ArticlePDF AvailableLiterature Review

Stress-induced Reversal of MicroRNA Repression and mRNA P-body Localization in Human Cells

Authors:

Abstract and Figures

In metazoa, microRNAs (miRNAs) imperfectly base-pair with the 3'-untranslated region (3'UTR) of mRNAs and prevent protein accumulation by either repressing translation or inducing mRNA degradation. Examples of specific mRNAs undergoing miRNA-mediated repression are numerous, but whether the repression is a reversible process remains largely unknown. Here, we show that cationic amino acid transporter 1 (CAT-1) mRNA and reporters bearing the CAT-1 3'UTR or its fragments can be relieved from the miRNA miR-122-induced inhibition in human hepatoma cells in response to different stress conditions. The derepression of CAT-1 mRNA is accompanied by its release from cytoplasmic processing bodies (P bodies) and its recruitment to polysomes, indicating that P bodies act as storage sites for mRNAs inhibited by miRNAs. The derepression requires binding of HuR, an AU-rich-element-binding ELAV family protein, to the 3'UTR of CAT-1 mRNA. We propose that proteins interacting with the 3'UTR will generally act as modifiers altering the potential of miRNAs to repress gene expression.
Content may be subject to copyright.
miRNAs are 20- to 22-nucleotide-long regulatory
RNAs expressed in plants and metazoan animals. Current
estimates indicate that hundreds of different miRNAs are
encoded in individual genomes, with the number of
human miRNAs possibly reaching 1000. Approximately
30% of all human genes are predicted to be subject to
miRNA regulation. Specific functions and target mRNAs
have been assigned to only a few dozen miRNAs, but it is
apparent that miRNAs participate in the regulation of
many different biological processes. Changes in miRNA
expression are observed in human pathologies and some
miRNAs were shown to act as oncogenes or tumor
supressors (for review, see Ambros 2004; Bartel 2004;
Wienholds and Plasterk 2005).
miRNAs regulate gene expression posttranscriptionally,
by base-pairing to target mRNAs. In animals, most inves-
tigated miRNAs form imperfect hybrids with sequences in
the 3UTR, with the miRNA 5-proximal “seed” region
(positions 2–8) providing most of the pairing specificity
(for review, see Filipowicz 2005; Tomari and Zamore
2005). The miRNA association results in translational
repression, frequently accompanied by a considerable
degradation of the mRNA by a non-RNAi mechanism (for
review, see Pillai 2005; Valencia-Sanchez et al. 2006).
Argonaute (AGO) proteins are the essential and best-
characterized components of miRNPs. In mammals, only
one of the four AGO proteins, AGO2, is competent to cat-
alyze cleavage of mRNA in the RNA interference (RNAi)-
like mechanism (Liu et al. 2004; Meister et al. 2004). On the
other hand, all four AGO proteins, AGO1–4, appear to
function in miRNA repression (Liu et al. 2004; Meister et
al. 2004; Pillai et al. 2004). The mechanism of translational
inhibition by miRNAs is not well understood. Some natural
or model miRNAs interfere with the initiation of protein
synthesis (Humphreys et al. 2005; Pillai et al. 2005),
although others may affect more downstream steps in trans-
lation (Olsen and Ambros 1999; Petersen et al. 2006). The
AGO proteins and repressed mRNAs are enriched in the
cytoplasmic P bodies (Jakymiw et al. 2005; Liu et al. 2005;
Pillai et al. 2005). The P-body association may represent a
secondary event, which follows the translation inhibition
step. Since mRNA catabolic enzymes also reside in P bod-
ies, the relocation likely results in the reported mRNA
degradation (Pillai 2005; Valencia-Sanchez et al. 2006).
To date, miRNAs have been primarily identified as
negative regulators of expression of cellular mRNAs, and
it remains unknown whether the inhibition of a specific
mRNA can be effectively reversed. Clearly, the ability to
disengage miRNPs from the repressed mRNA, or render
them inactive, would make miRNA regulation much
more dynamic and also more responsive to specific cellu-
lar needs. In this work, we present evidence that CAT-1
mRNA, which encodes the high-affinity cationic amino
acid transporter and which is translationally repressed by
miR-122 in Huh7 hepatoma cells, can be relieved from
the miR-122 repression by subjecting Huh7 cells to dif-
ferent stress conditions. The derepression is accompanied
by the release of CAT-1 mRNA from P bodies and its
recruitment to polysomes, consistent with miR-122
inhibiting translational initiation. We provide evidence
that the stress-induced up-regulation is mediated by bind-
ing of HuR, an AU-rich element (ARE)-binding protein,
to the 3UTR of CAT-1 mRNA.
RESULTS
CAT-1 mRNA as a Model to Investigate
Reversibility of miRNA-mediated Repression
To investigate whether mRNA can be relieved from
miRNA-mediated repression, we looked at the mRNA
encoding the high-affinity cationic amino acid trans-
porter, CAT-1, a member of the CAT (or SLC7A1-4)
family of system y
+
transporters. CAT-1, which facili-
Stress-induced Reversal of MicroRNA Repression and mRNA
P-body Localization in Human Cells
S.N. BHATTACHARYYA,* R. HABERMACHER,* U. MARTINE,
E.I. CLOSS,
AND W. FILIPOWICZ*
*Friedrich Miescher Institute for Biomedical Research, 4002 Basel, Switzerland;
Department of Pharmacology,
Johannes Gutenberg University, 67, 55101 Mainz, Germany
In metazoa, microRNAs (miRNAs) imperfectly base-pair with the 3-untranslated region (3UTR) of mRNAs and prevent
protein accumulation by either repressing translation or inducing mRNA degradation. Examples of specific mRNAs under-
going miRNA-mediated repression are numerous, but whether the repression is a reversible process remains largely
unknown. Here, we show that cationic amino acid transporter 1 (CAT-1) mRNA and reporters bearing the CAT-1 3UTR
or its fragments can be relieved from the miRNA miR-122-induced inhibition in human hepatoma cells in response to
different stress conditions. The derepression of CAT-1 mRNA is accompanied by its release from cytoplasmic processing
bodies (P bodies) and its recruitment to polysomes, indicating that P bodies act as storage sites for mRNAs inhibited by
miRNAs. The derepression requires binding of HuR, an AU-rich-element-binding ELAV family protein, to the 3UTR of
CAT-1 mRNA. We propose that proteins interacting with the 3UTR will generally act as modifiers altering the potential
of miRNAs to repress gene expression.
Cold Spring Harbor Symposia on Quantitative Biology, Volume LXXI. © 2006 Cold Spring Harbor Laboratory Press 978-087969817-1 513
513-522_Bhattacharyya_Symp71.qxd 2/8/07 1:42 PM Page 513
tates uptake of arginine and lysine in mammalian cells, is
expressed ubiquitously, but its levels vary significantly in
different cells and tissues and its expression is known to
undergo extensive regulation at both transcriptional and
posttranscriptional levels (for review, see Hatzoglou et al.
2004). CAT-1 regulation has been studied intensively in
rat C6 glioma cells, where transcription of the gene and
stability and translation of the mRNA are up-regulated in
response to different types of cellular stress (Yaman et al.
2002; Haztoglou et al. 2004). In human hepatoma Huh7
cells, CAT-1 mRNA activity is regulated by a liver-spe-
cific miRNA, miR-122. The human CAT-1 3UTR con-
tains several potential target sites for miR-122 (see Fig.
2A). Experiments performed with endogenous CAT-1
mRNA (Bhattacharyya et al. 2006) and reporters contain-
ing its 3UTR (or fragments thereof) indicated that miR-
122 has a repressive effect on translation and, in the case
in some chimeric reporter mRNAs, may also cause lim-
ited destabilization of the target RNA (Chang et al. 2004;
Bhattacharyya et al. 2006). Maintenance of low CAT-1
activity in liver cells is important to avoid hydrolysis of
the plasma arginine by arginase, a highly expressed
enzyme in hepatocytes, which catalyzes the last step of
the urea cycle (hydrolysis of arginine to ornithine and
urea). However, under certain conditions, e.g., when urea
cycle enzymes are down-regulated or during liver regen-
eration, CAT-1 expression is induced, most likely to sus-
tain hepatocellular protein synthesis.
