ArticlePDF Available

Expressed microRNA associated with high rate of egg production in chicken ovarian follicles

Wiley
Animal Genetics
Authors:

Abstract and Figures

MicroRNA (miRNA) is a highly conserved class of small noncoding RNA about 19-24 nucleotides in length that function in a specific manner to post-transcriptionally regulate gene expression in organisms. Tissue miRNA expression studies have discovered a myriad of functions for miRNAs in various aspects, but a role for miRNAs in chicken ovarian tissue at 300 days of age has not hitherto been reported. In this study, we performed the first miRNA analysis of ovarian tissues in chickens with low and high rates of egg production using high-throughput sequencing. By comparing low rate of egg production chickens with high rate of egg production chickens, 17 significantly differentially expressed miRNAs were found (P < 0.05), including 11 known and six novel miRNAs. We found that all 11 known miRNAs were involved mainly in pathways of reproduction regulation, such as steroid hormone biosynthesis and dopaminergic synapse. Additionally, expression profiling of six randomly selected differentially regulated miRNAs were validated by quantitative real-time polymerase chain reaction (RT-qPCR). Some miRNAs, such as gga-miR-34b, gga-miR-34c and gga-miR-216b, were reported to regulate processes such as proliferation, cell cycle, apoptosis and metastasis and were expressed differentially in ovaries of chickens with high rates of egg production, suggesting that these miRNAs have an important role in ovary development and reproductive management of chicken. Furthermore, we uncovered that a significantly up-regulated miRNA-gga-miR-200a-3p-is ubiquitous in reproduction-regulation-related pathways. This miRNA may play a special central role in the reproductive management of chicken, and needs to be further studied for confirmation.
Content may be subject to copyright.
Expressed microRNA associated with high rate of egg production in
chicken ovarian follicles
N. Wu
1
, U. Gaur
1
, Q. Zhu, B. Chen, Z. Xu, X. Zhao, M. Yang and D. Li
Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu
610000, China.
Summary MicroRNA (miRNA) is a highly conserved class of small noncoding RNA about 1924
nucleotides in length that function in a specific manner to post-transcriptionally regulate
gene expression in organisms. Tissue miRNA expression studies have discovered a myriad of
functions for miRNAs in various aspects, but a role for miRNAs in chicken ovarian tissue at
300 days of age has not hitherto been reported. In this study, we performed the first miRNA
analysis of ovarian tissues in chickens with low and high rates of egg production using
high-throughput sequencing. By comparing low rate of egg production chickens with high
rate of egg production chickens, 17 significantly differentially expressed miRNAs were
found (P<0.05), including 11 known and six novel miRNAs. We found that all 11 known
miRNAs were involved mainly in pathways of reproduction regulation, such as steroid
hormone biosynthesis and dopaminergic synapse. Additionally, expression profiling of six
randomly selected differentially regulated miRNAs were validated by quantitative real-time
polymerase chain reaction (RT-qPCR). Some miRNAs, such as gga-miR-34b, gga-miR-34c
and gga-miR-216b, were reported to regulate processes such as proliferation, cell cycle,
apoptosis and metastasis and were expressed differentially in ovaries of chickens with high
rates of egg production, suggesting that these miRNAs have an important role in ovary
development and reproductive management of chicken. Furthermore, we uncovered that a
significantly up-regulated miRNAgga-miR-200a-3pis ubiquitous in reproduction-
regulation-related pathways. This miRNA may play a special central role in the
reproductive management of chicken, and needs to be further studied for confirmation.
Keywords egg-laying, illumina sequencing, Luhua chicken, reproduction regulation, RT-
qPCR
Introduction
MicroRNAs (miRNA), which are extensively expressed in a
variety of organisms and tissues, comprise a class of
endogenous small noncoding single-stranded RNA about
1924 nucleotides (nt) in length (Bartel 2004). Mature
miRNAs from the transcripts of miRNA genes are produced
through processing from the one arm of fold back precursors
(pre-miRNAs) to form approximately 70-nt hairpin sec-
ondary structures (Lee et al. 2004; Kim 2005; Carthew &
Sontheimer 2009). Cellular endogenous miRNA plays
important roles in regulating various biological and meta-
bolic processes, including organogenesis, cell proliferation,
differentiation, development, hematopoiesis, apoptosis,
tumorigenesis and many other cellular processes (Lim et al.
2003; Bartel 2004; Mansfield et al. 2004). According to an
assessment, one miRNA can regulate the expression of
hundreds of mRNAs, and the expression of one mRNA can be
regulated by hundreds of miRNAs (Krek et al. 2005). In other
words, miRNA construct networks of sophisticated regulator
control systems in organisms and play very significant roles.
Genome-wide miRNA expression has been explored in
gonads of cattle (Huang et al. 2011), mice (Ro et al. 2007;
Mishima et al. 2008), sheep (McBride et al. 2012) and pigs (Li
et al. 2011). These studies revealed that miRNAs have a
significantfunction in the development of mammalian gonads.
Some miRNAs have been identified in chicken, and expression
has been reported in several processes, such as embryo
development (Darnell et al. 2006; Glazov et al. 2008; Hicks
Address for correspondence
D. Li, Farm Animal Genetic Resources Exploration and Innovation Key
Laboratory of Sichuan Province, Sichuan Agricultural University,
Chengdu 610000, China.
E-mail: diyanli@sicau.edu.cn
1
These authors contributed equally to this work.
Accepted for publication 01 September 2016
doi: 10.1111/age.12516
1
©2016 Stichting International Foundation for Animal Genetics
et al. 2008, 2010; Bannister et al. 2009; Rathjen et al. 2009;
Wang et al. 2009; Li et al. 2012), immune organ function
(Hicks et al. 2009), germ cell development (Burnside et al.
2008; Lee et al. 2011) and disease (Yu et al. 2008; Tian et al.
2012; Wang et al. 2013). The chicken (Gallus gallus)isan
important agricultural and avian-model species, and it is a
major source of protein worldwide. However, a role for
miRNAs in chicken ovarian development has not hitherto
been reported clearly. The characteristics of ovarian tissue are
highly related to reproductive and economic traits of chicken,
so it is necessary to identify and characterize the miRNA in
ovarian tissue.
In recent years, the development of next-generation
sequencing, which is known as deep sequencing or high-
throughput sequencing technology, has provided a power-
ful, highly reproducible and cost-efficient tool for transcrip-
tomic research (Morozova & Marra 2008). In previous
studies, the catalog of chicken miRNA expression during
embryonic development was characterized using deep
sequencing (Glazov et al. 2008), providing a strong back-
ground of information for analysis of tissue-specific miRNA
signatures. In addition, the genome sequence is available for
chicken, enhancing its use as a model for functional
genomics studies during development. Thus, by means of
miRNA sequencing, we aimed to identify relevant expres-
sive miRNA associated with high rates of egg production
and to extend the repertoire of known miRNAs in chicken
ovarian tissue. Moreover, we can screen suitable miRNAs to
use them as molecular markers in the application of genetic
selection in the breeding programs of chicken.
The Luhua chicken breed (Gallus gallus domesticus), orig-
inally found mainly in Wenshang county of Shandong
province, China, has been recognized as a commercial dual-
purpose eggmeat type chicken (Ruan & Zheng 2011; Hu
et al. 2013). It is a common strain especially selected for its
superior reproductive performance such as high rates of egg
production. In poultry breeding programs, egg number at
300 days of age is usually used as the most valuable indicator
of total egg production potential (Li et al. 2013). In the
present study, we sampled the ovarian tissues of three low-
rate egg production (LP) and three high-rate egg production
(HP) Luhua chickens at age 300 days. Comprehensive
miRNA profiles of ovarian tissue from LP and HP chickens
were generated, and a comparative analysis of miRNA data
was performed. This miRNA data will be very useful in further
understanding the functions of chicken ovarian tissues and
will contribute to the investigation of the regulatory mech-
anism of miRNA in avian species.
Materials and methods
Experimental animals
Eight hundred Luhua chickens from the Experimental
Chicken Farm of Sichuan Agricultural University were used
in this study. A total of 200 LP chickens and 200 HP
chickens were bi-directionally selected according to their
number of eggs at 250 days of age. Six reproductive traits
(body weight at first egg, weight of first egg, age at first egg,
number of eggs at 300 days of age, body weight at 300 days
of age and egg weight at 300 days of age) were recorded for
both groups. The average number of eggs at 300 days of
age (mean SEM) was 111 2.1 and 144 4.3 for LP
and HP chickens respectively. Egg production cycles of these
chickens were also recorded every day. According to their
similar reproductive traits and regular egg production cycle
(about 2 hours prior to ovulation), three LP (B4, B5, B7)
and three HP (A2, A3, A9) chickens (Table S1) were
selected for tissue collection. Experimental procedures were
approved by the Committee on the Care and Use of
Laboratory Animals of the State-level Animal Experimental
Teaching Demonstration Center of Sichuan Agricultural
University (Approval ID: S20141010) and performed in
accordance with the Regulations for the Administration of
Affairs Concerning Experimental Animals (China, 1988) for
animal experiments. All efforts were made to minimize the
suffering of the chickens.
Tissue collection, RNA isolation and small RNA
sequencing
Six chickens were sacrificed by exsanguination at 300 days
of age, and the ovarian tissues, which contained the entire
collection of various-sized follicles (including hierarchical
and all prehierarchical follicles), were collected about
2 hours prior to ovulation. The egg yolks of the follicles
were removed in the process of sample collection. Samples
were quickly stored in RNAlater (Ambion
â
), frozen in liquid
nitrogen and stored at 80 °C until RNA extraction. Total
RNA was isolated from these tissues using an RNeasy Mini
Kit (Qiagen), in accordance with the manufacturer’s
instructions. The quantity and purity of total RNA were
monitored by a Qubit
â
3.0 Fluorometer (Thermo Fisher
Scientific) and formaldehydeagarose gel electrophoresis
and then stored at 80 °C.
To identify ovarian miRNAs in chickens, complementary
DNA (cDNA) libraries for small RNA from chicken ovarian
tissues were constructed according to published miRNA
cloning protocols (Lagos-Quintana et al. 2001; Lau et al.
