Skip to main content
PLOS ONE logoLink to PLOS ONE
. 2015 Oct 1;10(10):e0139514. doi: 10.1371/journal.pone.0139514

Transcriptomic Analysis of Ovaries from Pigs with High And Low Litter Size

Xiaodong Zhang 1,#, Long Huang 1,#, Tao Wu 1,#, Yifang Feng 1, Yueyun Ding 1, Pengfei Ye 1,¤, Zongjun Yin 1,*
Editor: Ramona Natacha PENA i SUBIRÀ2
PMCID: PMC4591126  PMID: 26426260

Abstract

Litter size is one of the most important economic traits for pig production as it is directly related to the production efficiency. Litter size is affected by interactions between multiple genes and the environment. While recent studies have identified some genes associated with prolificacy in pigs, transcriptomic studies of specific genes affecting litter size in porcine ovaries are rare. In order to identify candidate genes associated with litter size in swine, we assessed gene expression differences between the ovaries of Yorkshire pigs with extremely high and low litter sizes using the RNA-Seq method. A total of 1 243 differentially expressed genes were identified: 897 genes were upregulated and 346 genes were downregulated in high litter size ovary samples compared with low litter size ovary samples. A large number of these genes related to steroid hormone regulation in animal ovaries, including 59 Gene Ontology terms and 27 Kyoto Encyclopedia of Genes and Genomes pathways involved in steroid biosynthesis and ovarian steroidogenesis. From these differentially expressed genes, we identified a total of 11 genes using a bioinformatics screen that may be associated with high litter size in Yorkshire pigs. These results provide a list of new candidate genes for porcine litter size and prolificacy to be further investigated.

Introduction

Litter size is one of the most important economic factors in pig production and is affected by interactions between multiple genes and the environment [1, 2]. The size of litters varies between pigs in different breeding farms. Altering litter size by conventional breeding methods can be slow, and has shown low heritability. However, using marker assisted selection (MAS) can speed up genetic improvements in litter size traits [3]. As the ovary directly mediates ovulation, it has a significant impact on the fecundity of mammals, and therefore, genetic differences in the ovaries may contribute to the observed differences in litter size [4, 5].

There has been some recent progress in characterizing the major genes involved in the prolificacy of swine, such as the estrogen receptor (ESR) [6, 7], follicle-stimulating hormone beta subunit (FSHB) [8], retinol-binding protein 4 (RBP4) [9, 10], prolactin receptor (PRLR) [9], melatonin receptor 1a (MTNR1A) [11], osteopontin (OPN) [12], bone morphogenetic protein (BMP) families [13], and growth differentiation factor (GDF9) [14]. With the recent publication of the pig genome, more candidate genes or quantitative trait loci (QTLs) have been extensively investigated for their involvement in porcine litter size [3, 15, 16]; but genomic location, function and interaction of these genes requires further research.

RNA sequencing (RNA-Seq) can measure genes, both quantitatively and functionally, at the transcriptome level [17]. To date, RNA-Seq has been used to study specific ovarian genes of important livestock animals (e.g., yak [18], and goat [19, 20]), but transcriptomic studies of specific genes in pigs are rare, and have mainly focused on the molecular regulation mechanism of fat deposition and muscle development [2123]. Therefore, transcriptomic analysis of pig ovaries using RNA-Seq may explain heredity of porcine fecundity, and be used to identify key genes relating to litter size.

Yorkshire is one of the world's most famous lean pig breeds, and is known for its higher fertility and high litter size [24, 25]. However, in pig production, some Yorkshire pigs always have low litter sizes, which seriously restricts productivity. In the present study, we sampled ovaries of Yorkshire pigs with extremely high and low litter sizes, and analyzed the differential expression of genes (DEGs) using RNA-Seq. The aim of this analysis was to identify major genes that control prolificacy, and thus provide a molecular basis for genetic improvement of reproductive traits in swine.

Materials and Methods

Ethics statement

Yorkshire (a Danish lean-type breed) pigs were obtained from the Anhui Daziran Primary Pig Breeding Farm, Huaibei, Anhui, China. All experimental procedures and sample collection were performed according to the Regulations for the Administration of Affairs Concerning Experimental Animals (Ministry of Science and Technology, China; revised in June 2004) and approved by the Institutional Animal Care and Use Committee of Anhui Agricultural University, Hefei, China, under permit No. ZXD-P20140809. This report fully adhered to the ARRIVE Guidelines for the reporting of animal research [26]. A completed ARRIVE guideline checklist is included (S1 Checklist). The animals were reared in the same environment and fed the same diet ad libitum during the experimental period. Food was withheld from the animals on the night before they were slaughtered.

Animals and ovary collection

A total of twelve healthy female pigs were used in this study from two groups: the extreme high litter size group (YH: n = 6), and the extreme low litter size group (YL: n = 6) (Table 1), representing pigs with high and low fecundity, respectively. In order to reduce, as far as is possible, the effects of age and parity on litter size, three pigs of similar age and parity from each group were selected as biological replicates for RNA-Seq. Their intact ovaries were rapidly harvested from their carcasses and immediately frozen in liquid nitrogen. All tissue samples were stored at −80°C until the total RNA extraction procedure was performed.

Table 1. Prolificacy characteristics of Yorkshire pigs with high and low litter sizes.

Group Sample ID Months of age Total parity numbers TNB NBA Use
YH YH1 30 4 16.5 ± 1.8 13.0 ± 0.4 RNA-Seq and qPCR
YH2 30 4 16.5 ± 1.8 13.0 ± 0.4 RNA-Seq and qPCR
YH3 35 5 17.0 ± 1.1 15.2 ± 1.3 RNA-Seq and qPCR
YH4 45 7 14.6 ± 1.8 11.0 ± 1.2 qPCR
YH5 46 7 16.4 ± 1.4 13.6 ± 1.0 qPCR
YH6 45 7 14.6 ± 1.8 11.0 ± 1.2 qPCR
YL YL1 30 4 5.2 ± 1.9 5.2 ± 1.9 RNA-Seq and qPCR
YL2 30 4 5.0 ± 0.7 4.0 ± 0.4 RNA-Seq and qPCR
YL3 35 5 5.8 ± 2.1 5.0 ± 1.6 RNA-Seq and qPCR
YL4 30 4 6.3 ± 2.1 5.3 ± 1.4 qPCR
YL5 31 4 5.5 ± 1.7 4.6 ± 0.6 qPCR
YL6 34 5 5.3 ± 0.8 4.5 ± 0.9 qPCR

Values are means ± standard error. YH represents the extreme high litter size group, and YL represents the extreme low litter size group. TNB represents the total number born, and NBA represents the number born alive.

mRNA library preparation and sequencing

The ovaries were completely ground and total RNA was extracted using TRIzol (Invitrogen, Carlsbad, CA, USA). The quality of the total RNA was checked using the Agilent 2100 Bioanalyzer system (Santa Clara, CA, USA). A total amount of 3 μg RNA per sample was used as input material for the RNA sample preparations. Sequencing libraries were generated using NEBNext® UltraTM RNA Library Prep Kit for Illumina® (NEB, USA) following manufacturer’s recommendations. Briefly, mRNA was extracted from total RNA using oligo (dT) magnetic beads and sheared into short fragments of about 200 bases. These fragmented mRNAs were then used as templates for cDNA synthesis. The cDNAs were then PCR amplified to complete the library. The cDNA library was sequenced using an Illumina HiSeqTM 2000 platform.

