ArticlePDF Available

Neddylation inhibition affects early embryonic development by disrupting maternal-to-zygotic transition and mitochondrial function in mice

Authors:
Theriogenology 220 (2024) 1–11
Available online 28 February 2024
0093-691X/© 2024 Elsevier Inc. All rights reserved.
Original Research Article
Neddylation inhibition affects early embryonic development by disrupting
maternal-to-zygotic transition and mitochondrial function in mice
Mingxiao Liu
a
,
1
, Zhiming Ding
c
,
d
,
1
, Peihao Sun
a
, Shuo Zhou
a
, Hanxiao Wu
a
, Lijun Huo
a
,
b
,
Liguo Yang
a
,
b
, John S. Davis
e
, Aixin Liang
a
,
b
,
*
a
Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction of Ministry of Education, College of Animal Science and Technology, Huazhong
Agricultural University, Wuhan, 430070, PR China
b
Frontiers Science Center for Animal Breeding and Sustainable Production (Huazhong Agricultural University), Ministry of Education, Wuhan, 430070, PR China
c
Department of Obstetrics and Gynecology, The First Afliated Hospital of Anhui Medical University, Hefei, 230022, PR China
d
NHC Key Laboratory of Study on Abnormal Gametes and Reproductive Tract, Anhui Medical University, Hefei, 230032, PR China
e
Olson Center for Womens Health, Department of Obstetrics and Gynecology, University of Nebraska Medical Center, and Veterans Affairs Medical Center, Omaha, NE,
68198, USA
ARTICLE INFO
Keywords:
Neddylation
Embryonic development
MLN4924
Maternal-to-zygotic transition
Mitochondria
ABSTRACT
Post-translational modications (PTMs) are critical for early development in mice because early cleavage-stage
embryos are characterized by transcriptional inactivity. Neddylation is an important ubiquitin-like PTM that
regulates multiple biophysical processes. However, the exact roles of neddylation in regulating early embryonic
development remain largely unknown. In the present study, we found that inhibition of neddylation by specic
inhibitor MLN4924 led to severe arrest of early embryonic development. Transcriptomic analysis showed that
neddylation inhibition changed the expression of 3959 genes at the 2-cell stage. Importantly, neddylation in-
hibition blocked zygotic genome activation and maternal mRNA degradation, thus disrupting the maternal-to-
zygotic transition. Moreover, inhibition of neddylation induced mitochondrial dysfunction including aberrant
mitochondrial distribution, decreased mitochondrial membrane potential, and reduced ATP content. Further
analysis showed that inhibition of neddylation resulted in the accumulation of reactive oxygen species and su-
peroxide anion, thereby resulting in oxidative stress and severe DNA damage at the 2-cell stage. Overall, this
study demonstrates that neddylation is vital for early embryonic development in mice. Our ndings suggest that
proper neddylation regulation is essential for the timely inter-stage transition during early embryonic
development.
1. Introduction
The early development of animal embryos is guided by maternal
gene products. Then, the developmental control is transferred from
maternally supplied gene products to the zygotic genome. The maternal-
to-zygotic transition (MZT) is required for the acquisition of develop-
mental competence by cleavage-stage embryos. The MZT involves the
degradation of maternal transcripts and proteins that are unnecessary
for subsequent embryonic development, and the activation of zygotic
genes [1]. Zygotic genome activation (ZGA) is regulated in a precisely
timed manner, which varies among mammals. It mainly occurs at the
2-cell stage in mice, and at the 4-cell and 8-cell stages in humans [2].
Totipotency and further development of early embryos require the
establishment of specic gene expression patterns during ZGA [3].
Recently, several key regulators, such as RNA polymerase II [4] and
PRD-like homeobox transcription factors (TPRXs) [5], have been iden-
tied. Functional disruption of these regulators results in failure of ZGA
and arrest of early embryonic development in 2-cell stage embryos [4,
5].
Since the zygotic genome is inactive during the initial stages of early
embryonic development, post-translational modications (PTMs) of
proteins would appear to be particularly crucial. Various PTMs, such as
phosphorylation, methylation and ubiquitination, have been identied
to participate in the MZT [6]. Neddylation is another important
* Corresponding author. Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction of Ministry of Education, College of Animal Science and
Technology, Huazhong Agricultural University, No.1 Shizishan Street, Wuhan, 430070, PR China.
E-mail address: lax.pipi@mail.hzau.edu.cn (A. Liang).
1
These authors should be considered joint rst author.
Contents lists available at ScienceDirect
Theriogenology
journal homepage: www.theriojournal.com
https://doi.org/10.1016/j.theriogenology.2024.02.029
Received 1 October 2023; Received in revised form 27 February 2024; Accepted 27 February 2024
Theriogenology 220 (2024) 1–11
2
post-translational modication, involving the attachment of the
ubiquitin-like molecule NEDD8 to substrate proteins [7]. Like ubiquiti-
nation, neddylation is characterized by a series of enzymatic cascades
including NEDD8-activating enzyme (NAE) E1, NEDD8
ubiquitin-conjugating enzyme (UBC12) E2, and substrate-specic
NEDD8 ligase E3. Specically, NEDD8 is activated by NAE1 in an
ATP-dependent manner. Next, the activated NEDD8 is transferred to E2,
and then transferred to the targeted substrates by E3 [8]. Recent reports
indicate that neddylation is involved in the regulation of reproduction.
For example, neddylation deciency leads to defects in spermatogenesis
and sterility in male mice [9]. Disruption of neddylation in female mice
results in meiotic arrest in oocytes accompanied by the activation of the
spindle assembly checkpoint [10]. Importantly, inhibition of neddyla-
tion during the 4-cell to late 8-cell stage transition results in delayed
development of bovine embryos [11]. Further, Yang and colleagues
demonstrated that neddylation inhibition led to diminished embryo
quality in mice due to increased oxidative stress and reduced IL-1β levels
[12]. Despite these ndings, the precise role of neddylation in regulating
early embryonic development, especially regulating the MZT process,
remains unclear.
In this study, we used the specic neddylation inhibitor MLN4924 to
investigate the effects of neddylation on early murine embryonic
development. We also performed transcriptome analysis to uncover the
underlying mechanisms. The results showed that inhibition of neddy-
lation led to severe early arrest of embryonic development and MZT
defects. Moreover, the arrested embryos exhibited abnormal mito-
chondrial function, oxidative stress, and DNA damage.
2. Materials and methods
2.1. Animals and ethics statement
All SPF Kunming (KM) mice used in the experiments were obtained
from the Laboratory Animal Center of Huazhong Agricultural University
and housed in the SPF animal facility. All animal experiments were
approved by the Animal Ethics Committee of Huazhong Agricultural
University (HZAUMO-2022-0105).
2.2. Collection and culture of embryos
Female SPF KM mice (46 weeks old) were injected (i.p.) with 10 IU
of PMSG. After 48 h they were injected with 10 IU of hCG and mated
with adult males at a ratio of 1:1. Twenty hours later, the zygotes were
collected from the oviducts. Cumulus cells were disaggregated from
zygotes with 0.1% hyaluronidase (10176, Vitrolife, Sweden). Zygotes
were then cultured in G1 medium (10128, Vitrolife, Sweden) with or
without 0.1, 1, 10
μ
M of MLN4924 (S7109, Selleck, Houston, TX, USA)
dissolved by DMSO at 37 C in humidied atmosphere of 5% CO
2
.
