ArticlePDF Available

The mRNA-destabilizing protein Tristetraprolin targets “meiosis arrester” Nppc mRNA in mammalian preovulatory follicles

Authors:

Abstract and Figures

Significance The fertility of female mammals depends upon the well-orchestrated progression of oocyte meiosis, the female germ cell division that is essential for haploid oocyte formation. In preovulatory follicles, how LH induces a decrease in CNP and its encoding mRNA Nppc , a prerequisite for oocyte meiotic resumption, remains an outstanding question to understand the mechanism that controls oocyte meiosis. The present study shows that TTP, an mRNA-destabilizing protein, is one of the regulatory components responsible for the LH-induced rapid decrease in Nppc mRNA and oocyte meiotic resumption. This finding not only highlights the importance of posttranscriptional regulation in the follicle periphery to fine-tune oocyte meiosis but also provides an insight into CNP-dependent cGMP homeostasis in other physiological systems.
TTP is up-regulated in response to the LH/hCG surge and targets the Nppc 3′ UTR. (A and B) RT-qPCR detection of the expression dynamics of Nppc (A) and Zfp36 (B) mRNA in preovulatory MGCs in response to hCG administration. (C) Western blotting analysis for TTP in preovulatory MGCs at different time points after hCG administration. The upper and lower bands indicate phosphorylated (inactive) and unphosphorylated (active) forms of TTP, respectively. (D) Immunohistochemical staining of TTP in preovulatory follicles before and after hCG administration. (Right) Higher magnification of boxed regions. (Scale bar, 100 μm.) (E) RIP analysis for the direct binding of TTP to the Nppc 3′ UTR. The RIP sample was subjected to RT-PCR, and the amplified products were visualized using agarose gel electrophoresis. (F) Quantitative detection of the interaction between TTP and Nppc 3′ UTR by RIP-RT-qPCR. (G) Schematic diagram of the luciferase constructs containing full-length Nppc 3′ UTR, Tnf 3′ UTR (targeted control), or Actb 3′ UTR (nontargeted control). Relative luciferase activity of different luciferase vectors, which were cotransfected with a Zfp36 overexpression vector or empty vector in HEK293 cells. (H) Measurement of the stability of the luciferase mRNA that contained the Nppc 3′ UTR. The luciferase vector was cotransfected with the Zfp36 expression vector or empty vectors in HEK293 cells, which were treated with ActD to terminate transcription. Total RNAs were prepared at the indicated time points to detect the remaining luciferase mRNA levels after ActD treatment. (I) Schematic diagram depicting the location of canonical AU-rich elements (AREs, presented as red triangles) in the Nppc 3′ UTR. All data are presented as the mean ± SEM of three independent experiments. *P < 0.05, **P < 0.01, ***P < 0.001. In A, B, and C, * indicates a significant difference compared with data at 0 h post-hCG.
… 
TTP degrades endogenous Nppc mRNA in preovulatory MGCs. (A) Western blotting analysis for TTP, as well as phosphorylated and total EGFR, in preovulatory MGCs in response to administration of hCG and AG1478, a specific EGFR inhibitor. MGCs were isolated from in vitro cultured preovulatory follicles. (B) Expression dynamics of Nppc and Zfp36 in in vitro cultured MGCs in response to EGF administration. * indicates significant difference compared with data at 0 h post-hCG. (C) Effect of Zfp36 knockdown on the EGF-induced decrease in Nppc mRNA in in vitro cultured MGCs. Total RNAs were prepared at the indicated time points after EGF administration to detect Zfp36 (Upper) and Nppc (Lower) mRNA levels using RT-qPCR. (D) Effect of Zfp36 overexpression on the Nppc mRNA levels in in vitro cultured MGCs. Nppc expression was maintained at a high level before overexpressing Zfp36. Total RNAs were prepared at the indicated time points after transfection to detect Zfp36 (Upper) and Nppc (Lower) mRNA levels. (E) Measurement of the stability of Nppc mRNA in MGCs following transfection with the Zfp36 overexpression vector (pCAGGS-Zfp36) or empty vector. Total RNAs were prepared at the indicated time points to detect the remaining Nppc mRNA levels after ActD treatment. (F-H) The detection of Zfp36 knockout efficiency in MGCs at 2 h post-hCG, at which point Zfp36 should be fully activated in response to hCG administration. The expression levels of Zfp36/TTP in MGCs were detected at 0 h and 2 h post-hCG using RT-qPCR (F), immunohistochemistry (G), and Western blotting (H). (G, Lower) Higher magnification of boxed regions. (Scale bar, 100 μm.) (I) Effects of MGC-specific depletion of Zfp36 on Nppc mRNA levels at 0 h and 2 h post-hCG. (J and K) Evaluation of oocyte meiotic resumption scored as GVBD percentage at 6 h (J) and 9 h (K) post-hCG. Representative images show the morphology of oocytes isolated from preovulatory follicles of wild-type (WT) and Zfp36 gc−/− mice (Right), five to eight mice were used at each time point. (Scale bar, 100 μm.) Data show the means ± SEMs of three independent experiments. *P < 0.05, **P < 0.01, ***P < 0.001.
… 
Content may be subject to copyright.
The mRNA-destabilizing protein Tristetraprolin targets
meiosis arresterNppc mRNA in mammalian
preovulatory follicles
Guangyin Xi
a,1
, Lei An
a,1
, Wenjing Wang
a,1
, Jing Hao
a
, Qianying Yang
a
, Lizhu Ma
a
, Jinlun Lu
a
,
Yue Wang
a
, Wenjuan Wang
a
, Wei Zhao
a
, Juan Liu
a
, Mingyao Yang
a
, Xiaodong Wang
a
, Zhenni Zhang
a
,
Chao Zhang
a
, Meiqiang Chu
a
, Yuan Yue
a
, Fusheng Yao
a
, Meijia Zhang
a
, and Jianhui Tian
a,2
a
Key Laboratory of Animal Genetics, Breeding and Reproduction of the Ministry of Agriculture and Rural Affairs, National Engineering Laboratory for
Animal Breeding, College of Animal Science and Technology, China Agricultural University, 100193 Beijing, P. R. China
Edited by R. Michael Roberts, University of Missouri, Columbia, MO, and approved April 13, 2021 (received for review August 31, 2020)
C-natriuretic peptide (CNP) and its receptor guanylyl cyclase, natri-
uretic peptide receptor 2 (NPR2), are key regulators of cyclic gua-
nosine monophosphate (cGMP) homeostasis. The CNP-NPR2-cGMP
signaling cascade plays an important role in the progression of
oocyte meiosis, which is essential for fertility in female mammals.
In preovulatory ovarian follicles, the luteinizing hormone (LH)-in-
duced decrease in CNP and its encoding messenger RNA (mRNA)
natriuretic peptide precursor C (Nppc) are a prerequisite for oocyte
meiotic resumption. However, it has never been determined how
LH decreases CNP/Nppc. In the present study, we identified that
tristetraprolin (TTP), also known as zinc finger protein 36 (ZFP36), a
ubiquitously expressed mRNA-destabilizing protein, is the critical
mechanism that underlies the LH-induced decrease in Nppc mRNA.
Zfp36 mRNA was transiently up-regulated in mural granulosa cells
(MGCs) in response to the LH surge. Loss- and gain-of-function anal-
yses indicated that TTP is required for Nppc mRNA degradation in
preovulatory MGCs by targeting the rare noncanonical AU-rich ele-
ment harbored in the Nppc 3UTR. Moreover, MGC-specific knockout
of Zfp36, as well as lentivirus-mediated knockdown in vivo, impaired
the LH/hCG-induced Nppc mRNA decline and oocyte meiotic re-
sumption. Furthermore, we found that LH/hCG activates Zfp36/
TTP expression through the EGFR-ERK1/2dependent pathway. Our
findings reveal a functional role of TTP-induced mRNA degradation, a
global posttranscriptional regulation mechanism, in orchestrating the
progression of oocyte meiosis. We also provided a mechanism for
understanding CNP-dependent cGMP homeostasis in diverse cellular
processes.
TTP/Zfp36
|
CNP/Nppc
|
mRNA degradation
|
oocyte meiosis
|
mural granulosa cells
Cyclic guanosine monophosphate (cGMP) is a ubiquitous sec-
ond messenger that regulates myriads functions, such as the
cell cycle, differentiation, apoptosis, and metabolism (1). During
recent decades, C-natriuretic peptide (CNP), encoded by natriuretic
peptide precursor C (Nppc), has emerged as a critical regulator of
cGMP homeostasis by activating its transmembrane guanylyl cyclase
receptor, natriuretic peptide receptor 2 (NPR2), which increases the
intracellular cGMP concentration (2, 3). Increasing evidence from
mouse models of genetic depletion or mutation of Nppc or Npr2
and related clinical assays have indicated that the CNP-NPR2-
cGMP signaling cascade is critical for the development and function
of the cardiovascular (4, 5), skeletal (6, 7), nervous (8, 9), and female
reproductive systems (10). In particular, CNP-NPR2-cGMP signal-
ing is the central regulator of the progression of oocyte meiosis (11),
the female germ cell division that is essential for haploid oocyte
formation.
In mammals, oocytes are arrested at the diplotene stage of meiosis
until the surge of luteinizing hormone (LH) restarts meiosis and
initiates the ovulatory process. In preovulatory follicles, LH-induced
CNP-NPR2-cGMP signaling inhibition is the most prominent
hallmark for meiotic resumption. Although LH also activates cGMP
hydrolysis (12) and decreases guanylyl cyclase activity (13, 14) and
NPR2 affinity for CNP in ovarian follicles (15), mouse models that
prevent cGMP hydrolysis or/and NPR2 inactivation can reinitiate
meiosis (12, 16), suggesting that these alterations are not indis-
pensable for LH-induced meiotic resumption. These facts suggest a
notable change upstream of the CNP-NPR2-cGMP signaling cas-
cade; indeed, remarkable decreases in CNP and Nppc messenger
RNA (mRNA) have been reported in mouse (14, 1719), rat (13),
pig (20), and human (17) follicles, and the time course of the Nppc/
CNP decrease almost coincides with the resumption of meiosis
(14). Previous studies have established the critical role of low-level
CNP in decreasing cGMP and restarting meiotic progression. The
incubation of isolated cumulus-oocyte complexes (COCs) with
low-level CNP that recapitulates the follicular concentration after
LH administration resumes meiosis; however, high levels of CNP
maintain meiotic arrest (11, 20, 21). Thus, the LH-induced decrease
in CNP levels is thought to be important for fine-tuning meiotic
resumption (14, 22, 23). However, the mechanism underlying
decreased CNP is unknown and thus represents an unanswered
question in our understanding of the mechanism that controls
Significance
The fertility of female mammals depends upon the well-
orchestrated progression of oocyte meiosis, the female germ
cell division that is essential for haploid oocyte formation. In
preovulatory follicles, how LH induces a decrease in CNP and its
encoding mRNA Nppc, a prerequisite for oocyte meiotic re-
sumption, remains an outstanding question to understand the
mechanism that controls oocyte meiosis. The present study
shows that TTP, an mRNA-destabilizing protein, is one of the
regulatory components responsible for the LH-induced rapid
decrease in Nppc mRNA and oocyte meiotic resumption. This
finding not only highlights the importance of posttranscrip-
tional regulation in the follicle periphery to fine-tune oocyte
meiosis but also provides an insight into CNP-dependent cGMP
homeostasis in other physiological systems.