Analysis of RNA-Seq data

Raw RNA-Seq reads were processed through in-house perl scripts. Clean reads were obtained by removing reads containing low quality reads and/or adaptor sequences from raw reads [27], and mapped to the pig genome (Sus scrofa 10.2) using TopHat software [28], allowing up to two base mismatches. The gene expression level was then calculated using the reads per kilo bases per million reads (RPKM) method [29]. We considered the gene was expressed exclusively in one of the two groups if its RPKM ≥ 0.3 in one group and < 0.3 in the other group. Differential expression analysis was performed using the Benjamini and Hochberg’s approach for controlling the false discovery rate [30]. Genes with an adjusted P value < 0.05 were assigned as differentially expressed (DEG).

DEG lists were submitted to the databases of Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) for enrichment analysis of the significant overrepresentation of GO terms and KEGG-pathway categories [31, 32]. In all tests, P values were calculated using the Benjamini-corrected modified Fisher’s exact test and ≤ 0.05 was taken as a threshold of significance.

Quantitative PCR analysis

The RNA-Seq results were validated using an RNA samples from the YH group (n = 6) and the YL group (n = 6), respectively. Six DEGs enriched in the ovarian steroidogenesis pathway, two DEGs (one upregulated, and the other downregulated in the YH group) were enriched for metabolic pathways, two DEGs with the highest numbers of reads in both YH and YL, and two genes (one expressed only in the YH group, and the other expressed only in the YL group) were analyzed by quantitative PCR (qPCR) (S1 Table). Total RNA (1.0 mg) was used to synthesize first-strand cDNA using PrimeScript RT Master Mix (TaKaRa, Osaka, Japan). Q-PCR was performed using SYBR Premix Ex Taq (TaKaRa, Osaka, Japan) in the CFX96 Real-Time PCR Detection System (Bio-Rad, Hercules, CA, USA). The primers were designed using the Primer Express (Applied Biosystems) software. All the mRNA nucleotide sequences were obtained from the NCBI Entrez Nucleotide database (http://www.ncbi.nlm.nih.gov/sites/entrez?db=nuccore&itool=toolbar), or from the EMBL-EBI database (http://www.ebi.ac.uk). The primers used for qPCR are listed in S1 Table. The thermal cycling conditions were 95°C for 10 min, followed by 40 cycles of 95°C for 15 s and 60°C for 1 min. There were three replicates for each amplification. Cycle threshold (Ct) values were analysed using a linear mixed model as described below.

Ctjkl= μ +Lj+ Ak+ Pl+ ejkl;

in which L is the effect of fecundity, A is the fixed effect of age, and P is the fixed effect of parity of animal [33]. Then the Ct values were transformed to quantities using the comparative Ct method. Data was normalized using the porcine β-actin reference gene. Comparison of gene expression levels conditional on prolificacy levels was performed using the t-test, and correlations between qPCR and RNA-Seq measures were calculated.

Results

Overview of sequencing data

After removing the low quality and adaptor sequences, we obtained approximately 52 to 66 million (M) clean reads for six RNA-Seq libraries, and high percentages of mapped reads ranging from 79.15 to 80.68%. Most mapped reads were located within an exon (71.6 to 85.6%) while a smaller percentage of mapped reads (less than 19. 0%) were located within the introns and intergenic regions (Table 2). These results indicated that our six libraries were of high quality, and had high coverage of the pig genome. This allowed us to compare the ovary transcriptomes from pigs with high and low litter size. The data is available from the Sequence Read Archive (SRA) (Accession no. SRP058401, Bioproject: PRJNA283575).

Table 2. RNA sequencing results of mRNA from the ovaries of Yorkshire pigs with high and low litter sizes.

Sample ID 1 Clean reads, M 2 Mapping rate, % 3 exons, % Intron, % Intergenic, %
YH1 52.66 80.67 82.20 3.70 14.20
YH2 60.14 80.68 85.60 2.40 12.00
YH3 64.89 80.07 82.20 4.10 13.70
YL1 65.60 79.80 73.20 8.30 18.60
YL2 57.76 80.16 71.60 9.40 19.00
YL3 65.04 79.15 78.30 6.00 15.70

1. YH1, YH2, YH3 and YL1, YL2, YL3 are replicate from the YH and YL groups. YH represents the extreme high litter size group, and YL represents the extreme low litter size group.

2. Indicates millions of reads.

3. Uses the Sus scrofa 10.2 as the reference genome annotation to classify the mapping tags into the different regions. Ratio was calculated by the number of tags on each region divided by the total tags on the whole genome.

Differentially expressed genes between YH and YL groups

After mapping to the pig genome, a total of 17 485 and 19 178 genes were obtained from the YH and YL libraries, respectively. Four hundred and two genes were expressed only in YH, 2 059 genes were expressed only in YL, and 17 083 genes were co-expressed in both libraries (Fig 1A). A total of 1 243 genes were differentially expressed between the two groups, in which 897 genes were upregulated and 346 genes were downregulated in the YH group (Fig 1B and S2 Table). The 10 most differentially expressed genes (log2FoldChange ≥4) from the total of 1 243 DEGs identified between the high and low litter size samples were: homogentisate 1,2 dioxygenase (HGD); phosphoenylpyruvate carboxykinase (PCK1), HSD17B2; early growth response 4 (EGR4), a member of the ras oncogene family (RAB33A); solute carrier protein family 6 (SLC6A20B); zinc finger protein (GLI1); U6 spliceosomal RNA (U6); solute carrier protein family 7 (SLC7A11); and spectrin alpha chain eyrythrocytic 1 (SPTA1), respectively (Table 3).

Fig 1. Comparative results of gene expression levels and differentially expressed gene distributions between the ovaries of Yorkshire pigs with extremely high (YH) and low (YL) litter size.

Fig 1

(A) Venn diagram showing genes only expressed in the YH group (yellow circle), only expressed in the YL group (light red circle), and common to both groups (intersection). (B) Scatter plot of differentially expressed genes (YH vs. YL). Red points represent upregulated genes with log2 (fold change) > 1 and padj < 0.05 (–log10 (padj) ≥ 1.3); Blue points represent downregulated genes with log2 (fold change) < -1 and padj < 0.05 (–log10 (padj) ≥ 1.3). Green points represent genes with no significant difference. Fold change = gene normalized expression of the YH group / gene normalized expression of the YL group.

Table 3. Detailed information on the top 10 most differentially expressed genes.

