Next Article in Journal
Phylogenetic Relationships among TnpB-Containing Mobile Elements in Six Bacterial Species
Next Article in Special Issue
Genome-Wide Association Study on Reproductive Traits Using Imputation-Based Whole-Genome Sequence Data in Yorkshire Pigs
Previous Article in Journal
The Telomeric Repeats of HHV-6A Do Not Determine the Chromosome into Which the Virus Is Integrated
Previous Article in Special Issue
Novel Haplotype in the HHEX Gene Promoter Associated with Body Length in Pigs
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

Genome-Wide Association Analysis Identified Variants Associated with Body Measurement and Reproduction Traits in Shaziling Pigs

1
College of Animal Science and Technology, Hunan Agricultural University, Changsha 410128, China
2
Xiang Dong Experiment Station, Hunan Provincial Pig Industrial Technology System, Xiangtan 411100, China
3
Key Laboratory of Agro-Ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, Chinese Academy of Sciences; Hunan Provincial Engineering Research Center for Healthy Livestock and Poultry Production; Scientific Observing and Experimental Station of Animal Nutrition and Feed Science in South-Central, Ministry of Agriculture, Changsha 410125, China
4
Kunpeng Institute of Modern Agriculture at Foshan, Foshan 528226, China
*
Authors to whom correspondence should be addressed.
These authors contributed equally to this work.
Genes 2023, 14(2), 522; https://doi.org/10.3390/genes14020522
Submission received: 14 January 2023 / Revised: 9 February 2023 / Accepted: 16 February 2023 / Published: 18 February 2023
(This article belongs to the Special Issue Genetics and Genomics of Pig Breeding)

Abstract

:
With the increasing popularity of genomic sequencing, breeders pay more attention to identifying the crucial molecular markers and quantitative trait loci for improving the body size and reproduction traits that could affect the production efficiency of pig-breeding enterprises. Nevertheless, for the Shaziling pig, a well-known indigenous breed in China, the relationship between phenotypes and their corresponding genetic architecture remains largely unknown. Herein, in the Shaziling population, a total of 190 samples were genotyped using the Geneseek Porcine 50K SNP Chip, obtaining 41857 SNPs for further analysis. For phenotypes, two body measurement traits and four reproduction traits in the first parity from the 190 Shaziling sows were measured and recorded, respectively. Subsequently, a genome-wide association study (GWAS) between the SNPs and the six phenotypes was performed. The correlation between body size and reproduction phenotypes was not statistically significant. A total of 31 SNPs were found to be associated with body length (BL), chest circumference (CC), number of healthy births (NHB), and number of stillborns (NSB). Gene annotation for those candidate SNPs identified 18 functional genes, such as GLP1R, NFYA, NANOG, COX7A2, BMPR1B, FOXP1, SLC29A1, CNTNAP4, and KIT, which exert important roles in skeletal morphogenesis, chondrogenesis, obesity, and embryonic and fetal development. These findings are helpful to better understand the genetic mechanism for body size and reproduction phenotypes, while the phenotype-associated SNPs could be used as the molecular markers for the pig breeding programs.

1. Introduction

Domesticated pigs account for a high proportion of meat production in the world [1]. In the pig industry, reproductive performance is directly correlated with production efficiency and economic profits. The reproductive traits, especially litter traits, exhibited low heritability (less than 0.1) and were regulated by many genes and their interactions [2]. Traditional breeding programs for genetic improvement with low heritability, complex traits often progress slowly, which has undoubtedly elevated the need for molecular breeding [3]. Genomic selection (GS) and Molecular marker-assisted selection (MAS) have been demonstrated to have remarkable superiority in improving traits with low heritability [4]. Obtaining more accurate genetic information, such as quantitative trait loci (QTLs), single-nucleotide polymorphisms (SNPs), and candidate genes, related to reproductive traits will greatly increase the selection efficiency of GS and MAS.
From the perspective of breeders, phenotypic records for body measurement traits are vital for evaluating the production performance and making a future breeding schedule. The body measurement traits present low-to-moderate heritability [5,6,7,8]. The study revealed that several body measurement traits could be regarded as indicator traits for improving production traits [9]. Importantly, some recent studies pointed out the vital relationship between the body measurement traits and reproductive performance [10,11], hinting that the body conformation traits of female animals might be important for improving reproduction performance.
The last decade has witnessed the application of genome-wide association studies (GWAS) in identifying the genetic markers covering the whole genome for diverse body measurement and reproduction phenotypes. According to the latest data from PigQTLdb (https://www.animalgenome.org/cgi-bin/QTLdb/SS/index, accessed on 9 January 2023), there are 35,846 QTLs related with 693 different traits in pigs that have been mapped by 773 publications. Among the 1281 QTLs associated with litter traits, there were only 466 QTLs related to total numbers born (TNB), 374 QTLs related to number of healthy births (NHB), 138 QTLs related to numbers of stillborn (NSB), and 149 QTLs related to the number of mummified pigs (MUMM). Regarding the body measurement traits, 188 QTLs related to chest circumference (CC), and 219 QTLs related to body length (BL). The Shaziling pig is primarily raised in the Hunan Province of China and is characterized by low growth rate, good meat quality, and strong resistance to diseases [12]. Earlier genomics studies for Shaziling pigs were primarily focused on the genetic diversity [13], skeletal muscle growth [14], and umbilical hernia [15]. However, so far, identifying key molecular markers through the GWAS technique for improving production performance is still progressing slowly.
In this study, we aim to uncover the relationship between body measurement traits and reproduction phenotypes, and to clarify their associated genetic variations in Shaziling pigs. To this end, the body measurement traits (BL and CC) and reproductive traits (TNB, NHB, MUMM, and NSB) were measured and collected, respectively. Subsequently, based on the Geneseek Porcine 50K SNP Chip data, the SNPs were detected in those Shaziling pigs. The GWAS was conducted to identify potentially critical SNPs and QTL regions. This work paves the way for the future genetic improvement of pigs with efficient growth and reproduction performances.

2. Materials and Methods

2.1. Animals and Phenotypes

Our studied population (Shaziling pigs) were raised on the conservation farm (Xiangtan, Hunan, China). Animals were subjected to the same nutritional and management conditions. Firstly, the BL and CC of 190 sows (from 240 to 265 days) were measured according to the method applied in previous research [7]. Briefly, these two body size traits were measured by a tape or a meter ruler. Then, the same batch of 190 sows in the production stage was sampled and litter records were collected in the first parity. TNB, NHB, MUMM, and NSB were recorded for each parity. Finally, these six traits were used for the following analysis. The ethics committee of Hunan Agricultural University approved all the experimental procedures in this study (Permit Number: 20210701).

2.2. Animals and Genotypes

The ear tissues of 190 Shaziling pigs were collected for genomic DNA extraction according the protocol of Accurate SteadyPure Universal Genomic DNA Extraction Kit (Accurate Biology, Changsha, China). The concentration and quality of genomic DNA were testing by Nanodrop 2000 (Thermo Scientific, Waltham, MA, USA) and 1.5% agarose gel electrophoresis, respectively. The final qualified DNA samples were genotyped using Geneseek Porcine 50K SNP Chip (GeneSeek, Lincoln, NE, USA).

2.3. Data Quality Control

For phenotypes, the R (version 4.2.1) was used to perform descriptive statistical analyses to check the data quality including the mean, standard deviation, coefficient of variance, minimum and maximum values for each trait. The plot of normal distribution and correlation coefficients among phenotypic values were all visualized and calculated by the “GGally” package in R. For the genotypes, PLINK software (version 1.90 beta) was used to conduct quality control to achieve high-quality common SNPs [16]. Briefly, the filtered procedures were performed as follow: (1) individual call rate >90%; (2) minor allele frequency (MAF) >5%; and (3) p-value > 10−6 for the Hardy-Weinberg equilibrium (HWE) test. Furthermore, we excluded the SNPs located on the sex chromosome and unmatched regions.