Author contributions: G.X., L.A., M.Z., and J.T. designed research; G.X., Wenjing Wang,
J.H., Q.Y., L.M., J. Lu, Y.W., Wenjuan Wang, W.Z., J. Liu, M.Y., X.W., Z.Z., C.Z., M.C., Y.Y.,
and F.Y. performed research; G.X., L.A., and J.T. analyzed data; G.X., L.A., and J.T. wrote
the paper; and Wenjing Wang, X.W., Z.Z., and C.Z. were responsible for animal care and
management.
The authors declare no competing interest.
This article is a PNAS Direct Submission.
Published under the PNAS license.
1
G.X., L.A., and Wenjing Wang contributed equally to this work.
2
To whom correspondence may be addressed. Email: tianjh@cau.edu.cn.
This article contains supporting information online at https://www.pnas.org/lookup/suppl/
doi:10.1073/pnas.2018345118/-/DCSupplemental.
Published May 24, 2021.
PNAS 2021 Vol. 118 No. 22 e2018345118 https://doi.org/10.1073/pnas.2018345118
|
1of8
DEVELOPMENTAL
BIOLOGY
Downloaded by qianying yang on May 24, 2021
oocyte meiosis (22). Given that the turnover of CNP is very rapid
(24), a decrease in Nppc mRNA could rapidly decrease the amount
of CNP (14). In addition, the LH-induced decrease in CNP is in-
dependent of the natriuretic peptide clearance receptor NPR3
(18). Therefore, we focused on the possible mechanism underlying
the LH-induced decrease in Nppc mRNA.
By reanalyzing a previously published transcriptome that pre-
sented rapid effects of LH on gene expression in the mural gran-
ulosa cells (MGCs) of mouse preovulatory follicles (25), we noticed
that zinc finger protein 36 homolog (Zfp36) that encodes triste-
traprolin (TTP) showed an expression pattern that was inversely
related to the LH-induced decrease in Nppc mRNA. TTP is an
RNA-binding protein that regulates mRNA stability by binding
to AU-rich elements (AREs) in the 3untranslated regions (UTRs)
of target mRNAs and destabilizes mRNA via poly(A) tail removal or
deadenylation (26, 27). Furthermore, we noticed that Nppc mRNA
is a representative ARE-containing transcript. Based on these find-
ings, we hypothesized that TTP might target and degrade Nppc
mRNA and thus could be the key factor underlying the LH-induced
decrease in Nppc mRNA.
In the present study, we identified that TTP-induced Nppc mRNA
degradation is a regulatory component of meiotic progression in
preovulatory follicles. Notably, we reported that a rare noncanonical
ARE motif mediated the TTP-induced destabilization of Nppc
mRNA. Additionally, we found that LH transiently activated Zfp36
via the epidermal growth factor receptor (EGFR)-extracellular
regulated kinase (ERK)1/2-dependent pathway. Thus, focusing on
the upstream part of the signaling cascade, the present study de-
termined a mechanism by which CNP-NPR2-cGMP signaling is
inactivated during meiotic resumption and ovulation processes and
provides insight into CNP-dependent cGMP homeostasis in other
physiological systems.
Results
Zfp36/TTP Is Transiently Up-Regulated in Response to the LH/hCG
Surge and Targets the 3UTR of the Nppc mRNA. To explore the mech-
anism underlying the LH-induced decrease in Nppc mRNA, we tested
the previous notion that theLH-induceddecreaseinNppc mRNA is a
universal switch for CNP-NPR2-cGMP signaling inhibition. Female
Institute of Cancer Research (ICR) mice were administered with
human chorionic gonadotropin (hCG) to mimic the endogenous
LH surge (14, 1719), and then we detected the Nppc mRNA levels
in MGCs and ovaries at various time points. We found that the Nppc
mRNA levels decreased immediately in MGCs and ovaries to less
than 50% by 2 h post-hCG and further declined to 10% by 5 h post-
hCG (Fig. 1Aand SI Appendix,Fig.S1A). Our findings, together
with previous results obtained from mice with a different genetic
background (1719) and from pigs (20), indicated that the rapid
decrease in the Nppc mRNA levels is a hallmark event common to
different genotypes and species.
Next, to identify the potential factor controlling Nppc expression
levels, we reanalyzed previously published transcriptome data that
identified early response genes in MGCs at 1 h post-hCG. We
noticed that the expression of the Zfp36 gene, whose product, TTP,
is responsible for degrading mRNAs that have AU-rich 3UTRs,
was up-regulated by 3.3-fold within 1 h after LH/hCG adminis-
tration (25). Given that the up-regulation of Zfp36 coincided with
thedecreaseinNppc levels and that the Nppc 3UTR is consid-
erably AU-rich, we hypothesized that Zfp36/TTP might be impli-
cated in the decrease in Nppc mRNA. To this end, we isolated
MGCs from preovulatory follicles at various times after injection
with hCG and detected the expression dynamics of Zfp36 using
real-time reverse transcription quantitative PCR (RT-qPCR). In
agreement with the results of previous transcriptome data (25),
we found a transient and significant increase in Zfp36 mRNA, by
approximately sevenfold and tenfold at 1 and 2 h post-hCG,
respectively, in MGCs and ovaries, followed by a rapid decline to
the basal level (Fig. 1Band SI Appendix, Fig. S1B). In addition,
the transient activation of Zfp36 mRNA in MGCs was accom-
panied by a transiently increased abundance of TTP at 2 to 4 h in
response to hCG injection (Fig. 1C). Collectively, these results
suggested that Zfp36/TTP is activated in response to hCG during
a specific time window in preovulatory follicles. Moreover, by
analyzing the previously published transcriptome data of ovarian
MGCs pre- and post-LH/hCG administration from different
species (2831), we found that activation of Zfp36/TTP in pre-
ovulatory MGCs in response to the LH/hCG surge appears to be
common in these species (SI Appendix, Fig. S2).
Next, immunohistochemical detection indicated that the LH/
hCG-induced TTP is mainly localized in MGCs and cumulus
cells (Fig. 1D), which overlaps with the distribution of Nppc mRNA
in the preovulatory follicle (11). The spatiotemporally associated
patterns, together with the negatively correlated expression dynamics
between Nppc and Zfp36, suggested that Zfp36-encoded TTP might
be an inducible repressor of Nppc mRNA in response to the LH/
hCG surge. Therefore, we determined whether TTP could target the
3UTR of Nppc mRNA. Given that the high level of TTP protein
appeared by 2 and 3 h post-hCG and Nppc mRNA was significantly
decreased at 2 h post-hCG, we isolated preovulatory MGCs at 2 and
3 h post-hCG and used RNA immunoprecipitation (RIP) to test
the binding of TTP to the Nppc mRNA. In a TTP-specific pulldown,
Nppc mRNA, but not nontargeted beta actin (Actb) mRNA, was
significantly enriched compared with the Immunoglobulin G (IgG)
controls (Fig. 1 Eand Fand SI Appendix,Fig.S3A). The specificity of
the interaction was confirmed by sequencing the RIP-qPCR amplicon.
Having confirmed the binding of TTP to the Nppc 3UTR, we
next examined whether the Nppc 3UTR can mediate the TTP-
induced mRNA destabilization. We generated luciferase constructs
containing the full-length Nppc 3UTR, the tumor necrosis factor
(Tnf)3UTR, or the Actb 3UTR. The Tnf 3UTR was used as the
targeted control because Tnf mRNA is a well-accepted target of
TTP (32), whereas the Actb 3UTR was used as the nontargeted
control (Fig. 1G). Humanembryonic kidney (HEK293) cells were
cotransfected with the psiCHECK2 luciferase vector containing
Nppc 3UTR or the control 3UTRs, together with a Zfp36 over-
expression vector or empty vector. Compared with cells transfected
with the empty vector, overexpression of Zfp36 significantly reduced
the luciferase activity of the reporter containing the full-length Nppc
3UTR to the level comparable to that of the reporter containing
full-length Tnf 3UTR but did not alter the luciferase activity of the
reporter containing full-length Actb 3UTR (Fig. 1G). To further
determine the role of Nppc 3UTR in mediating TTP-induced de-
stabilization of target mRNA, we measured the luciferase mRNA
stability after blocking transcription using actinomycin D (ActD),
which is frequently used to detect mRNA stability by inhibiting
de novo mRNA synthesis, thus facilitating the measurement of the
remaining mRNA (3335). The results showed that TTP over-
expression significantly accelerated the destabilization of luciferase
mRNA containing the full-length Nppc 3UTR to approximately a
quarter of that of the control vector after 3 h (Fig. 1H), which is
comparable to the rate of decline of Nppc mRNA in MGCs fol-
lowing the LH/hCG surge. The reliability of the mRNA stability
assay was supported by the fact that TTP overexpression significantly
degraded luciferase mRNA containing the Tnf 3UTR but not that
containing the Actb 3UTR (SI Appendix,Fig.S3Band C). These
results indicated that TTP can destabilize the Nppc mRNA via its
3UTR. Similar results were also obtained using the bovine NPPC
3UTR (SI Appendix,Fig.S3Dand E). Collectively, these experi-
ments demonstrated that Zfp36/TTP can be activated in preovulatory
follicles in response to the LH/hCG surge and then targets the Nppc
3UTR, which might contribute to hCG-induced destabilization of
Nppc mRNA.
