ArticlePDF Available

Long non-coding MEG3 is a marker for skeletal muscle development and meat production traits in pigs

Authors:
Long non-coding MEG3 is a marker for skeletal muscle development
and meat production traits in pigs
X. Yu*
1
, Z. Wang
1
, H. Sun*, Y. Yang*, K. Li* and Z. Tang*
*Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing 100193, China.
Group of Pig Genomic Design and
Breeding, Agricultural Genome Institute at Shenzhen, Chinese Academy of Agricultural Sciences, Shenzhen 518124, China.
Department of
Computer Science, City University of Hong Kong, Tat Chee Avenue, Kowloon, Hong Kong.
Summary Long non-coding RNA maternally expressed gene 3 (lncRNA MEG3) plays an important role
in mammalian muscle development. Our previous transcriptome study showed that lncRNA
MEG3 is differentially expressed during postnatal skeletal muscle development in pigs. The
objective of the present study was to analyse the role of lncRNA MEG3 in prenatal and
postnatal skeletal muscle development and investigate the association of MEG3 with meat
production traits in pigs. We investigated the sequence conservation and temporal-spatial
expression of lncRNA MEG3 and identified its core promoter and single nucleotide
polymorphisms (SNPs). Our results show that MEG3 is conserved among pig, human and
mouse and is expressed in a tissue-specific manner with high expression levels in kidney and
leg and dorsal muscles. In addition, MEG3 is more abundant in prenatal muscle compared
to postnatal muscle, and its expression peaks at gestational day 60. Notably, we observed
almost no expression 40 days after birth. The core promoter of MEG3 is located upstream of
the transcription initiation site between 447 and 40 bp. In our SNP linkage
disequilibrium and association analyses, four of the 10 potential polymorphism sites were
found to be associated with corrected back fat thickness and age to reach 100 kg
(rs325797437, rs344501106, rs81286029 and rs318656749). In addition, three
haplotypes were found to be associated with differences in corrected age to reach 100 kg
(AAAT, AAAT/GGGC, GAAT/GGGC). Our results indicate that MEG3 regulates skeletal
muscle development and is a candidate gene for improving meat production traits in pigs.
Keywords lncRNA, maternally expressed gene 3, swine, association analysis, muscle
growth
Introduction
Advances in high-throughput sequencing platforms have
revealed an astounding number of noncoding RNAs that
play critical roles in gene regulation and developmental
processes (Mattick 2004). Long noncoding RNAs (lncRNAs)
are a class of RNA molecules of more than 200 nucleotides
that lack protein-coding capacity. Many lncRNAs, including
H19 (Kallen et al. 2013), linc-MD1 (Legnini et al. 2014),
lncmyod (Gong et al. 2015), lnc-31 (Ballarino et al. 2015),
linc-YY1 (Zhou et al. 2015) and lnc-mg (Zhu et al. 2017),
play crucial roles in skeletal muscle development.
Maternally expressed gene 3 (MEG3), a lncRNA first
identified in mice, is the orthologue of the trap locus 2
(GTL2) gene (Schuster-Gossler et al. 1998). The human
orthologue of MEG3,DLK1/MEG3, is located on chromo-
some 14q32.3 (da Rocha et al. 2008). The pig MEG3 gene,
located on chromosome 7, encodes for multiple alterna-
tively spliced transcripts and is expressed from the mater-
nally inherited chromosome. Although the role of MEG3 in
regulating autophagy in cancer is known (Wang et al.
2012), the function of MEG3 in livestock is unclear. MEG3
is involved in callipyge, a muscle hypertrophy phenotype in
sheep that causes significant changes in muscle develop-
ment, carcass composition, shape and meat quality
(Koohmaraie et al. 1995; Freking et al. 1998, 1999). These
studies imply that MEG3 is associated with livestock meat
production traits.
The pig is an important protein source for humans, and
meat production traits are economically significant traits.
Although development, growth and function of skeletal
Address for correspondence
Z. Tang, Institute of Animal Sciences, Chinese Academy of Agricultural
Sciences, Beijing 100193, China.
E-mail: tangzhonglin@caas.cn
1
These authors contributed equally to this work.
Accepted for publication 26 June 2018
doi: 10.1111/age.12712
571
©2018 Stichting International Foundation for Animal Genetics, 49, 571–578
muscle are dynamic processes critical to animal survival
(Sollero et al. 2011), the molecular mechanisms of skeletal
muscle development and meat production traits are largely
unknown. The role of lncRNA MEG3 in skeletal muscle
development and meat production traits in pigs is unde-
termined.
Previously, we performed comprehensive profiling of Sus
scrofa lncRNAs across nine organs and skeletal muscles
collected at three developmental stages. We identified 1405
lncRNAs that were differentially expressed during postnatal
skeletal muscle development (Tang et al. 2017). Of these,
MEG3, which shares homology with human and mouse,
was differentially expressed in postnatal skeletal muscle
development. To further explore the potential functions of
MEG3 in pigs, we investigated the sequence conservation
and temporal-spatial expression and identified the core
promoter and single nucleotide polymorphisms (SNPs) of
lncRNA MEG3. Our study indicates that lncRNA MEG3 is
highly conserved across mammals and is involved in
skeletal muscle development. We conclude from these
studies that lncRNA MEG3 is a valid target for improving
meat production traits in pigs.
Materials and methods
Animal and trait data collection
All animal procedures were performed according to the
guidelines of the Biological Studies Animal Care and Use
Committee, P. R. China. Longissimus dorsi muscles were
isolated from foetal Large White pigs on gestational days 33,
60, 65, 70, 75, 80, 95 and 105 (named E33, E60, E65, E70,
E75, E80, E95 and E105 respectively) and on postnatal days
20, 40, 60, 100, 140 and 160 (named D20, D40, D60, D100,
D140 and D160 respectively). Muscle samples collected at
the 14 different time points were used for expression
analysis. Each time point included independent biological
replicates from at least three individuals. In addition, nine
tissuesheart, liver, spleen, lung, kidney, intestine, stom-
ach, and leg and dorsal musclesfrom three Large White
pigs at D160 were harvested. All samples were stored
immediately in liquid nitrogen for RNA extraction.
For SNP linkage disequilibrium and association studies,
blood samples were collected from 297 independent indi-
viduals from one farm in the Shunyi district of Beijing.
Traits of the 297 Large White pigs, including body weight,
age, back fat thickness, eye muscle area, piglet birth weight
and weaned piglets, were recorded. The following indicators
were corrected based on standard methods used in China
(GB 22283-2008), as follows:
Corrected back fat thickness ¼back fat thickness CF
Corrected age to reach 100 kg
¼measured age ½ðmeasured weight 100Þ=CF;
where CF is the correction factor for each trait.
Total RNA extraction, reverse transcription PCR and
real-time quantitative PCR (RT-qPCR)
Total RNA was extracted with TRIzol Reagent (Invitro-
gen). Total RNA concentration was determined using
spectrophotometry to ensure that the OD260/OD280
ratio was between 1.8 and 2.0. The integrity of the RNA
was assessed by gel electrophoresis. Total RNA was
reverse transcribed using a RevertAid First Strand cDNA
Synthesis Kit (ThermoFisher), according to the manu-
facturer’s instructions. RT-PCR was performed using
routine PCR programs (T
m
=60 °C) with 35 amplifica-
tion cycles. The RT-qPCR reaction was performed on a
7500 FAST Real-Time PCR System (Applied Biosystems)
accordingtotheSYBR
â
Premix Ex Taq
TM
instructions.
All reactions were replicated three times. Porcine glyc-
eraldehyde-3-phosphate dehydrogenase (GAPDH)wasused
as an endogenous control. The expression levels of all
genes of interest were normalized to those of GAPDH
using the 2
ΔΔCt
method. All primer sequences were
designed using PRIMER 5.0 software and are listed in
Table S1.
Sequence conservation and core promoter analysis
Based on our previous transcriptome data (Gene Expres-
sion Omnibus database, GSE73763), we analysed gene
conservation across human, mouse, cow, opossum and
S. scrofa using the UCSC Comparative Genomics pipeline
(Washietl et al. 2014; Rosenbloom et al. 2015). We also
determined lncRNA sequences using pairwise alignment
with the S. scrofa 10.2 genome build to eliminate the
effects of genome assembly. We restricted the conservation
analysis of lncRNA to human or mouse by requiring that
50% of the nucleotides uniquely intersect with an align-
ment in the chain file. The conserved local secondary
structure of MEG3 was analysed via the Rfam model
(http://rfam.xfam.org/family/RF01872/alignment/html).