2.4. SNP-Based Heritability

The heritability estimates of phenotypes were calculated using a restricted genomic-relatedness-based restricted maximum-likelihood (GREML) analysis, as implemented in GCTA software (version 1.94) [17]. Briefly, based on the high-quality SNPs, the genetic relationship matrix (GRM) was used to estimate the genetic relationships between pairwise individuals from all of the autosomal SNPs, and then the GRM and phenotypes were included in the restricted maximum likelihood (REML) analysis to estimate the variance explained by the SNPs.

2.5. Population Structure and Kinship Analyses

Principal component analysis (PCA) was performed to identify the population structure of Shaziling pigs to determine whether principal components (PCs) should be included in the GWAS model. Simply, the eigenvalues and eigenvectors were calculated by GCTA software based on high quality filtered SNPs. The lollipop-plot of the top ten principal components were visualized using the “ggplot2” package in R. A heat map was plotted against the kinship matrix to exhibit the level of relatedness among all individuals within the Shaziling population.

2.6. GWAS

The association analysis between each SNP marker and phenotypic data was performed using the mixed linear model in the GEMMA software [18]. The model described was as follows:
y = Wα + Xβ + μ+ e
where y represents the vector of phenotypes (BL, CC, TNB, NHB, NSB, and MUMM) for each individual; Wα included the population structure effect and fixed effects, briefly, age (in days) was used as fixed effects for BL and CC, and the individual of boar used for mating was used as fixed effects for TNB, NHB, NSB, and MUMM; Xβ is the marker effect to be tested; μ~N (0, Kϕ2) represents the polygenic effect; e~N (0, 2) refers to the residual effect, and K is the kinship matrix generated from the SNPs. Similar to previous GWAS research [19,20], the Bonferroni-adjusted genome-wide significant threshold was set as 0.05/N, and the suggestive threshold was set as 1/N, N being the total number of filtered SNPs.

2.7. Candidate Gene Search and Functional Annotation

To identify the candidate genes of each significant SNP, we searched for annotated genes according to their physical positions (within 500 Kb upstream or downstream) on the pig reference genome (Sscrofa 11.1) (http://asia.ensembl.org/biomart/martview/, accessed on 18 October 2022). Gene Ontology (GO) functional annotation analysis and Kyoto Encyclopedia of Genes and Genomes (KEGG) enrichment analysis were performed in the enrichment module of KOBAS 3.0 [21], and then the significant threshold for the GO terms (biological process) and KEGG pathways were set at Benjamini-Hochberg adjusted p-value < 0.05.

3. Results

3.1. Description of Phenotypes and Genotypes

As shown in Figure 1, phenotypes, including BL, CC, TNB, and NHB, of 190 Shaziling pigs displayed approximately normal distributions in the current study. TNB and NHB were strongly and positively correlated (r = 0.826, p < 0.001). BL and CC were weakly correlated (r = 0.294, p < 0.001). However, there was no correlation between body measurement traits (BL and CC) and reproduction traits (TNB and NHB). Descriptive statistics for the traits of BL, CC, TNB, NHB, MUMM, and NSB analyzed in the current study are shown in Table 1. In the present study, the heritability of BL and CC was 0.15 and 0.14, respectively. Except for MUMM, the other three reproduction traits were less than 0.01. The coefficient of variation (CV) ranges from 7.96% (lowest one: BL) to 9.03% (highest one: CC).
In the Shaziling population, a total of 190 samples were genotyped using the Geneseek Porcine 50K SNP Chip. A total of 41,857 SNPs were obtained for further marker analysis after a series of quality filter procedures. The distribution of the SNP information on eighteen autosome chromosomes was presented in Figure 2A. According to the SNP numbers for GWAS analysis, the genome-wide significant and suggestive threshold of the Shaziling population were 1.19 × 10−6 (0.05/41,857) and 2.38 × 10−5 (1/41,857), respectively.

3.2. Results of Population Structure and Kinship Analyses

The principal component analysis was performed for the tested Shaziling individuals to avoid the false-positive results caused by population stratification, which was considered as an important influencing factor to the reliability of the GWAS result. The results showed that the first four principal components could be included in the GWAS model as covariates (Figure 2B, Figure S1). Besides, a kinship matrix was also used as the covariate in the fixed effects model for GWAS analysis. The kinship heat map was shown in Supplementary Figure S2.

3.3. GWAS Results

The GWAS results are displayed in Figure 3 and Table 2. In total, 8 SNPs surpassed the genome-wide significance level and 23 SNPs reached the suggestive threshold. In detail, on chromosome 7, the GLP1R gene corresponding to the significant SNP (ALGA0040227) was significantly associated with BL. Moreover, at the suggestive level, another 4 SNPs located on chromosome 7 were also associated with BL, including ALGA0039856, ASGA0032589, ALGA0040238, and INRA0024788, which were respectively mapped to genes SNRPC, NFYA, GLP1R, and GLP1R. Regarding the CC trait, 4 SNPs reached the significant level. Among these 4 SNPs, H3GA0003059 and ALGA0004562 on chromosome 1 were mapped to genes NANOG and COX7A2, respectively. ASGA0050356 and ALGA0065112 located on chromosomes 11 and 12 were respectively mapped to genes FAM216B and CACNG1. Furthermore, 9 SNPs (ASGA0057447, ALGA0111294, MARC0074335, MARC0090402, ASGA0073620, ALGA0024545, DRGA0000770, WU_10.2_12_884330, and WU_10.2_12_890035) at the suggestive-level located on chromosome 13, 8, 12, 1, 16, 4, 1, 12, and 12, respectively, were also detected to be associated with trait CC, and the genes closest to the 9 SNPs were FOXP1, BMPR1B, DNAH9, ENSSSCG00000060802, CCNG1, COX6C, RIMS1, CCDC57, and CCDC57, respectively.
For reproduction traits, the SMG1 gene on chromosome 3 was found to be nearest to suggestive SNPs (CASI0010189) related to TNB. In addition, 3 SNPs (ASGA0038253, ALGA0108557, and ALGA0040524) in significant-level located on chromosome 8, 1, and 7, respectively, were detected to be related with trait NSB, and the genes nearest to the 3 SNPs were ARAP2, GTF2H5, and SLC29A1, respectively. Moreover, 9 SNPs (WU_10.2_4_11278211, CASI0009401, H3GA0024817, DIAS0000597, WU_10.2_18_23986398, DBMA0000241, H3GA0048552, WU_10.2_4_11747027, and WU_10.2_7_7925995) in suggestive-level located on chromosome 4, 6, 8, 3, 18, 7, 17, 4, and 7, respectively, were also detected to be related with the NSB trait, and the genes nearest to the 8 SNPs were FAM49B, CNTNAP4, KIT, POR, SPAM1, TFAP2B, BPIFA3, ENSSSCG00000060440, and NEDD9. However, no suggestive or significant SNPs were found to relate to MUMM and NHB (Supplementary Figure S3).

3.4. Enrichment Analysis

In order to obtain more insights into the functions of the 18 and 13 SNPs respectively related to the indicators of the body measurement traits (BL and CC) and reproductive phenotypes (NHB and NSB) of the Shaziling population, the corresponding candidate gene was used for enrichment analysis. The significant GO terms mainly included “GO:0008528: G protein-coupled peptide receptor activity”, “GO:0002063: chondrocyte development”, “GO:0006687: glycosphingolipid metabolic process”, “GO:0043588: skin development”, and “GO:0005246: calcium channel regulator activity” (Figure 4A). On the other hand, the KEGG pathways are mainly related to signaling pathways regulating the pluripotency of stem cells, Glycosaminoglycan degradation, and MAPK signaling pathway (Figure 4B). Other detailed annotated information can be found in Supplementary Table S1.