Amino Acid Deprivation Induces Translational
Up-regulation of CAT-1 mRNA in Huh7 Cells
In cultured rat C6 glioma cells, CAT-1 protein expres-
sion increases in response to amino acid deprivation and
other forms of cellular stress. Both transcriptional and
posttranscriptional regulation were reported to contribute
to the increase (Hatzoglou et al. 2004). We investigated
the effect of amino acid starvation on expression of CAT-
1 protein and mRNA in hepatoma Huh7 cells. The CAT-
1 protein level increased markedly after 1 hour of
starvation and then remained unchanged during several
additional hours of amino acid depletion (Fig. 1A). In con-
514 BHATTACHARYYA ET AL.
Figure 1. Starvation of Huh7 cells induces expression of CAT-1 protein independent of transcription. (A) Stress-induced expression of
CAT-1 in Huh7 cells is a posttranscriptional event. Huh7 cells were cultured for 0–4 hours in amino-acid-depleted medium in the absence
or presence of indicated inhibitors. Proteins were treated with PGNase F and analyzed by western blotting using indicated antibodies.
Two lanes on the right contained protein extracts of DLD-1 cells, abundant in CAT-1. Positions of glycosylated and deglycosylated
CAT-1 are indicated by an arrowhead and arrow. (B) Analysis of RNA isolated from Huh7 cells starved of amino acids in the presence
of indicated inhibitors. (Upper panel) Real-time PCR quantification of CAT-1 mRNA. Normalized values are means (+/–
SD) from three
independent experiments, with GAPDH mRNA serving as an internal control. (Two lower panels) Northern analysis of miR-122 and
staining of the gel with ethidium bromide. Details of methodological procedures used for experiments described in this and other figures
are described in Bhattacharyya et al. (2006). (Reprinted, with permission, from Bhattacharyya et al. 2006 [© Elsevier].)
513-522_Bhattacharyya_Symp71.qxd 2/8/07 1:42 PM Page 514
trast, the substantial increase in CAT-1 mRNA level,
measured by either real-time polymerase chain reaction
(PCR) (Fig. 1B) or northern blotting (not shown)
(Bhattacharyya et al. 2006), was only detectable after 3–4
hours of starvation. In HepG2 hepatoma cells, which do
not express miRNA miR-122, no appreciable change in
either CAT-1 protein or mRNA level was observed during
4 hours of starvation (data not shown). The early induction
of the CAT-1 protein in Huh7 cells was independent of
RNA polymerase II transcription, since treatment with
inhibitors of RNA polymerase II, either actinomycin D
(ActD) or α-amanitin (α-Am), had no effect (Fig. 1A).
However, the protein accumulation was inhibited by
cycloheximide (CHX), an inhibitor of translational
elongation (Fig. 1A). Amino acid starvation and treatment
of cells with ActD or α-Am had no effect on the level of
miR-122 or Ago2, essential components of miRNPs (Fig.
1A,B). However, inclusion of either inhibitor prevented
the late (4-hour time point) accumulation of CAT-1
mRNA, demonstrating that they effectively inhibit RNA
polymerase II transcription in Huh7 cells.
It was important to exclude the possibility that CAT-1
protein turns over rapidly in cells grown in the medium rich
in amino acids, and its apparent induction by starvation is
due to an increase in protein stability. To this end, we
have performed western analysis of lysates prepared from
cells grown in either the presence or absence of CXH.
The analysis indicated that instead of stabilizing the
CAT-1 protein, the starvation increased its turnover
(Bhattacharyya et al. 2006). The accelerated decay of
CAT-1 in stressed cells provides a plausible explanation
for the observation that steady-state levels of the protein
do not continue to increase at times beyond 1 hour post-
starvation. Taken together, the data indicate that starva-
tion of Huh7 cells results in a de novo synthesis of the
CAT-1 protein from the preexisting mRNA pool.
Response to Amino Acid Starvation and Other
Types of Stress Is Mediated by the
CAT-1 3UTR and Involves miR-122
To test whether the translational induction described
above is mediated by the CAT-1 mRNA 3UTR, we
measured the effect of starvation on the activity of dif-
ferent RL-cat reporters (Fig. 2A) in Huh7 cells. In RL-
catA, the RL-coding region is fused to the 2.5-kb 3UTR
found in a short form of CAT-1 mRNA. In RL-catB, the
3-proximal 1-kb region (referred to as region D) of the
CAT-1 3UTR is deleted. The remaining part contains
three predicted miR-122-binding sites identified in the
REVERSAL OF
MIRNA-MEDIATED REPRESSION 515
Figure 2. Effect of different types of cellular stress on activity of RL reporters in Huh7 and HepG2 cells. (A) Schemes of reporters
bearing different segments of the CAT-1 3UTR fused to the RL-coding region. Positions of potential miR-122-binding sites (1–3)
and the approximately 1-kb region D and its AU-rich subregion ARD are indicated. (B) Expression of the RL-catA reporter is specif-
ically up-regulated in stressed Huh7 cells (upper panel) but not HepG2 cells (lower panel). Cells transfected with indicated reporters
were starved for 2 hours (Starved) or grown in the presence of either thapsigargin (TG; 2 hr) or arsenite (Ars; 30 min). ActD or CHX
was added at the time of the shift to stress conditions. Values are normalized to activities in nonstressed (Fed) cells, which were set
to 1. (Reprinted, with permission, from Bhattacharyya et al. 2006 [© Elsevier].)
513-522_Bhattacharyya_Symp71.qxd 2/8/07 1:42 PM Page 515
2.5-kb CAT-1 3UTR. In RL-catC, the CAT-1 3UTR is
shortened further to eliminate the region containing the
miR-122 sites. Reporter activity, normalized to activity
of the coexpressed firefly luciferase (FL), was tested in
transfected Huh7 and HepG2 cells (Fig. 2B). In Huh7
cells, an approximately fourfold induction of RL activity
was observed upon starvation of cells transfected with
the reporter containing miR-122 sites, RL-catA, but not
with that devoid of miRNA sites, RL-catC. Starvation
increased expression of RL-catB by approximately 30%
(Fig. 2B). As in the case of endogenous CAT-1 protein,
the stress-induced expression of RL from RL-catA was
inhibited by addition of CHX but not ActD (Fig. 2B).
Likewise, other stress conditions such as the ER stress
(induced by thapsigargin) or oxidative stress (induced by
arsenite) stimulated expression of RL-catA approxi-
mately 2.5-fold, whereas the effect on other reporters
was either minimal or absent (Fig. 2B). Amino acid
starvation and treatment with thapsigargin or arsenite
had no effect on the level of RL-catA mRNA, indicating
that the effect was posttranscriptional (Bhattacharyya
et al. 2006).
To test whether the stress-mediated activation of RL-
catA indeed involves miR-122, we investigated whether
responses observed in Huh7 cells can be reproduced in
HepG2 cells, which do not express miR-122. Exposure of
HepG2 cells to different forms of stress had no effect on
expression of any RL-cat reporter (Fig. 2B, lower panel).
However, when HepG2 cells were cotransfected with
miR-122, up-regulation of RL-catA was clearly evident
in starved cells, with activity of RL-catB and RL-catC
remaining unchanged (Bhattacharyya et al. 2006).
HuR Protein Interacts with CAT-1 3UTR and
Is Essential for the Derepression
The stress-inducible RL-catA reporter differs from
the reporter not undergoing activation, RL-catB, by the
presence of an additional segment (region D) of the
CAT-1 3UTR (Fig. 2A). We found that region D is
essential for the stress-induced relief of the miR-122-
mediated repression in Huh7 cells. Moreover, this
region can confer stress inducibility on a reporter inhib-
ited by let-7 RNA, an abundant miRNA expressed in
HeLa cells. The let-7-specific RL-3xBulge reporter
(Pillai et al. 2005) was used to further dissect CAT-1
region D. It was found that the central part of region D,
containing sequences rich in A+U and U residues and
referred to hereafter as region ARD (see Fig. 2A), is
essential for mediating the stress-induced reversal of
miRNA repression (Bhattacharyya et al. 2006).
HuR is an AU-rich element (ARE)-binding protein, a
member of the ELAV family of proteins, implicated in
different aspects of posttranscriptional regulation (for
review, see Brennan and Steitz 2001; Katsanou et al.
2005; Lal et al. 2005). In response to various types of
cellular stress, HuR is mobilized from the nucleus to the
cytosol, where it may modulate translation or increase
the stability of different target mRNAs, including CAT-
1 mRNA in rat glioma cells (Yaman et al. 2002). We
found that HuR relocates from the nucleus to the cytosol
upon amino acid starvation also in Huh7 cells (Fig. 3A)
and that the RNA-mediated depletion of HuR, using two
different small interfering RNAs (siRNAs) (Fig. 3B),
eliminates the RL-catA response in comparison to con-
trol cells or cells treated with control siRNA (Fig. 3C).
