordinate regula-
tion of mRNA subpopulations is mediated by common cis-acting
elements in their 3 untranslated regions (UTRs). This is the case for
the cytoplasmic polyadenylation element (CPE), which is present in
hundreds of mRNAs involved in cell proliferation, chromosome seg-
regation and cell differentiation6–9. CPE is bound by the CPE binding
proteins (CPEBs)10,11. CPEBs can act as either translational repressors
or activators9–13 to regulate mitotic and meiotic cell cycles and senes-
cence9,14–16, although no direct link between cancer and CPEBs has
been found. To investigate the oncogenic potential of CPEBs, we ana-
lyzed pancreatic ductal adenocarcinoma (PDA) and glioblastoma as
model systems. PDA and glioblastomas are the solid tumors with the
worst clinical prognoses, and they have been mainly studied at the
level of transcriptional regulation17,18.
We report that CPEB4 is overexpressed in PDA and that its down-
regulation in mouse xenograft models results in a marked reduction
of tumorigenic properties, primarily proliferation and vascularization,
resulting in increased survival. We also show that CPEB4 is asso-
ciated with a large number of transcripts in cancer cells, and, in
the case of tissue plasminogen activator (tPA) mRNA, the tumor-
associated overexpression of CPEB4 results in poly(A) tail elongation
and translational activation, leading to abnormal overexpression of
tPA in tumors. Our results indicate that the high amounts of CPEB4
in PDA result in a pathological reprogramming of gene expression
that drives the translational activation of mRNAs encoding factors
that sustain tumor progression. In addition, we found that CPEB4 is
also overexpressed in glioblastoma and that overexpression supports
tumoral proliferation and angiogenesis, arguing that this is a general
mechanism of gene expression regulation in cancer. These findings
may therefore hold diagnostic and therapeutic implications for PDA,
glioblastoma and, possibly, other tumor types.
RESULTS
CPEB4 is overexpressed in human PDA
To address whether CPEB-mediated mRNA regulation could have a
role in PDA, we first assessed CPEB expression in a panel of human
cell lines and tissues. Of the CPEB proteins tested, only CPEB4 was
clearly associated with the tumoral phenotype, with five out of seven
tumoral cell lines showing high amounts of CPEB4 mRNA and
1Cancer Research Programme, Hospital del Mar Research Institute (IMIM), Barcelona, Spain. 2Molecular Pathology Programme, Spanish National Cancer
Research Centre (CNIO), Madrid, Spain. 3Department of Experimental and Health Sciences, Universitat Pompeu Fabra (UPF), Barcelona, Spain. 4Center for
Genomic Regulation (CRG), Barcelona, Spain. 5Institute for Research in Biomedicine (IRB Barcelona), Barcelona, Spain. 6Department of Pathology, Hospital del
Mar, Parc de Salut Mar, Universitat Autónoma de Barcelona (UAB), Barcelona, Spain. 7Computational Genomics Group, Universitat Pompeu Fabra (UPF), Barcelona,
Spain. 8Institució Catalana de Recerca i Estudis Avançats (ICREA), Barcelona, Spain. Present addresses: Richard Dimbleby Department of Cancer Research,
King’s College London, London, UK (E.O.-Z.) and Department of Anatomy and Structural Biology, Albert Einstein College of Medicine, Bronx, New York, USA (C.E.).
Correspondence should be addressed to R.M. (raul.mendez@irbbarcelona.es) or P.N. (pnavarro@imim.es).
Received 7 February; accepted 28 September; published online 4 December 2011; doi:10.1038/nm.2540
Key contribution of CPEB4-mediated translational
control to cancer progression
Elena Ortiz-Zapater1–3, David Pineda1,4, Neus Martínez-Bosch1, Gonzalo Fernández-Miranda5,
Mar Iglesias1,6, Francesc Alameda6, Mireia Moreno1, Carolina Eliscovich4, Eduardo Eyras7,8,
Francisco X Real1–3, Raúl Méndez4,5,8 & Pilar Navarro1
Malignant transformation, invasion and angiogenesis rely on the coordinated reprogramming of gene expression in the cells
from which the tumor originated. Although deregulated gene expression has been extensively studied at genomic and epigenetic
scales, the contribution of the regulation of mRNA-specific translation to this reprogramming is not well understood. Here we
show that cytoplasmic polyadenylation element binding protein 4 (CPEB4), an RNA binding protein that mediates meiotic mRNA
cytoplasmic polyadenylation and translation, is overexpressed in pancreatic ductal adenocarcinomas and glioblastomas, where
it supports tumor growth, vascularization and invasion. We also show that, in pancreatic tumors, the pro-oncogenic functions
of CPEB4 originate in the translational activation of mRNAs that are silenced in normal tissue, including the mRNA of tissue
plasminogen activator, a key contributor to pancreatic ductal adenocarcinoma malignancy. Taken together, our results document
a key role for post-transcriptional gene regulation in tumor development and describe a detailed mechanism for gene expression
reprogramming underlying malignant tumor progression.
npg © 2012 Nature America, Inc. All rights reserved.
articles
84  VOLUME 18 | NUMBER 1 | JANUARY 2012 nature medicine
protein compared to a non-tumorigenic pancreatic cell line (HPDE)
(Fig. 1a,b). We also assessed CPEB4 concentrations in vivo using a set
of 190 samples from a cohort of 58 patients (Fig. 1c,d). The samples
we analyzed were from normal pancreas (n = 47), tissue with pancrea-
titis (n = 51), pancreatic intraepithelial neoplasia (PanIN) lesions (54)
and pancreatic adenocarcinomas (n = 38, with 27 well or moderately
differentiated adenocarcinomas and 11 undifferentiated adenocar-
cinomas) (Supplementary Table 1). In normal pancreas, CPEB4
expression was restricted to the islets (100%) and, occasionally, to
ductal cells (25.5%). In the inflammatory atrophic pancreas, CPEB4
was expressed in 19.6% of ducts. Notably, the expression of CPEB4
in ductal cells increased during malignant transformation, showing a
low-to-moderate frequency (34.6%) in low-grade PanINs and stronger
expression and higher frequency in high-grade PanINs (71.4%) and
in moderately differentiated or well-differentiated PDAs (88.8%).
CPEB4 expression was reduced in undifferentiated tumors (36.3%).
Taken together, these results show in vitro and in vivo correlations
between CPEB4 expression and tumoral phenotype in human PDA,
suggesting a function for this protein during tumorigenesis.
Downregulation of CPEB4 reduces malignancy in vitro
To address the relevance of CPEB4 overexpression in tumorigenesis,
we knocked down its expression in the tumoral pancreatic cell line
RWP-1 by transfection with CPEB4 shRNA (shCPEB4). We used cells
transfected with scrambled shRNA (shCtrl) or parental cells (untrans-
fected) as controls. We monitored the efficiency of the CPEB4 down-
regulation by quantitative RT-PCR (qRT-PCR) and western blotting
(Fig. 2a,b). The proliferation rate was similar in untransfected, shCtrl
and shCPEB4 cells (Fig. 2c). In contrast, both Matrigel invasion
(Fig. 2d) and soft-agar colony formation (Fig. 2e) (used as indica-
tors of malignant transformation) were strongly reduced after CPEB4
knockdown, indicating that CPEB4 may favor tumor progression by
increasing invasiveness and anchorage-independent growth.
CPEB4 knockdown leads to reduced in vivo tumorigenicity
To determine whether CPEB4 is required for tumor progression
in vivo, we analyzed the tumorigenic properties of RWP-1 parental
cells, shCtrl cells and two different types of shCPEB4 cells (shCPEB4_2
and shCPEB4_4), which we injected subcutaneously in the flanks of
nude mice. We killed all animals 3 weeks after injection and confirmed
CPEB4 downregulation by immunohistochemistry (Supplementary
Fig. 1a,b). We found a 51% (shCPEB4_2) and a 57% (shCPEB4_4)
reduction in tumor weight and a 67% (shCPEB4_2) and an 80%
(shCPEB4_4) reduction in tumor volume after CPEB4 knockdown
(Fig. 3a). To rule out a cell-line–specific phenotype, we also down-
regulated CPEB4 expression in cells from the pancreatic cancer line
Capan-1 (Supplementary Fig. 2) and tested the effects on in vivo
tumorigenicity, which confirmed a significant decrease of tumor
weight and volume (Fig. 3b). These data indicate that CPEB4 down-
regulation in pancreatic tumoral cells results, directly or indirectly,
in slower tumor growth.
Peritoneal dissemination is one of the primary mechanisms in the
progression of pancreatic cancer19. Therefore, we analyzed tumor
formation after intraperitoneal injection of RWP-1 cells transduced
with luciferase (RWP-1 LUC cells). We killed the mice injected
with these cells when their tumors grew to over 1.5 cm in diameter
and confirmed CPEB4 downregulation by immunohistochemistr y
(Supplementary Fig. 1a,b). Luciferase activity detection showed a
clear delay of tumor formation in the mice injected with shCPEB4
cells (Fig. 3c and Supplementar y Fig. 3a). Even though they had
very different kinetics and size, our postmortem analysis showed
that both control and CPEB4 knockdown cells generated tumors
that were localized to the pancreas (with 84.2% frequency in both
groups). In the in vitro invasion samples (Fig. 2d), we consistently
found a clear trend in the control cells to develop tumors in other
organs (including the intestine, liver, kidney, stomach and ovary) at
a higher frequency than CPEB4 knockdown cells (73.6% compared
to 47.36%, respectively) (Supplementary Fig. 3b), suggesting an
impaired invasive capacity in the absence of CPEB4. Kaplan-Meier
survival curves showed that CPEB4 knockdown was associated with
a significant delay in mouse mortality (P = 0.024) (Fig. 3d). These
data show that pancreatic tumoral cells with low CPEB4 expression
have slower tumor growth and a lower capability to form tumors in
non-pancreatic tissues, resulting in a slower progression to death.
Figure 1 CPEB4 is overexpressed in PDA
cell lines and tumor tissues. (a) Expression
of CPEB4 mRNA detected by qRT-PCR in
a panel of human pancreatic cell lines.
Values, compared to hypoxanthine-guanine
phosphoribosyltransferase (HPRT) mRNA levels,
correspond to three independent experiments
Data are mean ± s.e.m. The P values
(determined by Student’s t test) are relative
to HPDE cells. *P 0.05, **P 0.01,
***P 0.001. (b) Analysis by immuno-
fluorescence of CPEB4 expression in
immortalized normal pancreatic ductal cells
(HPDE) and PDA (RWP-1 and SK-PC-1).