2001) and sequenced using an HiSeq2500 (Illumina)
followed by NEXTflexTM Small RNA-seq Kits (BIOO Scientific
Corp.).
Global annotation of miRNA sequences
The raw data, consisting of 1735 nt sequence adaptors,
first went through a data cleaning process to remove low
quality adaptors, having quality values less than 20, 50and
30primer contaminants, N adaptors, polyA adaptors,
sequences without insert tags and adaptors shorter than
©2016 Stichting International Foundation for Animal Genetics, doi: 10.1111/age.12516
Wu et al.2
17 nt and longer than 35 nt. Subsequently, standard
bioinformatic analyses were carried out to align or annotate
the clean adaptors. The clean reads were mapped to the
ensemble chicken genome galGal4 using NCBI MEGABLAST,
and rRNA, tRNA, miscRNA, snRNA and snoRNA were
discarded from the small RNA sequences; the remaining
sequences were again searched against the miRBase 21.0
database of known Gallus gallus miRNA sequences with
zero or one mismatch (http://www.mirbase.org/) were
continued (Ambros et al. 2003; Griffiths-Jones 2004; Grif-
fiths-Jones et al. 2006, 2008). The sequences matching
Gallus gallus miRBase were considered known miRNA
sequences, and the expression of all miRNAs was assayed.
Next, after filtering known miRNA sequences, the remain-
ing sequences were BLAST searched against the Gallus gallus
genome. The sequences matching chicken genome
sequences (except for one to three 50-or3
0-end nt) were
used to predict novel miRNAs by MIRDEEP2 (https://
www.mdcberlin.de/8551903/en/research/research_teams/
systems_biology_of_gene_regulatory_elements/projects/miR
Deep) using default parameters. These sequences were
considered as a potential novel miRNA, and expression of all
miRNAs was assayed.
Hierarchical cluster analysis of differentially expressed
miRNAs
Differential expression for known and novel miRNAs
were analyzed using EDGER (http://www.bioconductor.org/
packages/release/bioc/html/edgeR.html). Reads per million
miRNA mapped values were used to represent miRNA
expression levels. P-values were calculated using right-
tailed Fisher’s exact test. P<0.05 and |log
2
fold change|1
were used to screen differentially expressed miRNAs. A
hierarchical cluster analysis was performed for known and
novel miRNAs with similar expression patterns (Zhang et al.
2009).
MiRNA target gene prediction and functional
annotation
The miRNA targets were predicted by analyzing the
putative miRNA binding sites in the libraries. The miRNA
target prediction software MIRDB (http://mirdb.org/miRDB/
index.html) (Wong & Wang 2014) was used to predict the
binding sites of differentially expressed miRNA. The Tar-
getScan principle (http://www.targetscan.org/) was also
applied in the prediction procedures. The intersection of the
two prediction programs was selected for this study.
The BLAST2GO program was used to conduct gene ontology
(GO) annotations and GO functional classifications (Conesa
et al. 2005) of these predicted miRNA target genes. In GO
terms, P-value 0.001 was used to identify the significantly
enriched GO terms. These genes were also submitted to the
KEGG (Kyoto Encyclopedia of Genes and Genomes) database
for enrichment analyses. The P-value denotes the signifi-
cance of the pathway correlated to the conditions. The
lower the P-value, the more significant the metabolism
pathway (P-value cut-off was 0.05).
Validation of miRNA expression by real-time PCR (RT-
qPCR)
To validate the reliability of the Illumina analysis, we
randomly tested the expression of eight miRNAs, including
six differentially expressed miRNAs and two similarly
expressed miRNAs. Reverse transcription (RT) real-time
PCR was used to quantify the expression of eight mature
miRNAs. The RT-qPCR primers were designed using PRIMER
5.0 (http://downloads.fyxm.net/Primer-Premier-101178.
html) and are listed in Table S2 (miRNA-specific primers
were synthesized by Shanghai Biological Technology Co.
and universal primers were provided by miRcute miRNA
qPCR Detection kit, Aidlab).
Briefly, the total RNA of each sample was reverse-
transcribed with miRNA-specific RT primers using the First-
strand cDNA Synthesis Kit (Thermo Scientific Fermentas).
First, 2 llof109poly(A) polymerase buffer, 2 llof109
rATP Solution, 0.4 ll of E. coli Poly(A) Polymerase (5 U/ll)
and 2 ll of total RNA (10 pgll
1
~1.5 lgll
1
) were added
into ice-cold RNase-Free reaction tubes and supplemented
with RNase-Free ddH
2
O to a total volume of 20 ll. After
brief centrifugation, the mixture was incubated for 50 min
at 37 °C. Second, 3 ll of the Poly (A) reaction mixture
obtained in the first step was added to a solution containing
2llof25lmol RT Primer, 4 llof59RT Buffer Reaction
Mix, 0.8 ll of TUREscript H
RTase (200 U/ll) and 10.2 ll
of RNase-Free ddH
2
O. After brief centrifugation, the reac-
tion solutions were incubated at 42 °C for 50 min, at 70 °C
for 15 min and then held at 20 °C.
The expression of mature miRNAs was detected using the
SYBR green method. RT-qPCR was performed in a 96-well
plate using the Bio-Rad iQ5 Real-time PCR Detection
System, according to the protocol. In a 20-ll reaction
mixture, 1.0 ll of cDNA was used as a template, with 10 ll
of SYBR Select Master Mix (Applied Biosystems), 0.4 llof
specific forward primer and 0.4 ll of universal primer, with
the following program: 94 °C for 3 min, followed by 42
cycles of 94 °C for 20 s and the optimal temperature for
40 s. 5.8S rRNA, which has relatively stable expression in
most tissues, was used as an endogenous control (Elela &
Nazar 1997), and the expression level of 5.8S rRNA was
used to normalize the RT-qPCR results for each miRNA. All
reactions were run in three technical replicates, including
negative controls without template. Fold changes of miRNA
expression were calculated using the 2
DDCt
method (versus
5.8S rRNA) (Schmittgen & Livak 2008). All of the data
were expressed as the mean standard deviation, and a
statistical analysis using Student’s t-tests was performed
with SPSS 16.0 (SPSS Inc.).
©2016 Stichting International Foundation for Animal Genetics, doi: 10.1111/age.12516
Ovarian microRNA transcriptome of chicken 3
Accession code
The Illumina HiSeq 2500 sequencing data for the Luhua
chicken ovarian miRNA sequences has been deposited in
the NCBI Sequence Read Archive (SRA, http://
www.ncbi.nlm.nih.gov/Traces/sra) with accession no.
SRP062266.
Results
Overview of miRNA sequencing
A total of 30.20 million and 30.37 million raw reads were
obtained from ovarian tissues of LP and HP chickens
respectively. After filtering the low-quality sequences,
28 501 792 and 28 004 558 clean reads from the HP
(93.83% clean) and LP (92.72% clean) chicken ovary
libraries respectively were selected for further analysis
(Table S3). The total and unique read length distributions
of LP and HP chickens are shown in Fig. S1. Among the
selected reads, the size distribution of small RNAs for
sequencing was similar in six samples. The majority of these
total small RNAs ranged from 2024 nt, and unique reads
were distributed in mainly 2123 nt; there was no obvious
difference between the LP and HP chickens. The results are
consistent with the typical size range of small RNAs and
indicated that the clean reads included a large number of
miRNA sequences.
Annotation of small RNA sequences
Small RNA sequences identified by Illumina small RNA deep
sequencing were compared with known mature miRNAs
and precursors in miRBase 21.0. The unique and all small
RNAs were annotated as miRNA, snRNA, tRNA, rRNA,
snoRNA, miscRNA (repeat and polII-transcribed) and
unannotated for the LP and HP chickens (Table S4). The
HP chickens had a larger number of small RNAs and unique
small RNAs than did the LP chickens. Most small RNAs in
both groups were miRNAs (82%) and unannotated (16%)
for all small RNAs, whereas for unique small RNAs, HP
chickens had the greatest proportion of miRNAs (41%) and
LP chickens had only 33% of miRNAs as the second major
class (Fig. S2). This revealed that HP chickens produce more
unique miRNAs, but the difference is not significant
(P>0.05). Proportions of the remaining categories of small
RNAs, including tRNA, rRNA, snRNA, miscRNA or
snoRNA, were relatively lower (<1.2%).
Expression of known and novel miRNAs in chicken
ovaries
In the present study, 562 known miRNAs were identified in
all samples including 507 in HP and 496 in LP chickens
(Table S5). Among the 562 sequenced mature miRNAs,
441 (78.5%) unique miRNAs were expressed in all LP and
HP chickens; however, 66 (11.7%) and 55 (12.5%) were
specifically expressed in HP and LP chickens respectively. In
the miRNA expression profile, the read numbers of the top
10 miRNAs accounted for 81.93% of the total reads from
LP chickens and 78.3% of the total reads from HP chickens.
The miRNA expression profile revealed that most miRNAs
were expressed by a small portion of miRNA genes.
Interestingly, we found that the miRNAs with the 19
highest expression levels were the same for both groups.
Among them, miR-99a-5p and gga-miR-26a-5p displayed
more than 7 500 000 reads and had the highest expression
level, followed by gga-miR-199-5p and gga-miR-10a-5p,
which displayed about 2 500 000 reads each. Some
miRNAs (such as gga-miR-222b-3p and gga-miR-1729-
5p) displayed fewer than about 1 000 reads, thereby
indicating that expression varied significantly among
different miRNAs; this result is consistent with previous
studies (Gu et al. 2014).
A total of 470 novel miRNAs were predicted in this study,
and 389 and 384 novel miRNAs were identified in HP and
LP chickens respectively (Table S6). Among the 470
predicted novel miRNAs, 303 (64.5%) unique miRNAs
were expressed in both groups; however, 86 (18.3%) and
55 (17.2%) were specifically expressed in HP and LP
chickens respectively. The novel miRNA expression profile
also revealed that most novel miRNAs were expressed by a
small portion of miRNA genes. Moreover, we found that the
sequencing frequencies of novel miRNAs were much lower
compared to those of known miRNAs. The same expression
pattern has also been reported in other species (Chi et al.