We tested, by native gel analysis, if the
32
P-labeled ARD
fragment, which is essential for mediating the stress-
induced derepression, can interact with a recombinant
GST-HuR fusion protein. As shown in Figure 3D, puri-
fied GST-HuR but not GST, formed a complex with the
ARD fragment, and the complex was competed by an
excess of unlabeled fragment ARD but not the ΔARD
portion of region D. Additional experiments indicated
that CAT-1 mRNA can be specifically immunoselected
from the soluble cytoplasmic fraction of starved Huh7
cells by the anti-HuR antibody. We have also demon-
strated that HuR binding per se does not cause activation
of the RL reporter devoid of miR-122 sites and not
repressed by the miRNA (Bhattacharyya et al. 2006).
Taken together, the above experiments indicate that HuR
has a role in the stress-induced activation of CAT-1 mRNA
and RL reporters undergoing repression mediated by
either miR-122 or let-7 RNA.
Stress Induces HuR-dependent Relocation of
CAT-1 mRNA from P bodies
mRNA reporters repressed by miRNAs were found to
localize in P bodies (Liu et al. 2005; Pillai et al. 2005).
We analyzed the intracellular localization of the
endogenous CAT-1 mRNA and RL-cat reporters in cells
grown under different conditions. In situ hybridization
revealed that in nonstarved Huh7 cells, CAT-1 mRNA
is concentrated in P bodies, as demonstrated by its colo-
calization with the P-body marker, GFP-Dcp1a (Fig.
4A). P-body enrichment of the mRNA was abolished
when cells were transfected with the 2-O-methyl
oligonucleotide complementary to miR-122 but not
with control anti-miR-15 oligonucleotide (Fig. 4B),
indicating that the localization was dependent on miR-
122. Most importantly, in Huh7 cells grown for 2 hours
under amino acid deficiency, a condition that markedly
increases CAT-1 protein without an effect on the mRNA
level (see Fig. 1), CAT-1 mRNA was no longer
detectable in P bodies (Fig. 4A). Starvation did not pro-
duce an appreciable decrease in the miR-122 signal in P
bodies (Bhattacharyya et al. 2006), arguing for an effect
specific for the CAT-1 mRNA and possibly only a lim-
ited number of other mRNAs among the many regulated
by miR-122 in liver cells (Krutzfeldt et al. 2005). As
expected, CAT-1 mRNA did not colocalize to P bodies
in HepG2 cells. However, in HepG2 cells transfected
with miR-122, CAT-1 mRNA was enriched in P bodies,
further supporting the idea that the repression and P-
body localization of CAT-1 mRNA are controlled by
miR-122 (Bhattacharyya et al. 2006).
To find out whether the relocation of CAT-1 mRNA
from P bodies, like the activation of its expression, also
requires HuR, we studied CAT-1 distribution in Huh7
cells in which HuR protein had been knocked down by
RNAi. The knockdown had no effect on the P-body
516 BHATTACHARYYA ET AL.
513-522_Bhattacharyya_Symp71.qxd 2/8/07 1:42 PM Page 516
REVERSAL OF MIRNA-MEDIATED REPRESSION 517
Figure 3. HuR relocates from the nucleus to the cytoplasm in stressed Huh7 cells, interacts with the CAT-1 3UTR, and is required for the
stress-induced derepression of the RL-catA reporter. (A) Starvation-induced relocation of HuR. Huh7 cells, either nonstarved (Fed) or
starved for 2 hours (Starved) were fixed, and the localization of HuR (red) was determined using mouse anti-HuR monoclonal antibodies.
(B) Two different siRNAs effectively deplete HuR. Western blot was performed 72 hours after siRNA transfection. (C) Knockdown of
HuR prevents starvation-induced derepression of RL-catA. Huh7 cells were cotransfected with RL-cat reporters and indicated siRNAs;
72 hours after transfection, cells were transferred for an additional 2 hours to a medium either with or without amino acids. The values are
means from three transfections +/– standard deviation. (D) Recombinant GST-HuR protein interacts with the CAT-1 ARD fragment.
32
P-labeled ARD RNA was incubated with either purified GST-HuR or GST alone in the absence or presence of indicated cold competi-
tors, and complexes were analyzed on a native gel. (Reprinted, with permission, from Bhattacharyya et al. 2006 [© Elsevier].)
enrichment of CAT-1 mRNA in control cells. However,
it prevented mobilization of the mRNA from these struc-
tures upon amino acid starvation. In cells transfected with
nonspecific siRNA, the relocation did take place, simi-
larly as in nontreated Huh7 cells (Fig. 4C).
We also determined the intracellular localization of
different RL reporters repressed by either miR-122 or
let-7 miRNA. The repressed reporters localized to P bod-
ies, and their relocalization from these structures in
response to amino acid depletion was dependent on the
CAT-1 region D present downstream from the miRNA-
binding sites. Reporters devoid of region D remained
enriched in P bodies in both nonstarved and starved cells
(Bhattacharyya et al. 2006).
In starved Huh7 cells, CAT-1 mRNA becomes
extractable from cells permeabilized with digitonine,
very likely as a consequence of the relocation of mRNA
from P bodies to the cytosol (Pillai et al. 2005;
Bhattacharyya et al. 2006). As shown in Figure 5A, the
amount of CAT-1 mRNA present in the cytosol prepared
from permeabilized Huh7 cells increased already after 20
minutes following the shift to the amino-acid-depleted
medium. Importantly, the shift of the CAT-1 mRNA to
the cytosol upon stressing the cells occurred with a kinet-
ics similar to the appearance of HuR in this fraction and
also paralleled the accumulation of CAT-1 protein (Fig.
5). Hence, the stress-induced derepression of the CAT-1
mRNA translation appears to be a rapid event.
Together, the results demonstrate that CAT-1 mRNA
and RL reporters are concentrated in P bodies when
repressed by the miRNA but are rapidly mobilized from
these structures under conditions, including cellular
stress, that preclude miRNA repression. The experiments
further support a role for HuR and the CAT-1 mRNA
region D in mediating the response induced by the amino
acid stress. Moreover, they indicate that such a response
may be a more general phenomenon, applying to differ-
ent cell types and miRNA–mRNA combinations.
513-522_Bhattacharyya_Symp71.qxd 2/8/07 1:42 PM Page 517
Stress-induced Relocation of CAT-1 mRNA from
P Bodies Is Accompanied by Its Entry to Polysomes
We found previously that repression of protein syn-
thesis by let-7 RNA in HeLa cells is accompanied by a
less effective entry of target reporters to polysomes,
indicative of the translation initiation block (Pillai et al.
2005). Gradient analysis of Huh7 cell extracts indicated
that amino acid starvation results in an increase in the
fraction of CAT-1 mRNA associated with polysomes. In
contrast, β-tubulin mRNA moved toward the top of the
gradient in response to starvation, consistent with a gen-
eral inhibitory effect of stress on translation (Fig. 6A,B).
Treatment of Huh7 cells with anti-miR-122 but not con-
trol anti-let-7a 2-O-methyl oligonucleotide resulted in
the CAT-1 mRNA shift to polysomes similar to that
induced by starvation (Fig. 6C). Of note, a fraction of
the “repressed” CAT-1 mRNA sedimented faster than
the polysome-associated CAT-1 mRNA present in
stressed or anti-miR-122-treated cells. Possibly, this
material represents P-body aggregates containing CAT-
1 mRNA.
Taken together, these data indicate that relocation of
CAT-1 mRNA from P bodies and relief of the miRNA-
mediated repression is accompanied by recruitment of
CAT-1 mRNA to polysomes, consistent with miR-122
inhibiting translational initiation.
CONCLUSIONS
Our work demonstrates that CAT-1 mRNA, and
reporters bearing the CAT-1 3UTR or its fragments,
can be relieved from miR-122-mediated repression in
Huh7 cells subjected to amino acid deprivation or the
ER or oxidative stress. Observations that the response to
the amino acid starvation stress can be recapitulated in
other cell lines by either an ectopic supply of miR-122
or the use of chimeric reporters targeted by another
miRNA argue for a general importance of this type of
regulation. We also demonstrate that repressed CAT-1
and reporter mRNAs accumulate in P bodies in an miR-
122-dependent process and that the derepression is
accompanied by the release of the mRNAs from these
structures. The demonstrated stress-induced mobiliza-
tion of mRNAs from P bodies provides evidence that
metazoan P bodies represent sites not only of mRNA
turnover, but also of storage of translationally
repressed mRNAs. So far, such evidence was available
for baker’s yeast, an organism lacking miRNA regula-
tion (Brengues et al. 2005). Time-course experiments
measuring the appearance of CAT-1 mRNA in a soluble
cytosolic fraction and the formation of CAT-1 protein
suggest that reactivation of mRNAs sequestered in P
bodies is a very rapid process. Hence, it is tempting to
speculate that P bodies may act as general storage sites
for mRNAs, which need to be quickly mobilized into
polysomes under specific cellular conditions.