TTP Targets the Noncanonical ARE Motif in the Nppc 3UTR. Lucif-
erase assays demonstrated that the full-length Nppc 3UTR
contains functional repressive motifs that respond to TTP. AREs
2of8
|
PNAS Xi et al.
https://doi.org/10.1073/pnas.2018345118 The mRNA-destabilizing protein Tristetraprolin targets meiosis arresterNppc mRNA in
mammalian preovulatory follicles
Downloaded by qianying yang on May 24, 2021
are essential for recruiting TTP to the 3UTR of target mRNA;
therefore, we attempted to determine the potent ARE(s) that
direct TTP-induced Nppc mRNA degradation. As expected, we
found four scattered AUUUA motifs, the canonical ARE sequence,
locatedintheNppc 3UTR (Fig. 1Iand SI Appendix,Fig.S4A). We
then constructed a range of luciferase reporters that contained dif-
ferent mutant ARE motifs (Fig. 2A); however, none of the detected
canonical ARE motifs were essential for mediating TTP-induced
repression of any reporters (Fig. 2B). To exclude possible synergis-
tic or additive effects of these canonical ARE motifs, we further
constructed a reporter containing the 3UTR in which all putative
ARE motifs were mutated (Fig. 2A). Unexpectedly, in line with the
results of each single mutation, the tetrad mutation also led to a
significant decrease in luciferase activity (Fig. 2B), indicating that the
reporter still retains significant AU richness in the 3UTR, and
implying that other unidentified motif(s) might underlie the TTP-
induced degradation of Nppc mRNA. This unexpected observation
was reminiscent of the presence of noncanonical, but AU-rich
3UTR motifs, also known as non-AUUUA AREs. Previous stud-
ies have shown that canonical ARE motifs are dispensable for rapid
mRNA degradation; however, noncanonical ARE motifs can func-
tionally mediate the rapid destabilization of target mRNA (36, 37).
Thus, we screened the Nppc 3UTR and characterized multiple non-
AUUUA but AU-rich motifs. We hypothesized that the Nppc
3UTR contained repressive noncanonical AREs that could medi-
ate TTP-induced destabilization of Nppc mRNA.
To confirm the presence and location of repressive noncanonical
ARE motifs, we next constructed a range of luciferase reporters
that contained different truncated fragments from the proximal to
distal portions of the Nppc 3UTR (Fig. 2C). Similar to the results
using the full-length Nppc 3UTR, overexpression of TTP signifi-
cantly decreased the luciferase activity of the reporter constructs
containing longer proximal regions (1 to 365, 1 to 275), even
though the truncated region contained canonical ARE sites (Fig. 2D).
However, when shorter fragments (1 to 160, 1 to 97) were retained,
the luciferase activity of these reporters were not repressed by TTP
overexpression, which was comparable to the full-length Nppc
3UTR deletion reporter construct (Fig. 2D). This result suggested
that the uncharacterized repressive motifs were located in the 160 to
275 region of the Nppc 3UTR. Additionally, we observed an in-
creased trend of luciferase activity when distal fragments (down-
stream from 275) were deleted (Fig. 2D), implying that weak
repressive AU-rich motifs were located in this region.
Next, we performed a more detailed analysis of the 160 to 275
region of the Nppc 3UTR and confirmed the location of the non-
canonical repressive ARE, which was characterized by UU/UA di-
nucleotide clusters (231 to 257) (Fig. 2E). More importantly, we
provided direct evidence that this non-AUUUA ARE motif could
functionally mediate TTP-induced destabilization of target mRNA
(Fig. 2F). Interestingly, by screening the Nppc 3UTR of different
mammalian species, we found that the UU/UA dinucleotide clusters
are highly conserved (SI Appendix,Fig.S4B). Collectively, these
Fig. 1. TTP is up-regulated in response to the LH/hCG surge and targets the Nppc 3UTR. (Aand B) RT-qPCR detection of the expression dynamics of Nppc (A)
and Zfp36 (B) mRNA in preovulatory MGCs in response to hCG administration. (C) Western blotting analysis for TTP in preovulatory MGCs at different time
points after hCG administration. The upper and lower bands indicate phosphorylated (inactive) and unphosphorylated (active) forms of TTP, respectively. (D)
Immunohistochemical staining of TTP in preovulatory follicles before and after hCG administration. (Right) Higher magnification of boxed regions. (Scale bar,
100 μm.) (E) RIP analysis for the direct binding of TTP to the Nppc 3UTR. The RIP sample was subjected to RT-PCR, and the amplified products were visualized
using agarose gel electrophoresis. (F) Quantitative detection of the interaction between TTP and Nppc 3UTR by RIPRT-qPCR. (G) Schematic diagram of the
luciferase constructs containing full-length Nppc 3UTR, Tnf 3UTR (targeted control), or Actb 3UTR (nontargeted control). Relative luciferase activity of
different luciferase vectors, which were cotransfected with a Zfp36 overexpression vector or empty vector in HEK293 cells. (H) Measurement of the stability of
the luciferase mRNA that contained the Nppc 3UTR. The luciferase vector was cotransfected with the Zfp36 expression vector or empty vectors in HEK293
cells, which were treated with ActD to terminate transcription. Total RNAs were prepared at the indicated time points to detect the remaining luciferase
mRNA levels after ActD treatment. (I) Schematic diagram depicting the location of canonical AU-rich elements (AREs, presented as red triangles) in the Nppc 3
UTR. All data are presented as the mean ±SEM of three independent experiments. *P<0.05, **P<0.01, ***P<0.001. In A,B, and C, * indicates a significant
difference compared with data at 0 h post-hCG.
Xi et al. PNAS
|
3of8
The mRNA-destabilizing protein Tristetraprolin targets meiosis arresterNppc mRNA in
mammalian preovulatory follicles
https://doi.org/10.1073/pnas.2018345118
DEVELOPMENTAL
BIOLOGY
Downloaded by qianying yang on May 24, 2021
observations showed that TTP can bind to the Nppc 3UTR via
the noncanonical non-AUUUA ARE motif to destabilize its
target mRNA.
Nppc mRNA in Preovulatory MGCs Is Degraded by Up-Regulated TTP.
Having established the correlation between TTP and the non-
canonical ARE motif in the Nppc 3UTR, we next aimed to test
whether TTP can regulate Nppc expression in preovulatory MGCs.
We used a well-established in vitro model using MGCs isolated
from preovulatory follicles, which maintain high levels of Nppc
mRNA via supplementation with estrogen (18, 38). Given that LH
failed to decrease Nppc mRNA in vitro because of the dilution of
LH-induced epidermal growth factor (EGF)-like peptides in the
culture medium (18), we recapitulated the LH-induced rapid de-
crease of Nppc mRNA by modulating EGFR signaling, as reported
in previous studies (18, 39). In addition, we found that the ad-
ministration of the EGFR kinase inhibitor, AG1478, completely
blocked the hCG-induced rapid up-regulation of TTP (Fig. 3A),
implying that the activation and function of TTP largely depends
on LH/hCG-activated EGFR signaling. Moreover, our results
showed that Zfp36 was rapidly and transiently up-regulated and
that Nppc mRNA was decreased in cultured mouse MGCs in
response to EGF supplementation (Fig. 3B), thus recapitulating
the LH/hCG-induced changes in ovaries and MGCs (Fig. 1 A
and Band SI Appendix, Fig. S1 Aand B). These findings support
the feasibility of the model to study the role of TTP in controlling
endogenous Nppc expression. Furthermore, the in vitro model has
the unique advantages of MGC-specific genetic manipulation of
Zfp36 and efficient transcriptional termination to assess mRNA
stability.
Using the in vitro model, we next determined if TTP up-regulation
was essential and sufficient to decrease Nppc mRNA levels via loss-
and gain-of-function studies. As expected, when the expression of
Zfp36/TTP was reduced to basal level via small interfering (siRNA)-
mediated knockdown (Fig. 3Cand SI Appendix,Fig.S5A), EGF
supplementation failed to induce the rapid decrease in Nppc mRNA
but maintained the steady-state expression levels in preovulatory
MGCs, which contrasted with the groups transfected with the blank-
control and negative-control siRNAs (Fig. 3C). In contrast, over-
expression of TTP was sufficient to maintain Nppc mRNA at low
levels in preovulatory MGCs (Fig. 3Dand SI Appendix,Fig.S5B).
This result was consistent with the observation from the in vivo
Fig. 2. TTP targets the noncanonical ARE motifs in the Nppc 3UTR. (A) Schematic diagram of the luciferase reporters that contained single or all mutant
canonical ARE motifs in the Nppc 3UTR. (B) Relative luciferase activity of luciferase reporters containing different mutant ARE motifs, which were
cotransfected with the Zfp36 overexpression vector (pCAGGS-Zfp36) or empty vector (pCAGGS) in HEK293 cells. The reporter containing the Actb 3UTR was
detected as a nontargeted control. (C) Schematic diagram of the luciferase reporters that contained different truncated regions from the proximal to distal
portions of the Nppc 3UTR. Canonical ARE sites located in the Nppc 3UTR are indicated by blue arrows. (D) Relative luciferase activity of luciferase reporters
containing different Nppc 3UTR truncations, which were cotransfected with the Zfp36 overexpression vector or empty vectors in HEK293 cells. The reporters
that contained full-length or deleted Nppc 3UTRs were used as positive or negative control, respectively. (E) The sequence of the Nppc 3UTR containing
putative noncanonical ARE motifs (highlighted in red). (F) Relative luciferase activity of luciferase reporters containing further truncated Nppc 3UTR
fragments. All data are presented as the mean ±SEM of at least three independent experiments. **P<0.01, ***P<0.001.
4of8
|
PNAS Xi et al.
https://doi.org/10.1073/pnas.2018345118 The mRNA-destabilizing protein Tristetraprolin targets meiosis arresterNppc mRNA in
mammalian preovulatory follicles
Downloaded by qianying yang on May 24, 2021
experiment that down-regulated Zfp36 was associated with failure
to decrease the Nppc mRNA level (Fig. 4C). Next, we attempted
to confirm that TTP-induced rapid decrease in Nppc mRNA is
dependent on mRNA destabilization. We found that Nppc mRNA
maintained a relatively stable level until 4 h after ActD treatment;
however, Zfp36 overexpression significantly accelerated the desta-
bilization of Nppc mRNA, similar to that of the targeted control Tnf
mRNA (Fig. 3Eand SI Appendix,Fig.S5C). By contrast, accelerated
destabilization of nontargeted control Actb mRNA was not ob-
served following Zfp36 overexpression (SI Appendix, Fig. S5D).
Using cultured bovine MGCs as the model, our results also
suggest that the physiological role of EGF-induced TTP in degrading
NPPC mRNA might be common to different species (SI Appendix,
Fig. S5 EI).