Primers were designed to amplify different segments of
MEG3 (Table S1) according to the 50flanking sequence
region of porcine lncRNA MEG3 in GenBank. Potential
transcription factors were predicted by TFSEARCH soft-
ware (available online: http://www.cbrc.jp/research/
db/TFSEARCH.html).
To identify the active promoter regions and determine
the DNA elements regulating lncRNA MEG3 expression,
we amplified eight short segments of different lengths
(2566, 2127, 1736, 1372, 951, 533, 217 and 126 bp)
using PCR. Recombinant vectors containing the amplified
segments, named PG-MEG3-1, PGL-MEG3-2, PGL-MEG3-
3, PG-MEG3-4, PG-MEG3-5, PG-MEG3-6, PG-MEG3-7
and PG-MEG3-8, were transfected into porcine kidney
cells (PK15) and premature intestinal epithelial cells
(PIEC). We detected changes in dual luciferase activity
48 h after transfection.
©2018 Stichting International Foundation for Animal Genetics, 49, 571–578
Yu et al.572
SNP identification
Potential polymorphism sites in lncRNA MEG3 were
identified by sequencing PCR products from 30 mixed Large
White pigs. Whole genomic DNA of lncRNA MEG3 was
amplified by PCR with four primer pairs, named segment 1
4 (Table S1). The SNP sites were analysed by Beijing
Compass Biotechnology Co., Ltd. using mass spectrometry.
The primer pairs for genotyping the SNPs are displayed in
Table S1.
Statistical analyses
To compare gene frequencies, a chi-square analysis was
performed using Rversion 3.3.1 software (https://www.r-
project.org/). The meat production traits were analysed
using the GENERALIZED LINEAR MODELS (GLM) program within
R, according to the following linear mixed model (Ma et al.
2008):
Y¼XbþZb þe;
where Yrepresents the phenotype value, Xis the fixed
effects matrix for phenotype value, bis the fixed-effect
coefficients including two sexes (coded as 1 for male and 2
for female) and two parities (coded as 1 for primiparity and
2 for multiparity), Zis the design matrix for the random
effects of observations, bis the vector of random-effect
coefficients and ɛis the vector of errors for observations.
We also used HAPLOVIEW version 4.2 software with default
settings (Barrett et al. 2005) to analyse linkage disequilib-
rium of the regions with multiple significant SNPs clustered
around the peak SNP. A haplotype association study (Druet
& Farnir 2011) was also performed to identify genomic
regions associated with the correction of back fat thickness
value and correction of age to reach 100 kg value in 297
individuals.
Results
Sequence conservation among human, mouse and pig
We identified 10 813 putative lncRNAs from our previous
RNA-seq data in pigs. We analysed the pairwise alignments
between pig and four other species to explore the conser-
vation of lncRNAs. As shown in Fig. 1a, a large portion of
pig lncRNAs (>70%) aligned with closely related species
(cow, human and mouse), but the ratio dramatically
decreased (30%) when compared to distant species (opos-
sum). We found fewer conserved lncRNAs between pig and
cow compared to the number of conserved lncRNAs
between human or mouse and pig, although according to
the overall conservation of lncRNAs, cow is more closely
aligned with pig. We identified MEG3-like lncRNAs and
analysed their secondary structures, as shown in Fig. 1b.
Bases shown in light green are conserved among pig,
human and mouse; bases shown in dark green are
covariance base pairs, which differ among species but do
not affect the local structure. Overall, these results suggest
that lncRNA MEG3 is conserved among pig, human and
mouse.
Temporal-spatial expression patterns
To explore the temporal and spatial expression patterns of
lncRNA MEG3 in S. scrofa, we measured expression distri-
bution in nine different tissues (heart, liver, spleen, lung,
kidney, intestine, stomach, and leg and dorsal muscles). To
determine dynamic changes, we measured MEG3 expres-
sion in skeletal muscle across 14 developmental stages
(embryonic E33, E60, E65, E70, E75, E80, E95 and E105
and postnatal D20, D40, D60, D100, D140 and D160) in
Large White pigs (Fig. 1c). We found that lncRNA MEG3
was expressed in all nine tissues, with the highest expres-
sion levels in skeletal muscle (including leg and dorsal
muscles) and kidney. Low expression levels of MEG3 were
observed in the stomach, and moderate expression levels
were observed in the remaining tissues (lung, heart, liver,
spleen and intestine). As shown in Fig. 1d, lncRNA MEG3
was detected mainly in prenatal and neonatal skeletal
muscle (from E33 to D40) and expression peaked at E60.
Identifying the core promoter of lncRNA MEG3
To further understand the temporal-spatial expression
restrictions of lncRNA MEG3, we cloned the porcine
lncRNA MEG3 promoter and analysed the sequence. The
pig lncRNA MEG3 promoter contains several putative
transcription factor binding sites, including binding sites
for MyoD, SP1, NFY, USF and E47. The relative activities of
the 50flanking region fragments expressed in PIEC (black)
and PK15 cells (grey) are shown in Fig. 2a. Luciferase
assays revealed that promoter activities of the truncated
fragments PGL-MEG3-1, PGL-MEG3-2, PGL-MEG3-3, PGL-
MEG3-4, PGL-MEG3-5, PGL-MEG3-6 and PGL-MEG3-7
were significantly higher than was pGL3-basic in both cell
types (Table S2). Promoter activity gradually increased
between PGL-MEG3-1 and PGL-MEG3-5. In contrast,
promoter activity gradually decreased between PGL-
MEG3-5 and PGL-MEG3-8. Promoter activity of PGL-
MEG3-8 decreased significantly. These results indicate that
the region between 447 and 40 bp, upstream of the
transcriptional start site, was sufficient for regulating
transcription activity (Fig. 2b).
SNP linkage disequilibrium and association with meat
production traits
Direct sequencing of the porcine MEG3 genome was
performed to detect potential polymorphisms in 30 Large
White pigs. The following SNPs were identified: rs322438324
(T/G) in the 50-UTR region; rs25 (C/T), rs24 (T/G) and rs23
©2018 Stichting International Foundation for Animal Genetics, 49, 571–578
lncRNA-MEG3 is a marker in pig meat production 573
(A/G) in the first intron; and rs318656749 (T/C),
rs81286029 (A/G), rs344501106 (A/G), rs325797437
(A/G), rs334059356 (T/A), rs323571592 (G/A) and
rs322802425 (T/G) in the second exon (rs identifications
were artificially assigned).
We examined polymorphisms in 297 Large White pigs.
Sequence comparative analysis revealed 10 polymorphic
sites. Unfortunately, rs24 was not detected. Genetic diver-
sity analysis showed that genotypic distribution for
rs322802425, rs323571592, rs334059356, rs23, rs25
and rs322438324 deviated from HardyWeinberg equilib-
rium (P
HW
<0.05 or P
HW
=1), whereas the distribution for
rs325797437, rs344501106, rs81286029 and rs318656749
was in accordance with HardyWeinberg equilibrium
(Table 1). Moreover, D0/r
2
linkage disequilibrium analysis
indicated that four SNPs (rs325797437, rs344501106,
rs81286029 and rs318656749) showed linkage inheritance
that led to six haplotypes (GGGC, GAAT, AAAT, AAAT/
GGGC, GAAT/GGGC and GAAT/AAAT) (Fig. 3). Thus, we
selected these four SNP sites for further association analysis
with meat production traits.
The results of the association analysis between the four
SNP sites and meat production traits are shown in Table 2.
Three of the four SNP sites were significantly associated
with corrected back fat thickness. Specifically, individual
animals with the TT genotype in rs318656749 had
significantly thicker back fat than did those with the
CT genotype (P=0.0240). Animals with the AA geno-
type had significantly thicker back fat than did those
with the AG genotype in both rs81286029 (P=0.0487)
and rs344501106 (P=0.0471). For corrected age to
reach 100 kg, individuals with the GG genotype in
rs325797437 took approximately 3 days longer to reach
the target weight than did individuals with the AG
genotype (P=0.0341).