Scale bars, 40 µm. (c) CPEB4 expression
determined by immunohistochemistry in sections
of formalin-fixed paraffin-embedded normal
human pancreas, samples with pancreatitis,
low- and high-grade PanIN lesions and
differentiated and undifferentiated tumors.
Scale bars, 200 µm (for normal pancreas
and differentiated and undifferentiated
adenocarcinoma) or 50 µm (for pancreatitis and
low- and high-grade PanIN). (d) Quantification of CPEB4 protein expression in human pancreatic tissue samples according to histology. The stacked bars show
the percent contribution of high and low CPEB4-positive samples (Supplementary Table 1, and see the legend of Supplementary Table 1 for the classification
of high and low samples) relative to the total number of samples in each pancreatic lesion. ***P 0.001. Diff., differentiated; undiff., undifferentiated.
c
d
bHPDE RWP-1 SK-PC-1
120
100
80
60
40
Percentage of
CPEB4-positive samples
20
0
Normal
ducts
Atrophic
ducts
Low-grade
PanINs
High-grade
PanINs
Diff.
tumor
Undiff.
tumor
*** ***
High
Low
25
a
20
15
10 *
** **
*** *
*
5
Relative CPEB4
mRNA expression
0
HPDE
IMIM-PC-2
IMIM-PC-1
PANC-1
RWP-1
SK-PC-1
SK-PC-3
Capan-1
Normal pancreas
High-grade
PanIN
Pancreatitis
Differentiated
adenocarcinoma
Undifferentiated
adenocarcinoma
Low-grade PanIN
npg © 2012 Nature America, Inc. All rights reserved.
articles
nature medicineVOLUME 18 | NUMBER 1 | JANUARY 2012 85
CPEB4 knockdown reduces proliferation and vascularization
We next aimed to determine whether the differences in the growth
rates of the tumors were a result of the tumor architecture rather
than the intrinsic properties of the tumoral cells. A histopathological
characterization of tumors that developed after subcutaneous
(Fig. 4a,b) and intraperitoneal injection (Supplementary Fig. 4)
1.2
abcd
1.0
0.8
*****
***
*
*
0.6
0.4
Relative CPEB4 mRNA expression
Abs 570
Abs 590
0.2
0
2.50 0.8
0.6
0.4
0.2
0
2.00
1.50
1.00
0.50
0
1234
Time in culture (d)
5678
shCtrl 1 2 3
shCPEB4
4
CPEB490
Tubulin
Untransfected
shCtrl
shCPEB4_2
shCPEB4_4
Untransfected
shCtrl
shCPEB4_2
shCPEB4_4
Untransfected
shCtrl
shCPEB4_2
shCPEB4_4
55
e
**
**
Number of colonies
60
50
40
30
20
10
0
Untransfected
Untransfected
shCtrl
shCtrl
shCPEB4_2
shCPEB4_4
shCPEB4_2 shCPEB4_4
Figure 2 In vitro characterization of the effects of CPEB4 knockdown in RWP-1 cells.
(a) CPEB4 mRNA expression in parental RWP-1 cells and in cells transfected with shCtrl
or four different CPEB4-targeting shRNAs (shCPEB4_1 through shCPEB4_4). Values
correspond to three independent experiments. Data are mean ± s.e.m. The P values
(determined by Student’s t test) are relative to shCtrl cells. *P 0.05. shCPEB4_2 and
shCPEB4_4 showed the greatest reduction of CPEB4 concentrations and were selected for
further experiments. (b) Downregulation of CPEB4 protein concentrations in control cells
and in cells transfected with shCPEB4, as analyzed by western blot. Tubulin concentrations
are shown as the loading control. One representative experiment (n = 3) is shown. (c) Proliferation
rate of parental RWP-1 cells or cells harboring shCtrl or shCPEB4 analyzed using a 3-(4.5-
dimethylthiazol-2-yl)-2.5-diphenyltetrazolium bromide (MTT) assay (three independent
experiments). Data are mean ± s.e.m. Abs 570, absorbance at 570 nm. (d) Matrigel invasion
of parental, shCtrl and shCPEB4 RWP-1 cells. Invasion (after 72 h) was measured by absorbance at 590 nm (Abs 590) (three independent
experiments). Data are mean ± s.e.m. The P values (determined by Student’s t test) are relative to shCtrl cells. ***P 0.001. (e) Soft agar colony
formation of parental RWP-1 cells and cells expressing shCtrl or shCPEB4. The graph shows the number of colonies (mean ± s.e.m.) after 4 weeks
of three independent experiments. The P values (determined by Student’s t test) are relative to shCtrl cells. **P 0.01. Shown at the right are
representative images of the colonies formed by each type of cell.
Untransfected
a b
1.0 0.5
0.4
0.3
0.2
0.1
0
1.2
0.9
0.6
0.3
0
0.8
0.6 **
** **
**
***
0.4
Weight (g)
Weight (g)
Volume (cm3)
0.2
0
Untransfected
shCtrl shCPEB4_2 shCPEB4_4
shCtrl
shCPEB4_2
shCPEB4_4
Untransfected
shCtrl
shCPEB4_2
shCPEB4_4
Untransfected
shCtrl
shCPEB4_2
shCPEB4_4
1.2
0.9
0.6
0.3
0
**
**
Volume (cm3)
Untransfected
shCtrl
shCPEB4_2
shCPEB4_4
c d
*
Untransfected shCtrl
shCPEB4_2 shCPEB4_4
1.0
0.8
0.6
0.4
0.2
Control
shCPEB4
0
0 10 20 30 40
Time (d)
50 60
Cumulative survival
Figure 3 Effects of CPEB4 knockdown in vivo on tumorigenicity in nude mice.
(a) Tumorigenicity of untransfected, shCtrl, shCPEB4_2 and shCPEB4_4 RWP-1
cells (1 × 106) after subcutaneous injection in the flanks of nude mice (n = 10).
One representative mouse and the tumors it formed are shown. The quantification of
tumor weight and volume is shown on the right. Box and whisker plots show the mean,
quartiles and tenth and ninetieth percentiles of the data. The P values (determined
by Student’s t test) are relative to tumors formed by shCtrl cells. *P 0.05, **P 0.01,
***P 0.001. Scale bars, 1 cm. (b) Quantification of the tumor weight and volume
of untransfected, shCtrl, shCPEB4_2 and shCPEB4_4 Capan-1 cells (1 × 106) after
subcutaneous injection in the flanks of nude mice (n = 8). Box and whisker plots
show the mean, quartiles and tenth and ninetieth percentiles of the data. The P values
(determined by Student’s t test) are relative to tumors formed by shCtrl cells. *P 0.05,
**P 0.01. (c) RWP-1 LUC cells were injected intraperitoneally, and bioluminescent
activity was measured weekly. Two representative mice from each group (n = 10) after
3 weeks of injection are shown. Scale bars, 1 cm. (d) Kaplan-Meier survival curves in tumor-bearing nude mice injected with control or knockdown
CPEB4 (shCPEB4) RWP-1 LUC cells. Control denotes the group of mice injected with untransfected or shCtrl RWP-1 LUC cells (n = 19), and shCPEB4
denotes the group of mice injected with shCPEB4_2 and shCPEB4_4 RWP-1 LUC cells (n = 19). The P values (determined by log-rank test) are relative
to mice from the control group. *P 0.05.
npg © 2012 Nature America, Inc. All rights reserved.
articles
86  VOLUME 18 | NUMBER 1 | JANUARY 2012 nature medicine
showed that tumors from shCPEB4 cells had a reduced prolifera-
tion rate (determined by Ki67 labeling) and decreased microvessel
density (by von Willebrand factor (vWF) labeling) compared to
parental and shCtrl RWP-1 cells. Notably, tumors from shCPEB4
cells showed less ischemic necrotic areas and increased stroma, as
detected by α smooth muscle actin (α-SMA) staining (Fig. 4c). The
fact that we found a similar increase in stroma formation in tumors
derived from subcutaneous and intraperitoneal injection indicates
that this increase is not a consequence of tumor size (as, in the mice
injected intraperitoneally, we collected all tumors when they reached
~1.5 cm3) or the time required to develop the tumor (as, in the mice
injected subcutaneously, we killed them all at the same time) but,
rather, was an effect directly related to low CPEB4 expression that
resulted in an increased amount of tumor-associated fibrosis. We
also observed reduced cell proliferation, tumoral angiogenesis and
increased stroma formation in tumors from Capan-1 shCPEB4 cells
(Supplementary Fig. 5).
These results indicate that, although CPEB4 is not required in a
cell-autonomous manner for cellular proliferation, reduced amounts
of CPEB4 affect both the proliferation rate of tumors and their archi-
tecture by reducing vascularization and increasing stroma formation,
with the consequence of reduced tumor growth and invasion during
in vivo pancreatic tumorigenesis.
CPEB4 associates with a large number of CPE-containing mRNAs
We next aimed to determine whether the overexpression of CPEB4
in PDA resulted in the abnormal translational activation of mRNAs
encoding pro-tumoral’ factors. To obtain a genome-wide picture
of potential CPEB4 targets in PDA, we performed RNA immuno-
precipitation (RIP) of CPEB4 from RWP-1 cell extracts and iden-
tified the associated mRNAs by Illumina Solexa ultrasequencing
(Supplementar y Fig. 6). We identified 842 mRNAs significantly
(P < 0.05) associated with CPEB4 when compared with the control
RNAs immunoprecipitated with IgG (Supplementary Tables 2 and 3).
Untransfected
a
b
c
vWF
100 8
6
4
2
0
*** *** *
*
**
** **
80
60
40
20
Percentage of
Ki67-positive cells
Percentage of area with
positive vWF staining
0
Subcutaneous Intraperitoneal
Ki67
**
*
***
100 µm100 µm100 µm
100 µm100 µm
*
*
*
*
*
**
**
*
*
shCtrl shCPEB4_2 shCPEB4_4
Untransfected
shCtrl
shCPEB4_2
shCPEB4_4
Untransfected
shCtrl
shCPEB4_2
shCPEB4_4
Subcutaneous Intraperitoneal
Subcutaneous Intraperitoneal
Untransfected
shCtrl
shCPEB4_2
shCPEB4_4
Untransfected
shCtrl
shCPEB4_2
shCPEB4_4
Untransfected
H&E
α-SMAα-SMA H&E
shCtrl shCPEB4_2 shCPEB4_4
Figure 4 Effects of CPEB4 knockdown on proliferation, vessel density and stroma in xenograft tumors. (a) Immunostaining of Ki67 and vWF in tumors
formed by untransfected, shCtrl or shCPEB4 (shCPEB4_2 and shCPEB4_4) RWP-1 cells injected subcutaneously in nude mice. Scale bars, 100 µm.