4. Discussion

To our knowledge, this is the first study evaluating the relationship between body measurement and reproductive phenotypes for female Shaziling pigs, and their associated genetic architecture by GWAS analysis.
Previous studies have indicated that body measurement traits could be the indicator of reproduction potential in sows [22,23,24] and cows [25]. However, in the current study, we did not observe significant effects of BL and CC on reproduction traits. Similarly, previous studies in Danish crossbred sows (Yorkshire × Landrace) and the Duroc population showed no statistically significant associations between BL and reproduction traits like litter size [26,27]. These results implied that body measurement traits have little influence on reproduction performance. However, it is necessary to enlarge the Shaziling population and types of body measurement traits in future correlation analyses to validate our findings. On the other hand, studies verified that the BL and CC exhibited low to moderate heritability [28,29]. We have presented here the same tendency for heritability in the two traits, suggesting that it is practicable to improve the body dimensions in Shaziling pigs through genetic selection. In addition, the genomic heritability of reproduction traits (TNB, NHB, NSB, and MUMM) were low. As a typical quantitative trait, litter traits are co-regulated by the interactions of many genes [30]. The low heritability of reproduction traits implies that accurately identifying more candidate genes related to reproduction traits would be helpful for breeders to improve reproduction performance.
Body measurement traits such as BL and CC are usually associated with growth traits and meat production. For example, it has been reported that BL and CC were positively associated with body weight [31]. BL is a vital factor affecting livestock slaughter performance, as a longer carcass signifies longer loins and more meat production [32]. In addition, the swine carcass length was positively correlated with the total lean meat content [33]. In the current work, a series of candidate genes corresponding to different SNPs for BL and CC were identified. As for BL, one of the significant candidate genes was GLP1R, which was the nearest gene for three different SNPs on chromosome 7. According to a previous study, GLP1R was expressed in primary osteoclasts, bone marrow cells, and osteoblasts [34]. In addition, the study revealed that mice lacking GLP1R displayed negative effects on bone strength and quality, the apparent symptoms including impaired mechanical properties, a significant decrease in the bone’s outer diameter and cortical thickness [35]. Our GO functional annotation analysis revealed that GLP1R is related to the G protein-coupled peptide receptor activity. Interestingly, the G-protein coupled receptor family has been reported to correspond with regulating limb patterning and skeletal morphogenesis, as well as morphogenesis during embryonic development [36,37]. This indicated that GLP1R might be the critical regulator of skeletal bone mass, so it was supposed to be a strong candidate gene for BL. Except for GLP1R, we found that the candidate gene NFYA was also associated with BL. It has been demonstrated that NFYA was an essential modulator in governing embryonic cartilage growth [38]. Moreover, the overexpression of two NFYA isoforms exerts different functions on myoblasts, with NFYAs increasing cell proliferation and NFYAl promoting differentiation [39]. We contend that NFYA might play important roles in modulating the formation of myoblasts and cartilage that ultimately affect the development of BL. Regarding the CC, the candidate genes were NANOG, COX7A2, CACNG1, FOXP1, BMPR1B, DNAH9, CCNG1, COX6C, RIMS1, and CCDC57. Among them, researchers found that NANOG-expressing human bone marrow mesenchymal stem cells (MSCs) had much higher capabilities for expansion and osteogenesis [40]. A GWAS analysis in humans has reported that COX7A2 and RIMS1 were identified as candidate genes for osteoarthritis [41,42], and COX7A2 was highly expressed in cartilage [43]. In another study, CACNG1 is thought to participate in modulating postmenopausal osteoporosis via Ca2+ regulation during muscle contraction [44]. Go analysis results showed that CACNG1 is involved in the process of calcium channel regulator activity. In recent years, the forkhead box protein (FOX) family has been reported to play important roles in different kinds of physiological bone processes [45]. For example, during the process of osteogenic differentiation, the expression of FOXP1 was enhanced under the regulation of circFOXP1 [46]. Additionally, a study also indicated that a decrease in FOXP1 level was accompanied by the augmented expression of PPARγ, which serves as a master regulator in adipocyte differentiation [47]. Over-expression of the FOXP1 in adipose cells affects adaptive thermogenesis and boosts diet-induced obesity [48]. GO results showed that FOXP1 is involved in the process of osteoclast development. It has been established that fat is the primary factor impairing body size. FOXP1 might play essential roles in lipid metabolism and osteogenic differentiation, so the FOXP1 gene could be considered as a strong candidate gene for the CC. In sheep, BMPR1B was detected as a candidate gene for variation in mature size [49]. In addition, BMPR1B participates in skeletal patterning, and is mainly found in differentiated chondrocytes and osteoblasts, and is also expressed in mesenchymal pre-cartilage condensations [50,51]. COX6C was identified as a critical gene involved in regulating osteoblast mineralization and mitochondrial bioenergetics [52].
TNB and NSB are key indicators for measuring sow reproduction performance. In TNB, gene annotation results showed that SMG1 was the closest gene for SNPs CASI0010189 on chromosome 3. It has been reported that SMG1 is a necessary kinase for mouse embryogenesis. The absence of SMG1 would be fatal to embryonic development, mainly due to the profound developmental defects (e.g., brain and heart) [53]. In daily production, high proportions of NSB cause serious economic losses. We here found several candidate genes, including ARAP2, GTF2H5, SLC29A1, FAM49B, CNTNAP4, KIT, POR, SPAM1, TFAP2B, BPIFA3, and NEDD9, that were potentially associated with NSB. For ARAP2, a GWAS analysis in Chinese Holstein cows found that it was associated with loin strength (LS), a reproduction-associated body-shape trait [25]. Cows with a weak loin usually possess a sinking uterus, which could easily lead to reproductive system diseases [54]. Whole exome sequencing for a male neonate with premature rupture of membranes and intrauterine growth restriction showed that GTF2H5 gene mutations induced severe clinical manifestations including multiple-organ failure [55]. SLC29A1, also called ENT1, belongs to the solute carrier (SLC) family. It was initially found from a human placental DNA library [56]. According to previous studies, the change in SLC drug transporter expression impacts the drug’s disposition and pharmacokinetics in the placenta, and a variety of substrates including toxins, nutrients, and signaling molecules are transported by SLC transporters during pregnancy [57,58]. Although previous research has not provided evidence to support a causal association between them, SLC29A1 might be a potential causal biomolecule for NSB. CNTNAP4 is a member of the neurexin superfamily and is essential for synaptic function and neural development [59]. In addition, a study showed that CNTNAP4 is necessary for the proliferation of embryonic neural progenitor cells (NPCs) in mice [60]. Notably, a GWAS study in beef cattle found that the CNTNAP4 gene was associated with fertility traits [61]. In the KIT mutation murine model, a reduction in placental vascularization was present, which finally resulted in the placenta developing irregularly and embryos presenting with severe growth retardation and dying in the uterus [62,63]. Moreover, GO analysis found that the KIT gene participates in the glycosphingolipid metabolic process. Intriguingly, an earlier study indicated that glycosphingolipid composition is important for the growth and differentiation of trophoblast cells during pregnancy [64]. These results suggested that KIT might be the important regulator for healthy embryonic development. It should be regarded as a strong candidate gene for NSB.
Recently, a study of prenatal and postnatal deaths for three sibling fetuses revealed that the combination of CNV and SNV in the mutated POR gene was responsible for their death [65]. SPAM1, a hyaluronidase, played roles in binding to sperm during capacitation, elevating cumulus dispersal efficiency and the capacity of sperm to across the cumulus of oocytes [66,67]. KEGG results showed that SPAM1 is involved in glycosaminoglycan degradation pathways. Of note, in the developmental process of embryos and fetuses, glycosaminoglycans play essential roles in cell morphogenesis, growth, differentiation, and cell migration [68,69]. SPAM1 may affect the expression pattern of the glycosaminoglycan degradation pathway, which ultimately affects embryonic development. TFAP2B is a member of the transcription factor TFAP2 family, also called AP-2. Earlier researchers found that TFAP2A-deficient mice suffered from anomalies, such as anencephaly, congenital heart disease, and body-wall defects [70,71,72]. According to earlier reports, TFAP2B-deletion led to neonatal mice death due to the massively increased apoptotic cell death for renal epithelial cells [73,74]. A subsequent study demonstrated that TFAP2B−/− mice exhibited developmental defects in the ductus arteriosus and limbs [75]. Thus, TFAP2B could be regarded as a strong candidate gene for NSB, because of its central role in teratogenicity. NEDD9, a target gene of TGF-beta, was identified in Yorkshire sows as a candidate core gene for litter size [76]. Besides, NEDD9 regulates the invasion and proliferation of ectopic endometriotic stromal cells [77].
The duration of pigs’ farrowing and estrous cycles are considered as critical factors of reproduction traits. A short duration of farrowing is vital for piglet survival as a delay can elevate the number of stillborn [78]. Another research indicated that the estrous cycles might affect litter size by influencing the ovulation rate [79]. Besides, the reproduction records in multiparous sows are also important phenotypes for the GWAS study. However, owing to the effects of the African Swine fever virus, during that period, many sows’ mating was suspended in the Shaziling pig farm, so that limited information for reproduction traits were obtained from that Shaziling pig farm, and only the first-parity records were included in this GWAS study. In the future, these reproduction traits-related factors shall be included in the model and the results shall be verified in a larger Shaziling population. In addition, metabolism and metabolic biomarkers have been previously implicated in studies of sows’ fertility, and the metabolite biomarkers could be the important phenotype in terms of sows’ reproduction potential [80]. Moreover, a recent report in our group for comparisons of carcass traits, meat quality, and serum metabolome between Shaziling and Yorkshire pigs has suggested that a higher serum L-carnitine content is a promising indicator for better meat quality of pigs [12]. Therefore, serum metabolite should also be regarded as a key trait for GWAS research in future study.
Chip-based SNP markers have been widely used in genomic studies because of their abundance in the genome and their low cost [81]. In this study, we applied the Porcine SNP 50k Chip for genotyping samples. In the early phase of the project for Shaziling pigs, the main object of genotyping samples was to identify the pureblood individuals and pedigree relationships for the Shaziling population. Afterwards, based on the previous results, we further measured the body size and reproduction traits for the pure Shaziling pigs. Then, this first GWAS study for the Shaziling population was carried out. With the development of sequencing technology and an analysis method, genotyping-by sequencing (GBS) is also used for identifying variants nowadays because of its lower cost per data point (especially for low-pass sequencing data) and the avoidance of ascertainment bias during genotyping [81]. Recent studies have found that GWAS using low-pass sequencing data showed similar results to those with SNP chip data, but may require much larger sample sizes to show measurable advantages [82]. Compared with other genotyping methods like GBS, although the SNP chip data in this study may provide limited information throughout the whole genome, it provided preliminary and useful information for the future selection and breeding of Shaziling pigs. Meanwhile, we shall acknowledge that a platform such as GBS should also be used for the detection of candidate functional SNP markers in Shaziling pigs. In this study, we detected some genetic markers that may affect the body measurement and reproduction traits of the Shaziling population. Information on the genomic regions explored in this current work can accelerate the identification of candidate genes for measurement and reproduction phenotypes. However, we did not identify any significant SNPs for NHB and MUMM. This may be partly due to the inadequate sample size used in the current study. Eventually, these outcomes provide meaningful information for genomic selection in the Shaziling pigs for high-efficiency genetic improvement.