2.3. Immunouorescence and confocal microscopy assay
Embryos were briey xed with PBS containing 4% para-
formaldehyde and 0.5% Triton X-100 for 50 min. After blocking in PBS
containing 2% BSA, 0.1% Tween-20, and 0.01% Triton X-100 for 1 h at
room temperature, xed embryos were then incubated with rabbit anti-
NEDD8 (1:100, ab81264, Abcam, Cambridge, UK) or rabbit anti-Phos-
pho-Histone H2AX antibody (1:100, 9718, CST, Boston, MA, USA)
overnight at 4 C. After washing, embryos were incubated with Cy3-
labelled goat anti-rabbit antibody (1:500, AS007, Abclonal, Wuhan,
China) at 37 C for 1 h. DNA of embryos was labelled with DAPI (C0065,
Solarbio, Beijing, China) for 10 min at room temperature. Finally, em-
bryos were transferred onto glass slides, cover slipped with Antifade
Mounting Medium (P0128S, Beyotime, Shanghai, China), and observed
under a Zeiss confocal laser scanning microscope (LSM 800, Germany).
2.4. Low input RNA sequencing
For RNA sequencing library preparation, three sets of samples
(twenty embryos per sample) were collected in the MLN4924 treatment
and control groups, respectively. The RNA of embryos was amplied by
the Smart-Seq 2 method [13] and reverse transcribed using an Oligo-dT
primer to synthesize rst-strand cDNA. The resulting cDNA was ampli-
ed by PCR, and the PCR products were puried with magnetic beads
before analysis with Qubit® 3.0 Flurometer and Agilent 2100 Bio-
analyzer to ensure the length of cDNA to be around 1~2 kb prior to
shearing ultrasonically. The cDNA was then sheared randomly by ul-
trasonic wave. The cDNA library was constructed by Illumina protocols
including DNA fragmentation, end repair, 3
ends A-tailing, adapter
ligation, PCR amplication, and library validation. Afterwards, the
quality of constructed cDNA library was determined using PerkinElmer
LabChip® GX Touch and Step OnePlus Real-Time PCR System.
Qualied libraries were then loaded onto Illumina Hiseq platform for
PE150 sequencing. The differentially expressed genes (DEGs) in the
comparison of treatment group and control group were identied using
DESeq2 according to the thresholds of |log2Ratio|≥ 1 and a q-value
<0.05.
2.5. RNA extraction and quantitative real-time PCR (qRT-PCR)
The RNA was extracted with RNAprep Pure Micro Kit according to
the manufacturers protocol (DP420, TIANGEN, Beijing, China), and
cDNA was then synthesized using the HiScript II Q RT SuperMix (R222-
01,Vazyme, Nanjing, China). The qRT-PCR was performed with ChamQ
Universal SYBR qPCR Master Mix (Q711-02/03, Vazyme, Nanjing,
China) by using Bio-Rad uorescence quantitative PCR instrument
(CFX96, Bio-Rad, USA). Specic primers were designed by Primer Pre-
mier 5.0 and listed in Supplementary Table S1. The qRT-PCR conditions
were as follows: 95 C for 1 min, followed by 40 cycles of 95 C for 10 s
and 60 C for 30 s. Relative gene expression was calculated by 2
ΔΔCt
method and normalized to Gapdh.
2.6. Ethynyl uridine (EU) detection
The EU assay Kit (C10316-1, RiboBio, Guangzhou, China) was used
to examine RNA transcriptional activity. Embryos were incubated with
EU (500
μ
M) in G1 medium for 2 h at 37 C. After washing with PBS,
embryos were xed in PBS containing 4% paraformaldehyde and 0.5%
Triton X-100 for 30 min. Then, embryos were stained with Apollo for 30
min at room temperature, and protected from light. Afterwards, em-
bryos were washed with PBS containing 0.1% Tween-20 and 0.01%
Triton X-100 three times for 10 min each. DNA was labelled with DAPI
for 10 min at room temperature. Finally, embryos were transferred onto
glass slides containing Antifade Mounting Medium (P0128S, Beyotime,
Shanghai, China) and observed with a Zeiss confocal laser scanning
microscope (LSM 800, Germany).
2.7. Mitochondrion staining
Mito-Tracker Red (C1049B, Beyotime, Shanghai, China) was applied
to evaluate mitochondrial localization in the embryos. After washing
with PBS, embryos were incubated in G1 medium containing 200 nM
Mito-Tracker at 37 C incubator for 30 min. After washing, the embryos
were transferred to a laser confocal Petri dish and observed under Zeiss
confocal laser scanning microscope (LSM 800, Germany).
2.8. ATP content assay
The levels of ATP were measured using an ATP assay Kit (S0026,
Beyotime, Shanghai, China) following the manufacturers instructions.
In brief, embryos were placed into black 96-well culture plates (Lysis
buffer, 50
μ
L/well). Next, 50
μ
L of ATP working solution was added.
M. Liu et al.
Theriogenology 220 (2024) 1–11
3
Finally, ATP concentration in embryos was detected using an EnSpire®
Multimode Reader (PerkinElmer, USA) in luminescence mode.
2.9. Mitochondrial membrane potential detection
The mitochondrial membrane potential (MMP) assay Kit (C2006,
Beyotime, Shanghai, China) with JC-1 was used to examine MMP. After
washing with PBS, embryos were cultured in G1 medium containing JC-
1 working solution for 20 min at 37 C. After washing, the embryos were
transferred to a laser confocal Petri dish and observed with uorescence
inverted microscope (IX73, Olympus, Japan).
2.10. Superoxide anion and ROS detection
A Dihydroethidium (DHE) Kit (S0063, Beyotime, Shanghai, China)
and a ROS Assay Kit (S0033S, Beyotime, Shanghai, China) were used to
determinate superoxide anion and ROS levels, respectively, in 2-cell
embryos according to the manufacturer protocols. Embryos per group
were incubated in G1 medium containing dihydroethidium (10
μ
M) or
DCFH-DA (10
μ
M) for 30 min at 37 C. After washing in PBS, the em-
bryos were transferred to a suitable vessel for observation under uo-
rescence inverted microscope (IX73, Olympus, Japan).
2.11. Statistical analysis
All statistical data from at least three independent experiments are
presented as mean ±SEM and the numbers of analyzed embryos are
labelled in gure legends as (n). All uorescence signal images were
quantied by ImageJ software. ANOVA and Students t-test were per-
formed to examine the difference among groups. P <0.05 was consid-
ered as statistically signicant.