Finally, to determine the physiological consequences of TTP
deficiency in vivo, we crossed Zfp36
flox/flox
mice and Fshr-Cre mice
to knock out Zfp36 specifically in MGCs. We detected that in the
resulting Zfp36
gc/
mice, Zfp36 expression was significantly de-
creased in preovulatory MGCs before and after hCG administra-
tion (Fig. 3F). The reduction in TTP protein levels in MGCs was
Fig. 3. TTP degrades endogenous Nppc mRNA in preovulatory MGCs. (A) Western blotting analysis for TTP, as well as phosphorylated and total EGFR, in
preovulatory MGCs in response to administration of hCG and AG1478, a specific EGFR inhibitor. MGCs were isolated from in vitro cultured preovulatory
follicles. (B) Expression dynamics of Nppc and Zfp36 in in vitro cultured MGCs in response to EGF administration. * indicates significant difference compared
with data at 0 h post-hCG. (C) Effect of Zfp36 knockdown on the EGF-induced decrease in Nppc mRNA in in vitro cultured MGCs. Total RNAs were prepared at
the indicated time points after EGF administration to detect Zfp36 (Upper) and Nppc (Lower) mRNA levels using RT-qPCR. (D) Effect of Zfp36 overexpression
on the Nppc mRNA levels in in vitro cultured MGCs. Nppc expression was maintained at a high level before overexpressing Zfp36. Total RNAs were prepared
at the indicated time points after transfection to detect Zfp36 (Upper) and Nppc (Lower) mRNA levels. (E) Measurement of the stability of Nppc mRNA in
MGCs following transfection with the Zfp36 overexpression vector (pCAGGS-Zfp36) or empty vector. Total RNAs were prepared at the indicated time points to
detect the remaining Nppc mRNA levels after ActD treatment. (FH) The detection of Zfp36 knockout efficiency in MGCs at 2 h post-hCG, at which point Zfp36
should be fully activated in response to hCG administration. The expression levels of Zfp36/TTP in MGCs were detected at 0 h and 2 h post-hCG using RT-qPCR
(F), immunohistochemistry (G), and Western blotting (H). (G, Lower) Higher magnification of boxed regions. (Scale bar, 100 μm.) (I) Effects of MGC-specific
depletion of Zfp36 on Nppc mRNA levels at 0 h and 2 h post-hCG. (Jand K) Evaluation of oocyte meiotic resumption scored as GVBD percentage at 6 h (J)and
9h(K) post-hCG. Representative images show the morphology of oocytes isolated from preovulatory follicles of wild-type (WT) and Zfp36
gc/
mice (Right),
five to eight mice were used at each time point. (Scale bar, 100 μm.) Data show the means ±SEMs of three independent experiments. *P<0.05, **P<0.01,
***P<0.001.
Xi et al. PNAS
|
5of8
The mRNA-destabilizing protein Tristetraprolin targets meiosis arresterNppc mRNA in
mammalian preovulatory follicles
https://doi.org/10.1073/pnas.2018345118
DEVELOPMENTAL
BIOLOGY
Downloaded by qianying yang on May 24, 2021
further confirmed by immunohistochemical and Western blot-
ting detection at 2 h post-hCG (Fig. 3 Gand H), at which point
the high-level expression of Zfp36 should appear in preovulatory
follicles (Fig. 1Band SI Appendix,Fig.S1B). Incomplete up-
regulation of TTP/Zfp36, in turn, impaired the hCG-induced rapid
decrease in Nppc mRNA (Fig. 3I). When oocyte meiotic resump-
tion, scored as percent germinal vesicle breakdown (GVBD), was
evaluated, Zfp36
gc/
mice showed impaired meiotic resumption
compared with that in the wild-type controls (Fig. 3 Jand K). These
findings were also supported by results of lentivirus-mediated short
hairpin RNA (shRNA) interference of Zfp36 in preovulatory
follicles (SI Appendix, Fig. S6 AF). Collectively, our results from
in vitro and in vivo models demonstrated that TTP-dependent
mRNA degradation underlies the rapid decrease in Nppc mRNA
in preovulatory follicles, which plays a functional role in fine-tuning
the progression of oocyte meiosis.
LH/hCG Activates Zfp36 through the EGFR-ERK1/2Dependent Pathway.
Having confirmed the functional role of TTP-mediated mRNA
degradation of Nppc mRNA in preovulatory MGCs, we next
attempted to understand the mechanism underlying the transient
up-regulation of Zfp36 in response to the LH/hCG surge. Multiple
Fig. 4. LH/hCG activates Zfp36/TTP through the EGFR-ERK1/2dependent pathway. (A) Effect of the EGFR-ERK1/2 pathway on the expression of Zfp36 and its
associated transcription factors. mRNA expression levels of Zfp36,Nppc,Elk1,andEgr1 in in vitro cultured MGCs that were treated with either EGF or AG1478 (a
specific EGFR inhibitor), U0126 (a specific ERK inhibitor), or their various combinations were detected. AG1478 or U0126 was administered 1 h before EGF sup-
plementation. (B) Western blotting analysis for TTP, EGR1, and phosphorylated and total ERK1/2 or ELK1 in in vitro cultured MGCs that were treated with either
EGF, AG1478, U0126, or their various combinations for 20 min and 1 h. (C) Expression levels of Zfp36,Nppc,Elk1,andEgr1 mRNA in isolated preovulatory MGCs
from mice that were intraperitoneally administrated with hCG, with or without PD0325901 (a specific ERK inhibitor). MGCs were collected at 3 h after hCG
administration, and PD0325901 was injected at 4 h before hCG. (D) Western blotting analysis for TTP, EGR1, and phosphorylated and total EGFR, ERK1/2, or ELK1
in MGCs from in vitro cultured preovulatory follicles that were treated with either hCG, AG1478, U0126, or their various combinations. (E) Western blotting
analysis for TTP, EGR1, and phosphorylated and total EGFR, ERK1/2, or ELK1 in isolated preovulatory MGCs from mice that were injected intraperitoneally with
either PD0325901, AG1478, or their various combinations at 4 h before hCG administration. All data are presented as the mean ±SEM of at least three inde-
pendent expe riments. *P<0.05, **P<0.01, ***P<0.001. (F) Working model of the signaling pathways that regulate the CNP-NPR2-cGMP signaling cascade and
meiotic resumption in preovulatory follicles. Pathways reported previously and those identified in the present study are indicated in black and blue, respectively.
6of8
|
PNAS Xi et al.
https://doi.org/10.1073/pnas.2018345118 The mRNA-destabilizing protein Tristetraprolin targets meiosis arresterNppc mRNA in
mammalian preovulatory follicles
Downloaded by qianying yang on May 24, 2021
molecular and cellular evidence indicated that Zfp36 transcrip-
tion is induced by E26 transformation-specific (ETS) transcrip-
tion factor ELK1 (ELK-1) and early growth response 1 (EGR-1)
in the EGFR-ERK1/2dependent pathway in somatic cells (4043).
Given that LH-activated EGFR-ERK1/2 signaling in preovulatory
follicles is essential to initiate meiotic resumption (22, 44), we hy-
pothesized that the rapid induction of Zfp36/TTP in response to
LH/hCG might depend on the EGFR-ERK1/2 pathway. Therefore,
we examined the hypothesis using both in vitro and in vivo models.
In in vitro cultured MGCs, accompanied by the rapidly increased
Zfp36 and decreased Nppc mRNA levels, we observed that the
transcriptional activation of Egr1, but not Elk1, responded rapidly to
EGF stimulation through the EGFR-ERK1/2dependent pathway
(Fig. 4A). Furthermore, Western blotting and immunofluorescence
assays showed that ERK1/2, as well as transcription factor ELK1,
could be phosphorylated (activated) in response to EGF stimulation
within 20 min (Fig. 4Band SI Appendix,Fig.S7Aand B). Subse-
quently, a marked increase in EGR1 was observed after 1 h (Fig. 4B
and SI Appendix,Fig.S7C), along with significantly increased TTP
levels (Fig. 4B). All these changes in response to EGFR signaling
activation could be attenuated by AG1478 (an EGFR signaling-
specific inhibitor) or U0126 (an ERK1/2-specific inhibitor) (Fig. 4 A
and B). Moreover, to further confirm the dependence of TTP in-
duction on the ERK1/2 pathway, mice were intraperitoneally in-
jected with the ERK1/2 inhibitor 4 h before hCG administration.
Consistent with the results from the in vitro model, in vivo inhibition
of the ERK1/2 pathway significantly blocked the induction of Egr1
and Zfp36 expression, and the decrease in Nppc mRNA levels
(Fig. 4C). Thus, the results of in vitro and in vivo assays indicated
that the ERK1/2 pathway is essential for rapidly activating the
transcriptional induction system of Zfp36.
Given that EGFR is not the only mediator of LH/hCG-in-
duced ERK1/2 activation (4547), we next examined if EGFR
was indispensable in mediating the LH/hCG-induced activation
of the ERK1/2 pathway and downstream transcriptional induction
system of Zfp36. To this end, isolated mouse preovulatory follicles
were cultured and treated with hCG, which resulted in the hCG-
induced activation/expression of the ERK1/2 pathway, as well as
TTP (Fig. 4D). Additionally, the changes in transcription factor
levels could be completely reversed by blocking EGFR signaling
(Fig. 4D). ERK inhibition did not attenuate EGFR phosphory-
lation/activation (Fig. 4D). Notably, EGFR inhibition and ERK
inhibition, alone or in combination, showed a similar effect of
blocking hCG-induced TTP expression (Fig. 4D), suggesting that
the ERK1/2 pathway acts downstream of EGFR signaling. These
findings were further confirmed using in vivo models (Fig. 4E).
Collectively, these results demonstrated that the preovulatory LH
surge induces transient expression of Zfp36/TTP and its transcrip-
tional induction system via the EGFR-ERK1/2 signaling pathway.
Discussion
The progression of gonadotropin-controlled oocyte meiosis is
tightly regulated by the activity of the CNP-NPR2-cGMP signaling
cascade. Before the LH surge, CNP-NPR2-cGMP signaling main-
tains the active state. The arrester,CNP, is primarily synthesized
by MGCs and binds to NPR2 throughout the follicle to stimulate
cGMP production, thus maintaining meiotic arrest. Following the
LH surge, CNP-NPR2-cGMP signaling inhibition occurs consecu-
tively on multiple levels; however, it remains unclear what mecha-
nism maintains the inhibition of the cascade, achieving sustained
cGMP at low levels before and during meiotic resumption. Earlier
studies proposed that the LH-induced rapid dephosphorylation
(inactivation) of NPR2 guanylyl cyclase might contribute to
CNP-NPR2-cGMP signaling inhibition, thus causing the rapid
decrease in cGMP. However, further studies have indicated that
the NPR2 dephosphorylation-dependent rapid decrease in cGMP (13,
14) is dispensable for LH-induced meiotic resumption because the
mouse model that prevents NPR2 dephosphorylation (inactivation)
only attenuated part of the cGMP decrease, which resulted in
delayed, but not failed, meiotic resumption (16). Similarly, LH-
induced phosphodiesterase 5 (PDE5), which directly hydrolyzes
cGMP, only partially contributes to the rapid cGMP decrease
and does not affect the timing of meiotic resumption. More im-
portantly, mice carrying both active NPR2 and mutated PDE5
failed to completely block cGMP decrease (12). Therefore, these
results are reminiscent of upstream changes to the signaling cascade
(i.e., the LH/hCG-induced decrease in follicular ligand CNP and its
encoding mRNA Nppc), which was documented as an important
fail-safe mechanism that may ensure LH-induced CNP-NPR2-
cGMP signaling inhibition and meiotic resumption (22).