Gene Name readcount_ YH readcount_ YL log2FoldChange P val P adj Up/Down (YH/YL) Interpro Description
HGD 77.54 0.53 7.18 8.29E-05 0.00436 Up Homogentisate 1,2-dioxygenase
PCK1 417.45 3.35 6.96 0.000101 0.005071 Up Phosphoenolpyruvate carboxykinase,
HSD17B2 235.38 1.91 6.95 0.000661 0.019735 Up Hydroxysteroid (17-beta) dehydrogenase 2, mRNA.
EGR4 245.79 2.15 6.84 0.000716 0.020727 Up Early growth response 4
RAB33A 488.52 5.84 6.39 4.92E-11 1.96E-08 Up Member RAS oncogene family (RAB33A), mRNA
SLC6A20B 0.81 15.67 -4.27 0.000992 0.026366 Down Solute carrier family 6, member 20B
GLI1 0.81 16.24 -4.32 0.000719 0.020791 Down Zinc finger protein GLI1
U6 0.41 12.20 -4.91 0.002086 0.043381 Down U6 spliceosomal RNA
SLC7A11 0.39 16.53 -5.42 0.000196 0.008195 Down Solute carrier family 7, member 11
SPTA1 2.43 331.59 -7.09 7.43E-10 2.25E-07 Down Spectrin, alpha, erythrocytic 1

Functional enrichment analysis of differentially expressed genes

To define the biological functions of the 1 243 DEGs, GO and KEGG analysis were carried out. Fifty-nine significantly enriched GO terms (corrected P < 0.05) were identified, including single-organism metabolic process, lipid metabolic process, oxidation-reduction process, lipid biosynthetic process, organic acid metabolic process, carboxylic acid metabolic process, oxoacid metabolic process, small molecule metabolic process, fatty acid metabolic process, cellular lipid metabolic process, catalytic activity, steroid metabolic process, and glucose metabolic process (Table 4 and S3 Table). Meanwhile, 27 significantly enriched KEGG pathways were identified, including metabolic pathways, valine/leucine and isoleucine degradation, fatty acid metabolism, carbon metabolism, steroid biosynthesis, butanoate metabolism, ovarian steroidogenesis, biosynthesis of unsaturated fatty acids, PPAR signaling pathway, synaptic vesicle cycle, and biosynthesis of amino acids (Table 5 and S4 Table). Among these GO terms and KEGG pathways, the steroid biosynthesis and ovarian steroidogenesis pathways are the ones related to steroid hormone regulation in animal ovaries and therefore likely to be contributing to litter size. However, as most GO and KEGG assignments and distributions are related to reproduction, growth and development, and metabolism, our results indicate that the DEGs are involved in a wide range of regulatory functions in porcine ovaries.

Table 4. The top 10 most significantly enriched Gene Ontology (GO) terms from differentially expressed genes (DEGs) in Yorkshire pigs.

GO term Description P value Corrected P value DEGs
GO:0044710 single-organism metabolic process 4.91E-19 9.99E-16 221
GO:0055114 oxidation-reduction process 8.01E-15 8.15E-12 115
GO:0044281 small molecule metabolic process 1.03E-09 3.01E-07 97
GO:0006629 lipid metabolic process 3.56E-15 4.83E-12 82
GO:0006082 organic acid metabolic process 4.36E-10 1.37E-07 58
GO:0019752 carboxylic acid metabolic process 4.36E-10 1.37E-07 58
GO:0043436 oxoacid metabolic process 4.36E-10 1.37E-07 58
GO:0044255 cellular lipid metabolic process 4.83E-09 1.23E-06 54
GO:0008610 lipid biosynthetic process 3.86E-10 1.37E-07 51
GO:0006631 fatty acid metabolic process 3.07E-09 8.32E-07 22

Table 5. The top ten significantly enriched Kyoto Encyclopedia of Genes and Genomes pathways from differentially expressed genes (DEGs).

Pathway ID Pathway P value Corrected P value DGEs
ssc01100 Metabolic pathways 1.14E-18 2.90E-16 191
ssc01200 Carbon metabolism 2.64E-11 1.67E-09 35
ssc00280 Valine, leucine and isoleucine degradation 3.04E-12 3.86E-10 26
ssc01212 Fatty acid metabolism 1.05E-11 8.92E-10 25
ssc04913 Ovarian steroidogenesis 2.96E-08 1.08E-06 21
ssc00071 Fatty acid degradation 5.62E-07 1.59E-05 17
ssc00100 Steroid biosynthesis 2.08E-09 1.06E-07 16
ssc00650 Butanoate metabolism 5.30E-09 2.24E-07 16
ssc01040 Biosynthesis of unsaturated fatty acids 3.97E-07 1.26E-05 12
ssc00062 Fatty acid elongation 1.40E-06 3.56E-05 12

Validation of DEGs by qPCR

Twelve genes were used for qPCR analysis from the ovaries of the YH and YL groups to validate the expression profiles obtained by RNA-Seq. Consistent with the RNA-Seq findings, we verified that hydroxysteroid (17-beta) dehydrogenase 2 (HSD17B2), cytochrome P450 E class group 1 (CYP11A1), steroidogenic acute regulatory protein (STAR), low density lipoprotein receptor (LDLR), scavenger receptor class B member 1 (SCARB1), cytochrome c oxidase, subunit I domain (CO1), and cytochrome c oxidase subunit III domain (COX3) genes were all upregulated, while luteinizing hormone/choriogonadotopin receptor (LHCGR), and insulin-like growth factor 1 (IGF-1) genes were downregulated in high litter size samples. Matrix metallopeptidase 13 (MMP13) gene was not significant difference between the two groups. However, branched-chain amino acid aminotransferase II (BCAT2) and dopachrome tautomerase (DCT) genes were validated as different expression with P > 0.05 in YH versus YL groups (Table 6 and Fig 2). Therefore, results obtained from RNA-Seq were statistically confirmed for 83.3% of the tested genes by qPCR. In all cases, the relative fold change of gene expression was in the same direction between the RNA-Seq and qPCR data.

Table 6. Summary of qPCR and RNA-Seq results.

Gene qPCR results RNA-Seq Fold Change (H/L) Significant diffs. Confirmed Correlation RNA-Seq vs. qPCR
YH mean (SE) YL mean (SE) Fold Chang (H/L) P value (H/L)
HSD17B2 0.02372 0.00025 96.419 0.037 123.469 Up Yes 0.85
CYP11A1 0.64853 0.02040 31.789 0.003 15.909 Up Yes 0.83
STAR 1.23509 0.01774 69.642 0.008 11.766 Up Yes 0.56
LDLR 0.03103 0.00056 55.888 0.001 7.555 Up Yes 0.74
SCARB1 0.67635 0.01722 39.268 0.010 12.028 Up Yes 0.62
CO1 3.26406 0.35572 9.176 0.005 5.799 Up Yes 0.85
COX3 3.84001 0.37585 10.217 0.003 4.490 Up Yes 0.76
BCAT2 0.53987 0.16381 3.296 0.293 6.860 Up No 0.39
IGF-1 0.03046 0.25306 0.120 0.037 0.283 down Yes 0.82
LHCGR 0.02432 0.19728 0.123 0.009 0.244 down Yes 0.63
DCT 0.00006 0.00058 0.100 0.174 0.275 down No 0.28
MMP13 0.00003 0.00061 0.048 0.322 0.009 No Yes 0.65

Fig 2. Verification of the RNA-Seq results using the qPCR method.