3. Results
3.1. Inhibition of neddylation leads to early embryonic development arrest
NEDD8 localization was examined at different stages during early
embryonic development. We collected murine embryos at 42, 66, 80,
104, and 128 h post injection of hCG, which represent 2-cell, 4-cell, 8-
cell, morula, and blastocyst stages, respectively. Immunouorescence
staining showed that NEDD8 was mainly localized in the nucleus of 2-
cell and 4-cell stage embryos, and nuclear localization disappeared in
the later stages (Fig. 1A). MLN4924, a specic inhibitor of E1, which
suppresses the neddylation modication of proteins by forming an
adduct with NEDD8 [14], was employed to explore the roles of neddy-
lation during early embryonic development. We cultured zygotes with
different concentrations of MLN4924, and investigated embryonic
development at each stage. As shown in Fig. 1BC and S1, MLN4924
treatment signicantly inhibited early embryonic development. Treat-
ment of embryos with 1
μ
M or 10
μ
M MLN4924 blocked progression
from the 2-cell stage by approximately 50% in both treatment groups.
No signicant difference was observed in response to 0.1
μ
M MLN4924
compared to control. Notably, the developmental progress at the 8-cell
stage was signicantly arrested with both 1
μ
M and 10
μ
M MLN4924,
resulting in no morula and blastocyst formation. Considering the similar
effects of 1
μ
M and 10
μ
M MLN4924 on early embryonic development,
we selected 1
μ
M MLN4924 as the working concentration for the sub-
sequent analysis.
Importantly, the nuclear-specic localization of NEDD8 at the 2-cell
stage was lost after MLN4924 treatment (Fig. 1D), and the proportion of
embryos with abnormal NEDD8 localization was signicantly higher in
the MLN4924 treatment group than the control group (90.95 ±1.32%
vs 6.52 ±1.55%, P <0.001, Fig. 1E), suggesting that neddylation might
play an important role at the 2-cell stage.
3.2. Neddylation inhibition alters the transcriptome proles of 2-cell
embryos
To explore the underlying mechanism of neddylation inhibition on
early embryonic developmental arrest, we compared the transcriptome
of the MLN4924-treated 2-cell embryos with that of the control group.
Principal component analysis (PCA) showed a good inter-group sample
repeatability (Fig. 2A), which was supported by the sample-to-sample
Heatmap results (Fig. 2B). A total of 3959 DEGs were identied be-
tween the MLN4924 treatment and control groups, of which 1597 DEGs
were up-regulated (Fig. 2C, yellow dots), and 2362 DEGs were down-
regulated (Fig. 2C, blue dots). DEGs were found to be enriched in 3
categories of Gene Ontology (GO) terms related to cellular component,
biological process, and molecular function. Specically, the signicantly
enriched GO terms in each category mainly included RNA metabolic
process, mitochondrion organization, antioxidant activity, and tran-
scription regulator activity (Fig. 2D and Fig. S2). Kyoto Encyclopedia of
Genes and Genomes (KEGG) analysis further showed that the DEGs were
mainly enriched in pathways such as RNA transport, RNA polymerase,
Ribosome biogenesis in eukaryotes, mRNA surveillance, and Protea-
some (Fig. 2E). Furthermore, we observed that a total of 953 differen-
tially expressed transcription factors were identied between the
MLN4924 treatment and control groups, of which 338 transcription
factors were up-regulated and 615 transcription factors were down-
regulated (Fig. 2F). The protein families related to these differentially
expressed transcription factors are shown in Fig. 2G. Collectively, our
data strongly suggest that neddylation inhibition changes the gene
expression patterns associated with mRNA transcription in 2-cell
embryos.
3.3. Neddylation inhibition affects the ZGA process of 2-cell embryos
The MZT is one of the most important events in mouse 2-cell stage
embryos, comprising both degradation of maternal effect genes and
ZGA. As described above, the nuclear-specic localization of NEDD8 at
the 2-cell stage was lost after inhibition of neddylation (Fig. 1D), indi-
cating that nuclear gene expression might have been affected. KEGG
analysis showed that RNA polymerase, an enzyme involved in ZGA, was
enriched after neddylation inhibition (Fig. 2E). Notably, homeobox
transcription factors, as the key regulators of ZGA, were downregulated
after MLN4924 treatment (Fig. 2G). Thus, we speculated neddylation
inhibition adversely affects ZGA. To test our speculation, 2279 ZGA-
related genes were rst screened from different stages of mouse embryos
according to published RNA-seq data [15]. Venn analysis showed that
719 out of the 3959 DEGs identied in this study belonged to
ZGA-related genes (Fig. 3A), 31.5% of which were regulated by ned-
dylation. Furthermore, cluster analysis based on RNA-seq data showed
that the expression of typical ZGA transcripts was signicantly
decreased, except for the Taf9b gene (Fig. 3B). In addition, qRT-PCR
validation of decreased ZGA transcripts by qRT-PCR revealed results
consistent with transcriptomic results, suggesting the reliability of
transcriptomic data (Fig. 3C). Subsequently, EU staining was performed
to evaluate the effects of neddylation inhibition on transcriptional ac-
tivity. The results showed that the EU uorescence intensity was
signicantly lower in the MLN4924 treatment group than that in the
control group (116.7 ±6.88 vs 146.3 ±6.56, P <0.01, Fig. 3D and E).
Taken together, these results indicate that neddylation inhibition in-
duces developmental arrest in 2-cell embryos by disrupting transcrip-
tional activity.
3.4. Neddylation inhibition impairs degradation of maternal effect genes
in 2-cell embryos
To examine the effect of neddylation inhibition on the degradation of
maternal effect genes, 7661 maternal genes were screened from
different stages of mouse early embryonic development according to the
M. Liu et al.
Theriogenology 220 (2024) 1–11
4
Fig. 1. Inhibition of neddylation led to early embryonic development arrest. (A) NEDD8 localization in 2-cell embryos, 4-cell embryos, 8-cell embryos, morulae and
blastocysts. NEDD8, red; DNA, blue. Scale bar =20
μ
m. (B) Embryo morphologies after 1
μ
M MLN4924 treatment at the 2-cell stage. Scale bar =100
μ
m. (C) Rates of
different stages in early embryonic development after MLN4924 treatment (n =120 embryos per group). **P <0.01, ***P <0.001. (D) NEDD8 localization in 2-cell
embryos after MLN4924 treatment. NEDD8, red; DNA, blue. Scale bar =20
μ
m. (E) The proportion of embryos with abnormal NEDD8 localization in the control (n =
79 embryos) and MLN4924-treated (n =89 embryos) groups. ***P <0.001. (For interpretation of the references to color in this gure legend, the reader is referred to
the Web version of this article.)
M. Liu et al.
Theriogenology 220 (2024) 1–11
5
Fig. 2. Neddylation inhibition changed the transcriptome proles of 2-cell embryos. (A) The principle component analysis (PCA) analysis of 2-cell embryo RNA-seq
data comparing control with MLN4924-treated embryos. (B) The heatmap of differentially expressed genes (DEGs) comparing control with MLN4924-treated em-
bryos. (C) The volcano plot of DEGs comparing control with MLN4924-treated embryos. Down-regulated, blue; up-regulated, yellow. (D) GO analysis of DEGs
comparing control with MLN4924-treated embryos. (E) KEGG analysis of DEGs comparing control with MLN4924-treated embryos, the dot size represents the
enriched gene number, the larger the value, the greater the enrichment, and the dot color represents the adjusted p-value. (F) Differentially expressed transcription
factors comparing control with MLN4924-treated embryos. Down-regulated, yellow; up-regulated, dark blue. (G) The enriched protein family of differentially
expressed transcription factors. "C" represents "Control" and "T" represents "1
μ
M MLN4924 treatment". (For interpretation of the references to color in this gure
legend, the reader is referred to the Web version of this article.)