2011; Xu et al. 2013), which suggests that novel miRNAs
are usually weakly expressed whereas known miRNA genes
are highly expressed.
Hierarchical cluster analysis of differentially expressed
miRNAs
We first performed a correlation analysis to assess the
variance of miRNA expression across replicates. All samples
showed high reproducibility in the same group, with
Pearson correlation coefficients >0.85 (Fig. 1). Known and
novel miRNAs were compared between the two groups to
identify differentially expressed miRNAs. The results of the
analysis demonstrated that there were 17 significantly
differentially expressed miRNAs, which were divided into
two categories, the upper part comprising eight up-
regulated miRNAs and the lower half nine down-regulated
miRNAs in HP chickens (Table 1 & Fig. 2). In the cluster
analysis, miRNAs that showed similar patterns of differen-
tial expression in different sample pairs were clustered
together. Based on miRBase 21.0, these 17 miRNAs
comprised five clusters, of which one cluster contained
eight miRNAs, one cluster contained five miRNAs, one
©2016 Stichting International Foundation for Animal Genetics, doi: 10.1111/age.12516
Wu et al.4
cluster contained two miRNAs and two clusters contained
one miRNA (Fig. S3).
Target gene prediction and gene functional annotation
Target prediction is an important way to determine the
functions of miRNAs. We predicted a total of 1305 target
genes among the differentially expressed miRNAs
(Table S7). To probe the biological roles of the differentially
expressed miRNAs, all of the predicted targets in this study
were mapped to terms in the GO and KEGG databases. A
total of 128 significantly enriched GO terms and 18 KEGG
pathways regulated by 11 differentially expressed known
miRNAs were identified (Table S8). Among the top 30
significantly GO terms for biological process, most target
genes were associated with biological regulation, single-
organism cellular process, single-organism process and
regulation of metabolic process (Fig. 3). For cellular
component, target genes were significantly associated with
intracellular part. For molecular function, target genes
were significantly related to protein binding and ion
binding. Results of the KEGG analysis showed that these
target genes were involved mainly in endocytosis, miRNAs
in cancer, glycerol phospholipid metabolism, thyroid hor-
mone signaling pathway and ubiquitin mediated proteol-
ysis pathways (Fig. 4). Involvement in cancer pathways
suggest that the differentially expressed miRNAs play a
regulatory role in cell proliferation and cell cycle
progression. Notably, a specific enrichment of genes was
found in some reproduction regulation pathways, such as
steroid hormone biosynthesis, dopaminergic synapse,
GnRH signaling pathways, oxytocin signaling pathway,
oocyte meiosis, calcium signaling pathways, progesterone-
mediated oocyte maturation, endocrine and other factor-
regulated calcium reabsorption and MAPK signaling
pathway (Table S9).
A_ovary
B_ovary
A_ovary
B_ovary
A
3_ovary
A
2_ovary
A
9_ovary
B4_ovary
B5_ovary
B7_ovary
A3_ovar
y
A2_ovar
y
A9_ovar
y
B4_ovary
B5_ovary
B7_ovary
0.7 0.8 0.9 1
Value
Color key
Figure 1 Correlation analysis of miRNA
expression across samples of LP and HP
chickens.
Table 1 miRNAs significantly differentially expressed between low-
and high-rate egg production chickens.
miRNA logFC P-value Up-/down-regulated
gga-miR-34b-5p 3.73965 0.00003 Up
gga-miR-34c-5p 2.56597 0.00021 Up
gga-miR-34b-3p 1.74087 0.02398 Up
gga-novel-18-star 4.34580 0.03305 Up
gga-miR-34c-3p 1.93322 0.03626 Up
gga-miR-200a-3p 1.29779 0.03646 Up
gga-novel-31-mature 2.27375 0.03687 Up
gga-miR-1641 2.61869 0.04540 Up
gga-miR-1744-3p 3.16434 0.00086 Down
gga-novel-280-mature 2.55983 0.00944 Down
gga-miR-1655-5p 1.85425 0.02031 Down
gga-novel-73-star 2.66111 0.02283 Down
gga-miR-216b 2.75966 0.02540 Down
gga-miR-1734 3.19824 0.03786 Down
gga-novel-79-mature 4.02767 0.03809 Down
gga-novel-81-mature 4.02779 0.03823 Down
gga-miR-7465-3p 1.71936 0.04816 Down
©2016 Stichting International Foundation for Animal Genetics, doi: 10.1111/age.12516
Ovarian microRNA transcriptome of chicken 5
Verification of miRNA and targeted gene expression
RT-qPCR detection assays were used to confirm the
expression of certain differentially expressed miRNAs in
the LP and HP chickens. The expression of eight randomly
selected miRNAs was verified by real-time PCR (Table 2 &
Fig. 5). The expression of gga-miR-34b-3p, gga-miR-34c-3p
and gga-miR-200a-3p were up-regulated, whereas gga-
miR-1744-3p, gga-miR-216b and gga-miR-1655-5p were
down-regulated and gga-miR-99a-5p and gga-miR-26a-5p
showed no significant difference, which was in agreement
with the high-throughput sequencing results (Table 1). The
discrepancies with respect to ratio may be attributed to the
essentially different algorithms and sensitivities between the
two techniques.
We further investigated the potential roles of the 17
miRNAs in regulating targeted gene expression using RNA-
seq. Thirty-nine predicted targets of 10 miRNAs showed
evidence of differential expression between HP and LP
chickens (Fig. S4). Among them, most genes have been
demonstrated to play important roles in regulating organo-
genesis, development, tumorigenesis and many other pro-
cesses (Moreau et al. 1997; Sarbassov et al. 2004; Kuo et al.
2011; Kanda et al. 2016). Our results suggest that these
genes are also associated with reproductive traits of hens.
These results provide invaluable insights into candidate genes
for reproductive traits and selective breeding of chicken.
Discussion
The miRNA sequences we identified were generally close to
22 nt in length, in agreement with previous reports (Yuan
et al. 2014; Li et al. 2015), and annotation of RNA
0 5 10 15
−4 −2 0 2 4
MA plot
logCounts
logFC
−4 −2 0 2 4
01234
Volcano plot
logFC
−log10 (P value)
Figure 2 Scatter plot of the high-throughput
sequencing data. The high-throughput
sequencing data (differentially expressed
miRNAs) are graphed on the scatter plot to
visualize variations in miRNA expression
between HP and LP chickens. The graph
reflects the fold change value (HP/LP) distri-
bution in the differentially expressed miRNA
numbers. In both plots, red dots represent the
differentially expression miRNAs, and black
dots represent the similarly expressed miRNAs.
GO classification
Target gene number
0
200
400
600
800
1000
1200
Small GTPase mediated signal transduction
Intracellular signal transduction
Ras protein signal transduction
Regulation of response to stimulus
Regulation of signal transduction
Biological regulation
Cellular protein modification process
Protein modification process
Macromolecule modification
Single−organism cellular process
GTP metabolic process
Guanosine−containing compound metabolic process
Phosphorus metabolic process
Phosphate−containing compound metabolic process
GTP catabolic process
Single−organism process
Guanosine−containing compound catabolic process
Regulation of small GTPase mediated signal transduction
Regulation of catabolic process
Regulation of GTP catabolic process
Regulation of Rho protein signal transduction
Rho protein signal transduction
Regulation of Ras protein signal transduction
Regulation of metabolic process
Regulation of cell communication
Regulation of GTPase activity
Intracellular
Intracellular part
Protein binding
Ion binding
Biological_process
Cellular_component
Molecular_function
Figure 3 Selected top 30 GO categories of predicted target genes regulated by the differentially expressed miRNAs.
©2016 Stichting International Foundation for Animal Genetics, doi: 10.1111/age.12516
Wu et al.6
distribution showed that the clean reads included a myriad
of miRNA sequences. All of the above results suggest that
the major clean reads mapped to known miRNAs in
miRBase were highly enriched and that the deep sequenc-
ing data are representative of the miRNA expression profile
of ovarian tissues and would be reliable for subsequent
analyses and prediction of novel miRNAs.
In the ovary libraries, gga-miR-99a-5p and gga-miR-26a-
5p were the two most frequently sequenced miRNAs
(>7 500 000 reads). Previous studies demonstrated that
mir-99a and mir-99b can inhibit proliferation of c-Src-
transformed cells and prostate cancer cells by targeting
mTOR (Oneyama et al. 2011; Sun et al. 2011). They were
also identified as two novel target miRNA genes of
transforming growth factor-b (TGF-b) and played important
roles during TGF-b-induced epithelial to mesenchymal
transition (EMT) of NMUMG cells (Turcatel et al. 2012).
TGF-bis a secreted cytokine that regulates a variety of
processes in development and cancer including EMT (Bierie
& Moses 2006a,b). EMT is a key process during embryonic
development and disease development and progression.
miR-26a is reported to have anti-apoptotic effects on many
cancers (Garzon et al. 2006; Saito & Jones 2006; Zhang
et al. 2011). It was also found to play a role in normal tissue
growth and development and to have an impact on cell
proliferation and differentiation (Luzi et al. 2008; Wong &
Tellam 2008). One study showed that miR-26a regulates
osteoblast cell growth and differentiation in human adipose
tissue-derived stem cells (Luzi et al. 2008). Among other
miRNAs, we found that the let-7 miRNA family was
another abundant cluster with let-7a-5p being the most
abundantly expressed miRNA. The let-7 miRNA family was
also found to be abundantly expressed in ovary and oocyte
of bovines (Tripurani et al. 2010; Huang et al. 2011b; Miles
et al. 2012), as well as in murine ovaries and testis (Reid
et al. 2008). Furthermore, gga-miR-10a-5p, gga-miR-146c-
5p, gga-miR-21-5p, gga-miR-148a-3p, gga-miR-126-3p
and gga-miR-30d were abundant in our sequencing
libraries, as has been shown in other animal gonads
(Mishima et al. 2008; Md Munir et al. 2009; Tripurani et al.
2010; Kang et al. 2013). The significant biological func-
tions of these miRNAs imply that they have important roles
in the female reproductive physiology of chicken.