Our data further emphasized the importance of ovarian follicular
somatic cells in regulating cGMP homeostasis. To maintain cGMP at
high levels and sustain meiotic arrest, somatic cells not only produce
the ligand (CNP) that activates its guanylyl cyclase (NPR2) (11) but
also provide guanylyl substrate for de novo synthesis of cGMP (48).
After the LH surge, cGMP begins to decrease rapidly in somatic
cells via the synergistic effect of decreased activity and affinity of
NPR2 guanylyl cyclase (1416), direct hydrolysis of cGMP (12),
and decreased concentrations of CNP (1719). Functional redun-
dancy also suggests that the timing of meiotic resumption and ovu-
lation is critical, and multiple pathways might have evolved as a fail-
safe system to ensure LH-induced meiotic resumption. Compared
with the rapid decrease of NPR2 activity and activated hydrolysis of
cGMP that occurs within 30 min, the decrease of CNP is a relatively
slow event (50% decrease within 2 h and further decrease until the
nuclear envelope is completely broken down) but is concurrent with
the progression of meiotic resumption (14). Thus, the decrease of
CNP/Nppc in the preovulatory follicle was thought to potentially
contribute to driving meiotic resumption, and the mechanism un-
derlying the decrease was considered as an important and long-
standing unanswered question (14, 22). Our study supported the
idea that the delayed decrease in Nppc mRNA could impair the
progression of meiotic resumption. These results, together with
previous works that focused on NPR2 inactivation and cGMP hy-
drolysis (12, 14, 16), allow us to presume that NPR2 inactivation and
cGMP hydrolysis may preferentially contribute to the rapid initiation
of meiotic resumption, while Nppc mRNA decline is primarily re-
sponsible for the later meiotic progression.
In the present study, we revealed that the preovulatory LH surge
can stimulate Zfp36 expression via activation of ELK-1 and EGR-1
transcription factors through the EGFR-ERK1/2 signaling path-
way. Up-regulated TTP subsequently targets noncanonical ARE
motifs in the 3UTR of the Nppc mRNA, which induces its rapid
degradation (Fig. 4F). The turnover of CNP is rapid (24), and the
LH-induced decrease in the CNP peptide is independent of the na-
triuretic peptide clearance receptor NPR3 (18); therefore, the rapid
degradation of Nppc mRNA would lead to a decrease in CNP
peptide levels, thus maintaining CNP-NPR2-cGMP signaling inhibi-
tion during meiotic resumption.
We provided the functional link between TTP-mediated mRNA
degradation and oocyte meiotic progression. The involvement of
mRNA degradation-mediated posttranscriptional mechanism in
regulating CNP-NPR2-cGMP signaling has never been reported.
Furthermore, our results highlight the role of the rare noncanonical
AU-rich element in this process. Among the ARE-binding proteins
that regulate posttranscriptional mechanisms, TTP has the most
specific recognition and binding ARE sequences (i.e., canonical
AUUUA motifs) (26, 27). However, non-AUUUA, but AU-rich
motifs, can also destabilize target mRNA (36, 37, 49). The UU/UA
dinucleotide clusters observed in the Nppc 3UTR, although gener-
ally rare in vertebrate genomes, were shown to correlate positively
with TTP-induced mRNA destabilization (49). Our results, together
with those of previous studies, supported the idea that noncanonical
ARE motifs efficiently mediate the mRNA-destabilizing function.
The present study identified that TTP-induced mRNA degrada-
tion, a global posttranscriptional regulatory mechanism, acts as a
Xi et al. PNAS
|
7of8
The mRNA-destabilizing protein Tristetraprolin targets meiosis arresterNppc mRNA in
mammalian preovulatory follicles
https://doi.org/10.1073/pnas.2018345118
DEVELOPMENTAL
BIOLOGY
Downloaded by qianying yang on May 24, 2021
regulatory component of orchestrating the signals for oocyte meiotic
maturation. We report that Nppc mRNA is the functional target of
TTP, via rare noncanonical ARE motifs. This finding might shed
light on the mechanism of CNP-dependent cGMP homeostasis in
regulating other physiological processes, considering the significant
regulatory role of CNP-NPR2-cGMP signaling in the development
and function of the cardiovascular, skeletal, and nervous systems.
Materials and Methods
Animal Studies and Ethical Approval. All female ICR mice used in this study were
from Beijing Vital River Laboratory Animal Technology Co. Ltd. (Beijing, China).
Fshr-Cre mice and loxP-flanked Zfp36 mice (Zfp36
flox/flox
) were gifts from
Dr. Louis Dubeau (University of Southern California, Los Angeles, CA) and Dr. Perry
J. Blackshear (National Institute of Environmental Health Sciences, NIH), re-
spectively. All mice were maintained in a climate-controlled room on a 12 h
light/darkness cycle and allowed food and water ad libitum. The China Ag-
ricultural University Institutional Animal Care and Use Committee approved
this study, which was performed in accordance with the committees
guidelines. All efforts were made to minimize animal suffering.
Cell culture, isolation, and culture of follicles, Western blotting, immuno-
fluorescence and histologic analysis, RIP are described in SI Appendix,Materials
and Methods.
Data Availability. All study data are included in the article and/or SI Appendix.
ACKNOWLEDGMENTS. We thank Dr. Louis Dubeau (University of Southern
California) and Dr. Su Youqiang (Nanjing Medical University) for providing the
Fshr-Cre mice and breeding schemes. We also thank Dr. Perry J. Blackshear (Na-
tional Institute of Environmental Health Sciences, NIH) and Gu Ling (Nanjing
Agricultural University) for providing the Zfp36
flox/flox
mice and breeding
schemes. We thank Dr. Wang Chao (China Agricultural University) for his helpful
suggestions and for providing the lentiviral vectors. This work was supported by
grants from the National Natural Science Foundation of China (Grants 31972573
and 31672426), the National Key R&D Program (Grants 2017YFD0501901 and
2017YFD0501905), the Fundamental Research Funds for the Central Universities
(Grant 2020TC004), and the Beijing Innovation Consortium of Agriculture
Research System.
1. J. A. Beavo, L. L. Brunton, Cyclic nucleotide researchStill expanding after half a
century. Nat. Rev. Mol. Cell Biol. 3, 710718 (2002).
2. L. R. Potter, Natriuretic peptide metabolism, clearance and degradation. FEBS J. 278,
18081817 (2011).
3. L. R. Potter, S. Abbey-Hosch, D. M. Dickey, Natriuretic peptides, their receptors, and
cyclic guanosine monophosphate-dependent signaling functions. Endocr. Rev. 27,
4772 (2006).
4. A. J. Moyes et al., Endothelial C-type natriuretic peptide maintains vascular homeo-
stasis. J. Clin. Invest. 124, 40394051 (2014).
5. K. Ono et al ., A single-nucleotide polymorphism in C-type natriuretic peptide gene
may be associated with hypertension. Hypertens. Res. 25, 727730 (2002).
6. H. Chusho et al., Dwarfism and early death in mice lacking C-type natriuretic peptide.
Proc. Natl. Acad. Sci. U.S.A. 98, 40164021 (2001).
7. K. Miura et al., An overgrowth disorder associated with excessive production of cGMP
due to a gain-of-function mutation of the natriuretic peptide receptor 2 gene. PLoS
One 7, e42180 (2012).
8. Q. Ma, L. Zhang, C-type natriuretic peptide functions as an innate neuroprotectant in
neonatal hypoxic-ischemic brain injury in mouse via natriuretic peptide receptor 2.
Exp. Neurol. 304,5866 (2018).
9. G. Ter-Avetisyan et al., Loss of axon bifurcation in mesencephalic trigeminal neurons im-
pairs the maximal biting force in Npr2-deficient mice. Front. Cell. Neurosci. 12, 153 (2018).
10. H. Schmidt et al., C-type natriuretic peptide (CNP) is a bifurcation factor for sensory
neurons. Proc. Natl. Acad. Sci. U.S.A. 106, 1684716852 (2009).
11. M. Zhang, Y. Q. Su, K. Sugiura, G. Xia, J. J. Eppig, Granulosa cell ligand NPPC and its
receptor NPR2 maintain meiotic arrest in mouse oocytes. Science 330, 366369 (2010).
12. J. R. Egbert, S. P. Yee, L. A. Jaffe, Luteinizing hormone signaling phosphorylates and
activates the cyclic GMP phosphodiesterase PDE5 in mouse ovarian follicles, contrib-
uting an additional component to the hormonally induced decrease in cyclic GMP
that reinitiates meiosis. Dev. Biol. 435,614 (2018).
13. J. R. Egbert et al., Dephosphorylation and inactivation of NPR2 guanylyl cyclase in
granulosa cells contributes to the LH-induced decrease in cGMP that causes re-
sumption of meiosis in rat oocytes. Development 141,35943604 (2014).
14. J. W. Robinson et al., Luteinizing hormone reduces the activity of the NPR2 guanylyl
cyclase in mouse ovarian follicles, contributing to the cyclic GMP decrease that pro-
motes resumption of meiosis in oocytes. Dev. Biol. 366, 308316 (2012).
15. X. Hao et al., Epidermal growth factor-mobilized intracellular calcium of cumulus cells
decreases natriuretic peptide receptor 2 affinity for natriuretic peptide type C and
induces oocyte meiotic resumption in the mouse. Biol. Reprod. 95, 45 (2016).
16. L. C. Shuhaibar et al., Dephosphorylation of juxtamembrane serines and threonines of
the NPR2 guanylyl cyclase is required for rapid resumption of oocyte meiosis in re-
sponse to luteinizing hormone. Dev. Biol. 409, 194201 (2016).
17. K. Kawamura et al., Pre-ovulatory LH/hCG surge decreases C-type natriuretic peptide
secretion by ovarian granulosa cells to promote meiotic resumption of pre-ovulatory
oocytes. Hum. Reprod. 26, 30943101 (2011).
18. K. B. Lee et al., Hormonal coordination of natriuretic peptide type C and natriuretic
peptide receptor 3 expression in mouse granulosa cells. Biol. Reprod. 88, 42 (2013).
19. X. Liu, F. Xie, A. M. Zamah, B. Cao, M. Conti, Multiple pathways mediate luteinizing hor-
mone regulation of cGMP signaling in the mouse ovarian follicle. Biol. Reprod. 91, 9 (2014).
20. W. Zhang et al., Brain natriuretic peptide and C-type natriuretic peptide maintain
porcine oocyte meiotic arrest. J. Cell. Physiol. 230,7181 (2015).