Although miRNAs may affect gene expression in dif-
ferent ways (for review, see Pillai 2005; Valencia-
Sanchez et al. 2006), recent findings indicated that let-7
RNA and some model miRNAs in mammals inhibit
518 BHATTACHARYYA ET AL.
Figure 4. CAT-1 mRNA accumulation in P bodies is miR-122-
dependent, and its stress-induced relocation from P-bodies
requires HuR. Details of cell treatment are indicated at the left of
each row. P bodies were visualized by measuring GFP-Dcp1a
fluorescence (green), and CAT-1 by in situ hybridization with
Cy3-labeled probes (red). DAPI (blue) stained the nucleus. Cells
were starved for 2 hours prior to fixation. Exposure time of the
red channel for starved Huh7 cells (A), the anti-miR-122 row (B),
and starved siControl cells (C) was ten times longer than that for
other images. (Insets) Enlargements of indicated regions. For
quantification and discussion of overlap between CAT-1 mRNA
and GFP-Dcp1a foci, see Bhattacharyya et al. (2006). (A,B)
CAT-1 mRNA is mobilized from P bodies upon starvation of
Huh7 cells (A) or upon transfection with anti-miR-122 but not
anti-miR-15 oligonucleotide (B). Bar, 5 μm. (C) HuR is required
for the stress-induced mobilization of CAT-1 mRNA from
P bodies. Huh7 cells were cotransfected with pGFP-Dcp1a and
either anti-HuR or control siRNA. After 48 hours, a fraction of
the cells was starved for 2 hours. (Reprinted, with permission,
from Bhattacharyya et al. 2006 [© Elsevier].)
513-522_Bhattacharyya_Symp71.qxd 2/8/07 1:42 PM Page 518
REVERSAL OF MIRNA-MEDIATED REPRESSION 519
Figure 5. The stress-induced relocation of HuR
protein and CAT-1 mRNA to the cytosol and the
accumulation of CAT-1 protein are rapid events.
(A) Accumulation of CAT-1 mRNA and HuR
protein in the cytosol upon stressing Huh7 cells
occurs with a similar kinetics. Huh7 cells were
starved for amino acids for increasing time, and
levels of CAT-1 mRNA (upper panels) and HuR
protein (western; lower panels) were measured in
the cytosolic and total extracts of starved cells.
Cytosolic extracts were prepared by permeabiliz-
ing cells with digitonine (Bhattacharyya et al.
2006). (B) Kinetics of the CAT-1 protein accu-
mulation in Huh7 cells subjected to amino acid
deprivation stress, determined by western.
Figure 6. Stress- or anti-miR-122-induced relocation of CAT-1 mRNA from P bodies is accompanied by its recruitment to polysomes.
(A) Distribution of CAT-1 mRNA in extracts from cells fed with amino acids (upper panels) or starved for amino acids (lower pan-
els). RNA extracted from individual fractions was analyzed by northern blots with probes specific for CAT-1 and β-tubulin mRNAs.
Two lanes at the right represent input RNA isolated from fed and starved cells. (B) Quantification of distribution of mRNAs analyzed
in panel A, expressed as a percentage of total radioactivity present in each lane. (C) Distribution of CAT-1 mRNA in extracts from
cells treated with either anti-let-7a or anti-miR-122 (middle panels). A
260
profile of the anti-let-7a extract was similar to that shown
at the top, and distribution of β-tubulin mRNA in both gradients was similar to that shown in panel A, fed cells (data not shown).
Quantification of CAT-1 mRNA is at the bottom. (Reprinted, with permission, from Bhattacharyya et al. 2006 [© Elsevier].)
translation of reporter mRNAs at the initiation step
(Humphreys et al. 2005; Pillai et al. 2005) and that
repressed mRNAs localize to P bodies for either storage
or degradation (Liu et al. 2005; Pillai 2005; Pillai et al.
2005; Valencia-Sanchez et al. 2006). The data presented
in this work indicate that this scenario also applies to the
miRNA-mediated regulation of an endogenous mRNA.
The stress- or anti-miR-122-induced relocalization of
CAT-1 mRNA from P bodies was accompanied by its
increased association with polysomes, consistent with the
miRNA inhibition acting at the initiation step of transla-
tion. Importantly, activation of CAT-1 and reporter
mRNAs by exposing the cells to stress or transfecting
them with anti-miR-122 oligonucleotide had no apprecia-
ble effect on the mRNA level, indicating that miR-122 in
Huh7 cells controls CAT-1 mRNA mainly at the transla-
tional level and not the stability level.
Our results strongly argue for a role of HuR in the
stress-induced activation of mRNAs undergoing miR-
122-mediated repression. The HuR knockdown pre-
vented both the translational activation of repressed
mRNAs and their mobilization from P bodies.
513-522_Bhattacharyya_Symp71.qxd 2/8/07 1:42 PM Page 519
Moreover, a recombinant HuR interacted with the CAT-
1 3UTR fragment implicated in mediating the stimula-
tory effect of stress (see Fig. 3D), whereas the
endogenous HuR associated with all reporter mRNAs
undergoing derepression but not with their inactive vari-
ants bearing mutations in the predicted HuR-binding
sites (Bhattacharyya et al. 2006). HuR is a ubiquitously
expressed member of the ELAV family of proteins,
which also comprises three neuronal proteins. In
response to different types of cellular stress, HuR is
mobilized from the nucleus to the cytosol, where it may
modulate translation and/or stability of different
mRNAs (for review, see Brennan and Steitz 2001;
Katsanou et al. 2005; Lal et al. 2005). Our data suggest
that at least some of the known effects of HuR, both
translational and stability-related, may be due to the
interference of HuR with the function of miRNAs,
which would result in enhanced translation or stability
of mRNA. HuR shuttles between the nucleus and cyto-
plasm, and HuR has been suggested to bind some ARE-
containing mRNAs in the nucleus and chaperone them
to the cytoplasm (Gallouzi and Steitz 2001; Lal et al.
2005). The in situ experiments demonstrating CAT-1
mRNA and RL reporter localization in P bodies in
unstressed hepatoma and HeLa cells make it very
unlikely that redistribution of mRNA between the
nucleus and the cytoplasm contributes to the effects
described in our work.
The demonstration that mRNAs repressed by miRNAs
can depart P bodies to return to active translation indi-
cates that P bodies are dynamic structures, exchanging
their content rapidly with that of the cytosol (Andrei et
al. 2005; Brengues et al. 2005). It is possible that HuR,
following its relocation to the cytoplasm in stressed
cells, shifts the P-body-to-cytosol equilibrium of
repressed mRNAs by binding to AREs in the 3UTR.
Whether this is accompanied by the dissociation of
miRNPs from the mRNA or just prevents miRNPs from
acting as effectors in the repression remains to be estab-
lished (Fig. 7). It will also be interesting to study other
details of HuR involvement in relieving the miRNP
repression. The findings that several identified protein
ligands of HuR are protein phosphatase PP2A inhibitors
(Brennan et al. 2000) and that HuR can undergo methy-
lation (Li et al. 2002) or synergize with other RNA-bind-
ing proteins (Katsanou et al. 2005) indicate that HuR is a
part of an elaborated network involved in posttranscrip-
tional regulation of gene expression.
Other examples of the reversible action of miRNAs
have recently been identified in neuronal cells. In neu-
rons, many mRNAs are transported along the dendrites
as repressed mRNPs to become translated at the final
destination, dendritic spines, upon synaptic activation.
Such local translation is important for spine develop-
ment, learning, and memory (Sutton and Schuman
2005). miRNA miR-134 is implicated in translational
regulation of Limk1, a protein kinase important for
spine development, in cultured rat neurons. Limk1
mRNA appears to be relieved from the miR-134-medi-
ated repression in dendritic spines in response to extra-
cellular stimuli, in a process involving mTOR
(mammalian target of rapamycin) (Schratt et al. 2006).
In Drosophila, stimulation of olfactory neurons, which
leads to long-term memory formation, is associated with
proteolysis of Armitage, a protein essential for the
assembly of the RNA-induced silencing complex
(RISC)/miRNP complexes. As the result of Armitage
degradation, mRNAs that are normally repressed by
miRNAs, including the one encoding calcium/calmod-
ulin-dependent protein kinase II (CamKII), become
520 BHATTACHARYYA ET AL.
Figure 7. Model of the stress- and HuR-mediated relief of CAT-1 mRNA repression by miR-122.
513-522_Bhattacharyya_Symp71.qxd 2/8/07 1:42 PM Page 520
effectively translated in the synapse (Ashraf et al. 2006).