5. Conclusions

By the genome-wide association study in the Shaziling pig breed, we identified 31 SNPs in total that were potentially associated with body size and reproductive traits of interest. Eighteen functional genes, including GLP1R, NFYA, NANOG, COX7A2, CACNG1, FOXP1, BMPR1B, COX6C. SMG1, ARAP2, GTF2H5, SLC29A1, CNTNAP4, KIT, POR, SPAM1, TFAP2B, and NEDD9, were identified as important candidate genes that may regulate the underlying genetic architecture of porcine body size and reproductive traits. Although the current study is limited by its small sample size, it helps us to understand the genetic basis of porcine body measurement and reproduction traits and could be potentially applied in pig breeding programs.

Supplementary Materials

The following supporting information can be downloaded at: https://www.mdpi.com/article/10.3390/genes14020522/s1, Figure S1: The PCA chart of Shaziling pigs; Figure S2: The kinship relationships of Shaziling pigs; Figure S3: Manhattan and quantile-quantile (QQ) plots of the observed −log10(p-values) for MUMM and NHB in Shaziling pigs. The horizontal red and blue dashed lines in the Manhattan plots indicate the suggestive level (2.38 × 10−5) and significant level (1.19 × 10−6), respectively. The QQ plots show the observed −log10-transformed p-values (y-axis) and the expected −log10-transformed p-values (x-axis); Table S1: The detailed information for GO terms and KEGG pathways.

Author Contributions

Conceptualization, M.L. and J.H.; formal analysis, Q.L. and S.Q.; data curation, Q.D., Y.Z., Z.L. and S.Y.; investigation, H.T. and Y.L.; resources, Y.Y. and M.W.; funding acquisition, Q.D. and J.H.; writing-original draft: Q.L. and Q.D.; writing-review and editing: M.L. All authors have read and agreed to the published version of the manuscript.

Funding

This research was funded by Natural Science Foundation of Hunan Province (2022JJ40171), the National Natural Science Foundation of China (32002166), the Special Fund for the Construction of Innovative Provinces in Hunan (2021NK1009), the Funding of Major Scientific Research Tasks, Kunpeng Institute of Modern Agriculture at Foshan (KIMA-ZDKY2022001).

Institutional Review Board Statement

The animals and experimental protocols used in this study follow the guidelines of the Ministry of Agriculture of China and the Committee of Animal Care at Hunan Agricultural University (Changsha, China) (Permit Number: CACAHU 20210701).

Informed Consent Statement

Not applicable.

Data Availability Statement

The data presented in this study are available on request from the corresponding author.