Fig 2

Fold change values greater than 2 and P < 0.05 indicate overexpression in the YH group, and fold change values less than 0.5 and P < 0.05 indicate overexpression in the YL group. Genes with asterisk differ significantly (P < 0.05).

Identification of candidate genes for pig litter size

To accurately identify the key genes that may influence porcine litter size, we created a Venn diagram of the 10 most DEGs between high and low litter size samples (Table 3), the 10 most expressed genes in the ovaries of pigs with high litter size (Table 7), the 10 most expressed genes in the ovaries of pigs with low litter size (Table 7), and the 21 genes enriched in the steroid metabolic process and ovarian steroidogenesis of the above-mentioned (S5 Table). The results showed that 11 genes appeared in the two or more sets, including the CO1, glutathione peroxidase 3 (GPX3), beta-microseminoprotein (MSMB), COX3, tissue inhibitor of metalloproteinase 1 (TIMP1), cytochrome b (CYTB), STAR, 3-beta hydroxysteroid dehydrogenase (HSD3B), CYP11A1, SCARB1, and hydroxysteroid (17-beta) dehydrogenase 2 (HSD17B2) (Fig 3). These 11 genes may be candidates for porcine fecundity and litter size.

Table 7. Detailed information of the top 10 most expressed genes in the YH and YL groups.

Group Gene Name Readcount_YH Readcount_YL log2FoldChange P val P adj Up/Down(YH/YL) Interpro Description
YH CO1 1062714.38 183270.39 2.54 6.86E-07 8.59E-05 Up Cytochrome c oxidase subunit I domain
GPX3 829844.21 63398.00 3.71 2.35E-07 3.43E-05 Up Glutathione peroxidase 3
MSMB 462726.06 19382.82 4.58 1.90E-18 6.77E-15 Up Beta-microseminoprotein
COX3 444826.68 99075.58 2.17 1.34E-05 0.001048 Up Cytochrome c oxidase subunit III domain
TIMP1 407692.96 34203.54 3.58 7.45E-07 9.19E-05 Up Tissue inhibitor of metalloproteinases-like
STAR 350027.39 29747.05 3.56 1.76E-08 3.57E-06 Up Steroidogenic acute regulatory protein, animal
HSD3B 256334.52 26353.57 3.28 3.39E-11 1.45E-08 Up 3-beta hydroxysteroid dehydrogenase
CYTB 187523.02 46638.76 2.01 9.15E-05 0.00469 Up Cytochrome b
CYP11A1 148774.93 9351.70 3.99 2.42E-16 3.04E-13 Up Cytochrome P450 E class group 1
SCARB1 128598.74 10691.72 3.59 9.40E-14 7.03E-11 Up Scavenger receptor class B, member 1
YL CO1 1062714.38 183270.39 2.54 6.86E-07 8.59E-05 Up Cytochrome c oxidase, subunit I domain
COX3 444826.68 99075.58 2.17 1.34E-05 0.001048 Up Cytochrome c oxidase subunit III domain
COL1A2 25163.69 68810.42 -1.45 0.000946 0.02541 Down Collagen triple helix repeat
GPX3 829844.21 63398.00 3.71 2.35E-07 3.43E-05 Up Glutathione peroxidase 3
CYTB 187523.02 46638.76 2.01 9.15E-05 0.00469 Up Cytochrome b
TIMP1 407692.96 34203.54 3.58 7.45E-07 9.19E-05 Up Tissue inhibitor of metalloproteinases-like
STAR 350027.39 29747.05 3.56 1.76E-08 3.57E-06 Up Steroidogenic acute regulatory protein, animal
SEPP1 81114.92 29232.10 1.47 0.001024 0.027035 Up Selenoprotein P precursor 1
HSD3B 256334.52 26353.57 3.28 3.39E-11 1.45E-08 Up 3-beta hydroxysteroid dehydrogenase
MSMB 462726.06 19382.82 4.58 1.90E-18 6.77E-15 Up Beta-microseminoprotein

Fig 3. Venn diagram for identification of key genes influencing porcine litter size.

Fig 3

T10DEGs represents the top 10 most DEGs between the high and low litter size samples; T10YH represents the top 10 most expressed genes in the high litter size samples; T10YL represents the top 10 most expressed genes in the low litter size samples; E21OS represents the 21 genes enriched in the steroid metabolic process and ovarian steroidogenesis. Six, in the light green overlapping set surrounded by the red triangle, represents the CO1, GPX3, MSMB, COX3, TIMP1, and CYTB genes; two, in the brown overlapping set surrounded by the red triangle, represents the STAR and HSD3B genes; two, in the light brown overlapping set surrounded by the red triangle, represents the CYP11A1 and SCARB1 genes; and one, in the violet overlapping set surrounded by the red triangle, represents the HSD17B2 gene. Therefore, 11 genes (CO1, GPX3, MSMB, COX3, TIMP1, CYTB, STAR, HSD3B, CYP11A1, SCARB1, and HSD17B2) were present in two or more assemblies.

Discussion

Ovaries are one of the most important animal reproductive organs. They directly mediate ovulation and female hormone secretion, which has a significant impact on the fecundity of mammals [3436]. Some specific genes relating to animal fecundity have been reported using transcriptome analysis of certain characteristics in ovaries [18, 37], however similar studies on litter size in pigs had not been conducted to date. It is well-known that there is gene expression specificity in different tissues and cells. The ovaries contain a mixture of different tissue, and the expression of candidate genes may differ between them. We strove to ensure that we obtained intact ovaries and ground them completely for the purpose of RNA extraction, to ensure that the RNA-Seq results were representative of the complete porcine ovarian transcriptome. Additionally, the age of the sow is known to greatly affect litter size. In order to minimize this and the effect of parity, we selected six pigs of similar age and parity for RNA-Seq. Two sequencing libraries were constructed from extremely high and low litter size samples. High quality transcriptome data was generated (60 million clean reads; 80% genome mapping rate), which was sufficient for the quantitative analysis of gene expression.

Previous studies of goat ovaries suggested that the most differentially expressed genes identified by RNA-Seq were likely to be important for improving litter size [19]. Some of the top 10 most differentially expressed genes found in our study have been reported to be candidate genes involved in important metabolic processes. For example: the PCK1 gene has previously been shown to be associated with lipid metabolism in animals [3841]; the HSD17B2 gene has been identified as a key regulator of steroid hormone metabolism [4246]; the EGR4 gene is a candidate gene for reproductivity in animal ovary and testis [4750]; and the RAB33A gene may affect the risk of developing certain disease, such as tuberculosis [51, 52]. However, while there was an association between HGD and meat quality traits reported in Chinese red cattle [53, 54], these gene association studies have mainly concentrated on humans or animal models. Therefore, the role of these genes in pig fecundity requires further investigation.