Further association analysis on haplotypes indicated that
corrected age to reach 100 kg differed significantly among
the haplotype classes. The homozygous AAAT individuals
took about 10 days less than did heterozygous GAAT/GGGC
individuals (P=0.0301) to reach the target weight;
heterozygous AAAT/GGGC individuals took approximately
4.6 days less than did the heterozygous GAAT/GGGC
individuals to reach the target weight (P=0.0184)
(Table 3).
Discussion
To understand the role of lncRNAs in skeletal muscle
development and meat production traits, we analysed the
pairwise alignments between pig and four other species
(human, mouse, cow and opossum). The results revealed
that lncRNAs were highly conserved among mammals.
Figure 1 Conservation and spatial-temporal expression analysis of porcine lncRNA-MEG3. (a) Pig lncRNAs aligned with multiple species. (b)
Structure conservation of the lncRNA MEG3 in Sus scrofa. (c) RT-PCR tissue expression analysis of lncRNA MEG3. (d) Relative expression quantity of
lncRNA MEG3 at different developmental stages. The values in the graphs represent means standard error expressed as the expression level
relative to GAPDH.
©2018 Stichting International Foundation for Animal Genetics, 49, 571–578
Yu et al.574
Thousands of lncRNAs are evolutionary conserved, though
not to the same extent as many protein-coding genes
(Ponjavic et al. 2007; Guttman et al. 2009). We identified
MEG3 as a conserved lncRNA sequence. In agreement with
previous reports, MEG3 is evolutionary conserved between
the pig and closely related species (e.g. cow, human and
mouse) (Li et al. 2008; Hezroni et al. 2015).
LncRNAs are involved in various biological processes via
diverse mechanisms (Moran et al. 2012). LncRNA MEG3 is
associated with human diseases such as cancers and
diabetes (Benetatos et al. 2008; Qiu et al. 2016). However,
there are no reports on the involvement of MEG3 in pig
skeletal muscle development. Fleming-Waddell et al. (2009)
identified a number of genes that are regulated by DLK1 and
RTL1 and exert control over postnatal skeletal muscle
growth, which provides an important clue to the role of
lncRNA MEG3 in muscle development. The secondary
structures of DLK1 and RTL1 are highly similar to MEG3,
indicating that they have similar functions in pigs, e.g.
regulation of postnatal skeletal muscle growth. For
Figure 2 Relative luciferase activity in porcine kidney cells (PK15) and premature intestinal epithelial cells (PIEC) for different truncated fragments. (a)
MEG3 gene promoter activity detection in PIEC (black) and PK15 (grey). (b) Promoter activity of each truncated fragment recombinant vector in PIEC
(black) and PK15 (grey). Luciferase activities were normalized to co-expressed b-gal. Data for both panels are shown as the mean SD of a
representative experiment performed in triplicate.
Table 1 Genetic diversity analysis results. SNPs Real He Expected He Ho Ae P
HW
%Geno MAF
rs322802425 0 0 1 1 1 99.7 0
rs323571592 0 0 1 1 1 99.7 0
rs334059356 0.017 0.03 0.983 1.0173 0.0021 100 0.0125
rs325797437 0.192 0.2 0.808 1.238 0.6197 100 0.117
rs344501106 0.354 0.392 0.646 1.548 0.1179 100 0.276
rs81286029 0.358 0.388 0.642 1.558 0.2268 99.7 0.269
rs318656749 0.358 0.391 0.642 1.558 0.1816 99.7 0.274
rs23 0 0 1 1 1 100 0
rs25 0.003 0.01 0.997 1.00 0.0102 99.7 0.0035
rs322438324 0.003 0.01 0.997 1.00 0.0101 100 0.007
Ae, effective number of alleles; He, heterozygosity; Ho, homogeneity; MAF, minor allele
frequency; P
HW
,P-value of Hardy-Weinberg balance.
©2018 Stichting International Foundation for Animal Genetics, 49, 571–578
lncRNA-MEG3 is a marker in pig meat production 575
example, MEG3 is abundantly expressed in the paraxial
mesoderm, suggesting a role in myogenesis (Schuster-
Gossler et al. 1998). MEG3-knockout mice acquire skeletal
muscle developmental defects, and perinatal death occurs,
indicating that MEG3 is a nuclear, cis-acting lncRNA
regulating muscle development (Zhou et al. 2010).
The spatial expression analysis showed that MEG3 was
expressed in a tissue-specific manner with high levels in the
kidney and dorsal and leg muscles. Temporal expression
analysis showed that MEG3 was expressed mainly in
prenatal and early postnatal skeletal muscle. These results
indicate that MEG3 is involved in myogenesis and con-
tributes to prenatal skeletal muscle development in pigs.
The promoter plays an important role in tissue-specific gene
expression (Xu et al. 2010), and the core promoter region of
lncRNA MEG3 in different tissues will be the subject of
future studies.
Skeletal muscle, composed mainly of myofibres, plays a
key role in the determination of meat production. During the
development of the pig, body segments develop on embryo
days 1422. Primary myotubes are formed at E35, and cell
proliferation peaks at around E49. Secondary myotubes
begin to form when the primary myotubes proliferate, after
which the primary myotubes disappear. Secondary myo-
tubes proliferate at E90. The number of myofibres is
determined before birth during secondary myoduct forma-
tion, whereas the diameter and length of myofibres keep
increasing until D60 after birth (Ashmore et al. 1973; Tang
et al. 2007). According to our results, expression of MEG3
peaked at E60 (P<0.05), whereas almost no expression was
detected at postnatal D40. These results indicate that MEG3
is involved in the differentiation and maintenance of skeletal
muscle cells in early development.
Mutations in imprinting genes can directly or indirectly
affect important economic traits in livestock. In sheep, the
Figure 3 Linkage disequilibrium analysis of
lncRNA MEG3. Four (rs325797437,
rs344501106, rs81286029 and rs318656749)
of 10 potential polymorphism sites showed
linkage inheritance resulting in six haplotypes.
Table 2 Results of the relative analysis between the genotypes of the
SNPs.
SNPs
Geno-
type
Sample
size BFT CA100
rs318656749 CC 163 17.08 0.2737
ab
161.3 0.8843
TT 26 17.91 0.6124
a
157.7 1.630
CT 106 16.45 0.2617
b
160.7 0.9598
rs81286029 AA 25 17.71 0.6025
a
157.7 1.696
GG 164 17.07 0.2724
ab
161.3 0.8807
AG 106 16.46 0.2616
b
160.5 0.9567
rs344501106 AA 26 17.76 0.5808
a
157.4 1.649
GG 164 17.07 0.2724
ab
161.3 0.8807
AG 104 16.53 0.2704
b
160.7 0.9627
rs325797437 AA 7 18.15 1.240 152.4 2.871
ab
GG 233 17.00 0.2137 161.5 0.7123
a
AG 57 16.57 0.3992 158.2 1.158
b
BFT, corrected back fat thickness; CA100, corrected age to reach
100 kg.
The same superscript letter indicates no significant difference
(P>0.05); different superscript letters indicate significant differences
(P<0.05).
Table 3 Results of the relative analysis between the genotypes of the
haplotypes.
Haplotypes Sample size BFT CA100
GAAT 11 17.63 1.045 158.0 2.691
abc
AAAT 5 18.15 1.240 152.4 2.871
ac
GGGC 163 17.12 0.2703 161.4 0.8847
abc
AAAT/GGGC 46 16.47 0.4294 158.1 1.320
ac
GAAT/GGGC 59 18.49 1.953 162.7 1.322
b
GAAT/AAAT 10 17.69 0.8647 159.3 2.663
abc
BFT, corrected back fat thickness; CA100, corrected age to reach
100 kg.
The same superscript letter indicates no significant difference
(P>0.05); different superscript letters indicate significant differences
(P<0.05).
©2018 Stichting International Foundation for Animal Genetics, 49, 571–578
Yu et al.576
mutant MEG3 genotype, callipyge, is beneficial to muscle
development and lower fat content in the waist and
hindquarters (Cockett et al. 2005). In cattle, one SNP of
MEG3 influences production traits such as milk yield (Magee
et al. 2011). Another SNP, rs941576, is located in the sixth
intron of MEG3 and is associated with type 1 diabetes in
human beings, which is often accompanied by skeletal
muscle atrophy and recession (Wallace et al. 2010). In this
study, we identified four SNPs of MEG3 in Large White pigs.