The regulation in Drosophila differs from the examples
studied in mammalian cells, in that the Armitage deple-
tion most probably indiscriminately prevents the forma-
tion of repressed mRNPs, rather than causing
reactivation of specific mRNAs controlled by miRNAs.
In addition to HuR, three other ELAV proteins,
HuA, HuB, and HuD, are expressed in neurons, and a
role of HuD in stability and translation of some neu-
ronal mRNAs has already been documented (Perrone-
Bizzozero and Bolognani 2002). It will be interesting
to find out whether, similarly to HuR in hepatoma
cells, the other ELAV proteins modulate miRNA-
mediated regulation in neurons. Likewise, it will be
important to determine whether other classes of RNA-
binding proteins interacting with the 3UTR will act as
modifiers altering the potential of miRNAs to repress
gene expression.
ACKNOWLEDGMENTS
We thank C. Clayton, T. Hobman, Y. Nagamine, C.
Ender, R. Pillai, C. Artus, and E. Bertrand for providing
plasmids and/or antibodies. S.N.B. is a recipient of a long-
term HFSP fellowship. The Friedrich Miescher Institute is
supported by the Novartis Research Foundation.
REFERENCES
Andrei M.A., Ingelfinger D., Heintzmann R., Achsel T.,
Rivera-Pomar R., and Luhrmann R. 2005. A role for eIF4E
and eIF4E-transporter in targeting mRNPs to mammalian
processing bodies. RNA 11: 717.
Ambros V. 2004. The functions of animal microRNAs. Nature
431: 350.
Ashraf S.I., McLoon A.L., Sclarsic S.M., and Kunes S. 2006.
Synaptic protein synthesis associated with memory is regu-
lated by the RISC pathway in Drosophila. Cell 124: 191.
Bartel D.P. 2004. MicroRNAs: Genomics, biogenesis, mecha-
nism, and function. Cell 116: 281.
Bhattacharyya S.N., Habermacher R., Martine U., Closs E.I.,
and Filipowicz W. 2006. Relief of microRNA-mediated
translational repression in human cells subjected to stress.
Cell 125: 1111.
Brengues M., Teixeira D., and Parker R. 2005. Movement of
eukaryotic mRNAs between polysomes and cytoplasmic
processing bodies. Science 310: 486.
Brennan C.M. and Steitz J.A. 2001. HuR and mRNA stability.
Cell. Mol. Life Sci. 58: 266.
Brennan C.M., Gallouzi I.E., and Steitz J.A. 2000. Protein
ligands to HuR modulate its interaction with target mRNAs
in vivo. J. Cell Biol. 151: 11.
Chang J., Nicolas E., Marks D., Sander C., Lerro A., Buendia
M.A., Xu C., Mason W.S., Moloshok T., Bort R., et al.
2004. miR-122, a mammalian liver-specific microRNA, is
processed from hcr mRNA and may downregulate the high
affinity cationic amino acid transporter CAT-1. RNA Biol.
1: 106.
Filipowicz W. 2005. RNAi: The nuts and bolts of the RISC
machine. Cell 122: 17.
Gallouzi I.E. and Steitz J.A. 2001. Delineation of mRNA
export pathways by the use of cell-permeable peptides.
Science 294: 1895.
Hatzoglou M., Fernandez J., Yaman I., and Closs E. 2004.
Regulation of cationic amino acid transport: the story of the
CAT-1 transporter. Annu. Rev. Nutr. 24: 377.
Humphreys D.T., Westman B.J., Martin D.I., and Preiss T.
2005. MicroRNAs control translation initiation by inhibit-
ing eukaryotic initiation factor 4E/cap and poly(A) tail func-
tion. Proc. Natl. Acad. Sci. 102: 16961.
Jakymiw A., Lian S., Eystathioy T., Li S., Satoh M., Hamel
J.C., Fritzler M.J., and Chan E.K. 2005. Disruption of GW
bodies impairs mammalian RNA interference. Nat. Cell
Biol. 7: 1267.
Katsanou V., Papadaki O., Milatos S., Blackshear P.J.,
Anderson P., Kollias G., and Kontoyiannis D.L. 2005. HuR
as a negative posttranscriptional modulator in inflammation.
Mol. Cell 16: 777.
Krutzfeldt J., Rajewsky N., Braich R., Rajeev K.G., Tuschl T.,
Manoharan M., and Stoffel M. 2005. Silencing of
microRNAs in vivo with “antagomirs.” Nature 438: 685.
Lal A., Kawai T., Yang X., Mazan-Mamczarz K., and Gorospe
M. 2005. Antiapoptotic function of RNA-binding protein
HuR effected through prothymosin alpha. EMBO J. 24:
1852.
Li H., Park S., Kilburn B., Jelinek M.A., Henschen-Edman A.,
Aswad D., Stallcup M.R., and Laird-Offringa I.A. 2002.
Lipopolysaccharide-induced methylation of HuR, an
mRNA-stabilizing protein, by CARM1. J. Biol. Chem. 277:
44623.
Liu J., Valencia-Sanchez M.A., Hannon G.J., and Parker R.
2005. MicroRNA-dependent localization of targeted
mRNAs to mammalian P-bodies. Nat. Cell Biol. 7: 719.
Liu J., Carmell M.A., Rivas F.V., Marsden C.G., Thomson
J.M., Song J.J., Hammond S.M., Joshua-Tor L., and Hannon
G.J. 2004. Argonaute2 is the catalytic engine of mammalian
RNAi. Science 305: 1437.
Meister G., Landthaler M., Patkaniowska A., Dorsett Y., Teng
G., and Tuschl T. 2004. Human Argonaute2 mediates RNA
cleavage targeted by miRNAs and siRNAs. Mol. Cell 15:
185.
Olsen P.H. and Ambros V. 1999. The lin-4 regulatory RNA
controls developmental timing in Caenorhabditis elegans
by blocking LIN-14 protein synthesis after the initiation of
translation. Dev. Biol. 216: 671.
Perrone-Bizzozero N. and Bolognani F. 2002. Role of HuD
and other RNA-binding proteins in neural development and
plasticity. J. Neurosci. Res. 68: 121.
Petersen C.P., Bordeleau M.E., Pelletier J., and Sharp P.A.
2006. Short RNAs repress translation after initiation in
mammalian cells. Mol. Cell 21: 533.
Pillai R.S. 2005. MicroRNA function: Multiple mechanisms
for a tiny RNA? RNA 11: 1753.
Pillai R.S., Artus C.G., and Filipowicz W. 2004. Tethering of
human Ago proteins to mRNA mimics the miRNA-medi-
ated repression of protein synthesis. RNA 10: 1518.
Pillai R.S., Bhattacharyya S.N., Artus C.G., Zoller T., Cougot
N., Basyuk E., Bertrand E., and Filipowicz W. 2005.
Inhibition of translational initiation by let-7 miRNA in
human cells. Science 309: 1573.
Schratt G.M., Tuebing F., Nigh E.A., Kane C.G., Sabatini
M.E., Kiebler M., and Greenberg M.E. 2006. A brain-
specific miRNA regulates dendritic spine development.
Nature 439: 283.
Sutton M.A. and Schuman E.M. 2005. Local translational con-
trol in dendrites and its role in long-term synaptic plasticity.
J. Neurobiol. 64: 116.
Tomari Y. and Zamore P.D. 2005. Perspective: Machines for
RNAi. Genes Dev. 19: 517.
Valencia-Sanchez M.A., Liu J., Hannon G.J., and Parker R.
2006 Control of translation and mRNA degradation by
miRNAs and siRNAs. Genes Dev. 20: 515.
Wienholds E. and Plasterk R.H. 2005. MiRNA function in ani-
mal development. FEBS Lett. 579: 5911.
Yaman I., Fernandez J., Sarkar B., Schneider R.J., Snider
M.D., Nagy L.E., and Hatzoglou M. 2002. Nutritional con-
trol of mRNA stability is mediated by a conserved AU-rich
element that binds the cytoplasmic shuttling protein HuR. J.
Biol. Chem. 277: 41539.
REVERSAL OF MIRNA-MEDIATED REPRESSION 521
513-522_Bhattacharyya_Symp71.qxd 2/8/07 1:42 PM Page 521
513-522_Bhattacharyya_Symp71.qxd 2/8/07 1:42 PM Page 522
... Il a été proposé que ces condensats pourraient être le lieu où s'effectue le mécanisme d'interférence par l'ARN médié par le complexe RISC, les microARNs et les répresseurs transcriptionnels ou traductionnels (Decker and Parker, 2012). D'autres études s'accordent à montrer que ces condensats pourraient, à l'inverse, jouer un rôle uniquement de stockage provisoire des ARNm non traduits en vue de leur traduction ultérieure (Brengues et al., 2005;Bhattacharyya et al., 2006;Ayache et al., 2015). Une étude récemment publiée par notre groupe a montré que la transfection de cellules de glioblastome (U87MG) par des microARNs synthétiques à forte concentration conduit à une accumulation significative de ces microARNs dans les P-bodies et les GW-bodies (Simion et al., 2020). ...