From our GO and KEGG analysis, we found that the functions of the 10 most DEGs between high and low litter size samples were mainly in the metabolic pathways, ovarian steroidogenesis, citrate cycle (TCA cycle), pyruvate metabolism, and the PPAR signaling pathway. It is accepted that the molecular regulation of animal traits is very complex and the relationship between genes and traits is often that of “one-to-many” or “many-to-one”. The DEGs were not only enriched in reproduction-related pathways but also in those involved with lipid and fatty acid metabolism. This suggests that the genes may be associated with both reproduction and fat metabolism. It is easily appreciated that, in the life of organisms, all physiological processes interact with each other. The pathways mentioned above are, to a greater or lesser degree, involved in the development of follicular cells and oocytes. Consequently, functional studies should be performed with these DGEs in order to identify key candidate genes influencing reproductive traits in swine.

Previous research with gene expression microarrays and QTL mapping analysis, identified 27 candidate genes, co-localizing with QTL for porcine litter size traits, which fulfill biological, positional, and functional criteria [55, 56]. These genes, which include CYP19A1, CYP2E1, RBP4, MSRB2, and SLC16A3, mainly encode specific proteins, hormones and cytokines. CYP11A1 (which belongs to cytochrome P450 E class group 1), RBP4 (retinol-binding protein 4), and SLC5A10, SLC7A11 & SLC6A20B (which belong to the solute carrier family) were also identified in our study as DEGs in pigs, suggesting that they may also be relevant to porcine prolificacy (S2 Table). However, apart from these DEGs, the other candidate genes found previously to co-localize with QTL for porcine litter size were not identified in our study. This may be because of inter-species differences, or it may be a consequence of an insufficiently large sample size.

Genes that are highly expressed in reproductive tissues or cells may also indicate that the gene itself is activated, as shown in previous studies [5759]. Therefore, we identified the 10 most highly expressed genes in the ovaries of high and low litter size pigs, respectively (Table 7). Eight of these genes (CO1, GPX3, MSMB, COX3, TIMP1, STAR, HSD3B, and CYTB) were upregulated in the high litter size samples. Some of them may be involved in specific reproductive processes. For example, previous research has shown that MSMB participates in spermatogenesis [60], and that HSD3B and STAR regulate specific gonadal development and hormone metabolism pathways [6164]. The other most highly expressed genes may play other important roles in biological processes. CO1 and COX3 genes have been extensively utilized in polymorphic and phylogenetic analysis in some species [65, 66] and GPX3 is associated with certain cancers [67, 68].

Furthermore, in order to identify more candidate genes relate to porcine litter size, we emphatically analyzed the 21 candidate genes (S5 Table) that may be associated with ovarian steroid hormone secretion. We showed that 19 of these 21 genes were all upregulated in the ovaries of pigs with high litter size. This suggests that the ovarian steroidogenesis pathway may be activated. These 21 genes mainly contribute to ovarian steroidogenesis and development of follicular cells.

Finally, by comparing the most highly expressed genes in the high and low litter size samples with the 10 most DEGs, and the 21 genes enriched in the steroid metabolic process and ovarian steroidogenesis, we identified a total of 11 candidate genes (CO1, GPX3, MSMB, COX3, TIMP1, CYTB, STAR, HSD3B, CYP11A1, SCARB1, and HSD17B2) relating to porcine fecundity and litter size. Therefore, the reproductive roles of these 11 genes in pigs should be further investigated in specific ovarian cells (such as theca and granulosa cells) in order to determine their functions both in vitro and in vivo.

Conclusion

This study screened for DEGs in the ovarian tissues of extremely high and low litter size Yorkshire pigs using RNA-Seq. We identified 897 genes that were upregulated and 346 genes that were downregulated in the high litter size samples. After analyzing the function of these genes, we found 11 DEGs that may be relevant to the prolificacy of pigs. This new information provides a solid foundation for further studies of the molecular mechanisms underlying porcine prolificacy. In the future, biochemical and physiological analyses of these candidate genes will be conducted.

Supporting Information

S1 Checklist. Completed ARRIVE Guidelines Checklist for the reporting of animal data.

(PDF)

S1 Table. Primer pairs selected for RNA-Seq validation by qPCR.

(XLSX)

S2 Table. Detailed information of differentially expressed genes.

(XLS)

S3 Table. Significantly enriched GO terms among differentially expressed genes.

(XLS)

S4 Table. Significantly enriched KEGG pathways among differentially expressed genes.

(XLS)

S5 Table. Detailed information of 21 differentially expressed genes enriched in the ovarian steroidogenesis pathway.

(XLSX)

Acknowledgments

Authors are indebted to Qinsheng Chen from Anhui Daziran Primary Pig Breeding Farm for providing pigs for experiment and to animal molecular quantitative genetics team members from Anhui Agricultural University for experiment.

Data Availability

The authors confirm that all data underlying the findings are fully available without restriction. Data are available from the Sequence Read Archive (SRA) under Accession no. SRP058401, Bioproject: PRJNA283575.

Funding Statement

This research was supported by grants from the National Natural Science Foundation of China (Nos. 31402037 and 31171200), the Anhui Provincial Natural Science Foundation (Nos. 1408085QC55 and 1508085QC52), and the Project of Anhui Province Scientific Technology Plan (No. 1301031032). The funders had no role in study design, data collection and analysis, decision to publish, or preparation of the manuscript.