Among the significantly differentially expressed miRNAs,
gga-miR-34b and gga-miR-34c (both including 3p and 5p)
exhibited a significant increase in the HP ovary compared with
Alanine
Bacterial invasion of epithelial cells
Circadian rhythm
Circadian rhythm − fly
Endocytosis
Epithelial cell signaling in Helicobacter pylori infection
ErbB signaling pathway
Fc gamma R−mediated phagocytosis
Glycerophospholipid metabolism
Inositol phosphate metabolism
Lysine degradation
MicroRNAs in cancer
Mucin type O−glycan biosynthesis
Phosphatidylinositol signaling system
SNARE interactions in vesicular transport
Thyroid hormone signaling pathway
Ubiquitin mediated proteolysis
Vasopressin−regulated water reabsorption
0 5 10 15 20 25
Gene number
Pathway term
0.01
0.02
0.03
0.04
P value
Figure 4 Top 18 pathways of predicted target genes regulated by the differentially expressed miRNAs.
©2016 Stichting International Foundation for Animal Genetics, doi: 10.1111/age.12516
Ovarian microRNA transcriptome of chicken 7
the LP ovary. Previous studies identified the mir-34 family as a
p53 target and a potential tumor suppressor for regulating
processes such as proliferation, cell cycle, apoptosis and
metastasis (Bommer et al. 2007; He et al. 2007a,b; Hermeking
2009). The expression of miR-34a, b and c appears to be
correlated with p53 and was found in over 50% of human
cancers, including pancreatic cancer (Ji et al. 2009), lung
adenocarcinoma (Okada et al. 2014), breast cancer (Kato et al.
2009), gastric cancer (Ji et al. 2008), ovarian cancer (Corney
et al. 2010) and so on. Some studies also revealed that, in
Drosophila, miR-34 loss triggers a gene profile of accelerated
brain aging, late-onset brain degeneration and a catastrophic
decline in survival; miR-34 upregulation extends median
lifespan and mitigates neurodegeneration induced by human
pathogenic poly-glutamine disease protein; and miR-34 can
regulate age-associated events and long-term brain integrity to
modulate aging and neurodegeneration (Liu et al. 2012). In the
present study, gga-miR-34b and gga-miR-34c both had a
significant increase in HP ovary compared with LP ovary, and
gga-miR-34b-5p had the highest levela 13.36-fold increase
in HP ovary, implying that gga-miR-34 plays an important
role in reproductive management in hens.
Previous studies reported that miR-216b attenuated
nasopharyngeal carcinoma cell proliferation, invasion and
tumor growth in nude mice and that it mediated its tumor
suppressor function, at least in part, by suppressing
downstream pathways of KRAS, such as in the PI3K-AKT
and MEK-ERK pathways (Deng et al. 2011). Other studies
also showed that miR-216b inhibits cell proliferation,
migration and invasion of hepatocellular carcinoma by
regulating insulin-like growth factor 2 mRNA-binding
protein 2 and that it is regulated by the hepatitis B virus
x protein (Liu et al. 2015). In the present study, gga-miR-
216b was down-regulated 6.77-fold in HP ovary, sug-
gesting that the suppressing expression of gga-miR-216b
may be beneficial for the improvement of egg laying in hens.
Among the KEGG pathways, some pathways associated
with endocytosis, cancer, Fc gamma R-mediated phagocy-
tosis, snare interactions in vesicular transport, mucin type
o-glycan biosynthesis, lysine degradation, ubiquitin
mediated proteolysis, alanine and some signal transduction
pathways, such as the phosphatidylinositol signaling sys-
tem, thyroid hormone signaling pathway and ErbB signal-
ing pathway, were all significantly enriched. This indicates
a role of the differentially expressed miRNAs in the
regulation of cell motility, cell proliferation, cell nutrition,
nervous system development and function, communication
between cells and the extracellular matrix. In addition, a
small number of genes involved in the epithelial cell
signaling pathways in Helicobacter pylori infection and
bacterial invasion of epithelial cells suggests that the hens
were involved in a stage of immune regulation.
We also found that specific enrichment of predicted
targeted genes was involved in some reproduction-related
pathways, such as steroid hormone biosynthesis, dopamin-
ergic synapse and GnRH signaling pathways. Some of these
genes have been demonstrated to play important roles in
ovary development and reproductive management of hens;
for example, FSHB produces a pituitary glycoprotein
hormone, FSH, that plays a key role in the reproductive
system of chicken, including steroidogenesis, folliculogene-
sis and follicular maturation (Choi et al. 2005). Moreover,
we observed that all differentially expressed known miRNAs
were involved in reproduction-related pathways, and bio-
logical functions of most miRNAs have not been reported.
This provides us with a guideline to explore their unknown
roles in reproductive management of hens. Furthermore, we
uncovered a miRNAgga-miR-200a-3pthat is ubiqui-
tous in most reproduction-regulation-related pathways. The
miR-200a family was reported to play a critical role in
tumor progression and metastasis, specifically in the main-
tenance of the epithelial phenotype by targeting ZEB1, SIP1
and SIRT1, thus preventing the silencing of E-cadherin
Table 2 Expression profile of eight randomly selected miRNAs.
miRNA
Fold change (high
egg-laying/low
egg-laying) P-value
RNA-Seq RT-qPCR RNA-Seq RT-qPCR
gga-miR-1744-3p 0.11 0.16 0.0009 0.0148
gga-miR-216b 0.15 0.03 0.0254 0.0002
gga-miR-1655-5p 0.28 0.09 0.0203 0.0049
gga-miR-99a-5p 0.88 1.06 0.6960 0.3723
gga-miR-26a-5p 0.86 0.78 0.6440 0.2766
gga-miR-34b-3p 3.34 3.49 0.0240 0.0017
gga-miR-34c-3p 3.81 3.75 0.0363 0.0026
gga-miR-200a-3p 2.46 2.34 0.0365 0.0027
Relative expression
gga-miR-1744-3p
gga-miR-216b
gga-miR-1655-5p
gga-miR-99a-5p
gga-miR-26a-5p
gga-miR-34b-3p
gga-miR-34c-3p
gga-miR-200a-3p
0
1
2
3
4
5
LP
HP
**
***
*
*
ns ns
* *
*
*
Figure 5 Validation of the miRNA expression profile by RT-qPCR.
Relative expression of miRNAs was calculated according to the 2
DDCt
method using 5.8S rRNA as an internal reference RNA. The error bar
shows the standard deviation. *P<0.05, **P<0.01, ***P<0.001.
©2016 Stichting International Foundation for Animal Genetics, doi: 10.1111/age.12516
Wu et al.8
(Bracken et al. 2008; Eades et al. 2011). In several mes-
enchymal phenotype breast cancer cell lines, the miR-200a
family was found to be down-regulated (Gregory et al.
2008) and enforced expression of miR-200a family mem-
bers prevented TGF-b-induced epithelial to mesenchymal
transition (Eades et al. 2011). In breast cancer, in addition
to targeting ZEB1 and SIP1, the miR-200 family has been
shown to target phospholipase C-c1 and BMI1, reducing
EGF-driven motility and cancer stem cell self-renewal
respectively (Shimono et al. 2009; Uhlmann et al. 2010).
In the present study, gga-miR-200a-3p exhibited a signif-
icant 2.46-fold increase in HP ovary, implying that the
miRNA may play a special essential role in ovary develop-
ment and reproductive management of hens.
In previous work, genomic and transcriptomic studies
have identified that some genes and miRNAs are related to
egg production performance. Kang et al. (2009) revealed 18
known and eight unknown differentially expressed genes
detected in ovarian tissues from the pre-laying to the egg-
laying stages (Kang et al. 2009). Kang et al. (2013) iden-
tified 202 known miRNAs, 93 of which were found to be
significantly differentially expressed in sexually immature
and mature chicken ovaries. Luan et al. (2014) presented
the transcriptomic profiles of ovarian tissue from Huoyan
geese during the ceased and laying periods using RNA-Seq.
In the present study, we performed the first miRNA analysis
of low- and high-rate of egg production chicken ovarian
tissues using high-throughput sequencing. Compared with
other studies, we found some new significantly differentially
expressed ovarian miRNAs, such as gga-miR-1744-3p, gga-
miR-1655-5p, gga-miR-1734 and gga-miR-7465-3p, in the
high egg-laying chickens. These newly identified miRNAs
will be an important guideline for the future research.
Conflict of interest
All authors declared no conflict of interest exists.
Acknowledgements
This work was supported by the Program from Sichuan
Agricultural University (02920400), the National Natural
Science Foundation of China (NSFC31402063) and the
Sichuan Provincial Department of Science and Technology
Program (2015JQO023).
References
Ambros V., Bartel B., Bartel D.P. et al. (2003) A uniform system for
microRNA annotation. RNA 9, 2779.
Bannister S.C., Tizard M.L., Doran T.J., Sinclair A.H. & Smith C.A.
(2009) Sexually dimorphic microRNA expression during chicken
embryonic gonadal development. Biology of Reproduction 81,
16576.
Bartel D.P. (2004) MicroRNAs: genomics, biogenesis, mechanism,
and function. Cell 116, 28197.
Bierie B. & Moses H.L. (2006a) TGF-beta and cancer. Cytokine &
Growth Factor Reviews 17,2940.
Bierie B. & Moses H.L. (2006b) Tumour microenvironment:
TGFbeta: the molecular Jekyll and Hyde of cancer. Nature Reviews
Cancer 6, 50620.
Bommer G.T., Gerin I., Feng Y., Kaczorowski A.J., Kuick R., Love
R.E., Zhai Y., Giordano T.J., Qin Z.S. & Moore B.B. (2007) p53-
mediated activation of miRNA34 Candidate tumor-suppressor
genes. Current Biology 17, 1298307.
Bracken C.P., Gregory P.A., Kolesnikoff N., Bert A.G., Wang J.,
Shannon M.F. & Goodall G.J. (2008) A double-negative feedback
loop between ZEB1-SIP1 and the microRNA-200 family regulates
epithelial-mesenchymal transition. Cancer Research 68, 784654.
Burnside J., Ouyang M., Anderson A. et al. (2008) Deep sequencing
of chicken microRNAs. BMC Genomics 9, 185.