Thesis
Les microARNs jouent un rôle essentiel dans la morphogénèse et l’homéostasie cutanée. Cette classe d’ARN non codant contrôle plusieurs voies de signalisation en régulant l’expression de réseaux entiers de gènes cibles. Ils sont donc considérés comme des cibles biologiques de choix pour les stratégies de criblage de composés bioactifs. Au laboratoire, nous avons conçu une sonde d’imagerie bioluminescente inductible par les microARNs, dénommée RILES pour « RNAi-Inducible Luciferase Expression System » qui se prête particulièrement bien au criblage cellulaire de librairies de composés synthétiques. Au cours de ce doctorat, nous avons placé le système RILES sous contrôle de l’axe de régulation TGF-β1/microARN-21 choisi pour son rôle central dans la ré-épithélialisation cutanée. Le criblage d’une extractothèque de 37 extraits bruts de plantes nous a permis d’identifier trois extraits bruts de plantes, dont celui du Chardon Marie (Silybum marianum (L.) Gaertn) qui a fait l’objet d’études mécanistiques et fonctionnelles poussées.Nous avons montré que l’effet de l’extrait de Chardon-Marie sur le microARN-21 est dépendant d'un complexe contenant six flavonolignanes, appelé silymarine (SM). Des études d’immunoprécipitation de la protéine Argonaute 2 couplée à laRT-qPCR ont permis de révéler un mécanisme de régulation original du microARN-21 par cet extrait. Le séquençage àhaut débit du transcriptome (RNA-seq) des kératinocytes en réponse au traitement par le TGF-β1 et la SM a permis demettre en évidence trois signatures d’expression génique majeures associées à la différenciation kératinocytaire, aucycle cellulaire et de façon inattendue au métabolisme des lipides. Nous avons montré que la SM bloque le cycle cellulaire en phase G0/G1, inhibe la différenciation des kératinocytes via l’inhibition de l’expression de Notch3 et active la synthèse des lipides en inhibant la phosphorylation d’AMPK et en augmentant l’activité transcriptionnelle de PPARγ. Parailleurs, la SM ralentit la migration cellulaire en perturbant la transition épithélio-mésenchymateuse et inhibe les réponses inflammatoires en bloquant l’activité transcriptionnelle de NF-ƙB. Du fait de ces propriétés biologiques, pour certaines nouvelles, nous avons évalué l’effet thérapeutique de la SM contre le développement du psoriasis en plaques induit par l’imiquimod chez la souris. Nos résultats indiquent que la SM pourrait représenter une alternative prometteuse « naturelle » aux traitements pharmacologiques actuels pour la prise en charge de cette pathologie.
... For a long time, P-bodies were thought to mediate specific forms of mRNA decay, but the current picture is that they are more relevant for transient mRNA storage of translationally repressed mRNA during different forms of stress [60][61][62]. Later on, when the stress has ceased and cells resume function, specific mRNAs contained in P-bodies can be released to become degraded in the cytosol or to enter translation [63][64][65]. ...
Article
Full-text available
Simple Summary Gene expression regulated by the NF-κB transcription factor pathway is a multi-step process that finally results in the synthesis of proteins, many of which are eventually secreted. The NF-κB system is activated by cytosolic induction pathways (level 1) and then further modulated by cofactors and epigenetic mechanisms in the nucleus (level 2). This review discusses posttranscriptional events as the third level of NF-κB control with a focus on NF-κB-mediated regulation of mRNA metabolism and protein secretion. Abstract The NF-κB system is a key transcriptional pathway that regulates innate and adaptive immunity because it triggers the activation and differentiation processes of lymphocytes and myeloid cells during immune responses. In most instances, binding to cytoplasmic inhibitory IκB proteins sequesters NF-κB into an inactive state, while a plethora of external triggers activate three complex signaling cascades that mediate the release and nuclear translocation of the NF-κB DNA-binding subunits. In addition to these cytosolic steps (level 1 of NF-κB regulation), NF-κB activity is also controlled in the nucleus by signaling events, cofactors and the chromatin environment to precisely determine chromatin recruitment and the specificity and timing of target gene transcription (level 2 of NF-κB regulation). Here, we discuss an additional layer of the NF-κB system that manifests in various steps of post-transcriptional gene expression and protein secretion. This less-studied regulatory level allows reduction of (transcriptional) noise and signal integration and endows time-shifted control of the secretion of inflammatory mediators. Detailed knowledge of these steps is important, as dysregulated post-transcriptional NF-κB signaling circuits are likely to foster chronic inflammation and contribute to the formation and maintenance of a tumor-promoting microenvironment.
... The putative promoter region of the miR-183 cluster contains several binding sites for transcription factors known to regulate the circadian rhythm in the eye, including RORα (Xu et al., 2007), but there is currently no experimental data to confirm this transcriptional regulation. Genetic variants with abnormal processing of pre-miR-182 have been described (Saus et al., 2010) and neuronal miRNAs have been shown to have very quick turn-over ratios (Krol et al., 2010) compared to nonneuronal cells (Bhattacharyya et al., 2006;Hwang et al., 2007;Krol et al., 2010). Thus, the regulation of miRNA processing and/or degradation could also play important roles in its oscillatory behavior. ...
Article
Full-text available
A broad repertoire of transcription factors and other genes display oscillatory patterns of expression, typically ranging from 30 min to 24 h. These oscillations are associated with a variety of biological processes, including the circadian cycle, somite segmentation, cell cycle, and metabolism. These rhythmic behaviors are often prompted by transcriptional feedback loops in which transcriptional activities are inhibited by their corresponding gene target products. Oscillatory transcriptional patterns have been proposed as a mechanism to drive biological clocks, the molecular machinery that transforms temporal information into accurate spatial patterning during development. Notably, several microRNAs (miRNAs) -small non-coding RNA molecules-have been recently shown to both exhibit rhythmic expression patterns and regulate oscillatory activities. Here, we discuss some of these new findings in the context of the developing retina. We propose that miRNA oscillations are a powerful mechanism to coordinate signaling pathways and gene expression, and that addressing the dynamic interplay between miRNA expression and their target genes could be key for a more complete understanding of many developmental processes.
... They contain various components, including ribonucleoproteins (RNPs) and miRNA, which are both engaged in the translation suppression and mRNA decay machinery regulation. It has been reported that mRNA binding to the RNP or miRNA determines its destiny; that is, whether transcripts are going to be translated, stored, or degraded [16]. ...
Article
Full-text available
In recent years, processing bodies (P-bodies) formed by liquid–liquid phase separation, have attracted growing scientific attention due to their involvement in numerous cellular activities, including the regulation of mRNAs decay or storage. These cytoplasmic dynamic membraneless granules contain mRNA storage and decay components such as deadenylase and decapping factors. In addition, different mRNA metabolic regulators, including m⁶A readers and gene-mediated miRNA-silencing, are also associated with such P-bodies. Cancerous cells may profit from these mRNA decay shredders by up-regulating the expression level of oncogenes and down-regulating tumor suppressor genes. The main challenges of cancer treatment are drug resistance, metastasis, and cancer relapse likely associated with cancer stem cells, heterogeneity, and plasticity features of different tumors. The mRNA metabolic regulators based on P-bodies play a great role in cancer development and progression. The dysregulation of P-bodies mediators affects mRNA metabolism. However, less is known about the relationship between P-bodies mediators and cancerous behavior. The current review summarizes the recent studies on P-bodies mediators, their contribution to tumor development, and their potential in the clinical setting, particularly highlighting the P-bodies as potential drug-carriers such as exosomes to anticancer in the future.
... 249 These structures, made of entwined and coiled fine filaments, are mixed with adherent granules smaller than ribosome granules but measured up their subunit size. [250][251][252][253][254][255][256] The filamentous network that appeared outwardly of these structures were very thin macromolecules (less than 5 nm in diam.) and could be of cytoskeletal actin and/or of associated depolymerized proteinaceous structure(s) linked with the ongoing dispatching or trafficking of ribonucleoprotein transcripts [255][256][257] as they associated with the nuclear creases accumulated while some proteome repression occurred 257 as in the case of the S100 protein in PNS? 258 Alternatively, if one considers that a functional nucleus undergoes oscillating waves, huge amounts of transcripts disseminated in the surrounded restoration neuroplasm to be proteome/metabolome translations and, associated with transport cytoskeleton 'sudden' loads of ribonucleoproteins [258][259][260][261][262][263] with the nucleus topology changed (indents as spheroid), immersed in a viscous liquid flow, could act as sort of swinging watermill surrounded by those cytoskeletal macromolecules, mainly actin. 170,171,258 This oscillating, turbulent flow in the neuroplasm could induce one eddy at the opposite side of the axon hillock along with the less mobile, concentric organelles. ...