References

  • 1. Lee DG, Nam J, Kim SW, Kang YM, An HJ, Kim CW, et al. Proteomic analysis of reproduction proteins involved in litter size from porcine placenta. Bioscience, biotechnology, and biochemistry. 2015;79(9):1414–21. 10.1080/09168451.2015.1039478 [DOI] [PubMed] [Google Scholar]
  • 2. Kwon WS, Rahman MS, Lee JS, Yoon SJ, Park YJ, Pang MG. Discovery of predictive biomarkers for litter size in boar spermatozoa. Molecular & cellular proteomics: MCP. 2015;14(5):1230–40. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3. Du H, Chen J, Cui J, Wang X, Zhang X. Polymorphisms on SSC15q21-q26 Containing QTL for reproduction in Swine and its association with litter size. Genetics and molecular biology. 2009;32(1):69–74. 10.1590/S1415-47572009000100010 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4. Stephens SM, Moley KH. Follicular origins of modern reproductive endocrinology. American journal of physiology Endocrinology and metabolism. 2009;297(6):E1235–6. 10.1152/ajpendo.00575.2009 [DOI] [PubMed] [Google Scholar]
  • 5. Pan Z, Zhang J, Lin F, Ma X, Wang X, Liu H. Expression profiles of key candidate genes involved in steroidogenesis during follicular atresia in the pig ovary. Molecular biology reports. 2012;39(12):10823–32. 10.1007/s11033-012-1976-2 [DOI] [PubMed] [Google Scholar]
  • 6. Munoz G, Ovilo C, Estelle J, Silio L, Fernandez A, Rodriguez C. Association with litter size of new polymorphisms on ESR1 and ESR2 genes in a Chinese-European pig line. Genetics, selection, evolution: GSE. 2007;39(2):195–206. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7. Chen KF, Huang LS, Li N, Zhang Q, Luo M, Wu CX. The genetic effect of estrogen receptor(ESR) on litter size traits in pig. Yi chuan xue bao = Acta genetica Sinica. 2000;27(10):853–7. [PubMed] [Google Scholar]
  • 8. Zhao Y, Li N, Xiao L, Cao G, Chen Y, Zhang S, et al. FSHB subunit gene is associated with major gene controlling litter size in commercial pig breeds. Science in China Series C, Life sciences / Chinese Academy of Sciences. 1998;41(6):664–8. 10.1007/BF02882910 [DOI] [PubMed] [Google Scholar]
  • 9. Sun YX, Zeng YQ, Tang H, Fan XZ, Chen QM, Li H, et al. Relationship of genetic polymorphism of PRLR and RBP4 genes with litter size traits in pig. Yi chuan = Hereditas / Zhongguo yi chuan xue hui bian ji. 2009;31(1):63–8. [DOI] [PubMed] [Google Scholar]
  • 10. Spotter A, Muller S, Hamann H, Distl O. Effect of polymorphisms in the genes for LIF and RBP4 on litter size in two German pig lines. Reproduction in domestic animals = Zuchthygiene. 2009;44(1):100–5. 10.1111/j.1439-0531.2007.01004.x [DOI] [PubMed] [Google Scholar]
  • 11. Ramirez O, Tomas A, Barragan C, Noguera JL, Amills M, Varona L. Pig melatonin receptor 1a (MTNR1A) genotype is associated with seasonal variation of sow litter size. Animal reproduction science. 2009;115(1–4):317–22. 10.1016/j.anireprosci.2008.12.013 [DOI] [PubMed] [Google Scholar]
  • 12. Goluch D, Korwin-Kossakowska A, Prusak B, Pierzchala M, Urbanski P, Michaluk A, et al. The study of polymorphism within the promoter region of the osteopontin (OPN) gene in sows. Neuro endocrinology letters. 2009;30(4):525–9. [PubMed] [Google Scholar]
  • 13. Paradis F, Novak S, Murdoch GK, Dyck MK, Dixon WT, Foxcroft GR. Temporal regulation of BMP2, BMP6, BMP15, GDF9, BMPR1A, BMPR1B, BMPR2 and TGFBR1 mRNA expression in the oocyte, granulosa and theca cells of developing preovulatory follicles in the pig. Reproduction. 2009;138(1):115–29. 10.1530/REP-08-0538 [DOI] [PubMed] [Google Scholar]
  • 14. Shimizu T, Miyahayashi Y, Yokoo M, Hoshino Y, Sasada H, Sato E. Molecular cloning of porcine growth differentiation factor 9 (GDF-9) cDNA and its role in early folliculogenesis: direct ovarian injection of GDF-9 gene fragments promotes early folliculogenesis. Reproduction. 2004;128(5):537–43. [DOI] [PubMed] [Google Scholar]
  • 15. Rothschild MF, Hu ZL, Jiang Z. Advances in QTL mapping in pigs. International journal of biological sciences. 2007;3(3):192–7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16. Distl O. Mechanisms of regulation of litter size in pigs on the genome level. Reproduction in domestic animals = Zuchthygiene. 2007;42 Suppl 2:10–6. [DOI] [PubMed] [Google Scholar]
  • 17. Marioni JC, Mason CE, Mane SM, Stephens M, Gilad Y. RNA-seq: an assessment of technical reproducibility and comparison with gene expression arrays. Genome research. 2008;18(9):1509–17. 10.1101/gr.079558.108 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18. Lan D, Xiong X, Wei Y, Xu T, Zhong J, Zhi X, et al. RNA-Seq analysis of yak ovary: improving yak gene structure information and mining reproduction-related genes. Science China Life sciences. 2014;57(9):925–35. 10.1007/s11427-014-4678-2 [DOI] [PubMed] [Google Scholar]
  • 19. Ling YH, Xiang H, Li YS, Liu Y, Zhang YH, Zhang ZJ, et al. Exploring differentially expressed genes in the ovaries of uniparous and multiparous goats using the RNA-Seq (Quantification) method. Gene. 2014;550(1):148–53. 10.1016/j.gene.2014.08.008 [DOI] [PubMed] [Google Scholar]
  • 20. Chen HY, Shen H, Jia B, Zhang YS, Wang XH, Zeng XC. Differential Gene Expression in Ovaries of Qira Black Sheep and Hetian Sheep Using RNA-Seq Technique. PLOS one. 2015;10(3):e0120170 10.1371/journal.pone.0120170 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21. Sodhi SS, Song KD, Ghosh M, Sharma N, Lee SJ, Kim JH, et al. Comparative transcriptomic analysis by RNA-seq to discern differential expression of genes in liver and muscle tissues of adult Berkshire and Jeju Native Pig. Gene. 2014;546(2):233–42. 10.1016/j.gene.2014.06.005 [DOI] [PubMed] [Google Scholar]
  • 22. Sodhi SS, Park WC, Ghosh M, Kim JN, Sharma N, Shin KY, et al. Comparative transcriptomic analysis to identify differentially expressed genes in fat tissue of adult Berkshire and Jeju Native Pig using RNA-seq. Molecular biology reports. 2014;41(9):6305–15. 10.1007/s11033-014-3513-y [DOI] [PubMed] [Google Scholar]
  • 23. Jung WY, Kwon SG, Son M, Cho ES, Lee Y, Kim JH, et al. RNA-Seq approach for genetic improvement of meat quality in pig and evolutionary insight into the substrate specificity of animal carbonyl reductases. PLOS one. 2012;7(9):e42198 10.1371/journal.pone.0042198 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24. Wolf J. Heritabilities and genetic correlations for litter size and semen traits in Czech Large White and Landrace pigs. Journal of animal science. 2010;88(9):2893–903. 10.2527/jas.2009-2555 [DOI] [PubMed] [Google Scholar]