Three of the SNPs were significantly associated with changes
in back fat thickness, and two of the SNPs were significantly
associated with changes in corrected age to reach 100 kg
(P<0.05). Haplotypes composed of four SNPs
(rs318656749, rs81286029, rs344501106 and
rs325797437) were associated with changes in corrected
age to reach 100 kg. Corrected back fat and corrected age to
reach 100 kg shows a genetic correlation with lean meat
percentage (Dube et al. 2013; Cabling et al. 2015). Therefore,
we conclude that SNPs can influence skeletal muscle devel-
opment and lead to significant production trait differences by
changing the secondary structure of MEG3. Although our
work shows that some SNP variants in lncRNA MEG3 have
strong associations with meat-producing traits in pigs, the
underlying mechanisms need further investigation.
Conclusions
We found that lncRNA MEG3 is conserved among mam-
mals and exhibits specific temporal-spatial expression pat-
terns. We identified the core promoter region, which can be
used for further functional studies of MEG3. Our data
indicate that lncRNA MEG3 is involved in the differentia-
tion and maintenance of early skeletal muscle development.
In addition, lncRNA MEG3 SNPs are associated with
changes in meat production traits and can be used as
biomarkers in pig breeding. This study increases our
knowledge about the biological function of MEG3 in muscle
growth. However, experiments focused on molecular mech-
anisms are needed.
Acknowledgements
This work was supported by the National Key Project
(2016ZX08009-003-006), the National Natural Science
Foundation of China (31372295) and the Agricultural
Science and Technology Innovation Program (ASTIP-
AGIS5).
Author contributions
Z.T. conceived and designed the experiments. Z.T., S.H.,
Z.W. and Y.Y. performed animal work and collected
biological samples. X.Y., Z.W. and H.S. contributed reagents
and materials and analysed data. H.S. and Y.Y. performed
cell culture and molecular experiments. Z.T., X.Y. and Z.W.
contributed to manuscript writing, and K.L. contributed to
manuscript revision. All authors read and agreed to the
final manuscript.
References
Ashmore C.R., Addis P.B. & Doerr L. (1973) Development of muscle
fibers in the fetal pig. Journal of Animal Science 36, 108893.
Ballarino M., Cazzella V., D’Andrea D. et al. (2015) Novel long
noncoding RNAs (lncRNAs) in myogenesis: a miR-31 overlap-
ping lncRNA transcript controls myoblast differentiation. Molec-
ular and Cellular Biology 35, 72836.
Barrett J.C., Fry B., Maller J. & Daly M.J. (2005) HAPLOVIEW: analysis
and visualization of LD and haplotype maps. Bioinformatics 21,
2635.
Benetatos L., Dasoula A., Hatzimichael E., Georgiou I., Syrrou M. &
Bourantas K.L. (2008) Promoter hypermethylation of the MEG3
(DLK1/MEG3) imprinted gene in multiple myeloma. Clinical
Lymphoma and Myeloma 8, 1715.
Cabling M.M., Kang H.S., Lopez B.M., Jang M., Kim H.S., Nam K.C.,
Choi J.G. & Seo K.S. (2015) Estimation of genetic associations
between production and meat quality traits in Duroc pigs. Asian-
Australasian Journal of Animal Sciences 28, 10615.
Cockett N.E., Smit M.A., Bidwell C.A., Segers K., Hadfield T.L.,
Snowder G.D., Georges M. & Charlier C. (2005) The callipyge
mutation and other genes that affect muscle hypertrophy in
sheep. Genetics Selection Evolution 37 (Suppl 1), S6581.
Druet T. & Farnir F.P. (2011) Modeling of identity-by-descent
processes along a chromosome between haplotypes and their
genotyped ancestors. Genetics 188, 40919.
Dube B., Mulugeta S.D. & Dzama K. (2013) Genetic relationship
between growth and carcass traits in Large White pigs. South
African Journal of Animal Science 43, 48292.
Fleming-Waddell J.N., Olbricht G.R., Taxis T.M., White J.D.,
Vuocolo T., Craig B.A., Tellam R.L., Neary M.K., Cockett N.E. &
Bidwell C.A. (2009) Effect of DLK1 and RTL1 but not MEG3 or
MEG8 on muscle gene expression in Callipyge lambs. PLoS ONE
4, e7399.
Freking B.A., Keele J.W., Nielsen M.K. & Leymaster K.A. (1998)
Evaluation of the ovine callipyge locus: II. Genotypic effects on
growth, slaughter, and carcass traits. Journal of Animal Science
76, 254959.
Freking B.A., Keele J.W., Shackelford S.D., Wheeler T.L., Koohmar-
aie M., Nielsen M.K. & Leymaster K.A. (1999) Evaluation of the
ovine callipyge locus: III. Genotypic effects on meat quality traits.
Journal of Animal Science 77, 233644.
Gong C., Li Z., Ramanujan K., Clay I., Zhang Y., Lemire-Brachat S.
& Glass D.J. (2015) A long non-coding RNA, LncMyoD, regulates
skeletal muscle differentiation by blocking IMP2-mediated mRNA
translation. Developmental Cell 34, 18191.
Guttman M., Amit I., Garber M. et al. (2009) Chromatin signature
reveals over a thousand highly conserved large non-coding RNAs
in mammals. Nature 458, 2237.
Hezroni H., Koppstein D., Schwartz M.G., Avrutin A., Bartel D.P. &
Ulitsky I. (2015) Principles of long noncoding RNA evolution
derived from direct comparison of transcriptomes in 17 species.
Cell Reports 11, 111022.
©2018 Stichting International Foundation for Animal Genetics, 49, 571–578
lncRNA-MEG3 is a marker in pig meat production 577
Kallen A.N., Zhou X.B., Xu J. et al. (2013) The imprinted H19
lncRNA antagonizes let-7 microRNAs. Molecular Cell 52, 101
12.
Koohmaraie M., Shackelford S.D., Wheeler T.L., Lonergan S.M.
& Doumit M.E. (1995) A muscle hypertrophy condition in
lamb (callipyge): characterization of effects on muscle growth
and meat quality traits. Journal of Animal Science 73, 3596
607.
Legnini I., Morlando M., Mangiavacchi A., Fatica A. & Bozzoni I.
(2014) A feedforward regulatory loop between HuR and the long
noncoding RNA linc-MD1 controls early phases of myogenesis.
Molecular Cell 53, 50614.
Li X.P., Do K.T., Kim J.J., Huang J., Zhao S.H., Lee Y., Rothschild
M.F., Lee C.K. & Kim K.S. (2008) Molecular characteristics of the
porcine DLK1 and MEG3 genes. Animal Genetics 39, 18992.
Ma G., Huang J., Sun N., Liu X., Zhu M., Wu Z. & Zhao S. (2008)
Molecular characterization of the porcine GBP1 and GBP2 genes.
Molecular Immunology 45, 2797807.
Magee D.A., Berry D.P., Berkowicz E.W., Sikora K.M., Howard
D.J., Mullen M.P., Evans R.D., Spillane C. & MacHugh D.E.
(2011) Single nucleotide polymorphisms within the bovine
DLK1-DIO3 imprinted domain are associated with economi-
cally important production traits in cattle. JournalofHeredity
102, 94101.
Mattick J.S. (2004) RNA regulation: a new genetics? Nature Reviews
Genetics 5, 31623.
Moran V.A., Perera R.J. & Khalil A.M. (2012) Emerging functional
and mechanistic paradigms of mammalian long non-coding
RNAs. Nucleic Acids Research 40, 6391400.
Ponjavic J., Ponting C.P. & Lunter G. (2007) Functionality or
transcriptional noise? Evidence for selection within long noncod-
ing RNAs Genome Research 17, 55665.
Qiu G.Z., Tian W., Fu H.T., Li C.P. & Liu B. (2016) Long noncoding
RNA-MEG3 is involved in diabetes mellitus-related microvascu-
lar dysfunction. Biochemical and Biophysical Research Communica-
tions 471, 13541.
da Rocha S.T., Edwards C.A., Ito M., Ogata T. & Ferguson-Smith
A.C. (2008) Genomic imprinting at the mammalian Dlk1-Dio3
domain. Trends in Genetics 24, 30616.
Rosenbloom K.R., Armstrong J., Barber G.P. et al. (2015) The UCSC
Genome Browser database: 2015 update. Nucleic Acids Research
43, D67081.