Article
Full-text available
The development of a murine model of osmotic demyelinating syndrome (ODS) allowed to study changes incurred in extrapontine zones of the CNS and featured neuron and glial cell changes in the relay thalamic ventral posterolateral (VPL) and ventral posteromedial (VPM) nuclei before, during and after ODS induction, and characterized without immune response. There, the neuron Wallerian-type deteriorations were verified with fine structure modifications of the neuron cell body, including some nucleus topology and its nucleolus changes. Morphologic analyses showed a transient stoppage of transcriptional activities while myelinated axons in the surrounding neuropil incurred diverse damages, previously reported. Even though the regional thalamus myelin deterioration was clearly recognized with light microscopy 48h after osmotic recovery of ODS, ultrastructure analyses demonstrated that, at that time, the same damaged parenchyma regions contained nerve cell bodies that have already reactivated nucleus transcriptions and neuroplasm translations because peculiar accumulations of fibro-granular materials, similar to those detected in restored ODS astrocytes, were revealed in these restructuring nerve cell bodies. Their aspects suggested to be accumulations of ribonucleoproteins. The findings suggested that progressive neural function’s recovery in the murine model could imitate some aspects of human ODS recovery cases.
... Ces dernières révélations suggèrent que les ARNm présents au niveau des GS sont inhibés d'un point de vue traductionnel mais non sujet à une dégradation immédiate, contrairement aux ARNm des P-bodies (Cougot et al., 2004; Franks and Lykke-Andersen, 2007; Parker, 2003, 2006). Ainsi, certains ARNm confinés dans les GS mais aussi dans les P-bodies peuvent sous certaines conditions se ré engager dans la traduction après leur sortie(Bhattacharyya et al., 2006;Brengues et al., 2005; Cougot et 121 | P a g e al., 2004;Kedersha et al., 2000). Initialement décrits comme de larges agrégats cytoplasmiques d'ARN messagers devenant visibles microscopiquement lorsque la synthèse protéique est inhibée en réponse à différents stress, cette définition a été mise à jour lorsque les GS ont été présentées comme contenant des complexes de préinitiation de la traduction et des protéines ...
Thesis
La Néphronophtise (NPH) est une néphropathie tubulo-interstitielle autosomique récessive, caractérisée par la présence d'une fibrose interstitielle massive et la formation de kystes. Elle est la cause majeure d'insuffisance rénale terminale chez l'enfant ou le jeune adulte. Elle peut être isolée ou syndromique, associée à des atteintes extrarénales. La NPH appartient aux ciliopathies, un ensemble de maladies multisystémiques causées par des mutations dans des gènes codant pour des protéines du cil primaire. Le cil primaire agit, à la surface de la plupart des cellules, comme un mécano et chimiosenseur contrôlant des voies de signalisation essentielles au développement et à l'homéostasie tissulaire (Shh, Wnt, signalisation PC2/Ca2). Vingt-trois gènes responsables de la NPH ont été identifiés (dont 14 dans notre laboratoire). Ils codent pour des protéines assurant des fonctions ciliaires telles que le contrôle de la composition protéique et de la signalisation ciliaire, à la base du cil (zone de transition (TZ)), au niveau du compartiment Inversine (CI), ou lors du transport intraflagellaire. Nous avons identifié une mutation faux-sens homozygote dans un nouveau gène, responsable d'une NPH associée à une fibrose hépatique. ANKS3 code une protéine connue pour interagir avec plusieurs protéines NPHP de la TZ (NPHP1), du CI (ANKS6, NEK8) ainsi qu'avec BICC1, une ribonucléoprotéine mutée dans les dysplasies rénales kystiques. Mon projet de thèse a porté sur la caractérisation de la fonction d'ANKS3 et de l'impact de la mutation humaine dans les processus cellulaires altérés dans les ciliopathies rénales. Ces études ont été réalisées à partir de différents modèles cellulaires: fibroblastes de patients, cellules tubulaires rénales IMCD3 déplétés pour Anks3 (ANKS3_KD) ré-exprimant la forme sauvage ou mutée. Mes travaux ont ainsi montré que la mutation ANSK3 diminue son interaction avec les composants de la TZ, NPHP1 et du CI, NEK8. Par ailleurs, l'absence ou l'expression de la forme mutée de ANKS3 perturbe la taille des cils et la biogenèse du CI. En effet, les composants du CI sont anormalement relocalisés le long du cil. Par ailleurs, une perturbation de la signalisation ciliaire de la voie Shh et de la localisation de la protéine PC2 au cil ont été observée dans les cellules ANKS3_KD ou mutantes. Cette dernière observation est confirmée dans le modèle du poisson zèbre muté pour anks3 (TALEN), qui présente une diminution de la motilité ciliaire associée à des altérations de la voie calcique dans la vésicule de Kupffer, conduisant à des défauts de latéralité (situs inversus). Outre ces défauts ciliaires, la perte ou la mutation de ANKS3 entrainent des défauts de polarité apico-basale dans les cellules IMCD, avec une diminution de la hauteur des cellules et un retard de la formation des jonctions serrées, un phénotype déjà observé dans les modèles mutés pour NPHP1. De façon concordante avec ces résultats nous avons observé une diminution de la stabilité des transcrits Nphp1 dans les cellules ANKS3_KD et mutantes. La réexpression de NPHP1 dans ces cellules restaure les défauts de polarité et de longueur des cils démontrant l'implication de la régulation des transcrits Nphp1 par ANKS3 dans ces phénotypes. Afin de préciser le mécanisme par lequel ANKS3 régule la stabilité des ARNs de Nphp1 et possiblement d'autres ARN ciliaires, nous avons étudié l'implication de son partenaire BICC1, connu pour lier et réguler les ARNs. Des analyses transcriptomiques par RNAseq et RIPseq sur des cellules ANKS3-KD en présence ou non de BICC1, ont permis de démontrer le rôle majeur d'ANKS3 au sein du complexe ANKS3/BICC1 dans la régulation de la dégradation par RISC/AGO2 de nombreux transcrits ciliaires et gènes de ciliopathies. L'ensemble de ces résultats permet de mettre en évidence un nouveau mécanisme de régulation des transcrits ciliaires par le complexe ANKS3/BICC1, dont les mutations causent des ciliopathies rénales.
Article
Full-text available
Life represents a very complex phenomenon based on the organization and functions of cells, the elementary units of any living structure. The proper functioning of every cell in an organism assures its health. In addition, proper functioning also means the great ability of cells to adapt to changes in the environment and survive to the advantage of the organism they belong to. Sometimes, the functions of some cells can be so affected by certain stressors that they can deviate from a normal to a pathological state. Proper functioning of the cell is determined by its identity, which if changed can often induce pathology. This is the underlying reason for our interest in cell identity determination, increasingly critical for our understanding of cell biology and medicine. In this paper, we aim to analyze the concept of identity in general terms, starting with its approaches in philosophy and switching toward its practical, biological application in cell identity. We will advocate the need to balance various views on the concept of identity in order to find solutions in approaching cell identity
Article
Stress granules (SGs), hallmarks of the cellular adaptation to stress, promote survival, conserve cellular energy, and are fully dissolved upon the cessation of stress treatment. Different stresses can initiate the assembly of SGs, but arsenite and heat are the best studied of these stresses. The composition of SGs and posttranslational modifications of SG proteins differ depending on the type and severity of the stress insult, methodology used, cell line, and presence of overexpressed and tagged proteins. A group of 18 proteins showing differential localization to SGs in heat‐ and arsenite‐stressed mammalian cell lines is described. Upon severe and prolonged stress, physiological SGs transform into more solid protein aggregates that are no longer reversible and do not contain mRNA. Similar pathological inclusions are hallmarks of neurodegenerative diseases. SGs induced by heat stress are less dynamic than SGs induced by arsenite and contain a set of unique proteins and linkage‐specific polyubiquitinated proteins. The same types of ubiquitin linkages have been found to contribute to the development of neurodegenerative disorders such as Parkinson disease, Alzheimer disease, and amyotrophic lateral sclerosis (ALS). We propose heat stress‐induced SGs as a possible model of an intermediate stage along the transition from dynamic, fully reversible arsenite stress‐induced SGs toward aberrant SGs, the hallmark of neurodegenerative diseases. Stress‐ and methodology‐specific differences in the compositions of SGs and the transition of SGs to aberrant protein aggregates are discussed. This article is categorized under: RNA in Disease and Development > RNA in Disease RNA Interactions with Proteins and Other Molecules > RNA–Protein Complexes RNA Export and Localization > RNA Localization
Article
Full-text available
lin-4 encodes a small RNA that is complementary to sequences in the 3' untranslated region (UTR) of lin-14 mRNA and that acts to developmentally repress the accumulation of LIN-14 protein. This repression is essential for the proper timing of numerous events of Caenorhabditis elegans larval development. We have investigated the mechanism of lin-4 RNA action by examining the fate of lin-14 mRNA in vivo during the time that lin-4 RNA is expressed. Our results indicate that the rate of synthesis of lin-14 mRNA, its state of polyadenylation, its abundance in the cytoplasmic fraction, and its polysomal sedimentation profile do not change in response to the accumulation of lin-4 RNA. Our results indicate that association of lin-4 RNA with the 3' UTR of lin-14 mRNA permits normal biogenesis of lin-14 mRNA, and normal translational initiation, but inhibits step(s) thereafter, such as translational elongation and/or the release of stable LIN-14 protein.