Acknowledgments

We are grateful to Bidong Li and the related employees in the NeX Eco-Agriculture Technologies, Inc., who help on feeding the experimental Shaziling pigs’ population, collecting samples and measuring the phenotypes.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Lunney, J.K. Advances in swine biomedical model genomics. Int. J. Biol. Sci. 2007, 3, 179–184. [Google Scholar] [CrossRef] [Green Version]
  2. Ma, X.; Li, P.H.; Zhu, M.X.; He, L.C.; Sui, S.P.; Gao, S.; Su, G.S.; Ding, N.S.; Huang, Y.; Lu, Z.Q.; et al. Genome-wide association analysis reveals genomic regions on Chromosome 13 affecting litter size and candidate genes for uterine horn length in Erhualian pigs. Animal 2018, 12, 2453–2461. [Google Scholar] [CrossRef]
  3. Chen, P.; Baas, T.J.; Mabry, J.W.; Koehler, K.J.; Dekkers, J.C. Genetic parameters and trends for litter traits in U.S. Yorkshire, Duroc, Hampshire, and Landrace pigs. J. Anim. Sci. 2003, 81, 46–53. [Google Scholar] [CrossRef] [PubMed]
  4. Zhang, Z.; Ober, U.; Erbe, M.; Zhang, H.; Gao, N.; He, J.; Li, J.; Simianer, H. Improving the accuracy of whole genome prediction for complex traits using the results of genome wide association studies. PLoS ONE 2014, 9, e93017. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  5. Fukawa, K.; Sugiyama, T.; Kusuhara, S.; Kudoh, O.; Kameyama, K. Model selection and genetic parameter estimation for performance traits, body measurement traits and leg score traits in a closed population of Duroc pigs. Nihon Chikusan Gakkaiho 2001, 72, 97–106. [Google Scholar] [CrossRef]
  6. Ishida, T.; Kuroki, T.; Harada, H.; Fukuhara, R. Estimation of additive and dominance genetic variances in line breeding swine. Asian Australas. J. Anim. Sci. 2001, 14, 1–6. [Google Scholar] [CrossRef]
  7. Ogawa, S.; Yazaki, N.; Ohnishi, C.; Ishii, K.; Uemoto, Y.; Satoh, M. Maternal effect on body measurement and meat production traits in purebred Duroc pigs. J. Anim. Breed. Genet. 2021, 138, 237–245. [Google Scholar] [CrossRef]
  8. Ohnishi, C.; Satoh, M. Estimation of genetic parameters for performance and body measurement traits in Duroc pigs selected for average daily gain, loin muscle area, and backfat thickness. Livest. Sci. 2018, 214, 161–166. [Google Scholar] [CrossRef]
  9. Yazaki, N.; Ogawa, S.I.; Onishi, C.; Ishii, K.; Uemoto, Y.; Sato, M. Effectiveness of body measurement traits for improving production traits in Duroc pigs. Nihon Chikusan Gakkaiho 2020, 91, 9–16. [Google Scholar] [CrossRef]
  10. Tuomi, J. Mammalian reproductive strategies: A generalized relation of litter size to body size. Oecologia 1980, 45, 39–44. [Google Scholar] [CrossRef]
  11. Haldar, A.; Pal, P.; Datta, M.; Paul, R.; Pal, S.K.; Majumdar, D.; Biswas, C.K.; Pan, S. Prolificacy and Its Relationship with Age, Body Weight, Parity, Previous Litter Size and Body Linear Type Traits in Meat-type Goats. Asian Australas. J. Anim. Sci. 2014, 27, 628–634. [Google Scholar] [CrossRef]
  12. Song, B.; Zheng, C.; Zheng, J.; Zhang, S.; Zhong, Y.; Guo, Q.; Li, F.; Long, C.; Xu, K.; Duan, Y.; et al. Comparisons of carcass traits, meat quality, and serum metabolome between Shaziling and Yorkshire pigs. Anim. Nutr. 2022, 8, 125–134. [Google Scholar] [CrossRef]
  13. Ai, H.; Huang, L.; Ren, J. Genetic diversity, linkage disequilibrium and selection signatures in chinese and Western pigs revealed by genome-wide SNP markers. PLoS ONE 2013, 8, e56001. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  14. Yang, H.; Xu, X.L.; Ma, H.M.; Jiang, J. Integrative analysis of transcriptomics and proteomics of skeletal muscles of the Chinese indigenous Shaziling pig compared with the Yorkshire breed. BMC Genet. 2016, 17, 80. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  15. Li, X.; Xu, P.; Zhang, C.; Sun, C.; Li, X.; Han, X.; Li, M.; Qiao, R. Genome-wide association study identifies variants in the CAPN9 gene associated with umbilical hernia in pigs. Anim. Genet. 2019, 50, 162–165. [Google Scholar] [CrossRef] [PubMed]
  16. Purcell, S.; Neale, B.; Todd-Brown, K.; Thomas, L.; Ferreira, M.A.; Bender, D.; Maller, J.; Sklar, P.; de Bakker, P.I.; Daly, M.J.; et al. PLINK: A tool set for whole-genome association and population-based linkage analyses. Am. J. Hum. Genet. 2007, 81, 559–575. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  17. Gray, A.; Stewart, I.; Tenesa, A. Advanced complex trait analysis. Bioinformatics 2012, 28, 3134–3136. [Google Scholar] [CrossRef]
  18. Zhou, X.; Stephens, M. Genome-wide efficient mixed-model analysis for association studies. Nat. Genet. 2012, 44, 821–824. [Google Scholar] [CrossRef] [Green Version]
  19. Xue, Y.; Liu, S.; Li, W.; Mao, R.; Zhuo, Y.; Xing, W.; Liu, J.; Wang, C.; Zhou, L.; Lei, M.; et al. Genome-Wide Association Study Reveals Additive and Non-Additive Effects on Growth Traits in Duroc Pigs. Genes 2022, 13, 1454. [Google Scholar] [CrossRef]
  20. Shi, L.; Wang, L.; Fang, L.; Li, M.; Tian, J.; Wang, L.; Zhao, F. Integrating genome-wide association studies and population genomics analysis reveals the genetic architecture of growth and backfat traits in pigs. Front. Genet. 2022, 13, 1078696. [Google Scholar] [CrossRef]
  21. Bu, D.; Luo, H.; Huo, P.; Wang, Z.; Zhang, S.; He, Z.; Wu, Y.; Zhao, L.; Liu, J.; Guo, J.; et al. KOBAS-i: Intelligent prioritization and exploratory visualization of biological functions for gene enrichment analysis. Nucleic Acids Res. 2021, 49, W317–W325. [Google Scholar] [CrossRef]
  22. Thiengpimol, P.; Koonawootrittriron, S.; Suwanasopee, T. Genetic and phenotypic correlations between backfat thickness and weight at 28 weeks of age, and reproductive performance in primiparous Landrace sows raised under tropical conditions. Trop. Anim. Health Prod. 2022, 54, 43. [Google Scholar] [CrossRef] [PubMed]
  23. Bohlouli, M.; Brandt, H.; Konig, S. Genetic parameters for linear conformation, stayability, performance and reproduction traits in German local Swabian-Hall landrace sows. J. Anim. Breed. Genet. 2023, 140, 144–157. [Google Scholar] [CrossRef]
  24. Nikkila, M.T.; Stalder, K.J.; Mote, B.E.; Rothschild, M.F.; Gunsett, F.C.; Johnson, A.K.; Karriker, L.A.; Boggess, M.V.; Serenius, T.V. Genetic associations for gilt growth, compositional, and structural soundness traits with sow longevity and lifetime reproductive performance. J. Anim. Sci. 2013, 91, 1570–1579. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  25. Lu, X.; Abdalla, I.M.; Nazar, M.; Fan, Y.; Zhang, Z.; Wu, X.; Xu, T.; Yang, Z. Genome-Wide Association Study on Reproduction-Related Body-Shape Traits of Chinese Holstein Cows. Animals 2021, 11, 1927. [Google Scholar] [CrossRef]
  26. Nielsen, S.; Kristensen, A.; Moustsen, V.A. Litter size of Danish crossbred sows increased without changes in sow body di mensions over a thirteen year period. Livest. Sci. 2018, 209, 73–76. [Google Scholar] [CrossRef]