(b) Quantification of Ki67 and vWF immunohistochemistry staining shown by the percentage of positively stained cells compared to total number of
cells per field. The P values (determined by Student’s t test) relative to shCtrl are shown. *P 0.05, **P 0.01, ***P 0.001. Data are mean ± s.e.m.
(c) H&E and α-SMA staining of tissue sections of the different tumors after subcutaneous and intraperitoneal injection of untransfected, shCtrl or
shCPEB4 RWP-1 cells. Tumors from shCPEB4 cells showed less ischemic necrotic areas (asterisks) and more stromal component between tumoral cells
(arrowheads and α-SMA staining). Scale bars, 200 µm.
b
0.5 1.0 120
100
80
60
40
20
0
0.8
0.6
0.4
0.2
0
0.4
0.3
0.2
0.1
Percent of inputPercent of input
Percent of input
Percent of input
Percent of input
Percent of input
Percent of input
Percent of input
Percent of input
0
4.0
3.0
2.0
1.0
0
4.0
3.0
2.0
1.0
0
4.0
3.0
2.0
1.0
0
4.0 8.0
6.0
4.0
2.0
0
3.0
2.0
1.0
0
0.5
0.4
0.3
0.2
0.1
0
MT2A MT1E tPA OAZ2 RPS27
lgG
CPEB4
lgG
CPEB4
lgG
CPEB4
lgG
CPEB4
lgG
CPEB4
Percent of input
0.5
0.4
0.3
0.2
0.1
0
TMOD3PORCNORC4LH2AFVIDH1
lgG
CPEB4
lgG
CPEB4
lgG
CPEB4
lgG
CPEB4
lgG
CPEB4
0.9
a
0.8
0.7
Proportion above
enrichment score threshold
0.6
0.5
–4 –3 –2 –1 0
Enrichment score
1 2 3
0.9
0.8
0.7
Proportion above
enrichment score threshold
0.6
0.5
–4 –3 –2 –1 0
Enrichment score
1 2 3 4
Figure 5 Illumina Solexa sequencing analysis of mRNAs bound to CPEB4
in RWP-1 cells. (a) Representation of the proportion of mRNAs with
activation CPEs having an enrichment score greater than or equal to each
cutoff value on the x-axis. At right is a representation of the proportion
of mRNAs with repression CPEs having an enrichment score greater
than or equal to each cutoff value on the x-axis. The proportions were
calculated over the total number of mRNAs for which we could find a CPE
of either type. (b) Validation by qRT-PCR of mRNAs bound to CPEB4. The
fold enrichment of target sequences in the CPEB4 immunoprecipitates
compared to IgG and to the input fraction is shown. Data are mean ± s.e.m.
npg © 2012 Nature America, Inc. All rights reserved.
articles
nature medicineVOLUME 18 | NUMBER 1 | JANUARY 2012 87
The 3 UTRs of the mRNAs coimmunoprecipitated with CPEB4
mRNAs were enriched for activation arrangements of CPEs that are
able to recruit the polyadenylation complex6. In contrast, repression
CPE arrangements, which can assemble the translational repression
complex but not recruit the cytoplasmic polyadenylation machinery6,
were under-represented (Fig. 5a). A gene ontology analysis of the
CPEB4-associated mRNAs (Supplementary Fig. 7) showed a signifi-
cant (P 0.05) enrichment in cellular functions relevant to tumor-
igenesis (cell cycle, transcriptional regulation, apoptosis and DNA
damage); the most significant enrichment was for mRNAs encod-
ing general translation factors and ribosomal proteins, which are
differentially regulated and are key mediators of cellular transfor-
mation5,20. We further validated ten of the mRNAs associated with
CPEB4 with a validation rate of 90% (Fig. 5b and Supplementary
Table 4). Notably, the only mRNA identified in the original RIP
ultrasequencing that was not validated in the RIP qRT-PCR analysis
(RPS27) was also the only mRNA that did not contain CPEs. These
results indicate that, when overexpressed in PDA, CPEB4 is associ-
ated with a large number of mRNAs that are potential targets for
tumor-specific translational regulation.
tPA expression in pancreas is regulated by CPEB4
To test whether the association of these transcripts with CPEB4
resulted in their differential regulation in PDA, we next analyzed
in detail the relevance of CPEB4 in the translation of tPA mRNA,
one of the transcripts that was more enriched in the CPEB4
coimmunoprecipitation (Supplementary Table 2) and that is known
to be overexpressed in pancreatic tumors21–27. In addition, tPA mRNA
has been previously found to be regulated by CPEB1-mediated cyto-
plasmic polyadenylation in mouse oocytes and neurons28,29. When we
analyzed tPA mRNA expression in PDA compared to normal acinar
or ductal cells, we saw a clear discrepancy between the amounts
of protein and mRNA. Thus, tPA mRNA levels were high both in
normal ductal cells, in which tPA protein is absent, and in PDAs,
which express high amounts of tPA protein (Fig. 6a), suggesting that
tPA mRNA is stored silently in normal pancreatic ducts and is trans-
lationally activated in PDA.
To assess whether tPA mRNA was translationally regulated by cyto-
plasmic polyadenylation, we measured the length of the tPA mRNA
poly(A) tails in normal and tumoral samples by anchored RT-PCR6.
In normal pancreas, tPA mRNA had a short poly(A) tail, whereas
a b c
ef
d
3 UTR of tPA
ARE
PUM
Cyclin B1
CPE
tPA
CPE
CPE CPE HEX
CPE
CPE CPE
CPE HEX
ARE HEX
020 40 60 80 100 120 140
Translation stimulation + progesterone
rey or Renilla Luc
(percentage C: B1)
HEX
PUM
ARE
ARE
ARE
AREARE
ARE CPE CPE HEX
HEX
HEX
CPE
70
Relative tPA mRNA expression
60
50
40
30
20
10
Acinar Ductal
Normal
PDA
Tumor
Endocrine
64
shCtrl
shCPEB4_2
shCPEB4_4
tPA (supernatants)
tPA (total extracts)
Tubulin
64
55
Relative tPA mRNA
expression
1.2
1.0
0.8
0.6
shCtrl 2 4
shCPEB4
0.4
0.2
0
shCtrl
SubcutaneousIntraperitoneal
shCPEB4_2 shCPEB4_4
shCtrl 2 4
*** ***
shCPEB4
Percent of tPA
staining
120
100
80
60
40
20
0shCtrl 2 4
***
***
shCPEB4
Percent of tPA
staining
120
100
80
60
40
20
0
A90
A20
1 2
Normal Tumor
1 2
A90
A20
Wild type
RWP-1
SK-PC-1
Untransfected
shCtrl
shCPEB4_2
shCPEB4_4
Untransfected
shCtrl
shCPEB4_2
shCPEB4_4
tPA mRNA
tPA mRNA
polyadenylation
A90
A20
A0
Figure 6 tPA expression is regulated at the
translational level by CPEB4 in normal and
tumoral pancreatic tissues. (a) Relative tPA
mRNA levels, compared to HPRT, detected
by qRT-PCR in normal human acinar and
ductal pancreatic cells, PDA and endocrine
tumors. Data are mean ± s.e.m. (b) Analysis
of the polyadenylation of tPA transcripts from mouse normal pancreas and ductal tumor
from Ela-myc mice (top) or normal human pancreas and RWP-1 and SK-PC-1 cells (bottom).
At the top, the complementary DNA was detected by labeling with [32P] γ-dATP followed by autoradiography. 1 indicates the internal control of tPA
mRNA expression, and 2 indicates polyadenylation. A20 and A90 indicate the respective length of poly(A) tail. (c) A schematic representation of the
3 UTR of human tPA mRNA (top) and luciferase Renilla activity (bottom) of synthetic chimeric mRNAs containing the firefly luciferase coding sequence
fused upstream from the indicated tPA 3 UTRs injected into oocytes. The intact 3 UTR of cyclin B1 and mutants of the three CPEs therein were used
as controls. Translation of the nonrepressed control mRNA was adjusted to 100%. (d) Polyadenylation analysis of tPA transcripts from untransfected,
shCtrl or shCPEB4 RWP-1 cells. (e) tPA RNA and protein expression after CPEB4 knockdown in RWP-1 cells. HPRT (for RNA) or tubulin (protein) was
used for normalization. Data are mean ± s.e.m. (f) tPA immunohistochemistry in tumors from nude mice after subcutaneous or intraperitoneal injection
of RWP-1 untransfected, shCtrl or shCPEB4 cells. Scale bars, 100 µm. At the bottom is quantification of tPA knockdown in tumors, calculated by the
percentage of tPA staining intensity and compared to shCtrl tumors. The P values (determined by Student’s t test) relative to shCtrl are shown.
***P 0.001. Data are mean ± s.e.m. One representative experiment (n = 3) is shown (ad).
npg © 2012 Nature America, Inc. All rights reserved.
articles
88  VOLUME 18 | NUMBER 1 | JANUARY 2012 nature medicine
in ductal tumors and in PDA cell lines (RWP-1 and SK-PC-1), the
poly(A) tail was elongated (Fig. 6b), suggesting that the translation-
ally silent tPA mRNA in normal pancreas becomes polyadenylated in
pancreatic cancer. To further characterize these changes in poly(A)
tail length, we sought to identify the regulatory cis-acting elements in
the tPA mRNA 3 UTR microinjecting reporter transcripts of Xenopus
laevis oocytes.
The 3 UTR of tPA mRNA contains two putative CPEs in proxi-
mity to the hexanucleotide polyadenylation signal (HEX)6,10 and two
potential AU-rich elements (AREs). AREs are well known regula-
tors of mRNA deadenylation and stability7 (Fig. 6c). To determine
whether these elements were functional, we fused the tPA 3 UTR
or variants in which these cis-acting elements had been inactivated
downstream of the firefly luciferase open reading frame, and we
then injected them into stage IV oocytes together with Renilla firefly
mRNA. As a control, we used the 3 UTR of cyclin B1 and a variant in
which the three CPEs were mutated6. Stimulation with progesterone,
to activate CPEBs, resulted in a marked increase in luciferase activ-
ity when the full-length sequence of the 3 UTR of tPA mRNA was
present, but this stimulation was largely prevented when the CPE or
HEX sequences were deleted or mutated. In contrast, we observed
an increase in luciferase activity when the AREs were deleted from
chimeric mRNA, which is consistent with their role in deadenylation
(Fig. 6c). These results show that the 3 UTR of human tPA contains
functional CPEs and AREs, which may account for its translational
control and the changes in poly(A) tail length observed in cell cul-
tures and tissues.