  • 25. Arango J, Misztal I, Tsuruta S, Culbertson M, Herring W. Threshold-linear estimation of genetic parameters for farrowing mortality, litter size, and test performance of Large White sows. Journal of animal science. 2005;83(3):499–506. [DOI] [PubMed] [Google Scholar]
  • 26. Kilkenny C, Browne WJ, Cuthill IC, Emerson M, Altman DG. Improving bioscience research reporting: the ARRIVE guidelines for reporting animal research. PLOS biology. 2010;8(6):e1000412 10.1371/journal.pbio.1000412 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27. Kerpedjiev P, Frellsen J, Lindgreen S, Krogh A. Adaptable probabilistic mapping of short reads using position specific scoring matrices. BMC bioinformatics. 2014;15 10.1186/1471-2105-15-15 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28. Kim D, Salzberg SL. TopHat-Fusion: an algorithm for discovery of novel fusion transcripts. Genome Biol. 2011;12(8):R72 10.1186/gb-2011-12-8-r72 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29. Mortazavi A, Williams BA, Mccue K, Schaeffer L, Wold B. Mapping and quantifying mammalian transcriptomes by RNA-Seq. Nature methods. 2008;5(7):621–8. 10.1038/nmeth.1226 [DOI] [PubMed] [Google Scholar]
  • 30. Benjamini Y, Drai D, Elmer G, Kafkafi N, Golani I. Controlling the false discovery rate in behavior genetics research. Behav Brain Res. 2001;125(1–2):279–84. [DOI] [PubMed] [Google Scholar]
  • 31. Ashburner M, Ball CA, Blake JA, Botstein D, Butler H, Cherry JM, et al. Gene Ontology: tool for the unification of biology. Nat Genet. 2000;25(1):25–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32. Kanehisa M, Araki M, Goto S, Hattori M, Hirakawa M, Itoh M, et al. KEGG for linking genomes to life and the environment. Nucleic acids research. 2008;36:D480–D4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33. Steibel JP, Poletto R, Coussens PM, Rosa GJM. A powerful and flexible linear mixed model framework for the analysis of relative quantification RT-PCR data. Genomics. 2009;94(2):146–52. 10.1016/j.ygeno.2009.04.008 [DOI] [PubMed] [Google Scholar]
  • 34. Weiner S, Wright KH, Wallach EE. Studies on the function of the denervated rabbit ovary: human chorionic gonadotropin-induced ovulation. Fertility and sterility. 1975;26(4):363–8. [PubMed] [Google Scholar]
  • 35. Paeschke KD. Ovulation preliminaries and ovulation. I. Generative function of the ovary and ascorbic acid metabolism during the ovarian cycle. Fortschritte der Geburtshilfe und Gynakologie. 1970;43:1–58. [PubMed] [Google Scholar]
  • 36. Rojas FJ. Ovulation induction. Effects of ovulation induction with gonadotrophins on the ovary and uterus and their implications for assisted reproduction. Human reproduction. 1995;10(9):2219–24. [DOI] [PubMed] [Google Scholar]
  • 37. Chen L, Liu K, Zhao Z, Blair HT, Zhang P, Li D, et al. Identification of sheep ovary genes potentially associated with off-season reproduction. Journal of genetics and genomics = Yi chuan xue bao. 2012;39(4):181–90. 10.1016/j.jgg.2012.03.002 [DOI] [PubMed] [Google Scholar]
  • 38. Metges CC, Gors S, Lang IS, Hammon HM, Brussow KP, Weitzel JM, et al. Low and high dietary protein:carbohydrate ratios during pregnancy affect materno-fetal glucose metabolism in pigs. The Journal of nutrition. 2014;144(2):155–63. 10.3945/jn.113.182691 [DOI] [PubMed] [Google Scholar]
  • 39. Guo F, Zhang Y, Su L, Ahmed AA, Ni Y, Zhao R. Breed-dependent transcriptional regulation of phosphoenolpyruvate carboxylase, cytosolic form, expression in the liver of broiler chickens. Poultry science. 2013;92(10):2737–44. 10.3382/ps.2013-03189 [DOI] [PubMed] [Google Scholar]
  • 40. Duan J, Shao F, Shao Y, Li J, Ling Y, Teng K, et al. Androgen inhibits abdominal fat accumulation and negatively regulates the PCK1 gene in male chickens. PLOS one. 2013;8(3):e59636 10.1371/journal.pone.0059636 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41. Hsieh CW, Huang C, Bederman I, Yang J, Beidelschies M, Hatzoglou M, et al. Function of phosphoenolpyruvate carboxykinase in mammary gland epithelial cells. Journal of lipid research. 2011;52(7):1352–62. 10.1194/jlr.M012666 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42. Wang CL, Ying SJ, Wang ZY, Xing HJ, Wang LZ, He DY, et al. Molecular cloning and expression of 17beta-hydroxysteroid dehydrogenase type 2 gene in Hu sheep. Molecular biology reports. 2013;40(2):1073–80. 10.1007/s11033-012-2149-z [DOI] [PubMed] [Google Scholar]
  • 43. Gao H, Yallampalli U, Yallampalli C. Gestational protein restriction reduces expression of Hsd17b2 in rat placental labyrinth. Biology of reproduction. 2012;87(3):68 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44. Shen Z, Peng Z, Sun Y, Vaananen HK, Poutanen M. Overexpression of human hydroxysteroid (17beta) dehydrogenase 2 induces disturbance in skeletal development in young male mice. Journal of bone and mineral research: the official journal of the American Society for Bone and Mineral Research. 2008;23(8):1217–26. [DOI] [PubMed] [Google Scholar]
  • 45. Rantakari P, Strauss L, Kiviranta R, Lagerbohm H, Paviala J, Holopainen I, et al. Placenta defects and embryonic lethality resulting from disruption of mouse hydroxysteroid (17-beta) dehydrogenase 2 gene. Molecular endocrinology. 2008;22(3):665–75. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46. Zhongyi S, Rantakari P, Lamminen T, Toppari J, Poutanen M. Transgenic male mice expressing human hydroxysteroid dehydrogenase 2 indicate a role for the enzyme independent of its action on sex steroids. Endocrinology. 2007;148(8):3827–36. [DOI] [PubMed] [Google Scholar]
  • 47. Wang J, Zhao Y, Gu K, Yu P, Zhang B, Wang W, et al. The novel porcine gene early growth response 4 (Egr4) is differentially expressed in the ovaries of Erhualian and Pietrain pigs. Reproduction, fertility, and development. 2014;26(4):587–98. 10.1071/RD12380 [DOI] [PubMed] [Google Scholar]
  • 48. Hogarth CA, Mitchell D, Small C, Griswold M. EGR4 displays both a cell- and intracellular-specific localization pattern in the developing murine testis. Developmental dynamics: an official publication of the American Association of Anatomists. 2010;239(11):3106–14. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49. Tourtellotte WG, Nagarajan R, Bartke A, Milbrandt J. Functional compensation by Egr4 in Egr1-dependent luteinizing hormone regulation and Leydig cell steroidogenesis. Molecular and cellular biology. 2000;20(14):5261–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50. Tourtellotte WG, Nagarajan R, Auyeung A, Mueller C, Milbrandt J. Infertility associated with incomplete spermatogenic arrest and oligozoospermia in Egr4-deficient mice. Development. 1999;126(22):5061–71. [DOI] [PubMed] [Google Scholar]