Carthew R.W. & Sontheimer E.J. (2009) Origins and mechanisms of
miRNAs and siRNAs. Cell 136, 64255.
Chi W., Tong C., Gan X. & He S. (2011) Characterization and
comparative profiling of MiRNA transcriptomes in bighead carp
and silver carp. PLoS One 6, e23549.
Choi J.H., Choi K.C., Auersperg N. & Leung P.C. (2005) Gon-
adotropins upregulate the epidermal growth factor receptor
through activation of mitogen-activated protein kinases and
phosphatidyl-inositol-3-kinase in human ovarian surface epithe-
lial cells. Endocrine-Related Cancer 12, 31424.
Conesa A., Gotz S., Garcia-Gomez J.M., Terol J., Talon M. & Robles M.
(2005) BLAST2GO: a universal tool for annotation, visualization and
analysis in functional genomics research. Bioi nformatics 21, 36746.
Corney D.C., Hwang C.I., Matoso A., Vogt M., Fleskennikitin A.,
Godwin A.K., Kamat A.A., Sood A.K., Ellenson L.H. & Hermeking
H. (2010) Frequent downregulation of miR-34 family in human
ovarian cancers. Clinical Cancer Research 16, 111928.
Darnell D.K., Kaur S., Stanislaw S., Konieczka J.H., Yatskievych
T.A. & Antin P.B. (2006) MicroRNA expression during chick
embryo development. Developmental Dynamics 235, 315665.
Deng M., Tang H., Zhou Y., Zhou M., Xiong W., Zheng Y., Ye Q.,
Zeng X., Liao Q. & Guo X. (2011) miR-216b suppresses tumor
growth and invasion by targeting KRAS in nasopharyngeal
carcinoma. Journal of Cell Science 124, 29973005.
Eades G., Yao Y., Yang M., Zhang Y., Chumsri S. & Zhou Q. (2011)
miR-200a regulates SIRT1 expression and epithelial to mes-
enchymal transition (EMT)-like transformation in mammary
epithelial cells. Journal of Biological Chemistry 286, 259926002.
Elela S.A. & Nazar R.N. (1997) Role of the 5.8 S rRNA in ribosome
translocation. Nucleic Acids Research 25, 178894.
Garzon R., Fabbri M., Cimmino A., Calin G.A. & Croce C.M. (2006)
MicroRNA expression and function in cancer. Trends in Molecular
Medicine 12, 5807.
Glazov E.A., Cottee P.A., Barris W.C., Moore R.J., Dalrymple B.P. &
Tizard M.L. (2008) A microRNA catalog of the developing
chicken embryo identified by a deep sequencing approach.
Genome Research 18, 95764.
Gregory P.A., Bert A.G., Paterson E.L., Barry S.C., Tsykin A.,
Farshid G., Vadas M.A., Khew-Goodall Y. & Goodall G.J. (2008)
The miR-200 family and miR-205 regulate epithelial to mes-
enchymal transition by targeting ZEB1 and SIP1. Nature Cell
Biology 10, 593601.
©2016 Stichting International Foundation for Animal Genetics, doi: 10.1111/age.12516
Ovarian microRNA transcriptome of chicken 9
Griffiths-Jones S. (2004) The microRNA registry. Nucleic Acids
Research 32, D10911.
Griffiths-Jones S., Grocock R.J., van Dongen S., Bateman A. &
Enright A.J. (2006) miRBase: microRNA sequences, targets and
gene nomenclature. Nucleic Acids Research 34, D1404.
Griffiths-Jones S., Saini H.K., van Dongen S. & Enright A.J. (2008)
miRBase: tools for microRNA genomics. Nucleic Acids Research
36, D1548.
Gu Y., Zhang L. & Chen X. (2014) Differential expression analysis of
Paralichthys olivaceus microRNAs in adult ovary and testis by deep
sequencing. General and Comparative Endocrinology 204,1814.
He X., He L. & Hannon G.J. (2007a) The guardian’s little helper:
microRNAs in the p53 tumor suppressor network. Cancer
Research 67, 11099101.
He L., He X., Lim L.P. et al. (2007b) A microRNA component of the
p53 tumour suppressor network. Nature 447, 11304.
Hermeking H. (2009) The miR-34 family in cancer and apoptosis.
Cell Death & Differentiation 17, 1939.
Hicks J.A., Tembhurne P. & Liu H.C. (2008) MicroRNA expression
in chicken embryos. Poultry Science 87, 233543.
Hicks J.A., Tembhurne P.A. & Liu H.C. (2009) Identification of
microRNA in the developing chick immune organs. Immuno-
genetics 61, 23140.
Hicks J.A., Trakooljul N. & Liu H.C. (2010) Discovery of chicken
microRNAs associated with lipogenesis and cell proliferation.
Physiological Genomics 41, 18593.
Hossain M.M., Ghanem N., Hoelker M., Rings F., Phatsara C.,
Tholen E., Schellander K. & Tesfaye D. (2009) Identification and
characterization of miRNAs expressed in the bovine ovary. BMC
Genomics 10, 443.
Hu W., Chen S., Zhang R. & Lin Y. (2013) Single nucleotide
polymorphisms in the upstream regulatory region alter the
expression of myostatin. In Vitro Cellular & Developmental Biology-
Animal 49, 41723.
Huang J., Ju Z., Li Q. et al. (2011) Solexa sequencing of novel and
differentially expressed microRNAs in testicular and ovarian
tissues in Holstein cattle. International Journal of Biological Sciences
7, 101626.
Ji Q., Hao X., Meng Y., Zhang M., Desano J., Fan D. & Xu L. (2008)
Restoration of tumor suppressor miR-34 inhibits human p53-
mutant gastric cancer tumorspheres. BMC Cancer 8,112.
Ji Q., Hao X., Zhang M. et al. (2009) MicroRNA miR-34 inhibits
human pancreatic cancer tumor-initiating cells. PLoS One 4,
e6816.
Kanda M., Shimizu D., Tanaka H. et al. (2016) Metastatic pathway-
specific transcriptome analysis identifies MFSD4 as a putative
tumor suppressor and biomarker for hepatic metastasis in
patients with gastric cancer. Oncotarget 7, 1366779.
Kang B., Guo J.R., Yang H.M., Zhou R.J., Liu J.X., Li S.Z. & Dong
C.Y. (2009) Differential expression profiling of ovarian genes in
prelaying and laying geese. Poultry Science 88, 197583.
Kang L., Cui X., Zhang Y., Yang C. & Jiang Y. (2013) Identification
of miRNAs associated with sexual maturity in chicken ovary by
Illumina small RNA deep sequencing. BMC Genomics 14,1.
Kato M., Paranjape T., M
uller R.U., Nallur S., Gillespie E., Keane K.,
Esquela-Kerscher A., Weidhaas J.B. & Slack F.J. (2009) The mir-
34 microRNA is required for the DNA damage response in vivo in
C. elegans and in vitro in human breast cancer cells. Oncogene 28,
241924.
Kim V.N. (2005) MicroRNA biogenesis: coordinated cropping and
dicing. Nature Reviews Molecular Cell Biology 6, 37685.
Krek A., Grun D., Poy M.N. et al. (2005) Combinatorial microRNA
target predictions. Nature Genetics 37, 495500.
Kuo M.W., Wang C.H., Wu H.C., Chang S.J. & Chuang Y.J. (2011)
Soluble THSD7A is an N-glycoprotein that promotes endothelial
cell migration and tube formation in angiogenesis. PLoS One 6,
e29000.
Lagos-Quintana M., Rauhut R., Lendeckel W. & Tuschl T. (2001)
Identification of novel genes coding for small expressed RNAs.
Science 294, 8538.
Lau N.C., Lim L.P., Weinstein E.G. & Bartel D.P. (2001) An
abundant class of tiny RNAs with probable regulatory roles in
Caenorhabditis elegans.Science 294, 85862.
Lee Y., Kim M., Han J., Yeom K.H., Lee S., Baek S.H. & Kim V.N.
(2004) MicroRNA genes are transcribed by RNA polymerase II.
The EMBO Journal 23, 405160.
Lee S.I., Lee B.R., Hwang Y.S., Lee H.C., Rengaraj D., Song G., Park
T.S. & Han J.Y. (2011) MicroRNA-mediated posttranscriptional
regulation is required for maintaining undifferentiated properties
of blastoderm and primordial germ cells in chickens. Proceedings
of the National Academy of Sciences of the United States of America
108, 1042631.
Li M., Liu Y., Wang T. et al. (2011) Repertoire of porcine
microRNAs in adult ovary and testis by deep sequencing.
International Journal of Biological Sciences 7, 104555.
Li T., Wang S., Wu R., Zhou X., Zhu D. & Zhang Y. (2012)
Identification of long non-protein coding RNAs in chicken
skeletal muscle using next generation sequencing. Genomics 99,
2928.
Li D.Y., Zhang L., Smith D.G., Xu H.L., Liu Y.P., Zhao X.L., Wang Y.
& Zhu Q. (2013) Genetic effects of melatonin receptor genes on
chicken reproductive traits. Czech Journal of Animal Science 58,
5864.
Li R., Zhang C.L., Liao X.X. et al. (2015) Transcriptome microRNA
profiling of bovine mammary glands infected with Staphylococcus
aureus.International Journal of Molecular Sciences 16, 49975013.
Lim L.P., Glasner M.E., Yekta S., Burge C.B. & Bartel D.P. (2003)
Vertebrate microRNA genes. Science 299, 1540.
Liu N., Landreh M., Cao K., Abe M., Hendriks G.J., Kennerdell J.,
Zhu Y., Wang L.S. & Bonini N.M. (2012) The microRNA miR-34
modulates aging and neurodegeneration in Drosophila. Nature
482, 51923.
Liu F.Y., Zhou S.J., Deng Y.L., Zhang Z.Y., Zhang E.L., Wu Z.B.,
Huang Z.Y. & Chen X.P. (2015) MiR-216b is involved in
pathogenesis and progression of hepatocellular carcinoma
through HBx-miR-216b-IGF2BP2 signaling pathway. Cell Death
& Disease 6, e1670.