M. Liu et al.
Theriogenology 220 (2024) 1–11
6
Fig. 3. Neddylation inhibition affected the zygotic genome activation (ZGA) process of 2-cell embryos. (A) Venn analysis of the ZGA genes and DEGs. ZGA genes,
yellow; DEGs, blue. (B) The cluster analysis of DEGs related to ZGA. (C) The relative mRNA levels of some ZGA genes, Gapdh was set as the internal reference gene.
*P <0.05, **P <0.01. (D) Fluorescence image of EU staining in the 2-cell embryos after MLN4924 treatment. EU, red; DNA, blue. Scale bar =20
μ
m. (E) Fluo-
rescence intensity of EU in the control (n =81 embryos) and MLN4924 treatment (n =81 embryos) groups, **P <0.01. (For interpretation of the references to color
in this gure legend, the reader is referred to the Web version of this article.)
M. Liu et al.
Theriogenology 220 (2024) 1–11
7
published RNA-seq data [15]. Venn analysis results showed that 1442
out of the 3959 DEGs identied in our study are maternal-related genes
(Fig. 4A). Maternal genes fall into three main categories according to the
degradation period: genes whose degradation mainly occurs during the
transition of GV oocytes to the zygote stage (Cluster 1), genes whose
degradation mainly occurs from the zygote to the 2-cell stage (Cluster 2),
genes whose degradation mainly occurs from GV oocytes to the 2-cell
stage (Cluster 3) [15]. Venn analysis results showed that Clusters 1, 2,
and 3 exhibited 27, 483, and 49 overlapping genes, respectively, with
783 up-regulated maternal effect genes (Fig. 4B). According to the above
maternal gene classication, the 483 genes identied in Cluster 2
overlapped with the 783 up-regulated maternal effect genes that should
be degraded in 2-cell stage embryos. However, RNA-seq data showed
that the expression of the representative genes from Cluster 2 (483
genes) were increased after neddylation inhibition, indicating that these
genes were not degraded at the 2-cell stage embryos (Fig. 4C). Our
qRT-PCR results were consistent with transcriptomic data (Fig. 4D),
further conrming these ndings. Overall, our results suggest that in-
hibition of neddylation leads to developmental arrest in 2-cell stage
embryos by disrupting the degradation of maternal effect genes.
3.5. Neddylation inhibition causes mitochondrial dysfunction in 2-cell
embryos
Transcriptome results showed that neddylation inhibition induced
the downregulation of several mitochondrial-related genes (Fig. 5A),
except for the Dnm1l gene, thus potentially interfering with mitochon-
drial function. Therefore, we investigated the distribution of mito-
chondria in 2-cell embryos, and found that in the control embryos,
mitochondria were evenly distributed in the cytoplasm, while mito-
chondria were aggregated and clustered in the MLN4924-treated em-
bryos (Fig. 5B). The rate of abnormal distribution was signicantly
higher in the MLN4924-treated embryos than that in the control em-
bryos (71.6 ±6.17% vs 9.68 ±1.87%, P <0.001, Fig. 5C). Next, we
investigated the changes of mitochondrial membrane potential (MMP)
by JC-1 staining. As shown in Fig. 5DE, MMP was signicantly lower in
the MLN4924-treated embryos than that in the control embryos (P <
0.001). Moreover, the ATP content was signicantly lower in the
MLN4924-treated embryos compared to the control embryos (Fig. 5F, P
<0.05). These ndings suggest that neddylation inhibition results in
mitochondrial dysfunction.
Fig. 4. Neddylation inhibition impaired the degradation of maternal effect genes in 2-cell embryos. (A) Venn analysis of the DEGs and maternal genes. Maternal
genes, yellow; DEGs, blue. (B) Venn analysis of up-regulated maternal genes of DEGs and cluster 13 genes. (C) The cluster analysis of DEGs related to maternal
genes. (D) The relative mRNA levels of several maternal genes, Gapdh was set as the internal reference gene. *P <0.05, **P <0.01, ***P <0.001. (For interpretation
of the references to color in this gure legend, the reader is referred to the Web version of this article.)
M. Liu et al.
Theriogenology 220 (2024) 1–11
8
Fig. 5. Neddylation inhibition resulted in mitochondrial dysfunction in 2-cell embryos. (A) The cluster analysis of DEGs related to mitochondrial function. (B)
Fluorescence image of mitochondria staining after MLN4924 treatment. Mitochondria, red. Scale bar =20
μ
m. (C) The proportion of embryos with abnormal
mitochondria in the control (n =93 embryos) and MLN4924 treatment (n =89 embryos) groups. ***P <0.001. (D) Fluorescence image of JC-1 staining after
MLN4924 treatment. Scale bar =100
μ
m. (E) The uorescence intensity of Red/Green in the control (n =95 embryos) and MLN4924 treatment (n =114 embryos)
groups. ***P <0.001. (F) The changes of ATP content (fold change) in the control (n =90 embryos) and MLN4924 treatment (n =90 embryos) groups. *P <0.05.
(For interpretation of the references to color in this gure legend, the reader is referred to the Web version of this article.)
M. Liu et al.
Theriogenology 220 (2024) 1–11
9
3.6. Neddylation inhibition induces oxidative stress and DNA damage in
2-cell embryos
Mitochondrial dysfunction has been reported to induce ROS accu-
mulation. RNA-seq data showed that the expression of several genes
required for the oxidation-reduction process and oxidoreductase activity
was altered in the MLN4924 treatment group (Fig. 6A). Considering this,
we investigated whether ROS was elevated in 2-cell embryos upon
treatment with the neddylation inhibitor. As expected, uorescence
staining results showed that ROS levels were globally increased (P <
0.001, Fig. 6B and D). Moreover, our results indicated that superoxide
anion was also signicantly stronger in the MLN4924-treated embryos
than that in the control embryos (P <0.001, Fig. 6C and E). These results
suggest that neddylation inhibition induces oxidative stress in 2-cell
embryos.
Since the cluster analysis results indicated that the expression of
several DNA damage-related genes were changed (Fig. 6F), we specu-
lated that neddylation inhibition might cause DNA damage in 2-cell
embryos. To test this, we determined the levels of γH2AX immunou-
orescence as a measure of DNA damage in 2-cell embryos. As shown in
Fig. 6GH, strong γH2AX uorescence signals were observed in the
nucleus of MLN4924-treated embryos compared to the control group (P
<0.001), suggesting that neddylation inhibition results in DNA damage
in 2-cell embryos.