21. G. Xi et al., Natriuretic peptide receptor 2 (NPR2) localized in bovine oocyte underlies
a unique mechanism for C-type natriuretic peptide (CNP)-induced meiotic arrest.
Theriogenology 106, 198209 (2018).
22. L. A. Jaffe, J. R. Egbert, Regulation of mammalian oocyte meiosis by intercellular
communication within the ovarian follicle. Annu. Rev. Physiol. 79, 237260 (2017).
23. L. C. Shuhaibar et al., Intercellular signaling via cyclic GMP diffusion through gap
junctions restarts meiosis in mouse ovarian follicles. Proc. Natl. Acad. Sci. U.S.A. 112,
55275532 (2015).
24. P. J. Hunt, A. M. Richards, E. A. Espiner, M. G. Nicholls, T. G. Yandle, Bioactivity and
metabolism of C-type natriuretic peptide in normal man. J. Clin. Endocrinol. Metab.
78, 14281435 (1994).
25. M. Z. Carletti, L. K. Christenson, Rapid effects of LH on gene expression in the mural
granulosa cells of mouse periovulatory follicles. Reproduction 137,843855 (2009).
26. D. Beisang, P. R. Bohjanen, Perspectives on the ARE as it turns 25 years old. Wiley
Interdiscip. Rev. RNA 3, 719731 (2012).
27. S. A. Brooks, P. J. Blackshear, Tristetraprolin (TTP): Interactions with mRNA and proteins, and
current thoughts on mechanisms of action. Biochim. Biophys. Acta 1829,666679 (2013).
28. L. K. Christenson et al., Research resource: Preovulatory LH surge effects on follicular
theca and granulosa transcriptomes. Mol. Endocrinol. 27, 11531171 (2013).
29. M. Jo et al., Development and application of a rat ovarian gene expression database.
Endocrinology 145, 53845396 (2004).
30. M. L. Wissing et al., Identification of new ovulation-related genes in humans by
comparing the transcriptome of granulosa cells before and after ovulation triggering
in the same controlled ovarian stimulation cycle. Hum. Reprod. 29, 9971010 (2014).
31. F. Xu et al., Dynamics of the transcriptome in the primate ovulatory follicle. Mol.
Hum. Reprod. 17, 152165 (2011).
32. E. Carballo, W. S. Lai, P. J. Blackshear, Feedback inhibition of macrophage tumor
necrosis factor-alpha production by tristetraprolin. Science 281, 10011005 (1998).
33. H. H. Lee et al., Tristetraprolin regulates expression of VEGF and tumorigenesis in
human colon cancer. Int. J. Cancer 126, 18171827 (2010).
34. K. Masuda et al., Arid5a controls IL-6 mRNA stability, which contributes to elevation
of IL-6 level in vivo. Proc. Natl. Acad. Sci. U.S.A. 110, 94099414 (2013).
35. E. Van Tubergen et al., Tristetraprolin regulates interleukin-6, which is correlated
with tumor progression in patients with head and neck squamous cell carcinoma.
Cancer 117, 26772689 (2011).
36. S. S. Peng, C. Y. Chen, A. B. Shyu, Functional characterization of a non-AUUUA AU-rich
element from the c-jun proto-oncogene mRNA: Evidence for a novel class of AU-rich
elements. Mol. Cell. Biol. 16, 14901499 (1996).
37. B. Sarkar, Q. Xi, C. He, R. J. Schneider, Selective degradation of AU-rich mRNAs pro-
moted by the p37 AUF1 protein isoform. Mol. Cell. Biol. 23, 66856693 (2003).
38. J. Yang et al., Transforming growth factor-βis involved in maintaining oocyte meiotic
arrest by promoting natriuretic peptide type C expression in mouse granulosa cells.
Cell Death Dis. 10, 558 (2019).
39. T. Tsuji, C. Kiyosu, K. Akiyama, T. Kunieda, CNP/NPR2 signaling maintains oocyte
meiotic arrest in early antral follicles and is suppressed by EGFR-mediated signaling in
preovulatory follicles. Mol. Reprod. Dev. 79, 795802 (2012).
40. M. Florkowska et al., EGF activates TTP expression by activation of ELK-1 and EGR-1
transcription factors. BMC Mol. Biol. 13, 8 (2012).
41. W. S. Lai, M. J. Thompson, G. A. Taylor, Y. Liu, P. J. Blackshear, Promoter analysis of
Zfp-36, the mitogen-inducible gene encoding the zinc finger protein tristetraprolin.
J. Biol. Chem. 270, 2526625272 (1995).
42. W. S. Lai, M. J. Thompson, P. J. Blackshear, Characteristics of the intron involvement in
the mitogen-induced expression of Zfp-36. J. Biol. Chem. 273, 506517 (1998).
43. C. Sobolewski, S. Sanduja, F. F. Blanco, L. Hu, D. A. Dixon, Histone deacetylase in-
hibitors activate tristetraprolin expression through induction of early growth re-
sponse protein 1 (EGR1) in colorectal cancer cells. Biomolecules 5, 20352055 (2015).
44. M. Zhang, H. Ouyang, G. Xia, The signal pathway of gonadotrophins-induced mam-
malian oocyte meiotic resumption. Mol. Hum. Reprod. 15, 399409 (2009).
45. H. Y. Fan et al., MAPK3/1 (ERK1/2) in ovarian granulosa cells are essential for female
fertility. Science 324, 938941 (2009).
46. B. Menon, M. Franzo-Romain, S. Damanpour, K. M. Menon, Luteinizing hormone
receptor mRNA down-regulation is mediated through ERK-dependent induction of
RNA binding protein. Mol. Endocrinol. 25, 282290 (2011).
47. S. Panigone, M. Hsieh, M. Fu, L. Persani, M. Conti, Luteinizing hormone signaling in
preovulatory follicles involves early activation of the epidermal growth factor re-
ceptor pathway. Mol. Endocrinol. 22, 924936 (2008).
48. K. Wigglesworth et al., Bidirectional communication between oocytes and ovarian
follicular somatic cells is required for meiotic arrest of mammalian oocytes. Proc. Natl.
Acad. Sci. U.S.A. 110, E3723E3729 (2013).
49. M. Al-Saif, K. S. Khabar, UU/UA dinucleotide frequency reduction in coding regions
results in increased mRNA stability and protein expression. Mol. Ther. 20, 954959
(2012).
8of8
|
PNAS Xi et al.
https://doi.org/10.1073/pnas.2018345118 The mRNA-destabilizing protein Tristetraprolin targets meiosis arresterNppc mRNA in
mammalian preovulatory follicles
Downloaded by qianying yang on May 24, 2021
... A temporary delay in meiosis resumption during IVM could have beneficial effects. C-type natriuretic peptide (CNP), which is expressed at high levels in granulosa cells (GCs), can temporarily maintain the meiotic arrest of oocytes (Zhang et al. 2010;Xi et al. 2018Xi et al. , 2021. High levels of cAMP within the oocyte maintain meiotic arrest. ...
... Pre-IVM with 100 and 50 nM sustains meiotic arrest for 4 h in adult goat and 24 h in mouse (Wei et al. 2017), respectively. It is reported that CNP binding to its receptor NPR2 in CCs plays a functional role in maintaining the arrest of meiosis (Zhang et al. 2010;Xi et al. 2018;Xi et al. 2021). NPR2 mRNA is significantly decreased during IVM of COCs and promotes oocyte meiotic resumption (Yang et al. 2016). ...
Article
Full-text available
The effects of C-type natriuretic peptide (CNP) pre-treatment on the in vitro maturation (IVM) and developmental competence of yak (Bos grunniens) oocytes were evaluated. The results showed that 100 nM CNP could effectively maintain meiotic arrest of yak oocytes in vitro within 6 h. Yak oocytes matured in two-step IVM (pre-IVM with 100 nM CNP for 6 h followed by conventional IVM for 28 h) yielded a significantly (P < .05) increased cleavage rate, blastocyst rate after in vitro fertilization with cattle spermatozoa compared to the conventional one-step IVM method. The density of transzonal projections (TZPs) decreased after 6 h standard IVM (P < .05), whereas, COCs pre-IVM with 100 nM CNP for 6 h maintained the same density of TZPs compared to the fresh COCs. This pre-IVM resulted in lesser reactive oxygen species level (P < .05). Two-step IVM resulted in the increased glutathione levels, mRNA expression levels of BCL2, BMP15, and GDF9 (P < .05) in oocytes. In conclusion, these results indicate that CNP treatment (100 nM, 6 h) before IVM could enhance the maturation of yak oocytes and developmental competence of their crossbred embryos. This may have important applications in in vitro production of yak crossbred embryos.
... [43][44][45][46] A more recent advance has been made by our discovery that mRNA-destabilizing protein Tristetraprolin-induced Nppc mRNA degradation, is the upstream mechanism that underlies LH-induced CNP-NPR2-cGMP signaling inhibition, which is critical for controlling the timing of meiotic resumption. 47 Moreover, our own work 14 as well as the research of other groups 48-50 also support the notion that CNP is essential for coordinating synchronization of nuclear-cytoplasmic maturation by maintaining meiotic arrest, thus ensuring oocytes complete cytoplasmic maturation and acquire developmental potential. The optimal cytoplasmic maturation is essential for acquiring further developmental competence. ...
Article
Full-text available
C‐natriuretic peptide (CNP) is the central regulator of oocyte meiosis progression, thus coordinating synchronization of oocyte nuclear‐cytoplasmic maturation. However, whether CNP can independently regulate cytoplasmic maturation has been long overlooked. Mitochondrial DNA (mtDNA) accumulation is the hallmark event of cytoplasmic maturation, but the mechanism underlying oocyte mtDNA replication remains largely elusive. Herein, we report that CNP can directly stimulate oocyte mtDNA replication at GV stage, and deficiency of follicular CNP may contribute largely to lower mtDNA copy number in in vitro matured oocytes. The mechanistic study showed that cAMP‐PKA‐CREB1 signaling cascade underlies the regulatory role of CNP in stimulating mtDNA replication and upregulating related genes. Of interest, we also report that CNP‐NPR2 signaling is inhibited in aging follicles, and this inhibition is implicated in lower mtDNA copy number in oocytes from aging females. Together, our study provides the first direct functional link between follicular CNP and oocyte mtDNA replication, and identifies its involvement in aging‐associated mtDNA loss in oocytes. These findings, not only update the current knowledge of the functions of CNP in coordinating oocyte maturation but also present a promising strategy for improving in vitro fertilization outcomes of aging females.
... The natriuretic peptide receptors (NPRs) in mammalian follicles play a critical role in regulating oocyte meiotic arrest. NPR2, a major member of the NPR family, is mainly located on CGCs, whereas its ligand, known as natriuretic peptide precursor C (NPPC), is expressed in MGCs in mice [41], humans [42], and pigs [43]. Natriuretic peptide precursor C inhibits spontaneous GVBD in COCs but exerts little inhibitory effect on DOs. ...