Luan X., Liu D., Cao Z., Luo L., Liu M., Gao M. & Zhang X. (2014)
Transcriptome profiling identifies differentially expressed genes in
Huoyan goose ovaries between the laying period and ceased
period. PLoS One 9, e113211.
Luzi E., Marini F., Sala S.C., Tognarini I., Galli G. & Brandi M.L.
(2008) Osteogenic differentiation of human adipose tissue-
derived stem cells is modulated by the miR-26a targeting of the
SMAD1 transcription factor. Journal of Bone & Mineral Research
23, 28795.
Mansfield J.H., Harfe B.D., Nissen R. et al. (2004) MicroRNA-
responsive ‘sensor’ transgenes uncover Hox-like and other
©2016 Stichting International Foundation for Animal Genetics, doi: 10.1111/age.12516
Wu et al.10
developmentally regulated patterns of vertebrate microRNA
expression. Nature Genetics 36, 107983.
McBride D., Carre W., Sontakke S.D., Hogg C.O., Law A., Donadeu
F.X. & Clinton M. (2012) Identification of miRNAs associated
with the follicular-luteal transition in the ruminant ovary.
Reproduction 144, 22133.
Miles J.R., Mcdaneld T.G., Wiedmann R.T., Cushman R.A.,
Echternkamp S.E., Vallet J.L. & Smith T.P.L. (2012) MicroRNA
expression profile in bovine cumulusoocyte complexes: possible
role of let-7 and miR-106a in the development of bovine oocytes.
Animal Reproduction Science 130,1626.
Mishima T., Takizawa T., Luo S.S. et al. (2008) MicroRNA (miRNA)
cloning analysis reveals sex differences in miRNA expression
profiles between adult mouse testis and ovary. Reproduction 136,
81122.
Moreau E.J., Matutes E., A’Hern R.P., Morilla A.M., Morilla R.M.,
Owusu-Ankomah K.A., Seon B.K. & Catovsky D. (1997)
Improvement of the chronic lymphocytic leukemia scoring
system with the monoclonal antibody SN8 (CD79b). American
Journal of Clinical Pathology 108, 37882.
Morozova O. & Marra M.A. (2008) Applications of next-generation
sequencing technologies in functional genomics. Genomics 92,
25564.
Okada N., Lin C.P., Ribeiro M.C., Biton A., Lai G., He X., Bu P.,
Vogel H., Jablons D.M. & Keller A.C. (2014) A positive feedback
between p53 and miR-34 miRNAs mediates tumor suppression.
Genes & Development 28, 43850.
Oneyama C., Ikeda J., Okuzaki D., Suzuki K., Kanou T., Shintani Y.,
Morii E., Okumura M., Aozasa K. & Okada M. (2011) MicroRNA-
mediated downregulation of mTOR/FGFR3 controls tumor
growth induced by Src-related oncogenic pathways. Oncogene
30, 3489501.
Rathjen T., Pais H., Sweetman D., Moulton V., Munsterberg A. &
Dalmay T. (2009) High throughput sequencing of microRNAs in
chicken somites. FEBS Letters 583, 14226.
Reid J.G., Nagaraja A.K., Lynn F.C., Drabek R.B., Muzny D.M.,
Shaw C.A., Weiss M.K., Naghavi A.O., Khan M. & Zhu H. (2008)
Mouse let-7 miRNA populations exhibit RNA editing that is
constrained in the 50-seed/ cleavage/anchor regions and stabilize
predicted mmu-let-7a:mRNA duplexes. Genome Research 18,
157181.
Ro S., Song R., Park C., Zheng H., Sanders K.M. & Yan W. (2007)
Cloning and expression profiling of small RNAs expressed in the
mouse ovary. RNA 13, 236680.
Ruan W.K. & Zheng S.J. (2011) Polymorphisms of chicken toll-like
receptor 1 type 1 and type 2 in different breeds. Poultry Science
90, 19417.
Saito Y. & Jones P.A. (2006) Epigenetic activation of tumor
suppressor microRNAs in human cancer cells. Cell Cycle 5,
22202.
Sarbassov D.D., Ali S.M., Kim D.H., Guertin D.A., Latek R.R.,
Erdjument-Bromage H., Tempst P. & Sabatini D.M. (2004) Rictor,
a novel binding partner of mTOR, defines a rapamycin-insensitive
and raptor-independent pathway that regulates the cytoskeleton.
Current Biology 14, 1296302.
Schmittgen T.D. & Livak K.J. (2008) Analyzing real-time PCR
data by the comparative CT method. Nature Proto cols 3,
11018.
Shimono Y., Zabala M., Cho R.W. et al. (2009) Downregulation of
miRNA-200c links breast cancer stem cells with normal stem
cells. Cell 138, 592603.
Sun D., Lee Y.S., Malhotra A., Kim H.K., Matecic M., Evans C., Jensen
R.V., Moskaluk C.A.& Dutta A. (2011) miR-99 familyof microRNAs
suppresses the expression of prostate-specific antigen and prostate
cancer cell proliferation. Cancer Research 71, 131324.
Tian F., Luo J., Zhang H., Chang S. & Song J. (2012) MiRNA
expression signatures induced by Marek’s disease virus infection
in chickens. Genomics 99, 1529.
Tripurani S.K., Xiao C., Salem M. & Yao J. (2010) Cloning and
analysis of fetal ovary microRNAs in cattle. Animal Reproduction
Science 120,1622.
Turcatel G., Rubin N., El-Hashash A. & Warburton D. (2012) MIR-
99a and MIR-99b modulate TGF-binduced epithelial to mes-
enchymal plasticity in normal murine mammary gland cells.
PLoS One 7, e31032.
Uhlmann S., Zhang J.D., Schwager A., Mannsperger H., Riazal-
hosseini Y., Burmester S., Ward A., Korf U., Wiemann S. &
Sahin O. (2010) miR-200bc/429 cluster targets PLCgamma1
and differentially regulates proliferation and EGF-driven inva-
sion than miR-200a/141 in breast cancer. Oncogene 29, 4297
306.
Wang Y., Brahmakshatriya V., Zhu H. et al. (2009) Identification of
differentially expressed miRNAs in chicken lung and trachea with
avian influenza virus infection by a deep sequencing approach.
BMC Genomics 10, 512.
Wang Q., Gao Y., Ji X., Qi X., Qin L., Gao H., Wang Y. & Wang X.
(2013) Differential expression of microRNAs in avian leukosis
virus subgroup J-induced tumors. Veterinary Microbiology 162,
2328.
Wong C.F. & Tellam R.L. (2008) MicroRNA-26a targets the histone
methyltransferase Enhancer of Zeste homolog 2 during myoge-
nesis. Journal of Biological Chemistry 283, 983643.
Wong N. & Wang X. (2015) miRDB: an online resource for
microRNA target prediction and functional annotations. Nucleic
Acids Res 43, D14652.
Xu Z., Chen J., Li X., Ge J., Pan J. & Xu X. (2013) Identification and
characterization of microRNAs in channel catfish (Ictalurus
punctatus) by using Solexa sequencing technology. PLoS One 8,
e54174.
Yu Y., Zhang H., Tian F., Bacon L., Zhang Y., Zhang W. & Song J.
(2008) Quantitative evaluation of DNA methylation patterns for
ALVE and TVB genes in a neoplastic disease susceptible and
resistant chicken model. PLoS One 3, e1731.
Yuan L., Zhang X., Li L., Jiang H. & Chen J. (2014) High-
throughput sequencing of microRNA transcriptome and expres-
sion assay in the sturgeon, Acipenser schrenckii.PLoS One 9,
e115251.
Zhang Y., Zhou X., Ge X., Jiang J., Li M., Jia S., Yang X., Kan Y.,
Miao X. & Zhao G. (2009) Insect-specific microRNA involved in
the development of the silkworm Bombyx mori.PLoS One 4,
e4677.
Zhang B., Liu X.X., He J.R., Zhou C.X., Guo M., He M., Li M.F.,
Chen G.Q. & Zhao Q. (2011) Pathologically decreased miR-
26a antagonizes apoptosis and facilitates carcinogenesis by
targeting MTDH and EZH2 in breast cancer. Carcinogenesis
32,29.
©2016 Stichting International Foundation for Animal Genetics, doi: 10.1111/age.12516
Ovarian microRNA transcriptome of chicken 11
Supporting information
Additional supporting information may be found online in
the supporting information tab for this article:
Figure S1 Length distribution of total small RNA in LP and
HP chicken ovary libraries.
Figure S2 Small RNAs percentages of (A) HP and (B) LP
chickens.
Figure S3 Cluster plots of the 17 differentially expressed
miRNAs.
Figure S4 Thirty-nine differentially expressed predicted
targets of 10 candidate miRNAs.
Table S1 Main reproductive traits of the selected LP and HP
samples.
Table S2 Primer sequences for real-time PCR.
Table S3 Overview of miRNA-seq data of all samples.
Table S4 Identities of various small RNA sequences.
Table S5 Expression profile of known miRNAs in ovarian
tissues of low and high rate of egg production chickens.
Table S6 Novel miRNAs expressed in ovarian tissues of low
and high rate of egg production chickens.
Table S7 Prediction of the differentially expressed miRNA
targets.
Table S8 GO and KEGG pathway annotations for the
miRNA targets.
Table S9 miRNAs and their predicted target genes involved
in the reproduction regulation process.
©2016 Stichting International Foundation for Animal Genetics, doi: 10.1111/age.12516
Wu et al.12
... miRNAs are small endogenous non-coding RNAs that play a critical role in the posttranscriptional regulation of gene expression 9 . The egg-laying performance of hens is tightly regulated by the interaction between multiple miRNAs and their target genes, which modulate avian follicular development, recruitment, maturation, selection, atresia, and regression [10][11][12][13] . While many studies have focused on the transcriptional expression of miRNAs, evidence supports the idea that miRNAs primarily affect ovarian function through their actions on ovarian granulosa cells 6,14 . ...