Schuster-Gossler K., Bilinski P., Sado T., Ferguson-Smith A. &
Gossler A. (1998) The mouse Gtl2 gene is differentially expressed
during embryonic development, encodes multiple alternatively
spliced transcripts, and may act as an RNA. Developmental
Dynamics 212, 21428.
Sollero B.P., Guimaraes S.E., Rilington V.D., Tempelman R.J.,
Raney N.E., Steibel J.P., Guimaraes J.D., Lopes P.S., Lopes M.S. &
Ernst C.W. (2011) Transcriptional profiling during foetal skeletal
muscle development of Piau and Yorkshire-Landrace cross-bred
pigs. Animal Genetics 42, 60012.
Tang Z., Li Y., Wan P., Li X., Zhao S., Liu B., Fan B., Zhu M., Yu M.
& Li K. (2007) LongSAGE analysis of skeletal muscle at three
prenatal stages in Tongcheng and Landrace pigs. Genome Biology
8, R115.
Tang Z., Wu Y., Yang Y. et al. (2017) Comprehensive analysis of
long non-coding RNAs highlights their spatio-temporal expres-
sion patterns and evolutional conservation in Sus scrofa.Scientific
Reports 7, 43166.
Wallace C., Smyth D.J., Maisuria-Armer M., Walker N.M., Todd J.A.
& Clayton D.G. (2010) The imprinted DLK1-MEG3 gene region
on chromosome 14q32.2 alters susceptibility to type 1 diabetes.
Nature Genetics 42,6871.
Wang P., Ren Z. & Sun P. (2012) Overexpression of the long non-
coding RNA MEG3 impairs in vitro glioma cell proliferation.
Journal of Cellular Biochemistry 113, 186874.
Washietl S., Kellis M. & Garber M. (2014) Evolutionary dynamics
and tissue specificity of human long noncoding RNAs in six
mammals. Genome Research 24, 61628.
Xu H.G., Ren W., Lu C. & Zhou G.P. (2010) Characterization of the
human IRF-3 promoter and its regulation by the transcription
factor E2F1. Molecular Biology Reports 37, 307380.
Zhou Y., Cheunsuchon P., Nakayama Y. et al. (2010) Activation of
paternally expressed genes and perinatal death caused by
deletion of the Gtl2 gene. Development,137, 264352.
Zhou L., Sun K., Zhao Y. et al. (2015) Linc-YY1 promotes myogenic
differentiation and muscle regeneration through an interaction
with the transcription factorYY1. Nature Communications 6, 10026.
Zhu M., Liu J., Xiao J. et al. (2017) Lnc-mg is a long non-coding
RNA that promotes myogenesis. Nature Communications 8,
14718.
Supporting information
Additional supporting information may be found online in
the Supporting Information section at the end of the article.
Table S1 Primer information for experiments.
Table S2 Electrophoresis charts for each truncated recom-
binant expression vector.
©2018 Stichting International Foundation for Animal Genetics, 49, 571–578
Yu et al.578
... Gong et al. (2015) demonstrated that lncMyoD regulated skeletal muscle differentiation by blocking IMP2-mediated mRNA translation. MEG3 (Maternally expressed gene 3), a lncRNA identified first in mice, was reported to be a regulator of skeletal muscle development (Yu et al., 2018). A previous study demonstrated that MEG3 promoted bovine skeletal muscle differentiation by interacting with miRNA-135 and MEF2C . ...
... Zhou et al. (2010) illustrated that maternal deletion of MEG3 resulted in perinatal death and skeletal muscle defects. MEG3 was expressed in eight different tissues including heart, liver, spleen, lung, kidney, intestine, stomach, and skeletal muscle in Large White pigs (Yu et al., 2018). The MEG3 expression was higher in skeletal muscle than in other tissues (Yu et al., 2018). ...
... MEG3 was expressed in eight different tissues including heart, liver, spleen, lung, kidney, intestine, stomach, and skeletal muscle in Large White pigs (Yu et al., 2018). The MEG3 expression was higher in skeletal muscle than in other tissues (Yu et al., 2018). Furthermore, previous researches suggested that MEG3 overexpression caused a decrease of cell proliferation and downregulations of proteins involved in proliferation, including CDK4, CDK6, and CyclinD1 (Ji and Li, 2019). ...
Article
Full-text available
Maternally expressed gene 3 (MEG3) is a long non-coding RNA that is a crucial regulator of skeletal muscle development. Some single-nucleotide polymorphism (SNP) mutants in MEG3 had strong associations with meat quality traits. Nevertheless, the function and mechanism of MEG3 mutants on porcine skeletal muscle development have not yet been well-demonstrated. In this study, eight SNPs were identified in MEG3 of fat- and lean-type pig breeds. Four of these SNPs (g.3087C > T, g.3108C > T, g.3398C > T, and g.3971A > C) were significantly associated with meat quality and consisted of the CCCA haplotype for fat-type pigs and the TTCC haplotype for lean-type pigs. Quantitative real-time PCR results showed that the expression of MEG3-TTCC was higher than that of MEG3-CCCA in transcription level (P < 0.01). The stability assay showed that the lncRNA stability of MEG3-TTCC was lower than that of MEG3-CCCA (P < 0.05). Furthermore, the results of qRT-PCR, Western blot, and Cell Counting Kit-8 assays demonstrated that the overexpression of MEG3-TTCC more significantly inhibited the proliferation of porcine skeletal muscle satellite cells (SCs) than that of MEG3-CCCA (P < 0.05). Moreover, the overexpression of MEG3-TTCC more significantly promoted the differentiation of SCs than that of MEG3-CCCA (P < 0.05). The Western blot assay suggested that the overexpression of MEG3-TTCC and MEG3-CCCA inhibited the proliferation of SCs by inhibiting PI3K/AKT and MAPK/ERK1/2 signaling pathways. The overexpression of the two haplotypes also promoted the differentiation of SCs by activating the JAK2/STAT3 signaling pathway in different degrees. These data are valuable for further studies on understanding the crucial role of lncRNAs in skeletal muscle development.
... For that instance, lncRNA MAR1 exhibits a potential positive impact on skeletal muscle hypertrophy and may also contribute to mitigating aging-related muscle atrophy [22]. During the pre-natal and early post-neonatal stages in porcine skeletal muscle, lncRNA MEG3 demonstrates elevated levels of expression, and it plays a crucial part in the development and upkeep of early skeletal muscle [23]. Many researchers have identified the crucial genes that influence muscle development through the comprehensive sequencing of lncRNAs across distinct species. ...
Article
Full-text available
The production performance of Jeryak, resulting from the F1 generation of the cross between Gannan yak and Jersey cattle, exhibits a significantly superior outcome compared with that of Gannan yak. Therefore, we used an RNA-seq approach to identify differentially expressed mRNAs (DEMs) and differentially expressed lncRNAs (DELs) influencing muscle growth and development in Gannan yaks and Jeryaks. A total of 304 differentially expressed lncRNAs and 1819 differentially expressed mRNAs were identified based on the screening criteria of |log 2 FC| > 1 and FDR < 0.05. Among these, 132 lncRNAs and 1081 mRNAs were found to be down-regulated, while 172 lncRNAs and 738 mRNAs were up-regulated. GO and KEGG analyses showed that the identified DELs and DEMs were enriched in the entries of pathways associated with muscle growth and development. On this basis, we constructed an lncRNA–mRNA interaction network. Interestingly, two candidate DELs (MSTRG.16260.9 and MSTRG.22127.1) had targeting relationships with 16 (MYC, IGFBP5, IGFBP2, MYH4, FGF6, etc.) genes related to muscle growth and development. These results could provide a basis for further studies on the roles of lncRNAs and mRNAs in muscle growth in Gannan yaks and Jeryak breeds.
... Previous research have proposed that MEG3 upregulates the expression of serum response factor by adsorbing miR-423-5p [20]. In addition, MEG3 exists in multiple single nucleotide loci in pigs and is significantly associated with meat production traits [21,22]. Therefore, it is of great significance to study the effect of MEG3 on myogenic differentiation. ...
Article
Full-text available
Long non-coding RNAs (lncRNAs) are involved in the process of muscle cell differentiation and play an important role. Previous studies have shown that lncRNA-MEG3 promotes the differentiation of porcine skeletal muscle satellite cells (PSCs), but the regulatory mechanism of MEG3 interaction with target protein has not been well studied. We demonstrated that MEG3 can bind dihydrolipoamide succinyltransferase (DLST) by RNA pull down and RIP-qPCR. Subsequently, knockdown and overexpression experiments showed that DLST promotes PSCs differentiation. Rescue experiments showed that the expression of DLST protein was significantly increased with MEG3 overexpression and decreased with MEG3 knockdown, while its mRNA expression was not changed. Furthermore, we have successfully predicted and validated that the transcription factor myogenic differentiation (MYOD) binds to the MEG3 core promoter though utilizing chromatin immunoprecipitation (CHIP) and luciferase reporter assays. The results indicated that MYOD acts as a transcription factor of MEG3 to promote MEG3 transcription. Knockdown of MEG3 in vivo indicated that MEG3 is involved in skeletal muscle regeneration. It is concluded that MYOD acts as a transcription factor to induce MEG3 expression. MEG3 acts as a molecular scaffold to bind and promote DLST protein expression. This paper provides a new molecular mechanism for MEG3 to promote the differentiation of PSCs.
... lncRNAs work as ceRNAs, which is the most common regulatory function. For example, lncRNA MEG3, as the "sponge" of miRNA-135, promoted bovine skeletal muscle differentiation via interacting with miRNA-135 and MEF2C (myocyte enhancer factor 2C) (Liu et al., 2019), and this lncRNA was also found in pig skeletal muscle (Yu et al., 2018). lncRNA-125b promoted skeletal muscle satellite cell differentiation by functioning as a ceRNA for miR-125b to positively regulate IGF2 (insulin-like growth factor 2) expression (Zhan et al., 2019). ...
Article
Full-text available
Introduction: As a valuable genetic resource, native birds can contribute to the sustainable development of animal production. Tibetan chickens, known for their special flavor, are one of the important local poultry breeds in the Qinghai–Tibet Plateau. However, Tibetan chickens have a slow growth rate and poor carcass traits compared with broilers. Although most of the research on Tibetan chickens focused on their hypoxic adaptation, there were fewer studies related to skeletal muscle development. Methods: Here, we performed the transcriptional sequencing of leg muscles from Tibetan chicken embryos at E (embryonic)10, E14, and E18. Results: In total, 1,600, 4,610, and 2,166 DE (differentially expressed) mRNAs, 210, 573, and 234 DE lncRNAs (long non-coding RNAs), and 52, 137, and 33 DE miRNAs (microRNAs) were detected between E10 and E14, E10 and E18, and E14 and E18, respectively. Functional prediction showed several DE mRNAs and the target mRNAs of DE lncRNAs and DE miRNAs were significantly enriched in sarcomere organization, actin cytoskeleton organization, myofibril, muscle fiber development, and other terms and pathways related to muscle growth and development. Finally, a lncRNA–miRNA–mRNA ceRNA (competing endogenous RNA) network associated with muscle growth and development, which contained 6 DE lncRNAs, 13 DE miRNAs, and 50 DE mRNAs, was constructed based on the screened DE RNAs by Gene Ontology (GO) enrichment. These DE RNAs may play a critical regulatory role in the skeletal muscle development of chickens. Discussion: The results provide a genomic resource for mRNAs, lncRNAs, and miRNAs potentially involved in the skeletal muscle development of chickens, which lay the foundation for further studies of the molecular mechanisms underlying skeletal muscle growth and development in Tibetan chickens.
... Increasing studies have begun to focus on the impact of SNPs on lncRNAs in animals. For example, an analysis of pigs showed that several SNPs in lncRNA MEG3 were associated with meat production (18). Another study in chicken also demonstrated that rs1914215137 in the lncRNA pouBW1 was associated with chicken growth and carcass (19). ...
Article
Full-text available
Long non-coding RNAs (lncRNAs) act as versatile regulators of many biological processes and play vital roles in various diseases. lncRNASNP is dedicated to providing a comprehensive repository of single nucleotide polymorphisms (SNPs) and somatic mutations in lncRNAs and their impacts on lncRNA structure and function. Since the last release in 2018, there has been a huge increase in the number of variants and lncRNAs. Thus, we updated the lncRNASNP to version 3 by expanding the species to eight eukaryotic species (human, chimpanzee, pig, mouse, rat, chicken, zebrafish, and fruitfly), updating the data and adding several new features. SNPs in lncRNASNP have increased from 11 181 387 to 67 513 785. The human mutations have increased from 1 174 768 to 2 387 685, including 1 031 639 TCGA mutations and 1 356 046 CosmicNCVs. Compared with the last release, updated and new features in lncRNASNP v3 include (i) SNPs in lncRNAs and their impacts on lncRNAs for eight species, (ii) SNP effects on miRNA−lncRNA interactions for eight species, (iii) lncRNA expression profiles for six species, (iv) disease & GWAS-associated lncRNAs and variants, (v) experimental & predicted lncRNAs and drug target associations and (vi) SNP effects on lncRNA expression (eQTL) across tumor & normal tissues. The lncRNASNP v3 is freely available at http://gong_lab.hzau.edu.cn/lncRNASNP3/.
... Wang et al. identified 1,932lncRNAs (760 novel ones) and found lnc_000414function as an inhibitor in the proliferation of porcine intramuscular adipocytes [28]. lncRNA MEG3 participated inthe regulation of skeletal muscle development [29]. Hainan black goats, an indigenous breed of China, is considered as a kind of excellent black goats with good meat quality [30]. ...
Article
Full-text available
It is deemed that meat quality of kids' is better than that of adults' for Hainan black goat. Generally, meat quality is affected by many indicators, such as intramuscular fat (IMF) content, muscle fiber diameter and shear force. It is indicated that long non-coding RNAs (lncRNAs) play essential roles in meat quality of goats. However, it is unclear whether and how lncRNAs and genes play their roles in meat quality of Hainan Black goats. Here, we firstly compared the meat quality between two-month-old kids (kids) and adult goats (adults). Then, the lncRNA-seq and RNA-seq data were integrated and analyzed to explore the potential functions of lncRNAs and genes. The results showed that adults' IMF content and muscle fiber diameter were extremely significantly higher than that of kids (P<0.01). For the sequenced data, average 84,970,398, and 83,691,250 clean reads were obtained respectively for Kids and adults, among which ~96% were mapped to the reference genome of goats. Through analyzing, 18,242 goat annotated genes, 1,429 goat annotated lncRNAs and 2,967 novel lncRNAs were obtained. Analysis of differential expression genes (DEGs) and lncRNAs (DELs) showed that 328 DEGs and 98 DELs existed between kids and adults. Furthermore, functional enrichment analysis revealed that a number of DEGs and DELs were mainly associated with IMF. Primarily, DGAT2 expressed higher in adults than that in kids and CPT1A expressed higher in kids than that in adults. Both of them were overlapped by DEGs and targets of DELs, suggesting the two DEGs and the DELs targeted by the two DEGs might be the potential regulators of goat IMF deposition. Taken together, our results provide basic support for further understanding the function and mechanism of lncRNAs and genes in meat quality of Hainan black goats.
... [5][6][7] Meanwhile, non-coding RNAs (ncRNAs), such as microRNAs (miRNAs) and long non-coding RNAs (lncRNAs), are emerging as regulators of skeletal muscle development and regeneration. [8][9][10] For instance, our previous study suggested that miR-148a promotes myogenic differentiation of both C2C12 myoblasts and primary muscle cells by targeting the Rho-associated, coiled-coil-containing protein kinase 1 (ROCK1) gene. 9 Circular RNAs (circRNAs) are a new type of endogenous ncRNA covalently closed by a non-canonical splicing event called backsplicing. ...
Article
Full-text available
Background Circular RNAs (circRNAs) represent a novel class of non‐coding RNAs formed by a covalently closed loop and play crucial roles in many biological processes. Several circRNAs associated with myogenesis have been reported. However, the dynamic expression, function, and mechanism of circRNAs during myogenesis and skeletal muscle development are largely unknown. Methods Strand‐specific RNA‐sequencing (RNA‐seq) and microarray datasets were used to profile the dynamic circRNAome landscape during skeletal muscle development and myogenic differentiation. Bioinformatics analyses were used to characterize the circRNAome and identify candidate circRNAs associated with myogenesis. Bulk and single‐cell RNA‐seq were performed to identify the downstream genes and pathways of circFgfr2. The primary myoblast cells, C2C12 cells, and animal model were used to assess the function and mechanism of circFgfr2 in myogenesis and muscle regeneration in vitro or in vivo by RT‐qPCR, western blotting, dual‐luciferase activity assay, RNA immunoprecipitation, RNA fluorescence in situ hybridization, and chromatin immunoprecipitation. Results We profiled the dynamic circRNAome in pig skeletal muscle across 27 developmental stages and detected 52 918 high‐confidence circRNAs. A total of 2916 of these circRNAs are conserved across human, mouse, and pig, including four circRNAs (circFgfr2, circQrich1, circMettl9, and circCamta1) that were differentially expressed (|log2 fold change| > 1 and adjusted P value < 0.05) in various myogenesis systems. We further focused on a conserved circRNA produced from the fibroblast growth factor receptor 2 (Fgfr2) gene, termed circFgfr2, which was found to inhibit myoblast proliferation and promote differentiation and skeletal muscle regeneration. Mechanistically, circFgfr2 acted as a sponge for miR‐133 to regulate the mitogen‐activated protein kinase kinase kinase 20 (Map3k20) gene and JNK/MAPK pathway. Importantly, transcription factor Kruppel like factor 4 (Klf4), the downstream target of the JNK/MAPK pathway, directly bound to the promoter of circFgfr2 and affected its expression via an miR‐133/Map3k20/JNK/Klf4 auto‐regulatory feedback loop. RNA binding protein G3BP stress granule assembly factor 1 (G3bp1) inhibited the biogenesis of circFgfr2. Conclusions The present study provides a comprehensive circRNA resource for skeletal muscle study. The functional and mechanistic analysis of circFgfr2 uncovered a circRNA‐mediated auto‐regulatory feedback loop regulating myogenesis and muscle regeneration, which provides new insight to further understand the regulatory mechanism of circRNAs.
... For example, SRA25 , lnc-ADINR (Xiao et al. 2015), lncRNA-ADNCR (Li et al. 2016) and lnc-BATE1 (Alvarez-Dominguez et al. 2015) were identified to mediate adipocyte differentiation. lncRNAs including MEG3 (Yu et al. 2018), SYISL (SYNPO2 intron sense-overlapping lncRNA; Jin et al. 2018) and AK143003 (Guo et al. 2017) have been proven to regulate skeletal muscle development. Numerous genomewide analyses of lncRNA have been performed in porcine subcutaneous adipose Yu et al. 2017;Liu et al. 2018;Kumar et al. 2019;Xing et al. 2019), IMF (Cheng et al. 2018;Huang et al. 2018;Miao et al. 2018) and muscle tissues (Cardoso et al. 2017;Sun et al. 2017;Yu et al. 2017;Shen et al. 2019;Wang et al. 2019). ...
Article
Full-text available
lncRNAs play crucial roles in fat metabolism in animals. Previously, we have compared the mRNA transcriptome profiles between seven fat‐type Chinese pig breeds and one lean‐type Western breed (Yorkshire, YY). The associations between differentially expressed (DE) genes and phenotypical traits were investigated. In the present study, to further explore the underlying regulatory mechanisms, lncRNAs were sequenced and compared between YY and Chinese indigenous breeds. The results showed 9114 and 7538 DE lncRNAs between at least one Chinese breed and the YY breed in the adipose and muscle tissue respectively. KEGG enrichment analysis revealed that the target genes of these DE lncRNAs mainly influenced the glucolipid metabolism, which is an important process affecting meat quality. Correlation analyses between the DE lncRNA and DE mRNA genes related to meat quality and growth traits were performed. The results showed that LTCONS_00073280 was associated with intramuscular fat content. Four lncRNAs (LTCONS_00101781, LTCONS_00037879, LTCONS_00088260 and LTCONS‐00128343) might mediate backfat thickness. Overall, this study provides candidate lncRNAs that potentially affect meat quality, which might be useful for molecular breeding of pig breeds in future.
Article
Long non-coding RNAs (LncRNAs) are generally longer than 200 bp in length and play an important regulatory role in the growth and development of skeletal muscle. In the previous work, the non-coding RNAs with abundant expression in bovine tissues were screened out. After quantitative real-time PCR (qPCR), 33 lncRNAs with differential expression in various bovine tissues were identified. Differential expression analysis base on tissue expression profiles of 33 lncRNAs, a long non-coding RNA LncRNA13, which may have effects on bovine muscle development, was found. The expression levels in embryo muscle and adult cattle muscle were significantly different (p < 0.01), so it is speculated that it may have a certain impact on the development of cattle muscle. It was named LncRNA 5.8S rRNA-OT1, and its overexpression vector pcDNA3.1-LncRNA 5.8S rRNA-OT1 was cloned and constructed. The purpose of this study is to further explore its impact on the proliferation and differentiation of bovine muscle cells and accumulate data to lay a foundation for further exploration of the function of LncRNA 5.8S rRNA-OT1 and add basic data for the study of the regulatory mechanism of lncRNA.
Article
Full-text available
Recent studies indicate important roles for long noncoding RNAs (lncRNAs) as essential regulators of myogenesis and adult skeletal muscle regeneration. However, the specific roles of lncRNAs in myogenic differentiation of adult skeletal muscle stem cells and myogenesis are still largely unknown. Here we identify a lncRNA that is specifically enriched in skeletal muscle (myogenesis-associated lncRNA, in short, lnc-mg). In mice, conditional knockout of lnc-mg in skeletal muscle results in muscle atrophy and the loss of muscular endurance during exercise. Alternatively, skeletal muscle-specific overexpression of lnc-mg promotes muscle hypertrophy. In vitro analysis of primary skeletal muscle cells shows that lnc-mg increases gradually during myogenic differentiation and its overexpression improves cell differentiation. Mechanistically, lnc-mg promotes myogenesis, by functioning as a competing endogenous RNA (ceRNA) for microRNA-125b to control protein abundance of insulin-like growth factor 2. These findings identify lnc-mg as a novel noncoding regulator for muscle cell differentiation and skeletal muscle development.
Article
Full-text available
Despite modest sequence conservation and rapid evolution, long non-coding RNAs (lncRNAs) appear to be conserved in expression pattern and function. However, analysis of lncRNAs across tissues and developmental stages remains largely uncharacterized in mammals. Here, we systematically investigated the lncRNAs of the Guizhou miniature pig (Sus scrofa), which was widely used as biomedical model. We performed RNA sequencing across 9 organs and 3 developmental skeletal muscle, and developed a filtering pipeline to identify 10,813 lncRNAs (9,075 novel). Conservation patterns analysis revealed that 57% of pig lncRNAs showed homology to humans and mice based on genome alignment. 5,455 lncRNAs exhibited typical hallmarks of regulatory molecules, such as high spatio-temporal specificity. Notably, conserved lncRNAs exhibited higher tissue specificity than pig-specific lncRNAs and were significantly enriched in testis and ovary. Weighted co-expression network analysis revealed a set of conserved lncRNAs that are likely involved in postnatal muscle development. Based on the high degree of similarity in the structure, organization, and dynamic expression of pig lncRNAs compared with human and mouse lncRNAs, we propose that these lncRNAs play an important role in organ physiology and development in mammals. Our results provide a resource for studying animal evolution, morphological complexity, breeding, and biomedical research.
Article
Full-text available
Little is known how lincRNAs are involved in skeletal myogenesis. Here we describe the discovery of Linc-YY1 from the promoter of the transcription factor (TF) Yin Yang 1 (YY1) gene. We demonstrate that Linc-YY1 is dynamically regulated during myogenesis in vitro and in vivo. Gain or loss of function of Linc-YY1 in C2C12 myoblasts or muscle satellite cells alters myogenic differentiation and in injured muscles has an impact on the course of regeneration. Linc-YY1 interacts with YY1 through its middle domain, to evict YY1/Polycomb repressive complex (PRC2) from target promoters, thus activating the gene expression in trans. In addition, Linc-YY1 also regulates PRC2-independent function of YY1. Finally, we identify a human Linc-YY1 orthologue with conserved function and show that many human and mouse TF genes are associated with lincRNAs that may modulate their activity. Altogether, we show that Linc-YY1 regulates skeletal myogenesis and uncover a previously unappreciated mechanism of gene regulation by lincRNA.