To further test whether CPEB4 regulates tPA synthesis, we per-
formed a poly(A) test assay of tPA mRNA from RWP-1 cells trans-
fected with shCPEB4. Downregulation of CPEB4 resulted in tPA
mRNA with a shorter poly(A) tail (Fig. 6d) and, accordingly, a
decrease of tPA protein expression without affecting tPA mRNA levels
(Fig. 6e). We confirmed these results in vivo, where subcutaneous and
intraperitoneal xenografts from shCPEB4 cells also had reduced tPA
expression (Fig. 6f). Together, these data show that tPA expression is
differentially regulated by CPEB4-mediated translational control in
tumoral cells compared to normal pancreas, leading to the accumula-
tion of tPA that is associated with PDA progression.
To determine the relative contribution of tPA translational regula-
tion to the tumoral phenotype observed after CPEB4 depletion, we
performed a rescue experiment using ectopic expression of tPA in
xenografted RWP-1 cells depleted of CPEB4, which resulted in an
increased number of tumors (Supplementary Fig. 8). This observa-
tion suggests that, although CPEB4 binds to hundreds of mRNAs
(Supplementary Table 2), tPA mRNA translation is a key con-
tributor to the decreased in vivo tumorigenicity that occurs after
CPEB4 downregulation.
CPEB4 promotes tumor growth and vascularization in gliomas
The crucial biological role of CPEB4 in PDA raised the question
of whether it also has a similar role in the development of other
tumor types. To explore this possibility, we analyzed the expres-
sion of CPEB4 in human glioblastomas. CPEB4 was absent from
normal astrocytes but was ver y abundant in high-grade gliomas
(Supplementary Fig. 9a). In addition, CPEB4 downregulation in cells
from the T98G line resulted in reduced tumor size (Supplementar y
Figs. 9b and 10), cellular proliferation (Supplementary Fig. 9c,d;
measurement of Ki67) and microvessel density (Supplementary
Fig. 9c,d; measurement of vWF). Thus, the contribution of CPEB4 to
tumor malignancy seems to extend to other tumor types, suggesting
that translational control by CPEB4 may be a general mechanism to
control gene expression during carcinogenesis.
DISCUSSION
PDA is a complex genetic disease resulting from the unbalanced
expression of many tumor-promoting or antitumoral factors imping-
ing on essential signal transduction pathways, for example, the K-Ras,
Hedgehog, TGF-β and Wnt-Notch pathways17. Although PDA develop-
ment has been associated with a number of genetic and epigenetic
alterations, it is now clear that this is only the first layer of genetic
reprogramming associated with tumor progression and that post-
transcriptional regulation may also participate in the cellular changes
that result in malignant transformation2–4,30–37. The altered expression
and activity of general translation factors has previously been observed
in tumors4,5. However, a direct functional link between the differen-
tial expression of mRNA-specific translational regulators and tumor
development has not been established. Here we show that CPEB4 is
specifically upregulated in high-grade PanIN lesions and in well or
moderately differentiated PDA. In tumoral cells, CPEB4 associates with
and translationally activates hundreds of mRNAs, engaging an addi-
tional level of gene regulation (Supplementar y Table 2). To test this
concept, we studied the expression of tPA, one of the transcripts that
is enriched in the CPEB4 immunoprecipitate. tPA is a member of the
plasminogen system, a family of proteins that are essential components
of tumor progression in several types of tumors, including PDA38–43.
tPA protein is absent in normal pancreas and is overexpressed in 85%
of PDA, and tPA protein has a role in tumor proliferation, migration,
invasion and angiogenesis21–27. However, tPA mRNA is similarly
expressed in both normal and tumoral pancreatic samples. Therefore,
translational activation of tPA mRNA as a result of the tumor-specific
overexpression of CPEB4 is consistent with its expression pattern and
its function in tumors. Indeed, we found that tPA mRNA is translation-
ally activated in tumors through the recruitment of CPEB4 to CPEs in
its 3 UTR. tPA mRNA is also translationally regulated by CPEB1 in
mouse oocytes28,44,45 and neurons29. However, in tumors, pathological
activation of tPA mRNA is mediated by CPEB4.
In addition to tPA transcript, CPEB4-associated mRNAs are
significantly (P 0.05) enriched in a number of cellular functions
that are relevant to tumorigenesis, including RAS-related mole-
cules, cell signaling components (Smad family member 3 (Smad3),
phosphoinositide-3-kinase (PI3K), calcium/calmodulin-dependent
protein kinase II (CamKII) and G-protein–coupled receptor), chro-
matin remodeling proteins (histone deacetylases and MYST histone
acetyltransferase), cyclins, apoptosis-related molecules (caspase 8,
apoptosis-related cysteine peptidase (CASP8) and B-cell CLL/lymphoma 2
(BCL2) binding component 3), stress and inflammation factors
(interleukin-32, hypoxia inducible domain family, member 1A (HIG1),
interferon receptor 2 and heat shock 70-kDa protein 4 (Hsp70)), meta-
bolic enzymes (isocitrate dehydrogenase 1 (IDH1)) and genes related
to cell migration and metastasis (encoding matrix metallopeptidase 7
(MMP-7), tPA, β-catenin and Twist) (Supplementary Table 2).
However, the most significant enrichment was for mRNAs encoding
translation factors (ribosomal proteins, eukaryotic translation initiation
factor 3, subunit E (eIF3e) and eIF3h), a cellular function that has been
previously associated with malignant transformation4,46–48. mRNA
from a number of pro-fibrogenic factors (MMP-7, SMAD3 and serpin
peptidase inhibitor, clade B (ovalbumin), member 1 (SerpinB1)) was
also associated with CPEB4. CPEB4-associated transcripts are enriched
in CPEs in proximity to the HEX, which is the hallmark of CPE-
mediated cytoplasmic polyadenylation and translational activation6.
npg © 2012 Nature America, Inc. All rights reserved.
articles
nature medicineVOLUME 18 | NUMBER 1 | JANUARY 2012 89
CPEB4-mediated reprogramming of gene expression in PDA has
a marked effect on at least three of the processes considered to be
hallmarks of cancer1. First, CPEB4 correlates with increased tumoral
growth, and its downregulation results in a substantial reduction
of cellular proliferation in the tumoral context but not in a cell-
autonomous manner. Second, CPEB4 knockdown results in a reduc-
tion of tumor angiogenesis, suggesting that this effect may contribute
to the changes in cell proliferation observed in vivo. Even though
reduced vascularization could explain the reduced tumoral size and
provide a clear link with the translational control of tPA, we cannot
rule out additional functions for CPEB4 in a tumoral-niche–specific
manner, for example, by regulating cell-cell attachment, tumor-stromal
interactions or other events. Third, cells overexpressing CPEB4 show
an advantage for tissue colonization and invasion, although further
studies will be required to better understand the role of CPEB4 in
this process. This work shows that overexpression of CPEB4 in PDA
activates the translation of a wide range of transcripts, subverting its
normal function in meiosis, and confers relevant advantages to cancer
cells for tumor growth and progression. Notably, ectopic expression
of genes normally related to germline traits was recently found to be
associated with tumor growth in Drosophila49.
CPEB4 overexpression and its contribution to tumor growth
and angiogenesis are not restricted to PDA: they also occur in glio-
blastomas, suggesting that CPEB4-mediated regulation of gene
expression might be a more general mechanism in cancer. Indeed,
gene profiling in other types of tumors has shown changes in mRNAs
encoding different members of the CPEB family of proteins50–55. In
addition, CPEB4 protein is overexpressed in a large variety of tumors
(17 out of a total of 20 tumor types listed at http://www.proteinatlas.
org/ENSG00000113742), suggesting that this overexpression might
be a general mechanism in tumoral development and a possible new
therapeutic target.
METHODS
Methods and any associated references are available in the online
version of the paper at http://www.nature.com/naturemedicine/.
Note: Supplementary information is available on the Nature Medicine website.
ACKNOWLEDGMENTS
The authors acknowledge F. Gebauer, S. Aznar-Benitah, A. García de
Herreros and J. Valcárcel for critical comments on the manuscript and for other
valuable contributions. We also thank S. Hahn (Department of Molecular
GI-Oncology, University of Bochum, Germany) and M. Buchholz (Department
of Gastroenterology, Endocrinolog y and Metabolism, Philipps-University of
Marburg, Marburg, Germany) for providing normal pancreas RNA,
O. Casanovas (Translational Research Laboratory, Catalan Institute of Oncology
(ICO)–Bellvitge Biomedical Research Institute (IDIBELL), L’Hospitalet
de Llobregat, Spain) for his help with the T98G nude mice injections,
J.R. González-Vallinas (Computational Genomics Group, Universitat Pompeu
Fabra, Parc de Recerca Biomèdica de Barcelona (PRBB)) for assistance with
Gene Ontology tools and E. Castillo from the Ultrasequencing Unit (Centre for
Genomic Regulation, PRBB), S. Mojal from the Statistics Unit (IMIM, PRBB)
and T. Lobato from the Histopathological Unit (IMIM, PRBB) for technical
assistance. This work was funded by the research grants Instituto de Salud
Carlos–Fondos Europeos de Desarrollo Regional (FEDER) (PI080421) from the
Ministerio de Ciencia e Innovación (MICINN) and grants from Fundació La
MaratóTV3 (051110), the American Institute for Cancer Research (AICR)
(11-0086) and Generalitat de Catalunya (2009SGR1409) to P.N.; grants
BFU2008-02373 and Consolider RNAREG CSD2009-00080 from the MICINN
and grants from Fundació La MaratóTV3 (051110), AICR (11-0086) and
Generalitat de Catalunya (2009SGR1436) to R.M.; grant SAF2007-60860
and Consolider ONCOBIO from the MICINN and a grant from the VI EU
Framework Programme MolDiag-PaCa project to F.X.R.; grants Consolider
RNAREG CSD2009-00080 and BIO2008-01091 from the MICINN to E.E.;
and grants from the Instituto de Salud Carlos III FEDER (RD09/0076/00036)
and the Xarxa de Bancs de tumors sponsored by the Pla Director d’Oncologia de
Catalunya to the MARBiobanc. P.N. is supported by the Instituto de Salud Carlos
III and the Departament de Sanitat de la Generalitat de Catalunya. D.P. holds
a Juan de la Cierva grant from the MICINN. N.M.-B. holds a grant from the
Fundación Ramón Areces. C.E. was supported by a fellowship from the DURSI
(Generalitat de Catalunya) and Fons Social Europeu (ESF).