Article
Full-text available
MicroRNAs (miRNAs) play a crucial role as transcription regulators in various aspects of follicular development, including steroidogenesis, ovulation, apoptosis, and gene regulation in poultry. However, there is a paucity of studies examining the specific impact of miRNAs on ovarian granulosa cells (GCs) across multiple grades in laying hens. Consequently, this study aims to investigate the roles of miRNAs in chicken GCs. By constructing miRNA expression profiles of GCs at 10 different time points, encompassing 4 pre-hierarchical, 5 preovulatory, and 1 postovulatory follicles stage, we identified highly expressed miRNAs involved in GC differentiation (miR-148a-3p, miR-143-3p), apoptosis (let7 family, miR-363-3p, miR-30c-5p, etc.), and autophagy (miR-128-3p, miR-21-5p). Furthermore, we discovered 48 developmentally dynamic miRNAs (DDMs) that target 295 dynamic differentially expressed genes (DDGs) associated with follicular development and selection (such as oocyte meiosis, progesterone-mediated oocyte maturation, Wnt signaling pathway, TGF-β signaling pathway) as well as follicular regression (including autophagy and cellular senescence). These findings contribute to a more comprehensive understanding of the intricate mechanisms underlying follicle recruitment, selection, and degeneration, aiming to enhance poultry’s reproductive capacity.
... The increased mRNA expressions of FSHb, LHb, FSHR, and LHR genes may explain the significant improvement of egg production traits in selected quails compared to the base generation. The mRNA expression pattern in ovarian tissue was also reported to be linked to egg production (Wu et al., 2017). In this context, Kurnia et al. (2021) recorded a positive correlation between the polymorphism of FSHR gene promoter fragment and egg number and egg weight in hybrid chickens. ...
Article
Full-text available
Investigating the impact of early egg production selection (the first 90 d of laying) on egg production features, cumulative selection response (CSR), and the mRNA expression of gonadotropins (FSHβ and LHβ), and their receptors (FSHR and LHR), in Japanese quails was the goal. The selection experiment involved 1293 females in all, 257 from the base group and 1036 from the 4 selected generations. Age and body weight at sexual maturity (ASM, BWSM), weight of the first egg (WFE), days to the first 10 eggs (DF10E), egg mass for the first 10 eggs (EMF10E), egg weight (EW), egg number at the first 90 d of laying (EN90D), and egg mass at the first 90 d of laying (EM90D) were all recorded. Most egg production traits had heritability estimates that were low to moderate and ranged from 0.17 to 0.33., where the highest estimates were reported for EN90D (0.33) and BWSM (0.32). With the exception of EN90D, low to moderate positive genetic correlations were observed between ASM and other egg production traits (0.17–0.44). The fourth generation showed significantly (P < 0.05) lower ASM and DF10E but higher BWSM, WFE, EN90D, EM10E, and EM90D when compared with the base generation. CSR were significant (P < 0.05) for ASM (−6.67 d), BWSM (27.13 g), WFE (0.93 g), DF10E (−1.25 d), EN90D (7.24 egg), EM10E (10.57 g), and EM90D (140.0 g). FSHβ, LHβ, FSHR, and LHR gene mRNA expression was considerably (P < 0.05) greater in the fourth generation compared to the base generation. In conclusion, selection programs depending on the efficiency of egg production (EN90D) could improve the genetic gain of egg production traits and upregulate the mRNA expression of FSHβ, LHβ, FSHR, and LHR genes in selected quails (fourth generation). These findings might help to enhance breeding plans and create commercial lines of high egg production Japanese quails.
... A series of studies demonstrated that miRNAs were involved in many biological regulatory processes, including ovarian function, follicular development and atresia, cell proliferation and apoptosis, steroid biosynthesis, and ovarian cancer [45][46][47]. For example, miR-26b increased the number of DNA breaks and facilitated apoptosis of porcine granulosa cell by targeting the ATM gene in vitro [48]. ...
Article
Background: As a crucial economic characteristic and a major indicator of reproductive performance in layers, egg production is controlled by a series of complex regulatory heredity basis. In particular, the interacting regulatory function between noncoding RNAs (ncRNAs) and coding RNA plays important roles in regulating laying performance. Methods: In this study, the RNA sequencing (RNA-seq) of ovarian tissues from Lohmann hens (n = 3) and Chengkou Mountain chicken (n = 3) under the laying peak period was performed to identify RNA transcriptional differences among different laying-performance populations. Results: Results showed that the expression level of 303 mRNAs, 68 long ncRNAs (lncRNAs), 533 circular RNAs (circRNAs), and 79 microRNAs (miRNAs) was significantly different among the groups. Functional enrichment analysis of these differentially expressed (DE) mRNAs revealed that the laying process was implicated in numerous significantly enriched pathways (p < 0.05), such as the neuroactive ligand-receptor interaction, steroid hormone biosynthesis, and calcium-signaling pathway. Furthermore, the lncRNA/circRNA-miRNA-mRNA regulatory networks related to the regulation of laying performance were constructed. Some randomly selective DE RNAs were verified by Real Time Quantitative (RT-qRCR), indicating that the bioinformatics analysis results of RNA-seq data were credible. Conclusions: This study could increase our understanding of the heredity basis of transcriptome in the laying performance of chicken.
... Noncoding RNAs also regulate the target genes related to endocrine and metabolism signaling pathways. A study reported that microRNAs present in avian ovarian tissue, such as gga-miR-34b, gga-miR-34c, and gga-miR-216b, were differently expressed in high-yield laying hens [10]. By binding to the target genes, noncoding RNAs regulate the synthesis, transport, and secretion of genes related to reproductive hormones downstream of signaling pathways, namely STAR, CYP11A1, and CYP19A1 [11,12]. ...
Article
Full-text available
Egg production is a vital biological and economic trait for poultry breeding. The ‘hypothalamic–pituitary–ovarian (HPO) axis’ determines the egg production, which affects the layer hens industry income. At the organism level, the HPO axis is influenced by the factors related to metabolic and nutritional status, environment, and genetics, whereas at the cellular and molecular levels, the HPO axis is influenced by the factors related to endocrine and metabolic regulation, cytokines, key genes, signaling pathways, post-transcriptional processing, and epigenetic modifications. MiRNAs and lncRNAs play a critical role in follicle selection and development, atresia, and ovulation in layer hens; in particular, miRNA is known to affect the development and atresia of follicles by regulating apoptosis and autophagy of granulosa cells. The current review elaborates on the regulation of the HPO axis and its role in the laying performance of hens at the organism, cellular, and molecular levels. In addition, this review provides an overview of the interactive network regulation mechanism of the HPO axis in layer hens, as well as comprehensive knowledge for successfully utilizing their genetic resources.
... In KEGG analysis, most of DEGs were significantly enriched in neuroactive ligand-receptor interaction, dopaminergic synapse, synaptic vesicle cycle, cholinergic synapse, and Cushing syndrome pathways. Studies have shown that the neuroactive ligand-receptor interaction [37], dopaminergic synapse [38], synaptic vesicle cycle [39], cholinergic synapse [40], and Cushing syndrome pathways [41] could play important roles in regulating reproductive performance. Further analysis showed that the five pathways have crosstalk, and DEGs enriched in the five pathways were up-regulated in HVS group, suggesting that these DEGs could play an important role in the epididymis regulating semen volume. ...
Article
Full-text available
Semen volume is an important factor in artificial insemination (AI) of ducks. In drakes, seminal plasma that is produced by the epididymis determines the semen volume. However, the mechanism of epididymis regulating semen volume of drakes remains unclear. Therefore, the aim of the present study was to preliminarily reveal the mechanism regulating the semen volume through comparing the epididymal histomorphology and mRNA expression profiles between drakes with high-volume semen (HVS) and low-volume semen (LVS). Phenotypically, drakes in the HVS group produced more sperm than drakes in the LVS group. In addition, compared with the HVS group, the ductal square of ductuli conjugentes (DC) and dutus epididymidis (DE) in epididymis was significantly smaller in the LVS group, and the lumenal diameter and epithelial thickness of DC/DE were significantly shorter in the LVS group. In transcriptional regulation, 72 different expression genes (DEGs) were identified from the epididymis between HVS and LVS groups. Gene Ontology (GO) analysis indicated that the DEGs were mainly related to hormone secretion, neurotransmitter synthesis/transport, transmembrane signal transduction, transmembrane transporter activity, and nervous system development (p < 0.05). Kyoto Encyclopedia of Genes and Genomes (KEGG) functional enrichment analysis showed that the DEGs were significantly enriched in pathways associated with hormone and neurotransmitter transmission (p < 0.05). In addition, further analysis of the top five pathways enriched by KEGG, nine key candidate genes (including SLC18A2, SNAP25, CACNA1B, GABRG2, DRD3, CAMK2A, NR5A1, and STAR) were identified, which could play a crucial role in the formation of semen. These data provide new insights into the molecular mechanism regulating semen volume of drakes and make feasible the breeding of drakes by semen volume.
Article
Full-text available
Follicular atresia occurs in every stage of ovarian development, which is relevant to female fertility. In the past decade, increasing studies have confirmed that miRNAs, a class of short non-coding RNAs, play an important role in follicular atresia by post-transcription regulation of their target genes. However, the function of miRNAs on follicular atresia initiation is unknown. In the present study, high-throughput small RNA sequencing was performed to analyze differential miRNA expression profiles between healthy (HF) follicles and early atretic (EAF) follicles. A total of 237 conserved miRNA were detected, and the miR-143 is the highest expressed in follicles. Meanwhile, we also found wide sequence variations (isomiRs) in porcine ovarian miRNA, including in 5′un-translation region, core seed sequences and 3′untranslation region. Furthermore, we identified 22 differentially expressed miRNAs in EAF groups compared to HF group, of which 3 miRNAs were upregulated, as well as 19 miRNAs were downregulated, and then the RT-PCR was performed to validate these profiles. The target genes of these differentially expressed miRNAs were predicted by using miRwalk, miRDB, and Targetscan database, respectively. Moreover, the gene ontology and KEGG pathway enrichment established that the regulating functions and signaling pathways of these miRNAs contribute to follicular atresia initiation and cell fate. In conclusion, this study provides new insights into the changes of miRNAs in early atretic follicles to demonstrate their molecular regulation in ovarian follicular atretic initiation.