  27. Ogawa, S.; Ohnishi, C.; Ishii, K.; Uemoto, Y.; Satoh, M. Genetic relationship between litter size traits at birth and body measurement and production traits in purebred Duroc pigs. Anim. Sci. J. 2020, 91, e13497. [Google Scholar] [CrossRef]
  28. Zhou, S.; Ding, R.; Zhuang, Z.; Zeng, H.; Wen, S.; Ruan, D.; Wu, J.; Qiu, Y.; Zheng, E.; Cai, G.; et al. Genome-Wide Association Analysis Reveals Genetic Loci and Candidate Genes for Chest, Abdominal, and Waist Circumferences in Two Duroc Pig Populations. Front. Vet. Sci. 2021, 8, 807003. [Google Scholar] [CrossRef]
  29. Li, C.; Duan, D.; Xue, Y.; Han, X.; Wang, K.; Qiao, R.; Li, X.L.; Li, X.J. An association study on imputed whole-genome resequencing from high-throughput sequencing data for body traits in crossbred pigs. Anim. Genet. 2022, 53, 212–219. [Google Scholar] [CrossRef]
  30. Manolio, T.A.; Collins, F.S.; Cox, N.J.; Goldstein, D.B.; Hindorff, L.A.; Hunter, D.J.; McCarthy, M.I.; Ramos, E.M.; Cardon, L.R.; Chakravarti, A.; et al. Finding the missing heritability of complex diseases. Nature 2009, 461, 747–753. [Google Scholar] [CrossRef] [Green Version]
  31. Jiang, N.; Liu, C.; Lan, T.; Zhang, Q.; Cao, Y.; Pu, G.; Niu, P.; Zhang, Z.; Li, Q.; Zhou, J.; et al. Polymorphism of VRTN Gene g.20311_20312ins291 Was Associated with the Number of Ribs, Carcass Diagonal Length and Cannon Bone Circumference in Suhuai Pigs. Animals 2020, 10, 484. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  32. Liu, Z.; Gao, Q.; Wang, T.; Chai, W.; Zhan, Y.; Akhtar, F.; Zhang, Z.; Li, Y.; Shi, X.; Wang, C. Multi-Thoracolumbar Variations and NR6A1 Gene Polymorphisms Potentially Associated with Body Size and Carcass Traits of Dezhou Donkey. Animals 2022, 12, 1349. [Google Scholar] [CrossRef] [PubMed]
  33. Stewart, T.; Schinckel, A. Genetic Parameters for Swine Growth and Carcass Traits; FAO: Rome, Italy, 1989. [Google Scholar]
  34. Pereira, M.; Jeyabalan, J.; Jorgensen, C.S.; Hopkinson, M.; Al-Jazzar, A.; Roux, J.P.; Chavassieux, P.; Orriss, I.R.; Cleasby, M.E.; Chenu, C. Chronic administration of Glucagon-like peptide-1 receptor agonists improves trabecular bone mass and architecture in ovariectomised mice. Bone 2015, 81, 459–467. [Google Scholar] [CrossRef] [Green Version]
  35. Mabilleau, G.; Mieczkowska, A.; Irwin, N.; Flatt, P.R.; Chappard, D. Optimal bone mechanical and material properties require a functional glucagon-like peptide-1 receptor. J. Endocrinol. 2013, 219, 59–68. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  36. Philipp, M.; Berger, I.M.; Just, S.; Caron, M.G. Overlapping and opposing functions of G protein-coupled receptor kinase 2 (GRK2) and GRK5 during heart development. J. Biol. Chem. 2014, 289, 26119–26130. [Google Scholar] [CrossRef] [Green Version]
  37. Hwang, S.H.; White, K.A.; Somatilaka, B.N.; Shelton, J.M.; Richardson, J.A.; Mukhopadhyay, S. The G protein-coupled receptor Gpr161 regulates forelimb formation, limb patterning and skeletal morphogenesis in a primary cilium-dependent manner. Development 2018, 145, dev154054. [Google Scholar] [CrossRef] [Green Version]
  38. Chen, Y.H.; Lin, Y.T.; Lee, G.H. Novel and unexpected functions of zebrafish CCAAT box binding transcription factor (NF-Y) B subunit during cartilages development. Bone 2009, 44, 777–784. [Google Scholar] [CrossRef]
  39. Basile, V.; Baruffaldi, F.; Dolfini, D.; Belluti, S.; Benatti, P.; Ricci, L.; Artusi, V.; Tagliafico, E.; Mantovani, R.; Molinari, S.; et al. NF-YA splice variants have different roles on muscle differentiation. Biochim. Biophys. Acta 2016, 1859, 627–638. [Google Scholar] [CrossRef] [Green Version]
  40. Go, M.J.; Takenaka, C.; Ohgushi, H. Forced expression of Sox2 or Nanog in human bone marrow derived mesenchymal stem cells maintains their expansion and differentiation capabilities. Exp. Cell Res. 2008, 314, 1147–1154. [Google Scholar] [CrossRef]
  41. arcOGEN Consortium; arcOGEN Collaborators. Identification of new susceptibility loci for osteoarthritis (arcOGEN): A genome-wide association study. Lancet 2012, 380, 815–823. [Google Scholar] [CrossRef] [Green Version]
  42. Zhu, M.; Yin, P.; Hu, F.; Jiang, J.; Yin, L.; Li, Y.; Wang, S. Integrating genome-wide association and transcriptome prediction model identifies novel target genes for osteoporosis. Osteoporos. Int. 2021, 32, 2493–2503. [Google Scholar] [CrossRef] [PubMed]
  43. Johnson, K.; Reynard, L.N.; Loughlin, J. Functional characterisation of the osteoarthritis susceptibility locus at chromosome 6q14.1 marked by the polymorphism rs9350591. BMC Med. Genet. 2015, 16, 81. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  44. Chen, L.R.; Ko, N.Y.; Chen, K.H. Medical Treatment for Osteoporosis: From Molecular to Clinical Opinions. Int. J. Mol. Sci. 2019, 20, 2213. [Google Scholar] [CrossRef] [Green Version]
  45. Ma, X.; Su, P.; Yin, C.; Lin, X.; Wang, X.; Gao, Y.; Patil, S.; War, A.R.; Qadir, A.; Tian, Y.; et al. The Roles of FoxO Transcription Factors in Regulation of Bone Cells Function. Int. J. Mol. Sci. 2020, 21, 692. [Google Scholar] [CrossRef] [Green Version]
  46. Shen, W.; Sun, B.; Zhou, C.; Ming, W.; Zhang, S.; Wu, X. CircFOXP1/FOXP1 promotes osteogenic differentiation in adipose-derived mesenchymal stem cells and bone regeneration in osteoporosis via miR-33a-5p. J. Cell. Mol. Med. 2020, 24, 12513–12524. [Google Scholar] [CrossRef] [PubMed]
  47. Li, H.; Liu, P.; Xu, S.; Li, Y.; Dekker, J.D.; Li, B.; Fan, Y.; Zhang, Z.; Hong, Y.; Yang, G.; et al. FOXP1 controls mesenchymal stem cell commitment and senescence during skeletal aging. J. Clin. Investig. 2017, 127, 1241–1253. [Google Scholar] [CrossRef] [Green Version]
  48. Liu, P.; Huang, S.; Ling, S.; Xu, S.; Wang, F.; Zhang, W.; Zhou, R.; He, L.; Xia, X.; Yao, Z.; et al. Foxp1 controls brown/beige adipocyte differentiation and thermogenesis through regulating beta3-AR desensitization. Nat. Commun. 2019, 10, 5070. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  49. Bolormaa, S.; Hayes, B.J.; van der Werf, J.H.; Pethick, D.; Goddard, M.E.; Daetwyler, H.D. Detailed phenotyping identifies genes with pleiotropic effects on body composition. BMC Genomics 2016, 17, 224. [Google Scholar] [CrossRef] [Green Version]
  50. Kausar, T.; Nayeem, S.M. Correlating interfacial water dynamics with protein-protein interaction in complex of GDF-5 and BMPRI receptors. Biophys. Chem. 2018, 240, 50–62. [Google Scholar] [CrossRef]
  51. Lin, S.; Svoboda, K.K.; Feng, J.Q.; Jiang, X. The biological function of type I receptors of bone morphogenetic protein in bone. Bone Res. 2016, 4, 16005. [Google Scholar] [CrossRef] [Green Version]
  52. Lin, P.I.; Tai, Y.T.; Chan, W.P.; Lin, Y.L.; Liao, M.H.; Chen, R.M. Estrogen/ERalpha signaling axis participates in osteoblast maturation via upregulating chromosomal and mitochondrial complex gene expressions. Oncotarget 2018, 9, 1169–1186. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  53. McIlwain, D.R.; Pan, Q.; Reilly, P.T.; Elia, A.J.; McCracken, S.; Wakeham, A.C.; Itie-Youten, A.; Blencowe, B.J.; Mak, T.W. Smg1 is required for embryogenesis and regulates diverse genes via alternative splicing coupled to nonsense-mediated mRNA decay. Proc. Natl. Acad. Sci. USA 2010, 107, 12186–12191. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  54. Ali, T.E.; Burnside, E.B.; Schaeffer, L.R. Relationship between external body measurements and calving difficulties in Canadian Holstein-Friesian cattle. J. Dairy Sci. 1984, 67, 3034–3044. [Google Scholar] [CrossRef]