AUTHOR CONTRIBUTIONS
E.O.-Z. performed the experiments shown in Figures 16 and in Supplementary
Figures 1, 3, 4 and 610, prepared the figures for the manuscript (except Fig. 5b)
and contributed to the experimental design of the study and the preparation of
the manuscript. D.P. generated the majority of the plasmids used in the study,
designed the primers for the RIP validation experiments, helped with the frog
surgery, injection of the oocytes, reporter analyses and in vivo experiments and
prepared Figures 5b, 6d and 6f and Supplementary Figure 2. N.M.-B. established
the RWP-1 LUC cell line, helped with the in vivo experiments, performed
immunohistochemistry analyses of the Capan-1 xenografts and performed all the
statistical analyses. D.P. and N.M.-B. prepared Figure 3 and Supplementary
Figure 5 and made useful contributions to the experimental design and the
interpretation of the data. G.F.-M. generated Capan-1 shCtrl and shCPEB4 cells
and contributed to the interpretation of the data. M.I. supplied all the human
pancreatic samples used in the study and helped with the histopathological
analyses of all the immunohistochemistries performed in samples from both
mice and humans. F.A. supplied human glioblastoma samples and helped with
the histopathological and immunohistochemistries analyses in tumors from both
mice and humans. M.M. helped with the in vivo experiments. C.E. generated the
data shown at the bottom of Figure 6b. E.E. performed the analysis of the data
generated in the Illumina Solexa sequencing, performed the motif analysis of the
3 UTR sequences shown in Figure 5a, generated the data shown in
Supplementary Figures 6 and 7 and contributed to the preparation of the
manuscript. F.X.R. contributed to the study design, data analysis and manuscript
preparation. P.N. and R.M. directed the study and wrote the manuscript, which all
authors approved.
COMPETING FINANCIAL INTERESTS
The authors declare no competing financial interests.
Published online at http://www.nature.com/naturemedicine/.
Reprints and permissions information is available online at http://www.nature.com/
reprints/index.html.
1. Hanahan, D. & Weinberg, R.A. The hallmarks of cancer. Cell 100, 57–70
(2000).
2. Mayr, C., Hemann, M.T. & Bartel, D.P. Disrupting the pairing between let-7 and
Hmga2 enhances oncogenic transformation. Science 315, 1576–1579 (2007).
3. Lee, Y.S. & Dutta, A. The tumor suppressor microRNA let-7 represses the HMGA2
oncogene. Genes Dev. 21, 1025–1030 (2007).
4. Sonenberg, N. & Hinnebusch, A.G. Regulation of translation initiation in eukaryotes:
mechanisms and biological targets. Cell 136, 731–745 (2009).
5. Silvera, D., Formenti, S.C. & Schneider, R.J. Translational control in cancer.
Nat. Rev. Cancer 10, 254–266 (2010).
6. Piqué, M., Lopez, J.M., Foissac, S., Guigo, R. & Mendez, R. A combinatorial code
for CPE-mediated translational control. Cell 132, 434–448 (2008).
7. Belloc, E. & Mendez, R. A deadenylation negative feedback mechanism governs
meiotic metaphase arrest. Nature 452, 1017–1021 (2008).
8. Eliscovich, C., Peset, I., Vernos, I. & Mendez, R. Spindle-localized CPE-mediated
translation controls meiotic chromosome segregation. Nat. Cell Biol. 10, 858–865
(2008).
9. Novoa, I., Gallego, J., Ferreira, P.G. & Mendez, R. Mitotic cell-cycle progression is
regulated by CPEB1- and CPEB4-dependent translational control. Nat. Cell Biol.
12, 447–456 (2010).
10. Mendez, R. & Richter, J.D. Translational control by CPEB: a means to the end.
Nat. Rev. Mol. Cell Biol. 2, 521–529 (2001).
11. Groppo, R. & Richter, J.D. Translational control from head to tail. Curr. Opin. Cell
Biol. 21, 444–451 (2009).
12. Igea, A. & Mendez, R. Meiosis requires a translational positive loop where CPEB1
ensues its replacement by CPEB4. EMBO J. 29, 2182–2193 (2010).
13. Huang, Y.S., Kan, M.C., Lin, C.L. & Richter, J.D. CPEB3 and CPEB4 in neurons:
analysis of RNA-binding specificity and translational control of AMPA receptor GluR2
mRNA. EMBO J. 25, 4865–4876 (2006).
14. Groisman, I. et al. Control of cellular senescence by CPEB. Genes Dev. 20,
2701–2712 (2006).
15. Richter, J.D. CPEB: a life in translation. Trends Biochem. Sci. 32, 279–285
(2007).
16. Burns, D.M. & Richter, J.D. CPEB regulation of human cellular senescence, energy
metabolism, and p53 mRNA translation. Genes Dev. 22, 3449–3460 (2008).
npg © 2012 Nature America, Inc. All rights reserved.
articles
90  VOLUME 18 | NUMBER 1 | JANUARY 2012 nature medicine
17. Jones, S. et al. Core signaling pathways in human pancreatic cancers revealed by
global genomic analyses. Science 321, 1801–1806 (2008).
18. Parsons, D.W. et al. An integrated genomic analysis of human glioblastoma
multiforme. Science 321, 1807–1812 (2008).
19. Iacobuzio-Donahue, C.A. et al. DPC4 gene status of the primary carcinoma correlates
with patterns of failure in patients with pancreatic cancer. J. Clin. Oncol. 27,
1806–1813 (2009).
20. van Riggelen, J., Yetil, A. & Felsher, D.W. MYC as a regulator of ribosome biogenesis
and protein synthesis. Nat. Rev. Cancer 10, 301–309 (2010).
21. Paciucci, R. et al. Isolation of tissue-type plasminogen activator, cathepsin H, and
non- specific cross-reacting antigen from SK-PC-1 pancreas cancer cells using
subtractive hybridization. FEBS Lett. 385, 72–76 (1996).
22. Paciucci, R., Tora, M., Diaz, V.M. & Real, F.X. The plasminogen activator system
in pancreas cancer: role of t-PA in the invasive potential in vitro. Oncogene 16,
625–633 (1998).
23. Díaz, V.M., Planaguma, J., Thomson, T.M., Reventos, J. & Paciucci, R. Tissue
plasminogen activator is required for the growth, invasion, and angiogenesis of
pancreatic tumor cells. Gastroenterology 122, 806–819 (2002).
24. Ryu, B. et al. Relationships and differentially expressed genes among pancreatic
cancers examined by large-scale serial analysis of gene expression. Cancer Res.
62, 819–826 (2002).
25. Iacobuzio-Donahue, C.A. et al. Highly expressed genes in pancreatic ductal
adenocarcinomas: a comprehensive characterization and comparison of the
transcription profiles obtained from three major technologies. Cancer Res. 63,
8614–8622 (2003).
26. Aguilar, S. et al. Tissue plasminogen activator in murine exocrine pancreas cancer:
selective expression in ductal tumors and contribution to cancer progression.
Am. J. Pathol. 165, 1129–1139 (2004).
27. Missiaglia, E. et al. Analysis of gene expression in cancer cell lines identifies
candidate markers for pancreatic tumorigenesis and metastasis. Int. J. Cancer 112,
100–112 (2004).
28. Huarte, J., Belin, D., Vassalli, A., Strickland, S. & Vassalli, J.D. Meiotic maturation
of mouse oocytes triggers the translation and polyadenylation of dormant tissue-type
plasminogen activator mRNA. Genes Dev. 1, 1201–1211 (1987).
29. Shin, C.Y., Kundel, M. & Wells, D.G. Rapid, activity-induced increase in tissue
plasminogen activator is mediated by metabotropic glutamate receptor-dependent
mRNA translation. J. Neurosci. 24, 9425–9433 (2004).
30. Gardina, P.J. et al. Alternative splicing and differential gene expression in colon
cancer detected by a whole genome exon array. BMC Genomics 7, 325
(2006).
31. Karni, R. et al. The gene encoding the splicing factor SF2/ASF is a proto-oncogene.
Nat. Struct. Mol. Biol. 14, 185–193 (2007).
32. Srebrow, A. & Kornblihtt, A.R. The connection between splicing and cancer. J. Cell
Sci. 119, 2635–2641 (2006).
33. Karni, R., Hippo, Y., Lowe, S.W. & Krainer, A.R. The splicing-factor oncoprotein
SF2/ASF activates mTORC1. Proc. Natl. Acad. Sci. USA 105, 15323–15327
(2008).
34. Cooper, C. et al. Increasing the relative expression of endogenous non-coding steroid
receptor RNA activator (SRA) in human breast cancer cells using modified
oligonucleotides. Nucleic Acids Res. 37, 4518–4531 (2009).
35. Pio, R. & Montuenga, L.M. Alternative splicing in lung cancer. J. Thorac. Oncol.
4, 674–678 (2009).
36. Mayr, C. & Bartel, D.P. Widespread shortening of 3 UTRs by alternative cleavage
and polyadenylation activates oncogenes in cancer cells. Cell 138, 673–684
(2009).
37. Ji, Z. & Tian, B. Reprogramming of 3 untranslated regions of mRNAs by alternative
polyadenylation in generation of pluripotent stem cells from different cell types.
PLoS ONE 4, e8419 (2009).
38. Blasi, F. & Sidenius, N. The urokinase receptor: focused cell surface proteolysis,
cell adhesion and signaling. FEBS Lett. 584, 1923–1930 (2010).
39. Ossowski, L. Plasminogen activator dependent pathways in the dissemination of
human tumor cells in the chick embryo. Cell 52, 321–328 (1988).
40. Aguirre-Ghiso, J.A., Liu, D., Mignatti, A., Kovalski, K. & Ossowski, L. Urokinase
receptor and fibronectin regulate the ERK(MAPK) to p38(MAPK) activity ratios that
determine carcinoma cell proliferation or dormancy in vivo. Mol. Biol. Cell 12,
863–879 (2001).
41. Smith, H.W. & Marshall, C.J. Regulation of cell signalling by uPAR. Nat. Rev. Mol.
Cell Biol. 11, 23–36 (2010).
42. Ulisse, S., Baldini, E., Sorrenti, S. & D’Armiento, M. The urokinase plasminogen
activator system: a target for anti-cancer therapy. Curr. Cancer Drug Targets 9,
32–71 (2009).
43. McMahon, B. & Kwaan, H.C. The plasminogen activator system and cancer.
Pathophysiol. Haemost. Thromb. 36, 184–194 (2008).
44. Huarte, J., Belin, D. & Vassalli, J.D. Plasminogen activator in mouse and rat oocytes:
induction during meiotic maturation. Cell 43, 551–558 (1985).
45. Huarte, J. et al. Transient translational silencing by reversible mRNA deadenylation.
Cell 69, 1021–1030 (1992).
46. Gandin, V. et al. Eukaryotic initiation factor 6 is rate-limiting in translation, growth
and transformation. Nature 455, 684–688 (2008).
47. Ruggero, D. et al. The translation factor eIF-4E promotes tumor formation and
cooperates with c-Myc in lymphomagenesis. Nat. Med. 10, 484–486 (2004).
48. Livingstone, M., Atas, E., Meller, A. & Sonenberg, N. Mechanisms governing the
control of mRNA translation. Phys. Biol. 7, 021001 (2010).
49. Janic, A., Mendizabal, L., Llamazares, S., Rossell, D. & Gonzalez, C. Ectopic
expression of germline genes drives malignant brain tumor growth in Drosophila.
Science 330, 1824–1827 (2010).
50. Gaspar, C. et al. Cross-species comparison of human and mouse intestinal polyps
reveals conserved mechanisms in adenomatous polyposis coli (APC)-driven
tumorigenesis. Am. J. Pathol. 172, 1363–1380 (2008).
51. Sääf, A.M. et al. Parallels between global transcriptional programs of polarizing
Caco-2 intestinal epithelial cells in vitro and gene expression programs in normal
colon and colon cancer. Mol. Biol. Cell 18, 4245–4260 (2007).
52. Read, T.A. et al. Identification of CD15 as a marker for tumor-propagating cells in
a mouse model of medulloblastoma. Cancer Cell 15, 135–147 (2009).
53. Chen, H.Y. et al. A five-gene signature and clinical outcome in non-small-cell lung
cancer. N. Engl. J. Med. 356, 11–20 (2007).
54. Israel, A., Sharan, R., Ruppin, E. & Galun, E. Increased microRNA activity in human
cancers. PLoS ONE 4, e6045 (2009).
55. Hansen, C.N. et al. Expression of CPEB, GAPDH and U6snRNA in cervical and
ovarian tissue during cancer development. APMIS 117, 53–59 (2009).
npg © 2012 Nature America, Inc. All rights reserved.
nature medicine
doi:10.1038/nm.2540
ONLINE METHODS
Cell culture, CPEB4 knockdown and tPA overexpression. Human pancreatic
cells have been previously described56 . T98G cells, derived from a human
glioblastoma tumor, were purchased from America Type Culture Collection.
All cells were cultured in DMEM supplemented with 10% FBS. All experi-
ments using human cell lines were approved by the Ethical Committee for
Clinical Research of IMIM–Hospital del Mar. shRNA against CPEB4 and the
corresponding scramble control (shCtrl) were obtained from Sigma (siMis-
sion) and transfected into RWP-1, RWP-1 LUC, Capan-1 or T98G cells with
Lipofectamine and Plus reagent (Invitrogen). Selection was performed for
3 d with puromycin (3 µg ml−1 for RWP-1 and RWP-1 LUC cells, 2 µg ml−1
for Capan-1 cells and 1 µg ml−1 for T98G cells). CPEB4 concentrations were
analyzed by western blot and qRT-PCR. For the in vivo rescue experiment,
RWP-1 shCtrl and RWP-1 shCPEB4_2 cells were transfected with an empty
vector, pcDNA3 or a pcDNA3 vector expressing the coding sequence of human
tPA26. Selection was performed for 5 d with neomycin (700 µg ml−1 ).
Generation of RWP-1 LUC cells. For the generation of RWP-1 LUC cells, the
pLHCX plasmid (a gift from C. Fillat, Center for Genomic Regulation, Barcelona,
Spain) was used to produce lentiviral particles in the Phoenix Ampho HEK293T
cell line. After transduction, stable clones expressing luciferase were selected
with hygromycin (0.3 mg ml−1); luciferase activity was measured using the Dual-
Luciferase Reporter Assay (Promega). A clone with a medium rate of luciferase
activity was selected.
MTT cell proliferation assay. Cell growth was determined by a MTT colori-
metric growth assay. Briefly, 1 × 103 cells per well were plated in 96-well plates
in complete medium and cultured to confluence. Each day, cell growth was
determined by adding MTT solution (1 mg ml−1) in DMEM without FBS for 4 h
at 37 °C. The dye was then extracted with a solution of 0.1 N HCl and 10% Triton
X-100 in isopropanol, and absorbance was measured with a 570-nm filter.
Cell invasion assays. For the invasion assays, Transwells (Costar) coated with
Matrigel (BD Bioscience) were used as previously described56. Briefly, 30,000
RWP-1 cells were seeded in DMEM without FBS and with 1% BSA and were
allowed to invade for 72 h. Cells that passed to the lower compartment of the
filter were fixed, and absorbance (at 590 nm) was quantified after crystal violet
staining for 5 min at 25 °C and 10% acetic acid extraction. The absorbance
measurements listed in Figure 2 are equivalent to the following number of
cells: 15,000 (untransfected), 16,600 (shCtrl), 7,230 (shCPEB4_2) and 8,840
(shCPEB4_4).
Soft-agar growth assay. Anchorage-independent growth was evaluated as
described57. Briefly, 1 × 104 cells were plated in complete DMEM containing
0.3% soft agar in 6-cm plates over a layer of solidified DMEM containing 0.7%
soft agar. Medium was added twice a week to maintain humidity. After 5 weeks,
colonies were stained with MTT (0.5 mg ml−1) for 3 h at 37 °C and counted.
In vivo tumorigenicity assay. RWP-1 (1 × 106), Capan-1 (1 × 106) or T98G
cells (6 × 106) were injected subcutaneously into the two posterior flanks of
male BALB/c nude mice (Charles River). Mice were followed weekly, and
when tumors in any of the groups (mice injected with untransfected, shCtrl,
shCPEB4_2 or shCPEB4_4 cells) reached around 1 cm3, determined by palpa-
tion (at 3 weeks), all mice were killed, and the tumors were weighed, measured
and processed. For intraperitoneal injections, 1 × 106 RWP-1 LUC cells were
injected in the peritoneal cavity, and tumor growth was assessed weekly by bio-
luminescence until mice were killed (when the bioluminescence signal indicated
a tumor size of 1.5 cm3 or when mice presented with symptoms of suffering
or health deterioration). For bioluminescence detection and quantification,
mice were anesthetized, and the substrate D-firefly-Luciferin (Xenogen) was
administered intraperitoneally (32 mg per kg of body weight). Luciferase activity
was visualized and quantified using an in vivo bioluminescent system (IVIS 50
Xenogen) and a Living Image 2.20.1 software overlay on Igor Pro4.06A software
(WaveMetrics) as previously described58. Luciferase activity was quantified from
non-saturated images by measuring the total amount of emitted light recorded
by the charge-coupled device camera. A total of ten (for RWP-1 cells and two
flanks of subcutaneous injection), eight (for Capan-1 cells and two flanks of sub-
cutaneous injection), four (for T98G cells and two flanks of subcutaneous injec-
tion) and ten (for RWP-1 LUC cells and one intraperitoneal injection) mice were
used for each condition (untransfected, shCtrl, shCEPB4_2 and shCPEB4_4).
For the analysis of tumor incidence in the pancreas and other organs (invasion)
and of survival, mice were clustered into two groups: a control group (mice
injected with untransfected and shCtrl RWP-1 LUC cells) and a shCPEB4 group
(mice injected with shCPEB4_2 or shCPEB4_4 RWP-1 LUC cells). All animal
procedures met the guidelines of European Community Directive and were
approved by the PRBB ethical committee.
Statistical analyses. All results were evaluated using the SPSS statistical soft-
ware package. Different statistical tests were used according to the type of data
analyzed (Student’s t test,
χ
2, Fisher’s or log-rank tests), as is indicated in figure
legends. P 0.05 was considered statistically significant.
Additional methods. Descriptions of the tissue samples, immunohistochemistry
and histopathological analyses, plasmid construction, oligonucleotides, immuno-
fluorescence microscopy, RNA extraction, qRT-PCR, translation of luciferase
reporters in X. laevis oocytes, poly(A) tail testing, western blot, zymography,
RNA immunoprecipitation, Illumina Solexa sequencing (RIP-Seq) and ultra-
sequencing analyses can be found in the Supplementary Methods.
56. Roda, O. et al. Galectin-1 is a novel functional receptor for tissue plasminogen
activator in pancreatic cancer. Gastroenterology 136, 1379–1390 (2009).
57. Rizzino, A. Behavior of transforming growth factors in serum-free media:
an improved assay for transforming growth factors. In Vitro 20, 815–822 (1984).
58. Huch, M. et al. Urokinase-type plasminogen activator receptor transcriptionally
controlled adenoviruses eradicate pancreatic tumors and liver metastasis in mouse
models. Neoplasia 11, 518–528 (2009).
npg © 2012 Nature America, Inc. All rights reserved.
... Rather, the association of efficient translation with short poly(A) tails in highly expressed genes was suggested (Lima et al., 2017). However, the conclusion remains controversial, since multiple studies using non-early embryonic cells have so far reported contrasting results, that translation of at least certain mRNAs positively correlates with tail length Burns and Richter, 2008;Chorghade et al., 2017;Kojima et al., 2012;Lackner et al., 2007;Novoa et al., 2010;Ortiz-Zapater et al., 2011;Peng et al., 2008;Shao et al., 2020;Shin et al., 2017;Yamagishi et al., 2016;Inagaki et al., 2020). Thus, it is important to carefully evaluate whether the uncoupling of poly(A) tail length and translation is universally applicable for all kinds of mRNAs or whether there are any exceptions. ...
... Poly(A) tail metabolism is well known to affect post-transcriptional gene expression; however, in the case of post-transcriptional poly(A) tail elongation, only a handful of examples have been reported in somatic cells (Burns and Richter, 2008;Novoa et al., 2010;Ortiz-Zapater et al., 2011;Shin et al., 2017;Yamagishi et al., 2016;Inagaki et al., 2020). Here we report a function of LARP1 in post-transcriptional mRNA polyadenylation. ...
Article
Full-text available
Translation of 5′ terminal oligopyrimidine (TOP) mRNAs encoding the protein synthesis machinery is strictly regulated by an amino-acid-sensing mTOR pathway. However, its regulatory mechanism remains elusive. Here, we demonstrate that TOP mRNA translation positively correlates with its poly(A) tail length under mTOR active/amino-acid-rich conditions, suggesting that TOP mRNAs are post-transcriptionally controlled by poly(A) tail-length regulation. Consistent with this, the tail length of TOP mRNAs dynamically fluctuates in response to amino acid availability. The poly(A) tail shortens under mTOR active/amino-acid-rich conditions, whereas the long-tailed TOP mRNAs accumulate under mTOR inactive/amino-acid-starved (AAS) conditions. An RNA-binding protein, LARP1, is indispensable for the process. LARP1 interacts with non-canonical poly(A) polymerases and induces post-transcriptional polyadenylation of the target. Our findings illustrate that LARP1 contributes to the selective accumulation of TOP mRNAs with long poly(A) tails under AAS, resulting in accelerated ribosomal loading onto TOP mRNAs for the resumption of translation after AAS.
... CPEB1 controls stem cell activation and muscle regeneration by attaching to CPEs in the 3′ untranslated regions of Myod1 [32]. Although CPEB4 is linked with abnormal posttranscriptional reprogramming in diseases such as cancer, liver disease, obesity, and neurodegenerative disorders [33][34][35][36][37], there is currently no evidence that CPEB4 serves any function in muscle. However, we found upregulation of CPEB4 after exercise and CR, as well as downregulation of Cpeb4 expression in the muscle of aging mice. ...
Article
Full-text available
Exercise and caloric restriction (CR) significantly increase longevity across a range of species and delay aging-related losses in organ function. Although both interventions enhance skeletal muscle function, the molecular mechanisms underlying these associations are unknown. We sought to identify genes regulated by CR and exercise in muscle, and investigate their relationship with muscle function. To do this, expression profiles of Gene Expression Omnibus datasets obtained from the muscle tissue of calorie-restricted male primates and young men post-exercise were analyzed. There were seven transcripts (ADAMTS1, CPEB4, EGR2, IRS2, NR4A1, PYGO1, and ZBTB43) that were consistently upregulated by both CR and exercise training. We used C2C12 murine myoblasts to investigate the effect of silencing these genes on myogenesis, mitochondrial respiration, autophagy, and insulin signaling, all of which are processes affected by CR and exercise. Our results show that in C2C12 cells, Irs2 and Nr4a1 expression were critical for myogenesis, and five genes (Egr2, Irs2, Nr4a1, Pygo1, and ZBTB43) regulated mitochondrial respiration while having no effect on autophagy. Cpeb4 knockdown increased the expression of genes involved in muscle atrophy and induced myotube atrophy. These findings suggest new resources for studying the mechanisms underlying the beneficial effects of exercise and calorie restriction on skeletal muscle function and lifespan extension.
... In turn, CPEBs regulate the translation and stability of mRNAs through changes in the length of their poly(A) tails (Fernandez-Miranda & Mendez, 2012;Ivshina et al, 2014). The stress-regulated member of the CPEB family, CPEB4, is required in a cellautonomous manner for the survival and proliferation of tumor cells (Ortiz-Zapater et al, 2011;Perez-Guijarro et al, 2016;Tsai et al, 2016;Maillo et al, 2017;Villanueva et al, 2017;Cao et al, 2018). ...
Article
Full-text available
Tumor growth is influenced by a complex network of interactions between multiple cell types in the tumor microenvironment (TME). These constrained conditions trigger the endoplasmic reticulum (ER) stress response, which extensively reprograms mRNA translation. When uncontrolled over time, chronic ER stress impairs the antitumor effector function of CD8 T lymphocytes. How cells promote adaptation to chronic stress in the TME without the detrimental effects of the terminal unfolded protein response (UPR) is unknown. Here, we find that, in effector CD8 T lymphocytes, RNA-binding protein CPEB4 constitutes a new branch of the UPR that allows cells to adapt to sustained ER stress, yet remains decoupled from the terminal UPR. ER stress, induced during CD8 T-cell activation and effector function, triggers CPEB4 expression. CPEB4 then mediates chronic stress adaptation to maintain cellular fitness, allowing effector molecule production and cytotoxic activity. Accordingly, this branch of the UPR is required for the antitumor effector function of T lymphocytes, and its disruption in these cells exacerbates tumor growth.
Article
Sorbin and SH3 domain‐containing 2 (SORBS2) is an RNA‐binding protein and has been implicated in the development of some cancers. However, its role in bladder cancer (BC) is yet to be established. The expression of SORBS2 in BC tissues was determined from the Gene Expression Omnibus and Gene Expression Profiling Interactive Analysis databases and collected paired tumor/normal samples. The effects of SORBS2 on BC cells were detected by CCK‐8, colony formation, Transwell, dual‐luciferase, RNA immunoprecipitation, chromatin immunoprecipitation, and DNA pull‐down assays. In vivo, BC cell growth and metastasis were studied by a xenograft subcutaneous model and a tail‐vein metastasis model. The results showed that SORBS2 expression was significantly decreased in BC tissues and cells. SORBS2 overexpression inhibited cell proliferation, migration, invasion, and epithelial–mesenchymal transition in vitro and tumor growth and metastasis in vivo, while silencing SORBS2 produced the opposite effect. Mechanistically, we found that SORBS2 enhanced the stability of tissue factor pathway inhibitor (TFPI) mRNA via direct binding to its 3′ UTR. Restoration of TFPI expression reversed SORBS2 knockdown‐induced malignant phenotypes of BC cells. In addition, SORBS2 expression was negatively regulated by the transcription factor specificity protein 1 (SP1). Conversely, SORBS2 can be transcriptionally regulated by SP1 and inhibit BC cell growth and metastasis via stabilization of TFPI mRNA, indicating SORBS2 may be a promising therapeutic target for BC.
Article
Most eukaryotic mRNAs and different non‐coding RNAs undergo a form of 3′ end processing known as polyadenylation. Polyadenylation machinery is present in almost all organisms except few species. In bacteria, the machinery has evolved from PNPase, which adds heteropolymeric tails, to a poly(A)‐specific polymerase. Differently, a complex machinery for accurate polyadenylation and several non‐canonical poly(A) polymerases are developed in eukaryotes. The role of poly(A) tail has also evolved from serving as a degradative signal to a stabilizing modification that also regulates translation. In this review, we discuss poly(A) tail emergence in prokaryotes and its development into a stable, yet dynamic feature at the 3′ end of mRNAs in eukaryotes. We also describe how appearance of novel poly(A) polymerases gives cells flexibility to shape poly(A) tail. We explain how poly(A) tail dynamics help regulate cognate RNA metabolism in a context‐dependent manner, such as during oocyte maturation. Finally, we describe specific mRNAs in metazoans that bear stem‐loops instead of poly(A) tails. We conclude with how recent discoveries about poly(A) tail can be applied to mRNA technology. This article is categorized under: RNA Evolution and Genomics > RNA and Ribonucleoprotein Evolution RNA Processing > 3′ End Processing RNA Turnover and Surveillance > Regulation of RNA Stability
Article
Full-text available
Glioblastoma is the most frequent form of malignant brain tumor. Cytoplasmic polyadenylation element binding protein 4 (CPEB4) is overexpressed and involved in the tumorigenesis and metastasis of glioblastoma. miR-130a-3p has been revealed to be aberrantly expressed in tumors and has aroused wide attention. In present study, we would like to investigate the effect and potential mechanism of miR-130a-3p on the proliferation and migration in glioblastoma. The relative expression levels of miR-130a-3p and CPEB4 in glioblastoma cell lines were detected by real-time quantitative polymerase chain reaction. Cell viability and migration were detected by methylthiazolyl tetrazolium assay and transwell assay, and cell cycle analysis was detected by flow cytometry. The expression of CPEB4 protein and epithelial-mesenchymal transition associated markers were detected by western blot. Bioinformatics and luciferase activity analysis were used to verify the targeting relationship between miR-130a-3p and CPEB4. We observed that the expression of CPEB4 was upregulated while that of miR-130a-3p was downregulated in glioblastoma cell lines. CPEB4 was validated as a target of miR-130a-3p by luciferase activity assay. Increased levels of miR-130a-3p inhibited the proliferation and migration of the glioblastoma cells and the overexpression of miR-130a-3p inhibited epithelial-mesenchymal transition. However, CPEB4 overexpression resisted the inhibitory effects of miR-130a-3p. Our study elucidates CPEB4 is upregulated because of the downregulated miR-130a-3p in glioblastoma, which enhances the glioblastoma growth and migration, suggesting a potential therapeutic target for the disease.
Article
Gynecological cancers pose a threat to women's health. Although early-stage gynecological cancers show good outcomes after standardized treatment, the prognosis of patients with advanced, met-astatic, and recurrent cancers is poor. RNA-binding proteins (RBPs) are important cellular proteins that interact with RNA through RNA-binding domains and participate extensively in post-transcriptional regulatory processes, such as mRNA alternative splicing, polyadenylation, intracellular localization and stability, and translation. Abnormal RBP expression affects the normal function of oncogenes and tumor suppressor genes in many malignancies, thus leading to the occurrence or progression of cancers. Similarly, RBPs play crucial roles in gynecological carcinogenesis. We summarize the role of RBPs in gynecological malignancies and explore their potential in the diagnosis and treatment of cancers. The findings summarized in this review may provide a guide for future research on the functions of RBPs.
Article
Background: Schizophrenia (SCZ) is caused by an interplay of polygenic risk and environmental factors, which might alter regulators of gene expression leading to pathogenic mis-expression of SCZ-risk genes. The CPEB family of RNA-binding proteins (CPEB1-4) regulates translation of target RNAs (approximately 40% of overall genes). We previously identified CPEB4 as a key dysregulated translational regulator in autism spectrum disorder (ASD), as its neuronal-specific microexon (exon4) is mis-spliced in ASD brains, causing underexpression of numerous ASD-risk genes. The genetic and pathogenic mechanisms shared between SCZ and ASD led us to hypothesize CPEB4 mis-splicing in SCZ, leading to underexpression of multiple SCZ-related genes. Methods: We performed MAGMA-enrichment analysis in Psychiatric Genomics Consortium GWAS data and analyzed RNA-seq data from the PsychENCODE Consortium. RT-PCR and Western blot were performed on post-mortem brain tissue in which presence/absence of antipsychotics was assessed through toxicological analysis. Finally, mice with mild overexpression of exon4-lacking CPEB4 (CPEB4Δ4) were generated and biochemically and behaviorally analyzed. Results: We first found enrichment of SCZ-associated genes for CPEB4-binder transcripts. We also found decreased usage of CPEB4 microexon in SCZ probands, correlating with decreased protein levels of CPEB4-target SCZ-associated genes, selectively in antipsychotics-free individuals. Interestingly, differentially expressed genes fit those reported for SCZ, specifically in the SCZ probands with decreased CPEB4-microexon inclusion. Finally, we demonstrate that mice with mild overexpression of CPEB4Δ4 show decreased protein levels of CPEB4-target SCZ genes and SCZ-linked behaviors. Conclusion: We identify aberrant CPEB4 splicing and downstream mis-expression of SCZ-risk genes as a novel etiological mechanism in SCZ.