Article
Full-text available
For the past 15 years, the UCSC Genome Browser (http://genome.ucsc.edu/) has served the international research community by offering an integrated platform for viewing and analyzing information from a large database of genome assemblies and their associated annotations. The UCSC Genome Browser has been under continuous development since its inception with new data sets and software features added frequently. Some release highlights of this year include new and updated genome browsers for various assemblies, including bonobo and zebrafish; new gene annotation sets; improvements to track and assembly hub support; and a new interactive tool, the “Data Integrator”, for intersecting data from multiple tracks. We have greatly expanded the data sets available on the most recent human assembly, hg38/GRCh38, to include updated gene prediction sets from GENCODE, more phenotype- and disease-associated variants from ClinVar and ClinGen, more genomic regulatory data, and a new multiple genome alignment.
Article
Full-text available
Data collected from 690 purebred Duroc pigs from 2009 to 2012 were used to estimate the heritability, and genetic and phenotypic correlations between production and meat quality traits. Variance components were obtained through the restricted maximum likelihood procedure using Wombat and SAS version 9.0. Animals were raised under the same management in five different breeding farms. The average daily gain, loin muscle area (LMA), backfat thickness (BF), and lean percent (LP) were measured as production traits. Meat quality traits included pH, cooking loss, lightness (L*), redness (a*), yellowness (b*), marbling score (MS), moisture content (MC), water holding capacity (WHC), and shear force. The results showed that the heritability estimates for meat quality traits varied largely from 0.19 to 0.79. Production traits were moderate to highly heritable from 0.41 to 0.73. Genotypically, the BF was positively correlated (p<0.05) with MC (0.786), WHC (0.904), and pH (0.328) but negatively correlated with shear force (-0.533). The results of genetic correlations indicated that selection for less BF could decrease pH, moisture content, and WHC and increase the shear force of meat. Additionally, a significant positive correlation was recorded between average daily gain and WHC, which indicates pork from faster-growing animals has higher WHC. Furthermore, selection for larger LMA and LP could increase MS and lightness color of meat. The meat quality and production traits could be improved simultaneously if desired. Hence, to avoid further deterioration of pork characteristics, appropriate selection of traits should be considered.
Article
Full-text available
The inability to predict long noncoding RNAs from genomic sequence has impeded the use of comparative genomics for studying their biology. Here, we develop methods that use RNA sequencing (RNA-seq) data to annotate the transcriptomes of 16 vertebrates and the echinoid sea urchin, uncovering thousands of previously unannotated genes, most of which produce long intervening noncoding RNAs (lincRNAs). Although in each species, >70% of lincRNAs cannot be traced to homologs in species that diverged >50 million years ago, thousands of human lincRNAs have homologs with similar expression patterns in other species. These homologs share short, 5'-biased patches of sequence conservation nested in exonic architectures that have been extensively rewired, in part by transposable element exonization. Thus, over a thousand human lincRNAs are likely to have conserved functions in mammals, and hundreds beyond mammals, but those functions require only short patches of specific sequences and can tolerate major changes in gene architecture. Copyright © 2015 The Authors. Published by Elsevier Inc. All rights reserved.
Article
Full-text available
Genetic relationships between growth and carcass traits in South African Large White pigs were estimated. Genetic parameters for growth and carcass traits were generated using a maternal effects model in ASREML. Data analysed were on 13 703 pigs from 28 herds tested between 1990 and 2007, and 4 128 carcasses from 21 herds evaluated between 1993 and 2007. The traits included in the study were backfat thickness (BFAT), test period weight gain (TPG), lifetime weight gain (LTG), test period feed conversion ratio (FCR), age at slaughter (AGES), lean percentage (LEAN), drip-free lean percentage (DLEAN), drip loss percentage (DRIP), carcass length (CRLTH), dressing percentage (DRESS), eye muscle area (AREA) and carcass fat (CFAT). Heritability estimates for growth traits ranged from 0.24 ± 0.03 for FCR to 0.45 ± 0.04 for BFAT, while those for carcass traits ranged from 0.14 ± 0.06 for DRIP to 0.55 ± 0.0.08 for AREA. Maternal genetic effects were significant in most traits, and were negatively correlated with direct heritabilities. Genetic correlations among growth traits ranged from –0.14 ± 0.08 between BFAT and LTG to –0.99 ± 0.01 between TPG and FCR, and LTG and AGES. For carcass traits, genetic correlations ranged from –0.02 ± 0.20 between DRIP and CRLTH to 0.99 ± 0.01 between LEAN and DLEAN. There is substantial genetic variation in growth and carcass traits; hence faster genetic improvement may be achieved through selection. Growth performance and carcass characteristics can be improved through selection for reduced backfat thickness and increased daily weight gain.
Article
We have isolated a novel mouse gene (Gtl2) from the site of a gene trap integration (Gtl2lacZ) that gave rise to developmentally regulated lacZ expression, and a dominant parental-origin–dependent phenotype. Heterozygous Gtl2lacZ mice that inherited the transgene from the father showed a proportionate dwarfism phenotype, whereas the penetrance and expressivity of the phenotype was strongly reduced in Gtl2lacZ mice that inherited the transgene from the mother. Gtl2 expression is highly similar to the β-galactosidase staining pattern, and is down-regulated but not abolished in mice carrying the Gtl2lacZ insertion. In early postimplantation embryos, Gtl2 is expressed in the visceral yolk sac and embryonic ectoderm. During subsequent development and organogenesis, Gtl2 transcripts are abundant in the paraxial mesoderm closely correlated with myogenic differentiation, in parts of the central nervous system, and in the epithelial ducts of developing excretory organs. The Gtl2 gene gives rise to various differentially spliced transcripts, which contain multiple small open reading frames (ORF). However, none of the ATG codons of these ORFs is in the context of a strong Kozak consensus sequence for initiation of translation, suggesting that Gtl2 might function as an RNA. Nuclear Gtl2 RNA was detected in a temporally and spatially regulated manner, and partially processed Gtl2 transcripts were readily detected in Northern blot hybridizations of polyadenylated RNA, suggesting that primary Gtl2 transcripts are differently processed in various cell types during development. Gtl2 transcript levels are present in parthenogenic embryos but may be reduced, consistent with the pattern of inheritance of the Gtl2lacZ phenotype. Dev. Dyn. 1998;212:214–228. © 1998 Wiley-Liss, Inc.
Article
Microvascular dysfunction is an important characteristic of diabetic retinopathy. Long non-coding RNAs (lncRNAs) play important roles in diverse biological processes. In this study, we investigated the role of lncRNA-MEG3 in diabetes-related microvascular dysfunction. We show that MEG3 expression level is significantly down-regulated in the retinas of STZ-induced diabetic mice, and endothelial cells upon high glucose and oxidative stress. MEG3 knockdown aggravates retinal vessel dysfunction in vivo, as shown by serious capillary degeneration, and increased microvascular leakage and inflammation. MEG3 knockdown also regulates retinal endothelial cell proliferation, migration, and tube formation in vitro. The role of MEG3 in endothelial cell function is mainly mediated by the activation of PI3k/Akt signaling. MEG3 up-regulation may serve as a therapeutic strategy for treating diabetes-related microvascular complications.
Article
Increasing evidence suggests that long non-coding RNAs (LncRNAs) represent a new class of regulators of stem cells. However, the roles of LncRNAs in stem cell maintenance and myogenesis remain largely unexamined. For this study, hundreds of intergenic LncRNAs were identified that are expressed in myoblasts and regulated during differentiation. One of these LncRNAs, termed LncMyoD, is encoded next to the Myod gene and is directly activated by MyoD during myoblast differentiation. Knockdown of LncMyoD strongly inhibits terminal muscle differentiation, largely due to a failure to exit the cell cycle. LncMyoD directly binds to IGF2-mRNA-binding protein 2 (IMP2) and negatively regulates IMP2-mediated translation of proliferation genes such as N-Ras and c-Myc. While the RNA sequence of LncMyoD is not well conserved between human and mouse, its locus, gene structure, and function are preserved. The MyoD-LncMyoD-IMP2 pathway elucidates a mechanism as to how MyoD blocks proliferation to create a permissive state for differentiation. Copyright © 2015 Elsevier Inc. All rights reserved.