  55. Michalska, E.; Koppolu, A.; Dobrzanska, A.; Płoski, R.; Gruszfeld, D. A case of severe trichothiodystrophy 3 in a neonate due to mutation in the GTF2H5 gene: Clinical report. Eur. J. Med. Genet. 2019, 62, 103557. [Google Scholar] [CrossRef]
  56. Griffiths, M.; Beaumont, N.; Yao, S.Y.; Sundaram, M.; Boumah, C.E.; Davies, A.; Kwong, F.Y.; Coe, I.; Cass, C.E.; Young, J.D.; et al. Cloning of a human nucleoside transporter implicated in the cellular uptake of adenosine and chemotherapeutic drugs. Nat. Med. 1997, 3, 89–93. [Google Scholar] [CrossRef] [PubMed]
  57. Pfeifer, E.; Parrott, J.; Lee, G.T.; Domalakes, E.; Zhou, H.; He, L.; Mason, C.W. Regulation of human placental drug transporters in HCV infection and their influence on direct acting antiviral medications. Placenta 2018, 69, 32–39. [Google Scholar] [CrossRef]
  58. Liu, X.; Pan, G. Drug Transporters in Drug Disposition, Effects and Toxicity; Springer: Berlin/Heidelberg, Germany, 2019. [Google Scholar]
  59. Zhang, W.; Zhou, M.; Lu, W.; Gong, J.; Gao, F.; Li, Y.; Xu, X.; Lin, Y.; Zhang, X.; Ding, L.; et al. CNTNAP4 deficiency in dopaminergic neurons initiates parkinsonian phenotypes. Theranostics 2020, 10, 3000–3021. [Google Scholar] [CrossRef]
  60. Yin, F.T.; Futagawa, T.; Li, D.; Ma, Y.X.; Lu, M.H.; Lu, L.; Li, S.; Chen, Y.; Cao, Y.J.; Yang, Z.Z.; et al. Caspr4 interaction with LNX2 modulates the proliferation and neuronal differentiation of mouse neural progenitor cells. Stem Cells Dev. 2015, 24, 640–652. [Google Scholar] [CrossRef] [Green Version]
  61. Forutan, M.; Engle, B.; Goddard, M.; Hayes, B. A Conditional Multi-Trait Sequence GWAS of Heifer Fertility in Tropically Adapted Beef Cattle. Available online: https://www.wageningenacademic.com/pb-assets/wagen/WCGALP2022/54_005.pdf (accessed on 7 December 2022).
  62. Kaiser, F.; Hartweg, J.; Jansky, S.; Pelusi, N.; Kubaczka, C.; Sharma, N.; Nitsche, D.; Langkabel, J.; Schorle, H. Persistent Human KIT Receptor Signaling Disposes Murine Placenta to Premature Differentiation Resulting in Severely Disrupted Placental Structure and Functionality. Int. J. Mol. Sci. 2020, 21, 5503. [Google Scholar] [CrossRef]
  63. Nardi, E.; Seravalli, V.; Serena, C.; Mecacci, F.; Massi, D.; Bertaccini, B.; Di Tommaso, M.; Castiglione, F. A study on the placenta in stillbirth: An evaluation of molecular alterations through next generation sequencing. Placenta 2022, 129, 7–11. [Google Scholar] [CrossRef]
  64. Itonori, S.; Shirai, T.; Kiso, Y.; Ohashi, Y.; Shiota, K.; Ogawa, T. Glycosphingolipid composition of rat placenta: Changes associated with stage of pregnancy. Biochem. J. 1995, 307 Pt 2, 399–405. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  65. Tzetis, M.; Konstantinidou, A.; Sofocleous, C.; Kosma, K.; Mitrakos, A.; Tzannatos, C.; Kitsiou-Tzeli, S. Compound heterozygosity of a paternal submicroscopic deletion and a maternal missense mutation in POR gene: Antley-bixler syndrome phenotype in three sibling fetuses. Birth Defects Res. A Clin. Mol. Teratol. 2016, 106, 536–541. [Google Scholar] [CrossRef] [PubMed]
  66. Griffiths, G.S.; Miller, K.A.; Galileo, D.S.; Martin-DeLeon, P.A. Murine SPAM1 is secreted by the estrous uterus and oviduct in a form that can bind to sperm during capacitation: Acquisition enhances hyaluronic acid-binding ability and cumulus dispersal efficiency. Reproduction 2008, 135, 293–301. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  67. Martin-DeLeon, P.A. Epididymal SPAM1 and its impact on sperm function. Mol. Cell. Endocrinol. 2006, 250, 114–121. [Google Scholar] [CrossRef] [PubMed]
  68. Yamada, S.; Sugahara, K.; Ozbek, S. Evolution of glycosaminoglycans: Comparative biochemical study. Commun. Integr. Biol. 2011, 4, 150–158. [Google Scholar] [CrossRef]
  69. Sugahara, K.; Kitagawa, H. Recent advances in the study of the biosynthesis and functions of sulfated glycosaminoglycans. Curr. Opin. Struct. Biol. 2000, 10, 518–527. [Google Scholar] [CrossRef]
  70. Schorle, H.; Meier, P.; Buchert, M.; Jaenisch, R.; Mitchell, P.J. Transcription factor AP-2 essential for cranial closure and craniofacial development. Nature 1996, 381, 235–238. [Google Scholar] [CrossRef]
  71. Zhang, J.; Hagopian-Donaldson, S.; Serbedzija, G.; Elsemore, J.; Plehn-Dujowich, D.; McMahon, A.P.; Flavell, R.A.; Williams, T. Neural tube, skeletal and body wall defects in mice lacking transcription factor AP-2. Nature 1996, 381, 238–241. [Google Scholar] [CrossRef]
  72. Brewer, S.; Jiang, X.; Donaldson, S.; Williams, T.; Sucov, H.M. Requirement for AP-2alpha in cardiac outflow tract morphogenesis. Mech. Dev. 2002, 110, 139–149. [Google Scholar] [CrossRef]
  73. Moser, M.; Pscherer, A.; Roth, C.; Becker, J.; Mucher, G.; Zerres, K.; Dixkens, C.; Weis, J.; Guay-Woodford, L.; Buettner, R.; et al. Enhanced apoptotic cell death of renal epithelial cells in mice lacking transcription factor AP-2beta. Genes Dev. 1997, 11, 1938–1948. [Google Scholar] [CrossRef] [Green Version]
  74. Moser, M.; Dahmen, S.; Kluge, R.; Grone, H.; Dahmen, J.; Kunz, D.; Schorle, H.; Buettner, R. Terminal renal failure in mice lacking transcription factor AP-2β. Lab. Investig. 2003, 83, 571–578. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  75. Zhao, F.; Bosserhoff, A.K.; Buettner, R.; Moser, M. A heart-hand syndrome gene: Tfap2b plays a critical role in the development and remodeling of mouse ductus arteriosus and limb patterning. PLoS ONE 2011, 6, e22908. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  76. Li, X.; Ye, J.; Han, X.; Qiao, R.; Li, X.; Lv, G.; Wang, K. Whole-genome sequencing identifies potential candidate genes for reproductive traits in pigs. Genomics 2020, 112, 199–206. [Google Scholar] [CrossRef] [PubMed]
  77. Yang, C.; Ma, J.; Lin, J. Depletion of NEDD9, a target gene of TGF-β, inhibits the proliferation and invasion of ectopic endometriotic stromal cells. Int. J. Clin. Exp. Pathol. 2017, 10, 2619–2627. [Google Scholar]
  78. Oliviero, C.; Heinonen, M.; Valros, A.; Peltoniemi, O. Environmental and sow-related factors affecting the duration of farrowing. Anim. Reprod. Sci. 2010, 119, 85–91. [Google Scholar] [CrossRef]
  79. Soede, N.M.; Langendijk, P.; Kemp, B. Reproductive cycles in pigs. Anim. Reprod. Sci. 2011, 124, 251–258. [Google Scholar] [CrossRef]
  80. Fletcher, L.; Akhtar, N.; Zhan, X.; Jafarikia, M.; Sullivan, B.P.; Huber, L.A.; Li, J. Identification of Candidate Salivary, Urinary and Serum Metabolic Biomarkers for High Litter Size Potential in Sows (Sus scrofa). Metabolites 2022, 12, 1045. [Google Scholar] [CrossRef]