4. Discussion
Preimplantation embryonic development is a sensitive period sus-
ceptible to the adverse effects of various internal and external stimuli,
which can trigger developmental arrest. Precise regulation of gene
transcription and translation in early mouse embryonic development is
essential. Post-translational modications (PTMs) are critical for early
embryonic development because transcription of the zygote genome is
non-existent during these stages [6,16,17]. Neddylation is an important
ubiquitin-like PTM, which appears to be a good candidate as an
important player during preimplantation mouse embryo development.
The present study documents that the intracellular location of Nedd8
changes during early stages of embryonic development, implying that
neddylation may play an important role in regulating early embryonic
development in mice. Treatment with MLN4924, a specic inhibitor of
neddylation, resulted in the arrest of embryonic development in vitro,
conrming the idea that neddylation may play an important role in
regulating early embryonic development. Furthermore, transcriptome
analysis revealed that MLN4924 treatment altered gene expression
proles in 2-cell stage embryos. Bioinformatics analysis using GO and
KEGG jointly showed that the DEGs were mainly involved in tran-
scription regulator activity, RNA transport, and RNA polymerase, which
are related to the maternal-to-zygotic transition.
The maternal-to-zygotic transition consists of two important molec-
ular processes, the degradation of maternal effect genes and zygotic
genome activation (ZGA) [1]. In our study, the inhibition of neddylation
resulted in the loss of nuclear-specic localization of NEDD8 at the 2-cell
stage. Notably, we found that transcription factor activity, RNA poly-
merase, and homeobox transcription factor family, which have been
recently reported to participate in ZGA [3,4], were enriched after ned-
dylation inhibition. Combing Venn analysis and EU staining, we
conrmed that inhibition of neddylation adversely affects ZGA. On the
other hand, clearance of maternal mRNA is important for early embry-
onic development and impairment of this clearance process can lead to
developmental defects or even death of the embryo [18]. Our results
revealed that neddylation inhibition disrupted the degradation of
maternal effect genes, especially degradation of a portion of
ZGA-dependent maternal effect genes. As far as we know, this is the rst
study to reveal that neddylation inhibition can delay the MZT in the
mouse. Previously, Yang et al. demonstrated that neddylation inhibition
resulted in poor embryo quality and failure of blastocyst hatching [12].
In bovine embryos, MLN4924 treatment delayed initiation of embryonic
genome activation (EGA) as evidenced by the reduction of EGA markers
[11], results which indirectly support our ndings. Besides neddylation,
previously reports indicate that other PTMs including phosphorylation,
methylation, and ubiquitination are involved in the MZT [6], suggesting
that the MZT is regulated by several PTMs.
Mitochondria are vital for numerous physiological processes, such as
energy production, metabolite synthesis, calcium signaling, as well as
cell proliferation and death [19]. A recent study demonstrated that
neddylation is involved in the regulation of morphology, trafcking, and
function of mitochondria [20]. Herein, cluster analysis based on
RNA-seq data indicated that MLN4924 treatment downregulated the
expression of mitochondrial function-related genes. Moreover,
MLN4924 treatment altered the intracellular localization of mitochon-
dria and decreased the membrane potential of mitochondria and ATP
content. All the above data suggest that neddylation inhibition induces
mitochondrial dysfunction of 2-cell stage embryos. Previous studies
suggest that mitochondrial dysfunction adversely affects embryonic
development and ZGA [21]. Notably, our ndings indicate that neddy-
lation inhibition-induced ZGA impairment may be associated with
mitochondrial dysfunction. However, the exact effects and related
mechanisms of neddylation inhibition on the mitochondrial activity are
complicated and remain to be further investigated.
Mitochondria are the main sources of ROS in living cells. Appropriate
levels of ROS are required for oocyte maturation, fertilization, cell di-
vision and blastulation [22,23]. Oxidative stress occurs when there is an
imbalance between ROS production and consumption [24], and an in-
crease in ROS has been reported to impair preimplantation embryonic
development exhibiting development arrest at the 2-cell stage and a
signicant decrease in blastocyst formation [25]. Since neddylation in-
hibition led to abnormal mitochondrial function, we further examined
the intracellular oxidative stress. Our results showed that MLN4924
treatment caused signicantly higher levels of superoxide anion and
ROS, which were accompanied by altered expression levels of oxidative
stress-related genes. A recent study reports that inhibition of neddyla-
tion resulted in the activation of oxidative stress, poor mouse embryo
quality, and blastocyst hatching failure [12]. These results are consistent
with the possibility that neddylation inhibition impairs early embryonic
development through oxidative stress.
ROS can affect adversely genomic and mitochondrial DNA, thus
leading to various types of DNA damage. DNA damage in turn can affect
the integrity of the biological genome, activating complex DNA repair
mechanisms to avoid further DNA damage [26]. DNA double-strand
break (DSB) is one of the most severe form of DNA damage, capable
of causing genetic mutations and impairing embryo development. In this
study, elevated levels of γH2AX were observed in MLN4924-treated
embryos, suggesting neddylation inhibition resulted in DNA damage,
which could lead to arrest of early embryonic development. Jia et al.
reported that MLN4924 treatment triggered DNA damage response as
observed by the formation of γH2AX, ultimately resulting in senescence
in cancer cells [27]. Remarkably, ROS can induce intracellular DNA
damage, while DNA damage in return can elevate ROS levels. Therefore,
neddylation plays a complex, rather than a simple, regulatory role in
embryonic development.
5. Conclusion
Our ndings reveal multiple roles of neddylation in early embryonic
development. Specically, neddylation inhibition causes developmental
arrest of mouse preimplantation embryos by altering gene expression
and impairing the MZT. Moreover, neddylation inhibition results in
mitochondrial dysfunction, oxidative stress, and DNA damage. Further
studies should reveal how downstream target genes of Nedd8 are spe-
cically affected during the MZT.
M. Liu et al.
Theriogenology 220 (2024) 1–11
10
Fig. 6. Neddylation inhibition induced oxidative stress and DNA damage in 2-cell embryos. (A) The cluster analysis of DEGs related to oxidation-reduction process
and oxidoreductase activity. (B) Fluorescence image of ROS staining after MLN4924 treatment. ROS, green. Scale bar =50
μ
m. (C) Fluorescence image of superoxide
anion staining after MLN4924 treatment. Superoxide anion, red. Scale bar =100
μ
m. (D) The uorescence intensity of ROS in the control (n =81 embryos) and
MLN4924 treatment (n =85 embryos) groups. ***P <0.001. (E) The uorescence intensity of superoxide anion in the control (n =78 embryos) and MLN4924
treatment (n =80 embryos) groups. ***P <0.001. (F) The cluster analysis of DEGs related to DNA damage. (G) Fluorescence image of γH2AX staining after MLN4924
treatment. γH2AX, red; DNA, blue. Scale bar =20
μ
m. (H) The uorescence intensity of γH2AX in the control (n =68 embryos) and MLN4924 treatment (n =65
embryos) groups. ***P <0.001. (For interpretation of the references to color in this gure legend, the reader is referred to the Web version of this article.)
M. Liu et al.
Theriogenology 220 (2024) 1–11
11
Funding information:
Fundamental Research Funds for the Central Universities
(2662023DKPY001) and National Natural Science Foundation of China
(32072729). JSD is supported by a Senior Research Career Scientist
Award from the Department of Veterans Affairs.
Data availability
Data will be made available on request.
CRediT authorship contribution statement
Mingxiao Liu: Writing review & editing, Validation, Methodology,
Investigation, Formal analysis, Data curation. Zhiming Ding: Writing
original draft, Supervision, Methodology, Investigation, Conceptualiza-
tion. Peihao Sun: Project administration, Investigation. Shuo Zhou:
Investigation. Hanxiao Wu: Investigation. Lijun Huo: Resources,
Conceptualization. Liguo Yang: Supervision, Conceptualization. John
S. Davis: Supervision, Conceptualization. Aixin Liang: Writing review
& editing, Writing original draft, Supervision, Funding acquisition.
Declaration of competing interest
The authors declare that they have no known competing nancial
interests or personal relationships that could have appeared to inuence
the work reported in this paper.
Appendix A. Supplementary data
Supplementary data to this article can be found online at https://doi.
org/10.1016/j.theriogenology.2024.02.029.
References
[1] Li L, Lu X, Dean J. The maternal to zygotic transition in mammals. Mol Aspect Med
2013;34:91938. https://doi.org/10.1016/j.mam.2013.01.003.
[2] Xue Z, Huang K, Cai C, Cai L, Jiang CY, Feng Y, Liu Z, Zeng Q, Cheng L, Sun YE,
Liu JY, Horvath S, et al. Genetic programs in human and mouse early embryos
revealed by single-cell RNA sequencing. Nature 2013;500:5937. https://doi.org/
10.1038/nature12364.
[3] Dang Y, Li S, Zhao P, Xiao L, Wang L, Shi Y, Luo L, Wang S, Wang H, Zhang K. The
lysine deacetylase activity of histone deacetylases 1 and 2 is required to safeguard
zygotic genome activation in mice and cattle. Development 2022;149:dev200854.
https://doi.org/10.1242/dev.200854.
[4] Liu B, Xu Q, Wang Q, Feng S, Lai F, Wang P, Zheng F, Xiang Y, Wu J, Nie J, Qiu C,
Xia W, et al. The landscape of RNA Pol II binding reveals a stepwise transition
during ZGA. Nature 2020;587:13944. https://doi.org/10.1038/s41586-020-
2847-y.
[5] Zou Z, Zhang C, Wang Q, Hou Z, Xiong Z, Kong F, Wang Q, Song J, Liu B, Liu B,
Wang L, Lai F, et al. Translatome and transcriptome co-proling reveals a role of
TPRXs in human zygotic genome activation. Science 2022;378:abo7923. https://
doi.org/10.1126/science.abo7923.
[6] Liu C, Ma Y, Shang Y, Huo R, Li W. Post-translational regulation of the maternal-to-
zygotic transition. Cell Mol Life Sci 2018;75:170722. https://doi.org/10.1007/
s00018-018-2750-y.
[7] Dubiel W, Chaithongyot S, Dubiel D, Naumann M. The COP9 signalosome: a multi-
DUB complex. Biomolecules 2020;10:1082. https://doi.org/10.3390/
biom10071082.
[8] Mendoza HM, Shen LN, Botting C, Lewis A, Chen J, Ink B, Hay RT. NEDP1, a highly
conserved cysteine protease that deNEDDylates Cullins. J Biol Chem 2003;278:
2563743. https://doi.org/10.1074/jbc.M212948200.
[9] Huang G, Kaufman AJ, Ryan RJH, Romin Y, Huryn L, Bains S, Manova-Todorova K,
Morris PL, Hunnicutt GR, Adelman CA, Petrini JHJ, Ramanathan Y, et al. Mouse
DCUN1D1 (SCCRO) is required for spermatogenetic individualization. PLoS One
2019;14:e0209995. https://doi.org/10.1371/journal.pone.0209995.
[10] Yang M, Jin Y, Fan S, Liang X, Jia J, Tan Z, Huang T, Li Y, Ma T, Li M. Inhibition of
neddylation causes meiotic arrest in mouse oocyte. Cell Cycle 2019;18:125467.
https://doi.org/10.1080/15384101.2019.1617453.
[11] Kinterova V, Kanka J, Petruskova V, Toralova T. Inhibition of Skp1-Cullin-F-box
complexes during bovine oocyte maturation and preimplantation development
leads to delayed development of embryos. Biol Reprod 2019;100:896906. https://
doi.org/10.1093/biolre/ioy254.
[12] Yang G, Chen J, He Y, Luo H, Yuan H, Chen L, Huang L, Mao F, Hu S, Qian Y,
Miao C, Feng R. Neddylation inhibition causes impaired mouse embryo quality and
blastocyst hatching failure through elevated oxidative stress and reduced IL-1β.
Front Immunol 2022;13:925702. https://doi.org/10.3389/mmu.2022.925702.
[13] Picelli S, Bj¨
orklund ÅK, Faridani OR, Sagasser S, Winberg G, Sandberg R. Smart-
seq2 for sensitive full-length transcriptome proling in single cells. Nat Methods
2013;10:10968. https://doi.org/10.1038/nmeth.2639.
[14] Nawrocki ST, Grifn P, Kelly KR, Carew JS. MLN4924: a novel rst-in-class
inhibitor of NEDD8-activating enzyme for cancer therapy. Expet Opin Invest Drugs
2012;21:156373. https://doi.org/10.1517/13543784.2012.707192.
[15] Sha QQ, Zhu YZ, Li S, Jiang Y, Chen L, Sun XH, Shen L, Ou XH, Fan HY.
Characterization of zygotic genome activation-dependent maternal mRNA
clearance in mouse. Nucleic Acids Res 2020;48:87994. https://doi.org/10.1093/
nar/gkz1111.
[16] Wu Y, Li M, Yang M. Post-translational modications in oocyte maturation and
embryo development. Front Cell Dev Biol 2021;9:645318. https://doi.org/
10.3389/fcell.2021.645318.
[17] Beaujean N. Histone post-translational modications in preimplantation mouse
embryos and their role in nuclear architecture. Mol Reprod Dev 2014;81:10012.
https://doi.org/10.1002/mrd.22268.
[18] Wu D, Dean J. BTG4, a maternal mRNA cleaner. J Mol Cell Biol 2016;8:36970.
https://doi.org/10.1093/jmcb/mjw031.
[19] Akbari M, Kirkwood TBL, Bohr VA. Mitochondria in the signaling pathways that
control longevity and health span. Ageing Res Rev 2019;54:100940. https://doi.
org/10.1016/j.arr.2019.100940.
[20] Zhou Q, Zheng Y, Sun Y. Neddylation regulation of mitochondrial structure and
functions. Cell Biosci 2021;11:55. https://doi.org/10.1186/s13578-021-00569-6.
[21] Li Y, Mei NH, Cheng GP, Yang J, Zhou LQ. Inhibition of DRP1 impedes zygotic
genome activation and preimplantation development in mice. Front Cell Dev Biol
2021;9:788512. https://doi.org/10.3389/fcell.2021.788512.