Article
Full-text available
In human female primordial germ cells, the transition from mitosis to meiosis begins from the fetal stage. In germ cells, meiosis is arrested at the diplotene stage of prophase in meiosis I (MI) after synapsis and recombination of homologous chromosomes, which cannot be segregated. Within the follicle, the maintenance of oocyte meiotic arrest is primarily attributed to high cytoplasmic concentrations of cyclic adenosine monophosphate (cAMP). Depending on the specific species, oocytes can remain arrested for extended periods of time, ranging from months to even years. During estrus phase in animals or the menstrual cycle in humans, the resumption of meiosis occurs in certain oocytes due to a surge of luteinizing hormone (LH) levels. Any factor interfering with this process may lead to impaired oocyte maturation, which in turn affects female reproductive function. Nevertheless, the precise molecular mechanisms underlying this phenomenon has not been systematically summarized yet. To provide a comprehensive understanding of the recently uncovered regulatory network involved in oocyte development and maturation, the progress of the cellular and molecular mechanisms of oocyte nuclear maturation including meiosis arrest and meiosis resumption is summarized. Additionally, the advancements in understanding the molecular cytoplasmic events occurring in oocytes, such as maternal mRNA degradation, posttranslational regulation, and organelle distribution associated with the quality of oocyte maturation, are reviewed. Therefore, understanding the pathways regulating oocyte meiotic arrest and resumption will provide detailed insight into female reproductive system and provide a theoretical basis for further research and potential approaches for novel disease treatments.
... Ovarian folliculogenesis in mammals is a remarkably complex, well-orchestrated process in three phases: primordial follicle activation, growth of the follicle, and the ovulatory phase [1][2][3]. Most follicles undergo growth arrest and atresia, and only a few follicles develop to ovulation [4]. ...
Article
Full-text available
Background ISGylation is a post-translational protein modification that regulates many life activities, including immunomodulation, antiviral responses, and embryo implantation. The exact contribution of ISGylation to folliculogenesis remains largely undefined. Results Here, Isg15 knockout in mice causes hyperfertility along with sensitive ovarian responses to gonadotropin, such as increases in cumulus expansion and ovulation rate. Moreover, ISG15 represses the expression of ovulation-related genes in an ISGylation-dependent manner. Mechanistically, ISG15 binds to ADAMTS1 via the ISG15-conjugating system (UBA7, UBE2L6, and HERC6), ISGylating ADAMTS1 at the binding sites Lys309, Lys593, Lys597, and Lys602, resulting in ADAMTS1 degradation via a 20S proteasome-dependent pathway. Conclusion Taken together, the present study demonstrates that covalent ISG15 conjugation produces a novel regulatory axis of ISG15-ADAMTS1 that enhances the degradation of ADAMTS1, thereby compromising ovulation and female fertility.
... The elevation of intracellular calcium by S1P might also cause NPR2 inactivity by dephosphorylation [6,29]. The depletion of Sphk1/2 in granulosa cells could not block the meiotic resumption of all oocytes within preovulatory follicles, possibly because the LH/hCG-induced decrease in NPPC levels in MGCs leads to a reduction in NPR2 function [30]. S1P, a physiological calcium elevator, can mobilize Ca 2+ from the ER by S1P receptors and non-receptor pathways [18]. ...
Article
Full-text available
Germ cell division and differentiation require intimate contact and interaction with the surrounding somatic cells. Luteinizing hormone (LH) triggers epidermal growth factor (EGF)-like growth factors to promote oocyte maturation and developmental competence by activating EGF receptor (EGFR) in somatic cells. Here, we showed that LH-EGFR signaling-activated sphingosine kinases (SphK) in somatic cells. The activation of EGFR by EGF increased S1P and calcium levels in cumulus-oocyte complexes (COCs), and decreased the binding affinity of natriuretic peptide receptor 2 (NPR2) for natriuretic peptide type C (NPPC) to release the cGMP-mediated meiotic arrest. These functions of EGF were blocked by the SphK inhibitor SKI-II, which could be reversed by the addition of S1P. S1P also activated the Akt/mTOR cascade reaction in oocytes and promoted targeting protein for Xklp2 (TPX2) accumulation and oocyte developmental competence. Specifically depleting Sphk1/2 in somatic cells reduced S1P levels and impaired oocyte meiotic maturation and developmental competence, resulting in complete female infertility. Collectively, SphK-produced S1P in somatic cells serves as a functional transmitter of LH-EGFR signaling from somatic cells to oocytes: acting on somatic cells to induce oocyte meiotic maturation, and acting on oocytes to improve oocyte developmental competence.
Article
Sphingosine‐1‐phosphate (S1P), a lipid messenger, propagates its signals by interacting with its intracellular targets or is transported to autocrine/paracrine to activate its cell surface receptors. In the female reproductive system, the homeostasis of S1P plays an important role in ovarian follicular development. Our recent studies show that S1P emerges as a functional mediator of LH‐EGFR signaling from cumulus cells to oocytes: elevating calcium levels in cumulus cells to induce oocyte meiotic maturation, and activating Akt/mTOR cascade reaction to promote oocyte developmental competence. Thus, S1P might be applied to promote oocyte maturation in animals and humans.
Article
Full-text available
Chronic stress is suspected to be a causal factor of female subfertility; however, the underlying mechanisms remain unclear. Here, we found that chronic stress inhibited the cyclic adenosine 3′,5′-monophosphate (cAMP) signaling pathway, leading to ovarian reserve decline in mice. A chronic stress model was constructed using restraint stress for 8 weeks. An elongated estrous cycle and a significant increase in the number of atretic follicles were observed in the stress group. We identified a significant increase in meiotic arrest failure (MAF) in oocytes in the stress group, characterized by condensed metaphase chromosomes, assembled spindles, or polar bodies in the oocytes. Whole-mount ovarian reserve estimation at the single-oocyte level using the CUBIC method (clear, unobstructed brain/body imaging cocktails and computational analysis) revealed a significant decrease in quiescent oocytes from 2,261/ovary in the control group to 1,373/ovary in the stress group. The number of growing oocytes also significantly decreased from 220/ovary in the control group to 150/ovary in the stress group. Real-time quantitative polymerase chain reaction (RT-qPCR) analysis of the meiotic arrest maintenance pathways revealed significant downregulation of Gpr3 , Nppc , and Npr2 in the stress group. These results indicate that blocking cAMP production contributes to MAF and a decline in ovarian reserve. Overall, we present new insights into the mechanisms underlying chronic-stress-induced oocyte loss and potential targets for ovarian reserve preservation.
Article
Full-text available
It has been clear that retinoic acid (RA), the most active vitamin A (VA) derivative, plays a central role in governing oocyte meiosis initiation. However, it has not been functionally determined if RA participates in luteinizing hormone (LH)-induced resumption from long-lasting oocyte meiotic arrest, which is essential for haploid oocyte formation. In the present study, using well-established in vivo and in vitro models, we identified that intrafollicular RA signaling is important for normal oocyte meiotic resumption. A mechanistic study indicated that mural granulosa cells (MGCs) are the indispensable follicular compartment for RA-prompted meiotic resumption. Moreover, retinoic acid receptor (RAR) is essential for mediating RA signaling to regulate meiotic resumption. Furthermore, we found zinc finger protein 36 (ZFP36) is the transcriptional target of RAR. Both RA signaling and epidermal growth factor (EGF) signaling were activated in MGCs in response to LH surge, and two intrafollicular signalings cooperate to induce rapid Zfp36 upregulation and Nppc mRNA decrease, which is critical to LH-induced meiotic resumption. These findings extend our understanding of the role of RA in oocyte meiosis: RA not only governs meiotic initiation but also regulates LH-induced meiotic resumption. We also emphasize the importance of LH-induced metabolic changes in MGCs in this process.
Article
Full-text available
The estrous cycle is regulated by rhythmic endocrine interactions of the nervous and reproductive systems, which coordinate the hormonal and ovulatory functions of the ovary. Folliculogenesis and follicle progression require the orchestrated response of a variety of cell types to allow the maturation of the follicle and its sequela, ovulation, corpus luteum formation, and ovulatory wound repair. Little is known about the cell state dynamics of the ovary during the estrous cycle and the paracrine factors that help coordinate this process. Herein, we used single-cell RNA sequencing to evaluate the transcriptome of >34,000 cells of the adult mouse ovary and describe the transcriptional changes that occur across the normal estrous cycle and other reproductive states to build a comprehensive dynamic atlas of murine ovarian cell types and states.
Article
Full-text available
Natriuretic peptide type C (NPPC) secreted by mural granulosa cells (MGCs) maintains oocyte meiotic arrest via the activation of guanylyl cyclase-linked natriuretic peptide receptor 2 (NPR2). Here, we investigated the effect of transforming growth factor (TGF)-β on NPPC expression in MGCs and oocyte maturation. TGF-β ligands (TGFB1 and TGFB3, but not TGFB2) and receptors (TGFBR1 and TGFBR2) were predominantly expressed in MGCs. The activation of the follicle-stimulating hormone (FSH) receptor by FSH/equine chorionic gonadotropin (eCG) increased the levels of TGFB1, TGFBR2, and TGF-β downstream SMAD proteins in MGCs, which were decreased following the activation of the luteinizing hormone (LH) receptor by human chorionic gonadotropin (hCG). TGF-β significantly increased the gene and protein levels of NPPC in cultured MGCs through SMAD3 binding to Nppc promoter regions. In the presence of FSH, TGF-β further increased NPPC levels and inhibited oocyte meiotic resumption of cumulus-oocyte complexes (COCs). Moreover, Tgfbr2-specific depletion in granulosa cells using Fshr-Cre mice reduced NPPC mRNA and protein levels, resulting in the weak maintenance of oocyte meiotic arrest within large antral follicles. Tgfbr2 depletion also impaired follicle development, ovulation, and female fertility. Taken together, TGF-β-promoted NPPC in MGCs is involved in maintaining oocyte meiotic arrest. FSH and LH could regulate NPPC levels in MGCs via TGF-β and then control the process of oocyte meiosis.