Article
Here, we ask how developing precursors maintain the balance between cell genesis for tissue growth and establishment of adult stem cell pools, focusing on postnatal forebrain neural precursor cells (NPCs). We show that these NPCs are transcriptionally primed to differentiate and that the primed mRNAs are associated with the translational repressor 4E-T. 4E-T also broadly associates with other NPC mRNAs encoding transcriptional regulators, and these are preferentially depleted from ribosomes, consistent with repression. By contrast, a second translational regulator, Cpeb4, associates with diverse target mRNAs that are largely ribosome associated. The 4E-T-dependent mRNA association is functionally important because 4E-T knockdown or conditional knockout derepresses proneurogenic mRNA translation and perturbs maintenance versus differentiation of early postnatal NPCs in culture and in vivo. Thus, early postnatal NPCs are primed to differentiate, and 4E-T regulates the balance between cell genesis and stem cell expansion by sequestering and repressing mRNAs encoding transcriptional regulators.
Article
Full-text available
Plasminogen activators (PAs) play an important role in tumor cell invasion. We have analysed the expression of tissue-type PA (t-PA), urokinase-type PA (u-PA), and their respective receptors, annexin II and u-PAR, in normal and neoplastic cultures of pancreatic cells, as well as in pancreatic tissues, and have examined their role in tumor invasiveness in vitro. Using Northern blotting, Western blotting, and ELISA, t-PA is detected in cultured pancreas cancer cells displaying a well differentiated phenotype but it is undetectable in less differentiated cells and in normal pancreatic cultures. In contrast, u-PA transcripts, protein, and enzymatic activity are detected both in cancer cells and in normal cultures. Higher levels of u-PAR and annexin II are present in cancer cells than in normal cultures and, in SK-PC-1 cells, both receptors are localized in the basolateral membrane. In vitro invasion assays indicate that both t-PA and u-PA contribute to the invasiveness of SK-PC-1 cells through reconstituted extracellular matrix. To determine the relevance of these studies to pancreas cancer, immunohistochemical assays have been used to examine the expression of t-PA, u-PA, and their receptors in normal and neoplastic tissues. t-PA is absent from normal pancreas and from tumor associated pancreatitis, whereas it is detected in the majority of pancreas cancer tissues (16/17). Annexin II is also overexpressed in some tumors (5/13). u-PAR is overexpressed in most tumor samples examined (14/15), while u-PA is weakly detected in a low number of cases (3/14); both u-PAR and u-PA are overexpressed in areas of tumor associated pancreatitis. Indirect evidences indicate that K-ras and p53 mutated proteins can regulate the expression of PAs. In pancreatic cancer we have found an association between codon 12 K-ras mutations and t-PA expression (P=0.04). These results support the contention that, in the exocrine pancreas, activation of t-PA is more specifically associated to neoplastic transformation and to the invasive phenotype, whereas the induction of u-PA/u-PAR system might be more relevant to inflammatory or non-neoplastic events.
Article
Full-text available
Meiotic progression is driven by the sequential translational activation of maternal messenger RNAs stored in the cytoplasm. This activation is mainly induced by the cytoplasmic elongation of their poly(A) tails, which is mediated by the cytoplasmic polyadenylation element (CPE) present in their 3' untranslated regions. Although polyadenylation in prophase I and metaphase I is mediated by the CPE-binding protein 1 (CPEB1), this protein is degraded during the first meiotic division. Thus, raising the question of how the cytoplasmic polyadenylation required for the second meiotic division is achieved. In this work, we show that CPEB1 generates a positive loop by activating the translation of CPEB4 mRNA, which, in turn, replaces CPEB1 and drives the transition from metaphase I to metaphase II. We further show that CPEB1 and CPEB4 are differentially regulated by phase-specific kinases, generating the need of two sequential CPEB activities to sustain cytoplasmic polyadenylation during all the meiotic phases. Altogether, this work defines a new element in the translational circuit that support an autonomous transition between the two meiotic divisions in the absence of DNA replication.
Article
Full-text available
The translation of cellular mRNA to protein is a tightly controlled process often deregulated in diseases such as cancer. Furthering our understanding of mRNA structural elements and the intracellular proteins and signaling pathways that affect protein expression is crucial in the development of new therapies. In this review, we discuss the current state-of-the-art of detecting and determining the role of mRNA sequence elements in regulating the initiation of mRNA translation and the therapeutic strategies that exploit this knowledge to treat disease.
Article
Full-text available
Meiotic and early-embryonic cell divisions in vertebrates take place in the absence of transcription and rely on the translational regulation of stored maternal messenger RNAs. Most of these mRNAs are regulated by the cytoplasmic-polyadenylation-element-binding protein (CPEB), which mediates translational activation and repression through cytoplasmic changes in their poly(A) tail length. It was unknown whether translational regulation by cytoplasmic polyadenylation and CPEB can also regulate mRNAs at specific points of mitotic cell-cycle divisions. Here we show that CPEB-mediated post-transcriptional regulation by phase-specific changes in poly(A) tail length is required for cell proliferation and specifically for entry into M phase in mitotically dividing cells. This translational control is mediated by two members of the CPEB family of proteins, CPEB1 and CPEB4. We conclude that regulation of poly(A) tail length is not only required to compensate for the lack of transcription in specialized cell divisions but also acts as a general mechanism to control mitosis.
Article
Full-text available
Remarkable progress has been made in defining a new understanding of the role of mRNA translation and protein synthesis in human cancer. Translational control is a crucial component of cancer development and progression, directing both global control of protein synthesis and selective translation of specific mRNAs that promote tumour cell survival, angiogenesis, transformation, invasion and metastasis. Translational control of cancer is multifaceted, involving alterations in translation factor levels and activities unique to different types of cancers, disease stages and the tumour microenvironment. Several clinical efforts are underway to target specific components of the translation apparatus or unique mRNA translation elements for cancer therapeutics.
Article
Full-text available
Background: The 3' untranslated regions (3'UTRs) of mRNAs contain cis elements involved in post-transcriptional regulation of gene expression. Over half of all mammalian genes contain multiple polyadenylation sites that lead to different 3'UTRs for a gene. Studies have shown that the alternative polyadenylation (APA) pattern varies across tissues, and is dynamically regulated in proliferating or differentiating cells. Generation of induced pluripotent stem (iPS) cells, in which differentiated cells are reprogrammed to an embryonic stem (ES) cell-like state, has been intensively studied in recent years. However, it is not known how 3'UTRs are regulated during cell reprogramming. Methods/main findings: Using a computational method that robustly examines APA across DNA microarray data sets, we analyzed 3'UTR dynamics in generation of iPS cells from different cell types. We found that 3'UTRs shorten during reprogramming of somatic cells, the extent of which depends on the type of source cell. By contrast, reprogramming of spermatogonial cells involves 3'UTR lengthening. The alternative polyadenylation sites that are highly responsive to change of cell state in generation of iPS cells are also highly regulated during embryonic development in opposite directions. Compared with other sites, they are more conserved, can lead to longer alternative 3'UTRs, and are associated with more cis elements for polyadenylation. Consistently, reprogramming of somatic cells and germ cells involves significant upregulation and downregulation, respectively, of mRNAs encoding polyadenylation factors, and RNA processing is one of the most significantly regulated biological processes during cell reprogramming. Furthermore, genes containing target sites of ES cell-specific microRNAs (miRNAs) in different portions of 3'UTR are distinctively regulated during cell reprogramming, suggesting impact of APA on miRNA targeting. Conclusions/significance: Taken together, these findings indicate that reprogramming of 3'UTRs by APA, which result from regulation of both general polyadenylation activity and cell type-specific factors and can reset post-transcriptional gene regulatory programs in the cell, is an integral part of iPS cell generation, and the APA pattern can be a good biomarker for cell type and state, useful for sample classification. The results also suggest that perturbation of the mRNA polyadenylation machinery or RNA processing activity may facilitate generation of iPS cells.
Article
Tissue plasminogen activator (tPA) is absent from normal human pancreas and is expressed in 95% of human pancreatic adenocarcinomas. We have analyzed the expression of components of the tPA system in murine pancreatic tumors and the role of tPA in neoplastic progression. Transgenic mice expressing T antigen and c-myc under the control of the elastase promoter (Ela1-TAg and Ela1-myc, respectively) were used. tPA was undetectable in normal pancreas, acinar dysplasia, ductal complexes, and in all acinar tumors. By contrast, it was consistently detected in Ela1-myc tumors showing ductal differentiation. Crossing transgenic Ela1-myc with tPA−/− mice had no effect on the proportion of ductal tumors, indicating that tPA is not involved in the acinar-to-ductal transition. Ela1-myc:tPA−/− mice showed an increased survival in comparison to control mice. All ductal tumors, and none of the acinar tumors, overexpressed the tPA receptor annexin A2, suggesting its participation in the effects mediated by tPA. Our findings indicate that murine and human pancreatic ductal tumors share molecular alterations in the tPA system that may play a role in tumor progression.
Article
We have used subtractive hybridization to isolate cDNAs overexpressed in SK-PC-1 pancreas cancer cells. Fortyfive independent clones corresponding to 11 genes were identified. Their expression in cultured pancreas cancer cells, normal pancreas tissue, and normal exocrine pancreas cultures was examined by Northern blotting. cDNA clones can be grouped into two broad categories: (1) those corresponding to genes expressed at high levels both in tumor cell lines and in primary cultures of normal pancreas, but not in normal tissue (i.e. thymosin β43,, cytokeratin 18, β-actin, pyruvate kinase and mitochondrial genes); and (2) those corresponding to genes expressed at high levels in pancreas cancer cultures but not in normal pancreas tissue or cultured cells (i.e. tissue-type plasminogen activator and cathepsin H). The overexpression of these proteases in pancreas cancers suggests that they play a role in the aggressive biological behavior of this tumor.
Article
Model organisms such as the fruit fly Drosophila melanogaster can help to elucidate the molecular basis of complex diseases such as cancer. Mutations in the Drosophila gene lethal (3) malignant brain tumor cause malignant growth in the larval brain. Here we show that l(3)mbt tumors exhibited a soma-to-germline transformation through the ectopic expression of genes normally required for germline stemness, fitness, or longevity. Orthologs of some of these genes were also expressed in human somatic tumors. In addition, inactivation of any of the germline genes nanos, vasa, piwi, or aubergine suppressed l(3)mbt malignant growth. Our results demonstrate that germline traits are necessary for tumor growth in this Drosophila model and suggest that inactivation of germline genes might have tumor-suppressing effects in other species.
Article
MYC regulates the transcription of thousands of genes required to coordinate a range of cellular processes, including those essential for proliferation, growth, differentiation, apoptosis and self-renewal. Recently, MYC has also been shown to serve as a direct regulator of ribosome biogenesis. MYC coordinates protein synthesis through the transcriptional control of RNA and protein components of ribosomes, and of gene products required for the processing of ribosomal RNA, the nuclear export of ribosomal subunits and the initiation of mRNA translation. We discuss how the modulation of ribosome biogenesis by MYC may be essential to its physiological functions as well as its pathological role in tumorigenesis.