[22] Ristow M, Schmeisser K. Mitohormesis: promoting health and lifespan by increased
levels of reactive oxygen species (ROS). Dose Response 2014;12:288341. https://
doi.org/10.2203/dose-response.13-035.Ristow.
[23] Jamil M, Debbarh H, Aboulmaouahib S, Aniq Filali O, Mounaji K, Zarqaoui M,
Saadani B, Louanjli N, Cadi R. Reactive oxygen species in reproduction: harmful,
essential or both? Zygote 2020;28:25569. https://doi.org/10.1017/
S0967199420000179.
[24] Cie´
slar-Pobuda A, Yue J, Lee HC, Skonieczna M, Wei YH. ROS and oxidative stress
in stem cells. Oxid Med Cell Longev 2017;2017:5047168. https://doi.org/
10.1155/2017/5047168.
[25] Kuscu N, Gungor-Ordueri NE, Sozen B, Adiguzel D, Celik-Ozenci C. FoxO
transcription factors 1 regulate mouse preimplantation embryo development.
J Assist Reprod Genet 2019;36:212133. https://doi.org/10.1007/s10815-019-
01555-1.
[26] Winship AL, Stringer JM, Liew SH, Hutt KJ. The importance of DNA repair for
maintaining oocyte quality in response to anti-cancer treatments, environmental
toxins and maternal ageing. Hum Reprod Update 2018;24:11934. https://doi.
org/10.1093/humupd/dmy002.
[27] Jia L, Li H, Sun Y. Induction of p21-dependent senescence by an NAE inhibitor,
MLN4924, as a mechanism of growth suppression. Neoplasia 2011;13:5619.
https://doi.org/10.1593/neo.11420.
M. Liu et al.
ResearchGate has not been able to resolve any citations for this publication.
Article
Full-text available
Mammalian blastocyst hatching is an essential prerequisite for successful embryo implantation. As the rate-limiting step of current assisted reproductive technology, understanding the key factors regulating blastocyst hatching would be significantly helpful to improve the performance of the assisted reproductive practice. In early embryo development, the fine-tuned elimination of maternal materials and the balanced protein turnover are inevitable for the competent to hatch and implant into endometrium. Neddylation, a ubiquitination-like protein modification, has been shown to be involved in oocyte maturation and early embryo development. In this study, aiming to discover an unknown role of neddylation in the blastocyst hatching process, we provided functional evidence of neddylation in mammalian embryo quality and blastocyst hatching. Treatment with MLN4924, a specific neddylation inhibitor, lowered the embryo quality and dramatically reduced the hatching rate in mouse blastocysts. The transcriptional profile showed the upregulation of oxidative stress-related genes and aberrant expression of immune-related genes. The elevated oxidative stress was validated by qPCR and markers of apoptosis, DNA damage, reactive oxygen species, and cytoskeleton. Moreover, we found the secreted IL-1β level was reduced in an NF-κB-independent manner, leading to the final poor embryo quality and blastocyst hatching failure. This is the first report of neddylation being of great importance in the mammalian blastocyst hatching process. Further investigations uncovering more detailed molecular mechanisms of neddylation regulation in blastocyst hatching would greatly promote not only the understanding of this crucial biological process but also the clinical application in reproductive centers.
Article
Full-text available
The genome is transcriptionally inert at fertilization and must be activated through a remarkable developmental process called zygotic genome activation (ZGA). Epigenetic reprogramming contributes significantly to the dynamic gene expression during ZGA, however the mechanism has yet to be resolved. Here, we find histone deacetylase 1 and 2 (HDAC1/2) can regulate ZGA through the lysine deacetylase activity. Notably, in mouse embryos, overexpression of HDAC1/2 dominant negative mutant leads to a developmental arrest at 2-cell stage. RNA-seq reveals that 64% of down-regulated genes are ZGA genes and 49% of up-regulated genes are developmental genes. Inhibition of the deacetylase activity of HDAC1/2 causes a failure of histone deacetylation at multiple sites including H4K5, H4K16, H3K14, H3K18, and H3K27. ChIP-seq analysis exhibits an increase and decrease of H3K27ac enrichment at promoters of up- and down-regulated genes, respectively. Moreover, HDAC1 mutants prohibited the removal of H3K4me3 via impeding KDM5s. Importantly, the developmental block can be greatly rescued through Kdm5b injection and expression of the majority of dysregulated genes partially corrected. Similar functional significance of HDAC1/2 is conserved in cattle embryos. Overall, we propose that HDAC1/2 is indispensable for ZGA via creating correct transcriptional repressive and active states in mouse and bovine embryos.
Article
Full-text available
Mitochondrion plays an indispensable role during preimplantation embryo development. Dynamic-related protein 1 (DRP1) is critical for mitochondrial fission and controls oocyte maturation. However, its role in preimplantation embryo development is still lacking. In this study, we demonstrate that inhibition of DRP1 activity by mitochondrial division inhibitor-1, a small molecule reported to specifically inhibit DRP1 activity, can cause severe developmental arrest of preimplantation embryos in a dose-dependent manner in mice. Meanwhile, DRP1 inhibition resulted in mitochondrial dysfunction including decreased mitochondrial activity, loss of mitochondrial membrane potential, reduced mitochondrial copy number and inadequate ATP by disrupting both expression and activity of DRP1 and mitochondrial complex assembly, leading to excessive ROS production, severe DNA damage and cell cycle arrest at 2-cell embryo stage. Furthermore, reduced transcriptional and translational activity and altered histone modifications in DRP1-inhibited embryos contributed to impeded zygotic genome activation, which prevented early embryos from efficient development beyond 2-cell embryo stage. These results show that DRP1 inhibition has potential cytotoxic effects on mammalian reproduction, and DRP1 inhibitor should be used with caution when it is applied to treat diseases. Additionally, this study improves our understanding of the crosstalk between mitochondrial metabolism and zygotic genome activation.
Article
Full-text available
Mammalian oocyte maturation and embryo development are unique biological processes regulated by various modifications. Since de novo mRNA transcription is absent during oocyte meiosis, protein-level regulation, especially post-translational modification (PTM), is crucial. It is known that PTM plays key roles in diverse cellular events such as DNA damage response, chromosome condensation, and cytoskeletal organization during oocyte maturation and embryo development. However, most previous reviews on PTM in oocytes and embryos have only focused on studies of Xenopus laevis or Caenorhabditis elegans eggs. In this review, we will discuss the latest discoveries regarding PTM in mammalian oocytes maturation and embryo development, focusing on phosphorylation, ubiquitination, SUMOylation and Poly(ADP-ribosyl)ation (PARylation). Phosphorylation functions in chromosome condensation and spindle alignment by regulating histone H3, mitogen-activated protein kinases, and some other pathways during mammalian oocyte maturation. Ubiquitination is a three-step enzymatic cascade that facilitates the degradation of proteins, and numerous E3 ubiquitin ligases are involved in modifying substrates and thus regulating oocyte maturation, oocyte-sperm binding, and early embryo development. Through the reversible addition and removal of SUMO (small ubiquitin-related modifier) on lysine residues, SUMOylation affects the cell cycle and DNA damage response in oocytes. As an emerging PTM, PARlation has been shown to not only participate in DNA damage repair, but also mediate asymmetric division of oocyte meiosis. Each of these PTMs and external environments is versatile and contributes to distinct phases during oocyte maturation and embryo development.