Article
Full-text available
Bifurcation of axons from dorsal root ganglion (DRG) and cranial sensory ganglion (CSG) neurons is mediated by a cGMP-dependent signaling pathway composed of the ligand C-type natriuretic peptide (CNP), the receptor guanylyl cyclase Npr2 and the cGMP-dependent protein kinase I (cGKI). Here, we demonstrate that mesencephalic trigeminal neurons (MTN) which are the only somatosensory neurons whose cell bodies are located within the CNS co-express Npr2 and cGKI. Afferents of MTNs form Y-shaped branches in rhombomere 2 where the ligand CNP is expressed. Analyzing mouse mutants deficient for CNP or Npr2 we found that in the absence of CNP-induced cGMP signaling MTN afferents no longer bifurcate and instead extend either into the trigeminal root or caudally in the hindbrain. Since MTNs provide sensory information from jaw closing muscles and periodontal ligaments we measured the bite force of conditional mouse mutants of Npr2 (Npr2flox/flox;Engr1 Cre ) that lack bifurcation of MTN whereas the bifurcation of trigeminal afferents is normal. Our study revealed that the maximal biting force of both sexes is reduced in Npr2flox/flox;Engr1 Cre mice as compared to their Npr2flox/flox littermate controls. In conclusion sensory feedback mechanisms from jaw closing muscles or periodontal ligaments might be impaired in the absence of MTN axon bifurcation.
Article
Full-text available
Natriuretic peptide type C (NPPC) activation of the guanylyl cyclase-linked natriuretic peptide receptor 2 (NPR2) maintains oocyte meiotic arrest. Luteinizing hormone (LH)-dependent epidermal growth factor (EGF) receptor signaling elevates calcium of cumulus cells to inactivate NPR2, resulting in meiotic resumption. This study investigated the regulatory mechanism of calcium on NPR2 inactivation. In mouse ovarian follicles, LH, through the activation of EGF receptor, significantly elevated calcium levels in cumulus cells, but decreased the binding affinity of NPR2 for NPPC. In cultured cumulus-oocyte complexes (COCs), the activation of EGF receptor by EGF mobilized intracellular calcium of cumulus cells to decrease NPR2 affinity and cyclic guanosine monophosphate (cGMP) levels, resulting in meiotic resumption. However, hormone treatments had not changed NPR2 protein levels. In addition, the removal of magnesium ions from the medium decreased the binding affinity of NPR2 for NPPC, resulting in cGMP levels decrease and meiotic resumption. It is concluded that magnesium ions are required to maintain functional NPR2, and LH-dependent EGF receptor signaling mobilizes intracellular calcium of cumulus cells to reduce NPPC-NPR2 interaction that is required for meiotic resumption.
Article
Full-text available
The RNA-binding protein tristetraprolin (TTP) promotes rapid decay of mRNAs bearing 3' UTR AU-rich elements (ARE). In many cancer types, loss of TTP expression is observed allowing for stabilization of ARE-mRNAs and their pathologic overexpression. Here we demonstrate that histone deacetylase (HDAC) inhibitors (Trichostatin A, SAHA and sodium butyrate) promote TTP expression in colorectal cancer cells (HCA-7, HCT-116, Moser and SW480 cells) and cervix carcinoma cells (HeLa). We found that HDAC inhibitors-induced TTP expression, promote the decay of COX-2 mRNA, and inhibit cancer cell proliferation. HDAC inhibitors were found to promote TTP transcription through activation of the transcription factor Early Growth Response protein 1 (EGR1). Altogether, our findings indicate that loss of TTP in tumors occurs through silencing of EGR1 and suggests a therapeutic approach to rescue TTP expression in colorectal cancer.
Article
Full-text available
Significance By imaging cyclic GMP (cGMP) in live ovarian follicles from mice, we show how luteinizing hormone signaling in the follicle periphery results in a rapid decrease in cGMP in the oocyte, thus reinitiating meiosis. Luteinizing hormone signaling lowers cGMP in the outer cells of the follicle, then cGMP in the oocyte decreases as a consequence of diffusion through gap junctions. These findings demonstrate directly that a physiological signal initiated by a stimulus in one region of an intact tissue can travel across many layers of cells via cyclic nucleotide diffusion through gap junctions.
Article
Neonatal hypoxia-ischemia (HI) is the most common cause of brain injury in neonates, which leads to high neonatal mortality and severe neurological morbidity in later life (Vannucci, 2000; Volpe, 2001). Yet the molecular mechanisms of neuronal death and brain damage induced by neonatal HI remain largely elusive. Herein, using both in vivo and in vitro models, we determine an endogenous neuroprotectant role of c-type natriuretic peptide (CNP) in preserving neuronal survival after HI brain injury in mouse pups. Postnatal day 7 (P7) mouse pups with CNP deficiency (Nppclbab/lbab) exhibit increased brain infarct size and worsened long-term locomotor function after neonatal HI compared with wildtype control (Nppc+/+). In isolated primary cortical neurons, recombinant CNP dose-dependently protects primary neurons from oxygen-glucose deprivation (OGD) insult. This neuroprotective effect appears to be mediated through its cognate natriuretic peptide receptor 2 (NPR2), in that antagonization of NPR2, but not NPR3, exacerbates neuronal death and counteracts the protective effect of CNP on primary neurons exposed to OGD insult. Immunoblot and confocal microscopy demonstrate the abundant expression of NPR2 in neurons of the neonatal brain and in isolated primary cortical neurons as well. Moreover, similar to CNP deficiency, administration of NPR2 antagonist P19 via intracerebroventricular injection prior to HI results in exacerbated neuronal death and brain injury after HI. Altogether, the present study indicates that CNP and its cognate receptor NPR2 mainly expressed in neurons represent an innate neuroprotective mechanism in neonatal HI brain injury.
Article
Prior to birth, oocytes within mammalian ovarian follicles initiate meiosis, but then arrest in prophase until puberty, when with each reproductive cycle, one or more follicles are stimulated by luteinizing hormone (LH) to resume meiosis in preparation for fertilization. Within preovulatory follicles, granulosa cells produce high levels of cGMP, which diffuses into the oocyte to maintain meiotic arrest. LH signaling restarts meiosis by rapidly lowering the levels of cGMP in the follicle and oocyte. Part of this decrease is mediated by the dephosphorylation and inactivation the NPR2 guanylyl cyclase in response to LH, but the mechanism for the remainder of the cGMP decrease is unknown. At least one cGMP phosphodiesterase, PDE5, is activated by LH signaling, which would contribute to lowering cGMP. PDE5 exhibits increased cGMP-hydrolytic activity when phosphorylated on serine 92, and we recently demonstrated that LH signaling phosphorylates PDE5 on this serine and increases its activity in rat follicles. To test the extent to which this mechanism contributes to the cGMP decrease that restarts meiosis, we generated a mouse line in which serine 92 was mutated to alanine (Pde5-S92A), such that it cannot be phosphorylated. Here we show that PDE5 phosphorylation is required for the LH-induced increase in cGMP-hydrolytic activity, but that this increase has only a modest effect on the LH-induced cGMP decrease in mouse follicles, and does not affect the timing of meiotic resumption. Though we show that the activation of PDE5 is among the mechanisms contributing to the cGMP decrease, these results suggest that another cGMP phosphodiesterase is also activated by LH signaling.
Article
Meiosis is of prime importance for successful gametogenesis, and insufficient maintenance of oocyte meiotic arrest compromises oocyte developmental competence. Recent studies have demonstrated that the C-type natriuretic peptide (CNP)-Natriuretic peptide receptor 2 (NPR2) pathway can inhibit mammalian oocyte meiotic resumption. In mouse and porcine, the inhibitory effect of mural granulosa cell (MGC)-derived CNP on oocyte meiotic resumption is mediated by NPR2 localized in cumulus cells (CCs) surrounding the oocytes. However, in the present study, we identified a novel mechanism for CNP-induced meiotic arrest that appears to be unique to bovine oocytes. Unlike mouse and porcine, bovine NPR2 not only localizes in CCs, but also in oocyte membranes. We also showed that CNP can directly activate intra-oocyte cGMP production via NPR2 localized in oocyte membranes, in parallel with the CC-mediated pathway. Furthermore, we demonstrated that Npr2 expression in bovine CCs and oocytes were synergistically regulated by estradiol and oocyte-derived growth factors. Finally, based on the profound inhibitory effect of CNP on meiotic resumption, we established a natural factor synchronized in vitro oocyte maturation (NFSOM) system, which can significantly improve the developmental competence of matured oocytes, thereby resulting in higher in vitro embryo production efficiency. Taken together, our study not only provides new insight into understanding the crosstalk between oocytes and follicular somatic cells in mammals, but also presents a promising strategy for improving the in vitro oocyte maturation systems of assisted reproductive technology (ART).
Article
Meiotic progression in mammalian preovulatory follicles is controlled by the granulosa cells around the oocyte. Cyclic GMP (cGMP) generated in the granulosa cells diffuses through gap junctions into the oocyte, maintaining meiotic prophase arrest. Luteinizing hormone then acts on receptors in outer granulosa cells to rapidly decrease cGMP. This occurs by two complementary pathways: cGMP production is decreased by dephosphorylation and inactivation of the NPR2 guanylyl cyclase, and cGMP hydrolysis is increased by activation of the PDE5 phosphodiesterase. The cGMP decrease in the granulosa cells results in rapid cGMP diffusion out of the oocyte, initiating meiotic resumption. Additional, more slowly developing mechanisms involving paracrine signaling by extracellular peptides (C-type natriuretic peptide and EGF receptor ligands) maintain the low level of cGMP in the oocyte. These coordinated signaling pathways ensure a fail-safe system to prepare the oocyte for fertilization and reproductive success. Expected final online publication date for the Annual Review of Physiology Volume 79 is February 10, 2017. Please see http://www.annualreviews.org/page/journal/pubdates for revised estimates.
Article
The meiotic cell cycle of mammalian oocytes starts during embryogenesis and then pauses until luteinizing hormone (LH) acts on the granulosa cells of the follicle surrounding the oocyte to restart the cell cycle. An essential event in this process is a decrease in cyclic GMP in the granulosa cells, and part of the cGMP decrease results from dephosphorylation and inactivation of the natriuretic peptide receptor 2 (NPR2) guanylyl cyclase, also known as guanylyl cyclase B. However, it is unknown whether NPR2 dephosphorylation is essential for LH-induced meiotic resumption. Here, we prevented NPR2 dephosphorylation by generating a mouse line in which the seven regulatory serines and threonines of NPR2 were changed to the phosphomimetic amino acid glutamate (Npr2-7E). Npr2-7E/7E follicles failed to show a decrease in enzyme activity in response to LH, and the cGMP decrease was attenuated; correspondingly, LH-induced meiotic resumption was delayed. Meiotic resumption in response to EGF receptor activation was likewise delayed, indicating that NPR2 dephosphorylation is a component of the pathway by which EGF receptor activation mediates LH signaling. We also found that most of the NPR2 protein in the follicle was present in the mural granulosa cells. These findings indicate that NPR2 dephosphorylation in the mural granulosa cells is essential for the normal progression of meiosis in response to LH and EGF receptor activation. In addition, these studies provide the first demonstration that a change in phosphorylation of a transmembrane guanylyl cyclase regulates a physiological process, a mechanism that may also control other developmental events.