Article
Full-text available
AU-rich elements (AREs) present in the 3' untranslated regions of many protooncogene, cytokine, and lymphokine messages target them for rapid degradation. HuR, a ubiquitously expressed member of the ELAV (embryonic lethal abnormal vision) family of RNA binding proteins, selectively binds AREs and stabilizes ARE-containing mRNAs in transiently transfected cells. Here, we identify four mammalian proteins that bind regions of HuR known to be essential for its ability to shuttle between the nucleus and the cytoplasm and to stabilize mRNA: SETalpha, SETbeta, pp32, and acidic protein rich in leucine (APRIL). Three have been reported to be protein phosphatase 2A inhibitors. All four ligands contain long, acidic COOH-terminal tails, while pp32 and APRIL share a second motif: rev-like leucine-rich repeats in their NH(2)-terminal regions. We show that pp32 and APRIL are nucleocytoplasmic shuttling proteins that interact with the nuclear export factor CRM1 (chromosomal region maintenance protein 1). The inhibition of CRM1 by leptomycin B leads to the nuclear retention of pp32 and APRIL, their increased association with HuR, and an increase in HuR's association with nuclear poly(A)+ RNA. Furthermore, transcripts from the ARE-containing c-fos gene are selectively retained in the nucleus, while the cytoplasmic distribution of total poly(A)+ RNA is not altered. These data provide evidence that interaction of its ligands with HuR modulate HuR's ability to bind its target mRNAs in vivo and suggest that CRM1 is instrumental in the export of at least some cellular mRNAs under certain conditions. We discuss the possible role of these ligands upstream of HuR in pathways that govern the stability of ARE-containing mRNAs.
Article
Full-text available
Transcription factors have traditionally been viewed as the main determinants of gene expression. Yet, in recent years it has become apparent that RNA-binding proteins also play a critical role in determining the levels of expression of a large number of genes. Once mRNAs are transcribed, RNA-binding proteins can control all subsequent steps in their function, from alternative splicing and translation to mRNA transport and stability. In the nervous system, a large number of genes are regulated post-transcriptionally via the interaction of their mRNAs with specific RNA-binding proteins. This type of regulation is particularly important in the control of the temporal and spatial pattern of gene expression during neural development. This review will discuss the function of the embryonic lethal abnormal vision (ELAV)/Hu family of nervous system-specific RNA-binding proteins, with a special emphasis on HuD, a member of this family that controls GAP-43 mRNA stability and expression. In addition, we will present recent findings on other neural RNA-binding proteins: the ribonucleoprotein K homology (KH)-domain proteins, Fragile X mental retardation protein (FMRP), quakinguiable protein (QKI), and Nova-1. Together with the ELAV/Hu family, these proteins are essential for proper neural development and in some cases for plasticity in the mature brain. The biological significance of these proteins is evident not only by their evolutionary conservation but also by the magnitude of problems arising from autoimmune reactions against them or from mutations affecting their expression or function.
Article
Full-text available
The cationic amino acid transporter, Cat-1, is a high affinity transporter of the essential amino acids, arginine and lysine. Expression of the cat-1 gene increases during nutritional stress as part of the adaptive response to starvation. Amino acid limitation induces coordinate increases in stability and translation of the cat-1 mRNA, at a time when global protein synthesis decreases. It is shown here that increased cat-1 mRNA stability requires an 11 nucleotide AU-rich element within the distal 217 bases of the 3'-untranslated region. When this 217-nucleotide nutrient sensor AU-rich element (NS-ARE) is present in a chimeric mRNA it confers mRNA stabilization during amino acid starvation. HuR is a member of the ELAV family of RNA-binding proteins that has been implicated in regulating the stability of ARE-containing mRNAs. We show here that the cytoplasmic concentration of HuR increases during amino acid starvation, at a time when total cellular HuR levels decrease. In addition, RNA gel shift experiments in vitro demonstrated that HuR binds to the NS-ARE and binding was dependent on the 11 residue AU-rich element. Moreover, HuR binding to the NS-ARE in extracts from amino acid-starved cells increased in parallel with the accumulation of cytoplasmic HuR. It is proposed that an adaptive response of cells to nutritional stress results in increased mRNA stability mediated by HuR binding to the NS-ARE.
Article
An important mechanism of posttranscriptional gene regulation in mammalian cells is the rapid degradation of messenger RNAs (mRNAs) signaled by AU-rich elements (AREs) in their 3' untranslated regions. HuR, a ubiquitously expressed member of the Hu family of RNA-binding proteins related to Drosophila ELAV, selectively binds AREs and stabilizes ARE-containing mRNAs when overexpressed in cultured cells. This review discusses mRNA decay as a general form of gene regulation, decay signaled by AREs, and the role of HuR and its Hu-family relatives in antagonizing this mRNA degradation pathway. The influence of newly identified protein ligands to HuR on HuR function in both normal and stressed cells may explain how ARE-mediated mRNA decay is regulated in response to environmental change.
Article
The transport of messenger RNAs (mRNAs) from the nucleus to the cytoplasm involves adapter proteins that bind the mRNA as well as receptor proteins that interact with the nuclear pore complex. We demonstrate the utility of cell-permeable peptides designed to interfere with interactions between potential adapter and receptor proteins to define the pathways accessed by particular mRNAs. We show that HuR, a protein implicated in the stabilization of short-lived mRNAs containing AU-rich elements (AREs), serves as an adapter for c-fos mRNA export through two pathways. One involves the HuR shuttling domain, HNS, which exhibits a heat shock-sensitive interaction with transportin 2 (Trn2); the other involves two protein ligands of HuR-pp32 and APRIL-which contain leucine-rich nuclear export signals (NES) recognized by the export receptor CRM1. Heterokaryon and in situ hybridization experiments reveal that the peptides selectively block the nucleocytoplasmic shuttling of their respective adapter proteins without perturbing the overall cellular distribution of polyadenylated mRNAs.
Article
The RNA-binding protein HuR stabilizes labile mRNAs carrying AU-rich instability elements. This mRNA stabilization can be induced by hypoxia, lipopolysaccharide, and UV light. The mechanism by which these stimuli activate HuR is unclear and might be related to post-translational modification of this protein. Here we show that HuR can be methylated on arginine. However, HuR is not a substrate for PRMT1, the most prominent protein-arginine methyltransferase in mammalian cells, which methylates a number of heterogeneous nuclear ribonucleoproteins. Instead, HuR is specifically methylated by coactivator-associated arginine methyltransferase 1 (CARM1), a protein-arginine methyltransferase previously shown to serve as a transcriptional coactivator. By analyzing methylation of specific HuR arginine-to-lysine mutants and by sequencing radioactively methylated HuR peptides, Arg(217) was identified as the major HuR methylation site. Arg(217) is located in the hinge region between the second and third of the three HuR RNA recognition motif domains. Antibodies against a methylated HuR peptide were used to demonstrate in vivo methylation of HuR. HuR methylation increased in cells that overexpressed CARM1. Importantly, lipopolysaccharide stimulation of macrophages, which leads to HuR-mediated stabilization of tumor necrosis factor alpha mRNA in these cells, caused increased methylation of endogenous HuR. Thus, CARM1, which plays a role in transcriptional activation through histone H3 methylation, may also play a role in post-transcriptional gene regulation by methylating HuR.
Article
MicroRNAs (miRNAs) are endogenous approximately 22 nt RNAs that can play important regulatory roles in animals and plants by targeting mRNAs for cleavage or translational repression. Although they escaped notice until relatively recently, miRNAs comprise one of the more abundant classes of gene regulatory molecules in multicellular organisms and likely influence the output of many protein-coding genes.