Article
Full-text available
Mitochondria are the powerhouse of a cell. The structure and function of mitochondria are precisely regulated by multiple signaling pathways. Neddylation, a post-translational modification, plays a crucial role in various cellular processes including cellular metabolism via modulating the activity, function and subcellular localization of its substrates. Recently, accumulated data demonstrated that neddylation is involved in regulation of morphology, trafficking and function of mitochondria. Mechanistic elucidation of how mitochondria is modulated by neddylation would further our understanding of mitochondrial regulation to a new level. In this review, we first briefly introduce mitochondria, then neddylation cascade, and known protein substrates subjected to neddylation modification. Next, we summarize current available data of how neddylation enzymes, its substrates (including cullins/Cullin-RING E3 ligases and non-cullins) and its inhibitor MLN4924 regulate the structure and function of mitochondria. Finally, we propose the future perspectives on this emerging and exciting field of mitochondrial research.
Article
Full-text available
Zygotic genome activation (ZGA) is the first transcription event in life¹. However, it is unclear how RNA polymerase is engaged in initiating ZGA in mammals. Here, by developing small-scale Tn5-assisted chromatin cleavage with sequencing (Stacc–seq), we investigated the landscapes of RNA polymerase II (Pol II) binding in mouse embryos. We found that Pol II undergoes ‘loading’, ‘pre-configuration’, and ‘production’ during the transition from minor ZGA to major ZGA. After fertilization, Pol II is preferentially loaded to CG-rich promoters and accessible distal regions in one-cell embryos (loading), in part shaped by the inherited parental epigenome. Pol II then initiates relocation to future gene targets before genome activation (pre-configuration), where it later engages in full transcription elongation upon major ZGA (production). Pol II also maintains low poising at inactive promoters after major ZGA until the blastocyst stage, coinciding with the loss of promoter epigenetic silencing factors. Notably, inhibition of minor ZGA impairs the Pol II pre-configuration and embryonic development, accompanied by aberrant retention of Pol II and ectopic expression of one-cell targets upon major ZGA. Hence, stepwise transition of Pol II occurs when mammalian life begins, and minor ZGA has a key role in the pre-configuration of transcription machinery and chromatin for genome activation.
Article
Full-text available
The COP9 signalosome (CSN) is a signaling platform controlling the cellular ubiquitylation status. It determines the activity and remodeling of ~700 cullin-RING ubiquitin ligases (CRLs), which control more than 20% of all ubiquitylation events in cells and thereby influence virtually any cellular pathway. In addition, it is associated with deubiquitylating enzymes (DUBs) protecting CRLs from autoubiquitylation and rescuing ubiquitylated proteins from degradation. The coordination of ubiquitylation and deubiquitylation by the CSN is presumably important for fine-tuning the precise formation of defined ubiquitin chains. Considering its intrinsic DUB activity specific for deneddylation of CRLs and belonging to the JAMM family as well as its associated DUBs, the CSN represents a multi-DUB complex. Two CSN-associated DUBs, the ubiquitin-specific protease 15 (USP15) and USP48 are regulators in the NF-κB signaling pathway. USP15 protects CRL1β-TrCP responsible for IκBα ubiquitylation, whereas USP48 stabilizes the nuclear pool of the NF-κB transcription factor RelA upon TNF stimulation by counteracting CRL2SOCS1. Moreover, the CSN controls the neddylation status of cells by its intrinsic DUB activity and by destabilizing the associated deneddylation enzyme 1 (DEN1). Thus, the CSN is a master regulator at the intersection between ubiquitylation and neddylation.
Article
Full-text available
An important event of the maternal-to-zygotic transition (MZT) in animal embryos is the elimination of a subset of the maternal transcripts that accumulated during oogenesis. In both invertebrates and vertebrates, a maternally encoded mRNA decay pathway (M-decay) acts before zygotic genome activation (ZGA) while a second pathway, which requires zygotic transcription, subsequently clears additional mRNAs (Z-decay). To date the mechanisms that activate the Z-decay pathway in mammalian early embryos have not been investigated. Here, we identify murine maternal transcripts that are degraded after ZGA and show that inhibition of de novo transcription stabilizes these mRNAs in mouse embryos. We show that YAP1-TEAD4 transcription factor-mediated transcription is essential for Z-decay in mouse embryos and that TEAD4-triggered zygotic expression of terminal uridylyltransferases TUT4 and TUT7 and mRNA 3'-oligouridylation direct Z-decay. Components of the M-decay pathway, including BTG4 and the CCR4-NOT deadenylase, continue to function in Z-decay but require reinforcement from the zygotic factors for timely removal of maternal mRNAs. A long 3'-UTR and active translation confer resistance of Z-decay transcripts to M-decay during oocyte meiotic maturation. The Z-decay pathway is required for mouse embryo development beyond the four-cell stage and contributes to the developmental competence of preimplantation embryos.
Article
Translational regulation plays a critical role during the oocyte-to-embryo transition (OET) and zygotic genome activation (ZGA). Here, we integrated ultra-low-input Ribo-seq with mRNA-seq to co-profile the translatome and transcriptome in human oocytes and early embryos. Comparison with mouse counterparts identified widespread differentially translated genes functioning in epigenetic reprogramming, transposon defense, and small RNA biogenesis, in part driven by species-specific regulatory elements in 3′ untranslated regions. Moreover, PRD-like homeobox transcription factors, including TPRXL, TPRX1, and TPRX2, are highly translated around ZGA. TPRX1/2/L knockdown leads to defective ZGA and preimplantation development. Ectopically expressed TPRXs bind and activate key ZGA genes in human embryonic stem cells. These data reveal the conservation and divergence of translation landscapes during OET and identify critical regulators of human ZGA.
Article
The process of embryonic development is crucial and radically influences preimplantation embryo competence. It involves oocyte maturation, fertilization, cell division and blastulation and is characterized by different key phases that have major influences on embryo quality. Each stage of the process of preimplantation embryonic development is led by important signalling pathways that include very many regulatory molecules, such as primary and secondary messengers. Many studies, both in vivo and in vitro , have shown the importance of the contribution of reactive oxygen species (ROS) as important second messengers in embryo development. ROS may originate from embryo metabolism and/or oocyte/embryo surroundings, and their effect on embryonic development is highly variable, depending on the needs of the embryo at each stage of development and on their environment ( in vivo or under in vitro culture conditions). Other studies have also shown the deleterious effects of ROS in embryo development, when cellular tissue production overwhelms antioxidant production, leading to oxidative stress. This stress is known to be the cause of many cellular alterations, such as protein, lipid, and DNA damage. Considering that the same ROS level can have a deleterious effect on the fertilizing oocyte or embryo at certain stages, and a positive effect at another stage of the development process, further studies need to be carried out to determine the rate of ROS that benefits the embryo and from what rate it starts to be harmful, this measured at each key phase of embryonic development.