Article
MicroRNAs (miRNAs) are involved in many physiological processes such as signal transduction, cell proliferation and apoptosis. Many studies have shown that miRNAs can regulate the process of follicular development. Our previous studies found that the expression of miR-29c-5p in buffalo atretic follicles was much higher than that in healthy follicles, suggesting that this miRNA may participate in the process of buffalo follicular atresia. In this study, we aim to explore to the role and molecular mechanisms of miR-29c-5p on the functions of buffalo granulosa cells (GCs). GCs cultured in vitro were transfected with miR-29c-5p mimics and its inhibitor, respectively, and it was found that the mimics significantly increased the apoptotic rate of GCs. They also inhibited the proliferation of GCs and the secretion of steroid hormones. The effect of the inhibitor was opposite to that of the mimics. MiR-29c-5p was subsequently shown to target the inhibin subunit beta A, (INHBA). Overexpression of INHBA could promote the production of activin A and inhibin A, and then reverse the effect of miR-29c-5p on buffalo GCs. In conclusion, these results suggest that miR-29c-5p promotes apoptosis and inhibits proliferation and steroidogenesis by targeting INHBA in buffalo GCs. This may ultimately promote atresia in buffalo follicles.
Article
Granulosa cells (GCs) are the ovary's most critical cells since they undergo cell differentiation and hormone synthesis changes closely associated with follicle development. While micro RNA 140-3p (miRNA-140-3p) has an apparent cell signaling role, particularly in cell proliferation, its biological role in chicken ovarian follicle growth and development remains elusive. This study explored miR-140-3p's effects on chicken GC proliferation and steroid hormone synthesis. MiR-140-3p dramatically increased GC proliferation, prevented apoptosis, increased progesterone synthesis, and enhanced gene expression related to steroid hormone synthesis. In addition, the anti-Müllerian hormone (AMH) gene was identified as a direct miR-140-3p target. MiR-140-3p abundance correlated negatively with AMH mRNA and protein levels in GCs. Our findings show that miR-140-3p influences chicken GC proliferation and steroid hormone synthesis by suppressing AMH expression.
Article
Full-text available
Gastric cancer (GC) with hepatic metastasis remains a fatal disease. Global expression profiling was conducted using tissues from patients who had GC with synchronous hepatic metastasis, and major facilitator superfamily domain containing 4 (MFSD4) was identified as a candidate biomarker for hepatic metastasis in GC. Functional and expression analyses of this molecule in GC cell lines and clinical samples were conducted. We analyzed MFSD4 expression, DNA methylation, and copy number. RNA interference experiments evaluated the effects of MFSD4 expression on cell phenotype and apoptosis. We analyzed tissues of 200 patients with GC to assess the diagnostic performance of MFSD4 levels for predicting hepatic recurrence, metastasis, or both. Differential expression of MFSD4 mRNA by GC cell lines correlated positively with the levels of NUDT13 and OCLN mRNAs and inversely with those of BMP2. Hypermethylation of the MFSD4 promoter was detected in cells with lower levels of MFSD4 mRNA. Inhibition of MFSD4 expression significantly increased the invasiveness and motility of GC cells but did not influence cell proliferation or apoptosis. MFSD4 mRNA levels in primary GC tissues were reduced in patients with concomitant hepatic metastasis or recurrence compared with those without. Low levels of MFSD4 mRNA in primary GC tissues were an independent risk factor of hepatic recurrence and metastasis. MFSD4 expression in gastric tissues may represent a useful biomarker for identification of patients at high risk for hepatic recurrence, metastasis, or both.
Article
Full-text available
The melatonin receptors are G protein-coupled receptors (GPCR) that bind melatonin. Three types of melatonin receptors have been cloned. The MTNR1A (or Mel1A or MT1) and MTNR1B (or Mel1B or MT2) receptor subtypes are present in humans and other mammals, while an additional melatonin receptor subtype MTNR1C (or Mel1C or MT3) has been identified in amphibians and birds. Previous research has shown that the three common melatonin receptors regulate physiological processes, including seasonal reproduction and ovarian physiology. However, whether or not any polymorphisms of the different melatonin receptor subtypes are associated with reproductive traits in chickens is not known. In this study, we performed candidate gene analysis to identify single-nucleotide polymorphisms (SNPs) in the MTNR1A, MTNR1B, and MTNR1C genes in the Erlang Mountain Chicken population. SNP discovery was achieved by sequencing pooled DNA samples. Direct PCR-sequencing, PCR-SSCP/PCR-sequencing, and PCR-RFLP method were used to genotype the MTNR1A, MTNR1B, and MTNR1C genes, respectively. The GLM Procedure was used to estimate the statistical significance of association between genotypes at each locus and reproductive traits of chickens. In a sample of 460 chickens, four novel polymorphisms (JQ249890:g.384T>C, JQ249891:g.387T>C, JQ249894:g.63C>T, and JQ249896:g.294G>A) were detected in the melatonin receptor genes MTNR1A, MTNR1B, and MTNR1C, respectively. A statistically significant association (P < 0.01) was found between two SNPs (MTNR1A SNP, MTNR1C SNP) and reproductive traits: egg number at 300 days of age (EN) and age at first egg (AFE).
Article
Full-text available
MicroRNAs (miRNAs) are important regulators of cell fate determination and homeostasis. Expression of these small RNA genes is tightly regulated during development and in normal tissues, but they are often misregulated in cancer. MiRNA expression is also affected by DNA damaging agents, such as radiation. In particular, mammalian miR-34 is upregulated by p53 in response to radiation, but little is known about the role of this miRNA in vivo. Here we show that Caenorhabditis elegans with loss-of-function mutations in the mir-34 gene have an abnormal cellular survival response to radiation; these animals are highly radiosensitive in the soma and radioresistant in the germline. These findings show a role for mir-34 in both apoptotic and non-apoptotic cell death in vivo, much like that of cep-1, the C. elegans p53 homolog. These results have been additionally validated in vitro in breast cancer cells, wherein exogenous addition of miR-34 alters cell survival post-radiation. These observations confirm that mir-34 is required for a normal cellular response to DNA damage in vivo resulting in altered cellular survival post-irradiation, and point to a potential therapeutic use for anti-miR-34 as a radiosensitizing agent in p53-mutant breast cancer.
Article
Full-text available
MicroRNAs are small non-coding RNA molecules that are important regulators of gene expression at the post-transcriptional level. miRNAs impact the processes of cell proliferation, differentiation and apoptosis. Thus, the regulation of miRNA expression profiles associated with mastitis will be conducive for its control. In this study, Staphylococcus aureus (S. aureus) was administered to the mammary gland of Chinese Holstein cows to construct a bacteria-type mastitis model. Total RNA was isolated from bovine mammary gland tissue samples from the S. aureus-induced mastitis group and controls. miRNAs were analyzed using Solexa sequencing and bioinformatics processing for the experimental group and control group. Two miRNA libraries were constructed respectively. A total of 370 known bovine miRNAs and 341 novel mi RNAs were detected for the S. aureus and 358 known bovine miRNAs and 232 novel miRNAs for control groups. A total of 77 miRNAs in the S. aureus group showed significant differences compared to the control group. GO (Gene Ontology) analysis showed these target genes were involved in the regulation of cells, binding, etc., while KEGG (Kyoto Encyclopedia of Genes and Genomes) analysis showed that these genes were enriched in endocytosis, and olfactory transduction pathways involved in cancer. These results provide an experimental basis to reveal the cause and regulatory mechanism of mastitis and also suggest the potential of miRNAs to serve as biomarkers for the diagnosis of mastitis in dairy cows.
Article
Full-text available
This study aims to investigate the expression status of miRNA-216b in familial hepatocellular carcinoma (HCC) and the correlation between miRNA-216b expression and pathogenesis, as well as the progression of HCC. The expression profile of miRNAs in plasma of peripheral blood between HCC patients with HCC family history and healthy volunteers without HCC family history was determined by microarray. Using real-time quantitative PCR to detect the expression in paired tissues from 150 patients with HCC, miR-216b was selected as its expression value in HCC patients was significantly lower compared with healthy volunteers. Next, miR-216b expression and the clinicopathological features of HCC were evaluated. The effect of miR-216b expression on tumor cells was investigated by regulating miR-216b expression in SMMC-7721 and HepG2 in vitro and in vivo. Finally, we explored mRNA targets of miR-216b. In 150 HCC, 37 (75%) tumors showed reduced miR-216b expression comparing with their adjacent liver tissues. The decreased expression of miR-216b was significantly correlated with tumor volume (P=0.044), HBV infection (P=0.026), HBV DNA quantitative (P=0.001) and vascular invasion (P=0.032). The 5-year disease-free survival and overall rates after liver resection in low expression and high expression groups of miR-216b are 62% and 54%, 25% and 20%, respectively. MiR-216b overexpression inhibited cell proliferation, migration and invasion, and miR-216b inhibition did the opposite. The expression of hepatitis B virus x protein (HBx) has tight correlation with downregulation of miR-216b. Furthermore, miR-216b downregulated the expression of insulin-like growth factor 2 mRNA-binding protein 2 (IGF2BP2) and exerted its tumor-suppressor function through inhibition of protein kinase B and extracellular signal-regulated kinase signaling downstream of IGF2. MiR-216b inhibits cell proliferation, migration and invasion of HCC by regulating IGF2BP2 and it is regulated by HBx.
Article
... Thomas D Schmittgen 1 & Kenneth J Livak 2 . ABSTRACT. ... N. Engl. J . Med. ... 32, e178 (2004). | Article | PubMed | ChemPort |; Livak , KJ & Schmittgen , TD Analysis of relative gene expression data using real - time quantitative PCR and the 2 (- Delta Delta C(T)) Method . ...
Article
Two different methods of presenting quantitative gene expression exist: absolute and relative quantification. Absolute quantification calculates the copy number of the gene usually by relating the PCR signal to a standard curve. Relative gene expression presents the data of the gene of interest relative to some calibrator or internal control gene. A widely used method to present relative gene expression is the comparative CT method also referred to as the 2−ΔΔCT method. This protocol provides an overview of the comparative CT method for quantitative gene expression studies. Also presented here are various examples to present quantitative gene expression data using this method.