  • 51. Nakazawa H, Sada T, Toriyama M, Tago K, Sugiura T, Fukuda M, et al. Rab33a mediates anterograde vesicular transport for membrane exocytosis and axon outgrowth. The Journal of neuroscience: the official journal of the Society for Neuroscience. 2012;32(37):12712–25. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52. Jacobsen M, Repsilber D, Gutschmidt A, Neher A, Feldmann K, Mollenkopf HJ, et al. Ras-associated small GTPase 33A, a novel T cell factor, is down-regulated in patients with tuberculosis. The Journal of infectious diseases. 2005;192(7):1211–8. [DOI] [PubMed] [Google Scholar]
  • 53. Zhou GL, Cao Y, Li M, Zhang LC, Yu YS, Jin HG. Meat quality and carcass traits in relation to HGD-BstXI and HGD-HaeIII PCR-RFLP polymorphism in Chinese red cattle. Meat Sci. 2010;85(2):270–3. 10.1016/j.meatsci.2010.01.011 [DOI] [PubMed] [Google Scholar]
  • 54. Zhou G, Dudgeon C, Li M, Cao Y, Zhang L, Jin H. Molecular cloning of the HGD gene and association of SNPs with meat quality traits in Chinese red cattle. Molecular biology reports. 2010;37(1):603–11. 10.1007/s11033-009-9860-4 [DOI] [PubMed] [Google Scholar]
  • 55. Fernandez-Rodriguez A, Munoz M, Fernandez A, Pena RN, Tomas A, Noguera JL, et al. Differential gene expression in ovaries of pregnant pigs with high and low prolificacy levels and identification of candidate genes for litter size. Biology of reproduction. 2011;84(2):299–307. 10.1095/biolreprod.110.085589 [DOI] [PubMed] [Google Scholar]
  • 56. Noguera JL, Rodriguez C, Varona L, Tomas A, Munoz G, Ramirez O, et al. A bi-dimensional genome scan for prolificacy traits in pigs shows the existence of multiple epistatic QTL. BMC genomics. 2009;10:636 10.1186/1471-2164-10-636 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57. Xia J, Yuan J, Xin L, Zhang Y, Kong S, Chen Y, et al. Transcriptome analysis on the inflammatory cell infiltration of nonalcoholic steatohepatitis in bama minipigs induced by a long-term high-fat, high-sucrose diet. PLOS one. 2014;9(11):e113724 10.1371/journal.pone.0113724 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58. Wang X, Zhou G, Xu X, Geng R, Zhou J, Yang Y, et al. Transcriptome profile analysis of adipose tissues from fat and short-tailed sheep. Gene. 2014;549(2):252–7. 10.1016/j.gene.2014.07.072 [DOI] [PubMed] [Google Scholar]
  • 59. Gan L, Xie L, Zuo F, Xiang Z, He N. Transcriptomic analysis of Rongchang pig brains and livers. Gene. 2015;560(1):96–106. 10.1016/j.gene.2015.01.051 [DOI] [PubMed] [Google Scholar]
  • 60. Anahi Franchi N, Avendano C, Molina RI, Tissera AD, Maldonado CA, Oehninger S, et al. beta-Microseminoprotein in human spermatozoa and its potential role in male fertility. Reproduction. 2008;136(2):157–66. 10.1530/REP-08-0032 [DOI] [PubMed] [Google Scholar]
  • 61. Sechman A, Antos P, Katarzynska D, Grzegorzewska A, Wojtysiak D, Hrabia A. Effects of 2,3,7,8-tetrachlorodibenzo-p-dioxin on secretion of steroids and STAR, HSD3B and CYP19A1 mRNA expression in chicken ovarian follicles. Toxicology letters. 2014;225(2):264–74. 10.1016/j.toxlet.2013.12.021 [DOI] [PubMed] [Google Scholar]
  • 62. Nakamoto M, Fukasawa M, Tanaka S, Shimamori K, Suzuki A, Matsuda M, et al. Expression of 3beta-hydroxysteroid dehydrogenase (hsd3b), star and ad4bp/sf-1 during gonadal development in medaka (Oryzias latipes). General and comparative endocrinology. 2012;176(2):222–30. 10.1016/j.ygcen.2012.01.019 [DOI] [PubMed] [Google Scholar]
  • 63. Tosser-Klopp G, Mulsant P, Yerle M. Regional localisations of VIM, HSD3b, ACTA1 and PGM1 in pigs. Animal genetics. 1998;29(1):23–6. [DOI] [PubMed] [Google Scholar]
  • 64. Sechman A, Pawlowska K, Hrabia A. Effect of 3,3',5-triiodothyronine and 3,5-diiodothyronine on progesterone production, cAMP synthesis, and mRNA expression of STAR, CYP11A1, and HSD3B genes in granulosa layer of chicken preovulatory follicles. Domestic animal endocrinology. 2011;41(3):137–49. 10.1016/j.domaniend.2011.05.007 [DOI] [PubMed] [Google Scholar]
  • 65. Moazeni M, Sharifiyazdi H, Izadpanah A. Characterization of Fasciola hepatica genotypes from cattle and sheep in Iran using cytochrome C oxidase gene (CO1). Parasitology research. 2012;110(6):2379–84. 10.1007/s00436-011-2774-9 [DOI] [PubMed] [Google Scholar]
  • 66. Tian Z, Liu G, Yin H, Luo J, Guan G, Xie J, et al. Cytochrome c oxidase subunit III (COX3) gene, an informative marker for phylogenetic analysis and differentiation of Babesia species in China. Infection, genetics and evolution: journal of molecular epidemiology and evolutionary genetics in infectious diseases. 2013;18:13–7. 10.1016/j.meegid.2013.04.002 [DOI] [PubMed] [Google Scholar]
  • 67. Zhao H, Li J, Li X, Han C, Zhang Y, Zheng L, et al. Silencing GPX3 Expression Promotes Tumor Metastasis in Human Thyroid Cancer. Current protein & peptide science. 2015;16(4):316–21. [DOI] [PubMed] [Google Scholar]
  • 68. Cao S, Yan B, Lu Y, Zhang G, Li J, Zhai W, et al. Methylation of promoter and expression silencing of GPX3 gene in hepatocellular carcinoma tissue. Clinics and research in hepatology and gastroenterology. 2015;39(2):198–204. 10.1016/j.clinre.2014.09.003 [DOI] [PubMed] [Google Scholar]

Associated Data

This section collects any data citations, data availability statements, or supplementary materials included in this article.

Supplementary Materials

S1 Checklist. Completed ARRIVE Guidelines Checklist for the reporting of animal data.

(PDF)

S1 Table. Primer pairs selected for RNA-Seq validation by qPCR.

(XLSX)

S2 Table. Detailed information of differentially expressed genes.

(XLS)

S3 Table. Significantly enriched GO terms among differentially expressed genes.

(XLS)

S4 Table. Significantly enriched KEGG pathways among differentially expressed genes.

(XLS)

S5 Table. Detailed information of 21 differentially expressed genes enriched in the ovarian steroidogenesis pathway.

(XLSX)

Data Availability Statement

The authors confirm that all data underlying the findings are fully available without restriction. Data are available from the Sequence Read Archive (SRA) under Accession no. SRP058401, Bioproject: PRJNA283575.


Articles from PLoS ONE are provided here courtesy of PLOS

RESOURCES