  81. Wen, W.; He, Z.; Gao, F.; Liu, J.; Jin, H.; Zhai, S.; Qu, Y.; Xia, X. A High-Density Consensus Map of Common Wheat Integrating Four Mapping Populations Scanned by the 90K SNP Array. Front. Plant Sci. 2017, 8, 1389. [Google Scholar] [CrossRef] [Green Version]
  82. Li, J.; Wang, Z.; Lubritz, D.; Arango, J.; Fulton, J.; Settar, P.; Rowland, K.; Cheng, H.; Wolc, A. Genome-wide association studies for egg quality traits in White Leghorn layers using low-pass sequencing and SNP chip data. J. Anim. Breed. Genet. 2022, 139, 380–397. [Google Scholar] [CrossRef]
Figure 1. The normal distribution and correlation analysis between body size and reproduction traits. “***” represents for p < 0.001. TNB: total number born. NHB: number of healthy births. CC: chest circumstance. BL: body length.
Figure 1. The normal distribution and correlation analysis between body size and reproduction traits. “***” represents for p < 0.001. TNB: total number born. NHB: number of healthy births. CC: chest circumstance. BL: body length.
Genes 14 00522 g001
Figure 2. The distribution of SNPs on autosomal chromosomes and the principal component analysis. (A), The distribution of SNPs within a 1 Mb window size. (B), The lollipop-plot of top ten principal components. Index means top ten principal components and por represents proportion of explained variance.
Figure 2. The distribution of SNPs on autosomal chromosomes and the principal component analysis. (A), The distribution of SNPs within a 1 Mb window size. (B), The lollipop-plot of top ten principal components. Index means top ten principal components and por represents proportion of explained variance.
Genes 14 00522 g002
Figure 3. Manhattan and quantile-quantile (QQ) plots of the observed −log10(p-values) for BL, CC, TNB, and NSB in Shaziling pigs. The horizontal red and blue dashed lines in the Manhattan plots indicate the suggestive level (2.38 × 10−5) and significant level (1.19 × 10−6), respectively. The QQ plots show the observed −log10-transformed P-values (y-axis) and the expected −log10-transformed p-values (x-axis).
Figure 3. Manhattan and quantile-quantile (QQ) plots of the observed −log10(p-values) for BL, CC, TNB, and NSB in Shaziling pigs. The horizontal red and blue dashed lines in the Manhattan plots indicate the suggestive level (2.38 × 10−5) and significant level (1.19 × 10−6), respectively. The QQ plots show the observed −log10-transformed P-values (y-axis) and the expected −log10-transformed p-values (x-axis).
Genes 14 00522 g003
Figure 4. GO annotation and KEGG pathway analysis for candidate genes. (A), biological process of GO terms. (B), KEGG pathways. The numbers in the outermost layer indicated the −log10 adjusted p-values.
Figure 4. GO annotation and KEGG pathway analysis for candidate genes. (A), biological process of GO terms. (B), KEGG pathways. The numbers in the outermost layer indicated the −log10 adjusted p-values.
Genes 14 00522 g004
Table 1. Summary statistics of body measurement and reproduction traits in the Shaziling population.
Table 1. Summary statistics of body measurement and reproduction traits in the Shaziling population.
PopulationTrait aNMeanSDMinimumMaximumh2CV (%)
ShazilingBL (cm)190133.210.61101580.157.96%
CC (cm)190132.912.01071620.149.03%
TNB1909.882.90220<0.01-
NHB1908.812.81219<0.01-
NSB1900.140.4802<0.01-
MUMM1900.931.60080.02-
a: BL = body length, CC = chest circumference, TNB = total number born, NHB = number of healthy births, NSB = number of stillborn, MUMM: number of mummified pigs.
Table 2. Potential single-nucleotide polymorphisms (SNPs) and candidate genes identified by the genome-wide association study for body size and reproduction traits.
Table 2. Potential single-nucleotide polymorphisms (SNPs) and candidate genes identified by the genome-wide association study for body size and reproduction traits.
TraitSSC 1SNPPositionp-ValueMAF 2β 3Distance (bp)Candidate Genes
BL7ALGA0040227348359869.01 × 10−80.41 (A/G)−6.49301630GLP1R
BL7ALGA0039856307194242.36 × 10−60.15 (A/G)−7.27withinSNRPC
BL7ASGA0032589363788252.76 × 10−60.22 (A/G)−6.247890NFYA
BL7ALGA0040238348566401.42 × 10−50.33 (G/A)−5.48322284GLP1R
BL7INRA0024788349783832.38 × 10−50.35 (A/G)−5.31444027GLP1R
CC1H3GA00030591686501042.56 × 10−90.02 (A/G)−4.69414030NANOG
CC1ALGA0004562906548905.95 × 10−70.06 (G/A)−2.5441856COX7A2
CC11ASGA0050356240887367.95 × 10−70.02 (A/G)−3.41224869FAM216B
CC12ALGA0065112135636451.05 × 10−60.01 (A/C)−4.8589321CACNG1
CC13ASGA0057447528937761.24 × 10−60.01 (T/A)−5.1916884FOXP1
CC8ALGA01112941241780841.47 × 10−60.04 (G/A)−2.56360686BMPR1B
CC12MARC0074335561831796.87 × 10−60.02 (A/G)−3.11withinDNAH9
CC1MARC0090402485288548.86 × 10−60.05 (A/G)−2.66175464ENSSSCG00000060802
CC16ASGA0073620604691631.08 × 10−50.01 (G/A)−3.85112426CCNG1
CC4ALGA0024545372109681.78 × 10−50.05 (G/A)−2.58121604COX6C
CC1DRGA0000770519780661.90 × 10−50.03 (G/A)−2.34withinRIMS1
CC12WU_10.2_12_8843308843301.91 × 10−50.04 (G/A)−2.22withinCCDC57
CC12WU_10.2_12_8900358900351.91 × 10−50.04 (A/G)−2.22withinCCDC57
TNB3CASI0010189268998621.60 × 10−50.03 (A/C)4.26129540SMG1
NSB8ASGA0038253276897682.75 × 10−70.02 (G/A)0.8881655ARAP2
NSB1ALGA010855788553725.59 × 10−70.02 (A/C)1.0861868GTF2H5
NSB7ALGA0040524392397035.87 × 10−70.01 (G/A)1.255018SLC29A1
NSB4WU_10.2_4_11278211112782113.18 × 10−60.01 (A/G)0.90418666FAM49B
NSB6CASI0009401109879864.47 × 10−60.03 (A/G)0.68329863CNTNAP4
NSB8H3GA0024817415955421.01 × 10−50.07 (A/G)0.45103236KIT
NSB3DIAS0000597102737541.16 × 10−50.02 (G/A)0.882896POR
NSB18WU_10.2_18_23986398239863981.46 × 10−50.08 (A/G)0.39396451SPAM1
NSB7DBMA0000241451731791.58 × 10−50.09 (G/A)0.44402885TFAP2B
NSB17H3GA0048552367465391.70 × 10−50.15 (A/G)0.31withinBPIFA3
NSB4WU_10.2_4_11747027117470271.81 × 10−50.01 (C/A)0.9645942ENSSSCG00000060440
NSB7WU_10.2_7_792599579259952.07 × 10−50.12 (A/C)0.3411934NEDD9
BL, body length; CC, chest circumference; TNB, total number born; NSB, number of stillborn. 1 Sus scrofa chromosome. 2 allele frequency of first listed marker. 3 allele substitution effect.
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Lan, Q.; Deng, Q.; Qi, S.; Zhang, Y.; Li, Z.; Yin, S.; Li, Y.; Tan, H.; Wu, M.; Yin, Y.; et al. Genome-Wide Association Analysis Identified Variants Associated with Body Measurement and Reproduction Traits in Shaziling Pigs. Genes 2023, 14, 522. https://doi.org/10.3390/genes14020522

AMA Style

Lan Q, Deng Q, Qi S, Zhang Y, Li Z, Yin S, Li Y, Tan H, Wu M, Yin Y, et al. Genome-Wide Association Analysis Identified Variants Associated with Body Measurement and Reproduction Traits in Shaziling Pigs. Genes. 2023; 14(2):522. https://doi.org/10.3390/genes14020522

Chicago/Turabian Style

Lan, Qun, Qiuchun Deng, Shijin Qi, Yuebo Zhang, Zhi Li, Shishu Yin, Yulian Li, Hong Tan, Maisheng Wu, Yulong Yin, and et al. 2023. "Genome-Wide Association Analysis Identified Variants Associated with Body Measurement and Reproduction Traits in Shaziling Pigs" Genes 14, no. 2: 522. https://doi.org/10.3390/genes14020522

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop